Affiliations
Division of Infectious Diseases, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
Division of General Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
Department of Biostatistics and Epidemiology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
Email
shahs@email.chop.edu
Given name(s)
Samir S.
Family name
Shah
Degrees
MD, MSCE

Parent and Stakeholder Engagement

Article Type
Changed
Mon, 01/02/2017 - 19:34
Display Headline
Strategies to engage stakeholders in research to improve acute care delivery

We believe that patients, families, and other stakeholders can provide meaningful contributions throughout the research process. Involvement of a diverse group of stakeholders is also encouraged by the Patient Centered Outcomes Research Institute (PCORI), which emphasizes research focused on patient‐ and family‐centered outcomes.[1] Patient and family engagement in healthcare, however, has generally focused on children and adults with chronic conditions.[1, 2] Engagement of families of children with serious acute illnesses is infrequent, and no studies have documented the feasibility or acceptability of different methods of family engagement.[3] Furthermore, stakeholders, such as nurses, may participate in study execution but rarely receive opportunities to inform the research process. In this Perspective, we describe our experiences with family engagement using a novel approach of serial, focused, short‐term engagement of stakeholders.

PRESTUDY WORK

In 2012, our institution introduced a nurse‐led transitional home‐visit program, an approach associated with reduced healthcare utilization in adults.[4] Patients hospitalized for acute illness received a 1‐time transitional home visit 24 to 72 hours after hospital discharge. We formed a multidisciplinary team, consisting of physicians, nurse scientists, home healthcare (HHC) nursing staff, patient families, and research staff to design a mixed‐methods study of the transitional home visit, which was funded by PCORI in 2014. This study, the Hospital‐to‐Home Outcomes (H2O) study, has 3 aims: (1) identify barriers to successful transitions home and outcomes of such transitions that are meaningful to families, (2) optimize the transitional home visits to address family‐identified barriers and outcomes, and (3) determine the efficacy of transitional home visits through a randomized control trial.

Two parents joined the study team during study development. Both had children hospitalized for acute illnesses, received a transitional home visit, and participated in a pilot focus group to provide insight into barriers families encounter during care transitions. These parents made valuable contributions, including recommending strategies for patient enrollment and retention. They also committed to participating in regularly scheduled study meetings and ad hoc discussions. However, feedback from the pilot focus group also highlighted a potential research engagement challenge; specifically, once the acute illness resolved, many families were primarily focused on the return to their normal routine and may not be easily engaged in research.

Based on family input, we included several mechanisms to engage caregivers of children with acute illness in the study design of H2O. Each design element allowed families and other stakeholders to contribute via short‐term focused approaches (eg, focus groups, phone surveys). These short‐term interactions drove iterative changes in study processes and approaches, including how to measure outcomes important to families. Rather than a small group of stakeholders making a series of recommendations over a long period of time, we had dozens of individual stakeholders make a few recommendations apiece that were quickly implemented and subsequently tested via feedback from the next few stakeholders (Figure 1).

jhm2492-fig-0001-m.png
Features that distinguish the new engagement model of short‐term, focused engagement from the traditional engagement model.

PATIENT AND STAKEHOLDER ENGAGEMENT IN THE H2O STUDY

Having the short‐term, focused engagement strategy built into the study proved beneficial, when the 2 parents who were part of the initial design team and had planned to participate longitudinally were no longer able to participate. Over time, their circumstances changed. One parent moved out of the area to pursue a professional opportunity, and the second parent became increasingly difficult to reach and unable to join planned study meetings, a situation anticipated by the pilot focus group participants. These 2 instances illustrate challenges with long‐term engagement of families in research when the potential primary driver of their engagement, their child's acute illness, has resolved.

Short‐Term Focused Engagement Via Focus Groups With Parents/Caregivers

The first aim of the H2O study used 15 focus groups and semistructured interviews with parents/caregivers of recently discharged patients to identify barriers to and metrics of successful transitions of care from the hospital to home. The focus group question guide was developed by the research team and adapted as the focus groups progressed to incorporate new issues raised by participants. Analysis of focus group data revealed opportunities to improve the transitional home visit and identified outcomes important to families, including the need for emotional reassurance in the immediate period after discharge and the impact on family finances.

Short‐Term Focused Engagement Via Phone Calls With Parents/Caregivers

To continuously improve study processes and the transitional home visit during the second aim of H2O, we relied on short‐term focused engagement from 2 stakeholder groups, families and field nurses. We completed 107 phone calls with families who received a transitional home visit during the visit optimization period. These calls, completed 3 to 7 days after the visit, assessed parental perceptions of the effect of recent visit modifications through a standardized survey documented in an electronic database. These data were utilized in plan‐do‐study‐act cycles,[5] every 1 to 2 weeks, to determine if additional modifications to the visits were necessary. A cycle ended when the calls no longer provided new information. The questions asked on the calls also changed over time as different interventions were tested.

As an example, in aim 1, families highlighted the lack of clarity of discharge instructions, particularly regarding when and why to return for medical care. Thus, we developed condition‐specific red flag reminder cards to be shared at transitional home visits to help families remember and recognize concerning signs and symptoms and understand when additional evaluation may be warranted (Figure 2). Families in postvisit calls endorsed the concept of red flags, but sometimes preferred electronic rather than paper versions of the red flag cards to facilitate sharing with family members. Thus, we tested and refined texting the red flag information to families. Subsequent calls strongly supported this practice, so we will continue to use it during the third aim, the randomized trial of the transitional home visit.

jhm2492-fig-0002-m.png
Example of red flag card for bronchiolitis, croup, or pneumonia.

The remaining calls (N=72) were completed 14 days after the visit to mirror the time frame for follow‐up calls in the planned randomized trial. These calls allowed us to test measurement of family‐identified outcomes and determine their usability in the trial. We used family feedback to shorten the survey and reorder questions. We also used feedback from these calls to develop an optimal call‐back strategy to maximize family contacts.

Short‐Term Focused Engagement Via Discussions With Nurses

We also incorporated feedback from HHC nurses on 60 visits to ensure that the visit modifications were feasible to implement. HHC nurse feedback, which aligned with aim 1 data from families, highlighted the potential benefits of standardizing the transitional home visit to be more condition specific. The nurses also provided ongoing ad hoc feedback on other changes to the transitional home visit, which indicated both when tests were successful and when they were challenging to implement. The study team wanted to ensure that the nurses performing the visits were involved in the modification process.

ONGOING H2O WORK AND CONCLUSION

The third aim, with ongoing patient enrollment, involves a randomized trial to determine the efficacy of the revised transitional home visit compared with standard of care as measured by subsequent healthcare utilization and outcomes suggested in aim 1 and refined during aim 2, such as parental coping, stress, and confidence in care. We have engaged 1 parent to provide longitudinal feedback during regularly scheduled meetings.

We believe that our short‐term, focused engagement strategies have allowed integration of the invaluable perspective of families and other stakeholders into our research questions, intervention design, outcome measurement, and study execution. Our approach combined short‐term engagement from many stakeholders, blending qualitative techniques with rapid‐cycle implementation methods to quickly react to stakeholder input. Given the challenge of sustaining longitudinal engagement of families in research focused on acute care questions, and the tendency for many families interested in such engagement to be well versed in the care system due to chronic conditions, we propose this short‐term focused approach to include the unique viewpoints of families and patients whose care experience is confined to an acute period. Similarly, we propose that such an approach can efficiently include and rapidly react to input from other hard‐to‐engage key stakeholders such as field nurses.

Disclosures

This work was supported by the Patient Centered Outcomes Research Institute(HIS‐1306‐0081, SSS). The Patient Centered Outcomes Research Institute had no role in the design, preparation, review, or approval of the manuscript or in the decision to submit the manuscript for publication. The authors have no financial relationships relevant to this article to disclose. The authors report no potential conflicts of interest. The H2O study team members include the following: Katherine A. Auger, MD, MSc, JoAnne Bachus, BSN, Andrew F. Beck, MD, MPH, Monica L. Borell, BSN, Stephanie A. Brunswick, BS, Lenisa Chang, MA, PhD, Jennifer M. Gold, BSN, Judy A. Heilman, RN, Joseph A. Jabour, BS, Jane C. Khoury, PhD, Margo J. Moore, BSN, CCRP, Rita H. Pickler, PNP, PhD, Susan N. Sherman, DPA, Lauren G. Solan, MD, MEd, Angela M. Statile, MD, MEd, Heidi J. Sucharew, PhD, Karen P. Sullivan, BSN, Heather L. Tubbs‐Cooley, RN, PhD, Susan Wade‐Murphy, MSN, and Christine M. White, MD, MAT.

Files
References
  1. Frank L, Forsythe L, Ellis L, et al. Conceptual and practical foundations of patient engagement in research at the patient‐centered outcomes research institute. Qual Life Res. 2015;24(5):10331041.
  2. Haine‐Schlagel R, Walsh NE. A review of parent participation engagement in child and family mental health treatment. Clin Child Fam Psychol Rev. 2015;18(2):133150.
  3. Domecq JP, Prutsky G, Elraiyah T, et al. Patient engagement in research: a systematic review. BMC Health Serv Res. 2014;14:89.
  4. Auger KA, Kenyon CC, Feudtner C, Davis MM. Pediatric hospital discharge interventions to reduce subsequent utilization: a systematic review. J Hosp Med. 2014;9:251260.
  5. Langley GL, Moen R, Nolan KM, Nolan TW, Norman CL, Provost LP. The Improvement Guide: A Practical Approach to Enhancing Organizational Performance. 2nd ed. San Francisco, CA: Jossey‐Bass; 2009.
Article PDF
Issue
Journal of Hospital Medicine - 11(2)
Publications
Page Number
123-125
Sections
Files
Files
Article PDF
Article PDF

We believe that patients, families, and other stakeholders can provide meaningful contributions throughout the research process. Involvement of a diverse group of stakeholders is also encouraged by the Patient Centered Outcomes Research Institute (PCORI), which emphasizes research focused on patient‐ and family‐centered outcomes.[1] Patient and family engagement in healthcare, however, has generally focused on children and adults with chronic conditions.[1, 2] Engagement of families of children with serious acute illnesses is infrequent, and no studies have documented the feasibility or acceptability of different methods of family engagement.[3] Furthermore, stakeholders, such as nurses, may participate in study execution but rarely receive opportunities to inform the research process. In this Perspective, we describe our experiences with family engagement using a novel approach of serial, focused, short‐term engagement of stakeholders.

PRESTUDY WORK

In 2012, our institution introduced a nurse‐led transitional home‐visit program, an approach associated with reduced healthcare utilization in adults.[4] Patients hospitalized for acute illness received a 1‐time transitional home visit 24 to 72 hours after hospital discharge. We formed a multidisciplinary team, consisting of physicians, nurse scientists, home healthcare (HHC) nursing staff, patient families, and research staff to design a mixed‐methods study of the transitional home visit, which was funded by PCORI in 2014. This study, the Hospital‐to‐Home Outcomes (H2O) study, has 3 aims: (1) identify barriers to successful transitions home and outcomes of such transitions that are meaningful to families, (2) optimize the transitional home visits to address family‐identified barriers and outcomes, and (3) determine the efficacy of transitional home visits through a randomized control trial.

Two parents joined the study team during study development. Both had children hospitalized for acute illnesses, received a transitional home visit, and participated in a pilot focus group to provide insight into barriers families encounter during care transitions. These parents made valuable contributions, including recommending strategies for patient enrollment and retention. They also committed to participating in regularly scheduled study meetings and ad hoc discussions. However, feedback from the pilot focus group also highlighted a potential research engagement challenge; specifically, once the acute illness resolved, many families were primarily focused on the return to their normal routine and may not be easily engaged in research.

Based on family input, we included several mechanisms to engage caregivers of children with acute illness in the study design of H2O. Each design element allowed families and other stakeholders to contribute via short‐term focused approaches (eg, focus groups, phone surveys). These short‐term interactions drove iterative changes in study processes and approaches, including how to measure outcomes important to families. Rather than a small group of stakeholders making a series of recommendations over a long period of time, we had dozens of individual stakeholders make a few recommendations apiece that were quickly implemented and subsequently tested via feedback from the next few stakeholders (Figure 1).

jhm2492-fig-0001-m.png
Features that distinguish the new engagement model of short‐term, focused engagement from the traditional engagement model.

PATIENT AND STAKEHOLDER ENGAGEMENT IN THE H2O STUDY

Having the short‐term, focused engagement strategy built into the study proved beneficial, when the 2 parents who were part of the initial design team and had planned to participate longitudinally were no longer able to participate. Over time, their circumstances changed. One parent moved out of the area to pursue a professional opportunity, and the second parent became increasingly difficult to reach and unable to join planned study meetings, a situation anticipated by the pilot focus group participants. These 2 instances illustrate challenges with long‐term engagement of families in research when the potential primary driver of their engagement, their child's acute illness, has resolved.

Short‐Term Focused Engagement Via Focus Groups With Parents/Caregivers

The first aim of the H2O study used 15 focus groups and semistructured interviews with parents/caregivers of recently discharged patients to identify barriers to and metrics of successful transitions of care from the hospital to home. The focus group question guide was developed by the research team and adapted as the focus groups progressed to incorporate new issues raised by participants. Analysis of focus group data revealed opportunities to improve the transitional home visit and identified outcomes important to families, including the need for emotional reassurance in the immediate period after discharge and the impact on family finances.

Short‐Term Focused Engagement Via Phone Calls With Parents/Caregivers

To continuously improve study processes and the transitional home visit during the second aim of H2O, we relied on short‐term focused engagement from 2 stakeholder groups, families and field nurses. We completed 107 phone calls with families who received a transitional home visit during the visit optimization period. These calls, completed 3 to 7 days after the visit, assessed parental perceptions of the effect of recent visit modifications through a standardized survey documented in an electronic database. These data were utilized in plan‐do‐study‐act cycles,[5] every 1 to 2 weeks, to determine if additional modifications to the visits were necessary. A cycle ended when the calls no longer provided new information. The questions asked on the calls also changed over time as different interventions were tested.

As an example, in aim 1, families highlighted the lack of clarity of discharge instructions, particularly regarding when and why to return for medical care. Thus, we developed condition‐specific red flag reminder cards to be shared at transitional home visits to help families remember and recognize concerning signs and symptoms and understand when additional evaluation may be warranted (Figure 2). Families in postvisit calls endorsed the concept of red flags, but sometimes preferred electronic rather than paper versions of the red flag cards to facilitate sharing with family members. Thus, we tested and refined texting the red flag information to families. Subsequent calls strongly supported this practice, so we will continue to use it during the third aim, the randomized trial of the transitional home visit.

jhm2492-fig-0002-m.png
Example of red flag card for bronchiolitis, croup, or pneumonia.

The remaining calls (N=72) were completed 14 days after the visit to mirror the time frame for follow‐up calls in the planned randomized trial. These calls allowed us to test measurement of family‐identified outcomes and determine their usability in the trial. We used family feedback to shorten the survey and reorder questions. We also used feedback from these calls to develop an optimal call‐back strategy to maximize family contacts.

Short‐Term Focused Engagement Via Discussions With Nurses

We also incorporated feedback from HHC nurses on 60 visits to ensure that the visit modifications were feasible to implement. HHC nurse feedback, which aligned with aim 1 data from families, highlighted the potential benefits of standardizing the transitional home visit to be more condition specific. The nurses also provided ongoing ad hoc feedback on other changes to the transitional home visit, which indicated both when tests were successful and when they were challenging to implement. The study team wanted to ensure that the nurses performing the visits were involved in the modification process.

ONGOING H2O WORK AND CONCLUSION

The third aim, with ongoing patient enrollment, involves a randomized trial to determine the efficacy of the revised transitional home visit compared with standard of care as measured by subsequent healthcare utilization and outcomes suggested in aim 1 and refined during aim 2, such as parental coping, stress, and confidence in care. We have engaged 1 parent to provide longitudinal feedback during regularly scheduled meetings.

We believe that our short‐term, focused engagement strategies have allowed integration of the invaluable perspective of families and other stakeholders into our research questions, intervention design, outcome measurement, and study execution. Our approach combined short‐term engagement from many stakeholders, blending qualitative techniques with rapid‐cycle implementation methods to quickly react to stakeholder input. Given the challenge of sustaining longitudinal engagement of families in research focused on acute care questions, and the tendency for many families interested in such engagement to be well versed in the care system due to chronic conditions, we propose this short‐term focused approach to include the unique viewpoints of families and patients whose care experience is confined to an acute period. Similarly, we propose that such an approach can efficiently include and rapidly react to input from other hard‐to‐engage key stakeholders such as field nurses.

Disclosures

This work was supported by the Patient Centered Outcomes Research Institute(HIS‐1306‐0081, SSS). The Patient Centered Outcomes Research Institute had no role in the design, preparation, review, or approval of the manuscript or in the decision to submit the manuscript for publication. The authors have no financial relationships relevant to this article to disclose. The authors report no potential conflicts of interest. The H2O study team members include the following: Katherine A. Auger, MD, MSc, JoAnne Bachus, BSN, Andrew F. Beck, MD, MPH, Monica L. Borell, BSN, Stephanie A. Brunswick, BS, Lenisa Chang, MA, PhD, Jennifer M. Gold, BSN, Judy A. Heilman, RN, Joseph A. Jabour, BS, Jane C. Khoury, PhD, Margo J. Moore, BSN, CCRP, Rita H. Pickler, PNP, PhD, Susan N. Sherman, DPA, Lauren G. Solan, MD, MEd, Angela M. Statile, MD, MEd, Heidi J. Sucharew, PhD, Karen P. Sullivan, BSN, Heather L. Tubbs‐Cooley, RN, PhD, Susan Wade‐Murphy, MSN, and Christine M. White, MD, MAT.

We believe that patients, families, and other stakeholders can provide meaningful contributions throughout the research process. Involvement of a diverse group of stakeholders is also encouraged by the Patient Centered Outcomes Research Institute (PCORI), which emphasizes research focused on patient‐ and family‐centered outcomes.[1] Patient and family engagement in healthcare, however, has generally focused on children and adults with chronic conditions.[1, 2] Engagement of families of children with serious acute illnesses is infrequent, and no studies have documented the feasibility or acceptability of different methods of family engagement.[3] Furthermore, stakeholders, such as nurses, may participate in study execution but rarely receive opportunities to inform the research process. In this Perspective, we describe our experiences with family engagement using a novel approach of serial, focused, short‐term engagement of stakeholders.

PRESTUDY WORK

In 2012, our institution introduced a nurse‐led transitional home‐visit program, an approach associated with reduced healthcare utilization in adults.[4] Patients hospitalized for acute illness received a 1‐time transitional home visit 24 to 72 hours after hospital discharge. We formed a multidisciplinary team, consisting of physicians, nurse scientists, home healthcare (HHC) nursing staff, patient families, and research staff to design a mixed‐methods study of the transitional home visit, which was funded by PCORI in 2014. This study, the Hospital‐to‐Home Outcomes (H2O) study, has 3 aims: (1) identify barriers to successful transitions home and outcomes of such transitions that are meaningful to families, (2) optimize the transitional home visits to address family‐identified barriers and outcomes, and (3) determine the efficacy of transitional home visits through a randomized control trial.

Two parents joined the study team during study development. Both had children hospitalized for acute illnesses, received a transitional home visit, and participated in a pilot focus group to provide insight into barriers families encounter during care transitions. These parents made valuable contributions, including recommending strategies for patient enrollment and retention. They also committed to participating in regularly scheduled study meetings and ad hoc discussions. However, feedback from the pilot focus group also highlighted a potential research engagement challenge; specifically, once the acute illness resolved, many families were primarily focused on the return to their normal routine and may not be easily engaged in research.

Based on family input, we included several mechanisms to engage caregivers of children with acute illness in the study design of H2O. Each design element allowed families and other stakeholders to contribute via short‐term focused approaches (eg, focus groups, phone surveys). These short‐term interactions drove iterative changes in study processes and approaches, including how to measure outcomes important to families. Rather than a small group of stakeholders making a series of recommendations over a long period of time, we had dozens of individual stakeholders make a few recommendations apiece that were quickly implemented and subsequently tested via feedback from the next few stakeholders (Figure 1).

jhm2492-fig-0001-m.png
Features that distinguish the new engagement model of short‐term, focused engagement from the traditional engagement model.

PATIENT AND STAKEHOLDER ENGAGEMENT IN THE H2O STUDY

Having the short‐term, focused engagement strategy built into the study proved beneficial, when the 2 parents who were part of the initial design team and had planned to participate longitudinally were no longer able to participate. Over time, their circumstances changed. One parent moved out of the area to pursue a professional opportunity, and the second parent became increasingly difficult to reach and unable to join planned study meetings, a situation anticipated by the pilot focus group participants. These 2 instances illustrate challenges with long‐term engagement of families in research when the potential primary driver of their engagement, their child's acute illness, has resolved.

Short‐Term Focused Engagement Via Focus Groups With Parents/Caregivers

The first aim of the H2O study used 15 focus groups and semistructured interviews with parents/caregivers of recently discharged patients to identify barriers to and metrics of successful transitions of care from the hospital to home. The focus group question guide was developed by the research team and adapted as the focus groups progressed to incorporate new issues raised by participants. Analysis of focus group data revealed opportunities to improve the transitional home visit and identified outcomes important to families, including the need for emotional reassurance in the immediate period after discharge and the impact on family finances.

Short‐Term Focused Engagement Via Phone Calls With Parents/Caregivers

To continuously improve study processes and the transitional home visit during the second aim of H2O, we relied on short‐term focused engagement from 2 stakeholder groups, families and field nurses. We completed 107 phone calls with families who received a transitional home visit during the visit optimization period. These calls, completed 3 to 7 days after the visit, assessed parental perceptions of the effect of recent visit modifications through a standardized survey documented in an electronic database. These data were utilized in plan‐do‐study‐act cycles,[5] every 1 to 2 weeks, to determine if additional modifications to the visits were necessary. A cycle ended when the calls no longer provided new information. The questions asked on the calls also changed over time as different interventions were tested.

As an example, in aim 1, families highlighted the lack of clarity of discharge instructions, particularly regarding when and why to return for medical care. Thus, we developed condition‐specific red flag reminder cards to be shared at transitional home visits to help families remember and recognize concerning signs and symptoms and understand when additional evaluation may be warranted (Figure 2). Families in postvisit calls endorsed the concept of red flags, but sometimes preferred electronic rather than paper versions of the red flag cards to facilitate sharing with family members. Thus, we tested and refined texting the red flag information to families. Subsequent calls strongly supported this practice, so we will continue to use it during the third aim, the randomized trial of the transitional home visit.

jhm2492-fig-0002-m.png
Example of red flag card for bronchiolitis, croup, or pneumonia.

The remaining calls (N=72) were completed 14 days after the visit to mirror the time frame for follow‐up calls in the planned randomized trial. These calls allowed us to test measurement of family‐identified outcomes and determine their usability in the trial. We used family feedback to shorten the survey and reorder questions. We also used feedback from these calls to develop an optimal call‐back strategy to maximize family contacts.

Short‐Term Focused Engagement Via Discussions With Nurses

We also incorporated feedback from HHC nurses on 60 visits to ensure that the visit modifications were feasible to implement. HHC nurse feedback, which aligned with aim 1 data from families, highlighted the potential benefits of standardizing the transitional home visit to be more condition specific. The nurses also provided ongoing ad hoc feedback on other changes to the transitional home visit, which indicated both when tests were successful and when they were challenging to implement. The study team wanted to ensure that the nurses performing the visits were involved in the modification process.

ONGOING H2O WORK AND CONCLUSION

The third aim, with ongoing patient enrollment, involves a randomized trial to determine the efficacy of the revised transitional home visit compared with standard of care as measured by subsequent healthcare utilization and outcomes suggested in aim 1 and refined during aim 2, such as parental coping, stress, and confidence in care. We have engaged 1 parent to provide longitudinal feedback during regularly scheduled meetings.

We believe that our short‐term, focused engagement strategies have allowed integration of the invaluable perspective of families and other stakeholders into our research questions, intervention design, outcome measurement, and study execution. Our approach combined short‐term engagement from many stakeholders, blending qualitative techniques with rapid‐cycle implementation methods to quickly react to stakeholder input. Given the challenge of sustaining longitudinal engagement of families in research focused on acute care questions, and the tendency for many families interested in such engagement to be well versed in the care system due to chronic conditions, we propose this short‐term focused approach to include the unique viewpoints of families and patients whose care experience is confined to an acute period. Similarly, we propose that such an approach can efficiently include and rapidly react to input from other hard‐to‐engage key stakeholders such as field nurses.

Disclosures

This work was supported by the Patient Centered Outcomes Research Institute(HIS‐1306‐0081, SSS). The Patient Centered Outcomes Research Institute had no role in the design, preparation, review, or approval of the manuscript or in the decision to submit the manuscript for publication. The authors have no financial relationships relevant to this article to disclose. The authors report no potential conflicts of interest. The H2O study team members include the following: Katherine A. Auger, MD, MSc, JoAnne Bachus, BSN, Andrew F. Beck, MD, MPH, Monica L. Borell, BSN, Stephanie A. Brunswick, BS, Lenisa Chang, MA, PhD, Jennifer M. Gold, BSN, Judy A. Heilman, RN, Joseph A. Jabour, BS, Jane C. Khoury, PhD, Margo J. Moore, BSN, CCRP, Rita H. Pickler, PNP, PhD, Susan N. Sherman, DPA, Lauren G. Solan, MD, MEd, Angela M. Statile, MD, MEd, Heidi J. Sucharew, PhD, Karen P. Sullivan, BSN, Heather L. Tubbs‐Cooley, RN, PhD, Susan Wade‐Murphy, MSN, and Christine M. White, MD, MAT.

References
  1. Frank L, Forsythe L, Ellis L, et al. Conceptual and practical foundations of patient engagement in research at the patient‐centered outcomes research institute. Qual Life Res. 2015;24(5):10331041.
  2. Haine‐Schlagel R, Walsh NE. A review of parent participation engagement in child and family mental health treatment. Clin Child Fam Psychol Rev. 2015;18(2):133150.
  3. Domecq JP, Prutsky G, Elraiyah T, et al. Patient engagement in research: a systematic review. BMC Health Serv Res. 2014;14:89.
  4. Auger KA, Kenyon CC, Feudtner C, Davis MM. Pediatric hospital discharge interventions to reduce subsequent utilization: a systematic review. J Hosp Med. 2014;9:251260.
  5. Langley GL, Moen R, Nolan KM, Nolan TW, Norman CL, Provost LP. The Improvement Guide: A Practical Approach to Enhancing Organizational Performance. 2nd ed. San Francisco, CA: Jossey‐Bass; 2009.
References
  1. Frank L, Forsythe L, Ellis L, et al. Conceptual and practical foundations of patient engagement in research at the patient‐centered outcomes research institute. Qual Life Res. 2015;24(5):10331041.
  2. Haine‐Schlagel R, Walsh NE. A review of parent participation engagement in child and family mental health treatment. Clin Child Fam Psychol Rev. 2015;18(2):133150.
  3. Domecq JP, Prutsky G, Elraiyah T, et al. Patient engagement in research: a systematic review. BMC Health Serv Res. 2014;14:89.
  4. Auger KA, Kenyon CC, Feudtner C, Davis MM. Pediatric hospital discharge interventions to reduce subsequent utilization: a systematic review. J Hosp Med. 2014;9:251260.
  5. Langley GL, Moen R, Nolan KM, Nolan TW, Norman CL, Provost LP. The Improvement Guide: A Practical Approach to Enhancing Organizational Performance. 2nd ed. San Francisco, CA: Jossey‐Bass; 2009.
Issue
Journal of Hospital Medicine - 11(2)
Issue
Journal of Hospital Medicine - 11(2)
Page Number
123-125
Page Number
123-125
Publications
Publications
Article Type
Display Headline
Strategies to engage stakeholders in research to improve acute care delivery
Display Headline
Strategies to engage stakeholders in research to improve acute care delivery
Sections
Article Source
© 2015 Society of Hospital Medicine
Disallow All Ads
Correspondence Location
Address for correspondence and reprint requests: Samir S. Shah, MD, 3333 Burnet Avenue, ML 9016, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229; Telephone: 513‐636‐6222; Fax: 513‐803‐9244; E‐mail: samir.shah@cchmc.org
Content Gating
Gated (full article locked unless allowed per User)
Gating Strategy
First Peek Free
Article PDF Media
Image
Disable zoom
Off
Media Files
Image
Disable zoom
Off

Febrile Infant Diagnosis Code Accuracy

Article Type
Changed
Tue, 05/16/2017 - 22:46
Display Headline
Accuracy of diagnosis codes to identify febrile young infants using administrative data

Fever is one of the most common reasons for emergency department (ED) evaluation of infants under 90 days of age.[1] Up to 10% to 20% of febrile young infants will have a serious bacterial infection (SBI),[2, 3, 4] but infants with SBI are difficult to distinguish from those without SBI based upon symptoms and physical examination findings alone.[5] Previously developed clinical prediction algorithms can help to identify febrile infants at low risk for SBI, but differ in age range as well as recommendations for testing and empiric treatment.[6, 7, 8] Consequently, there is widespread variation in management of febrile young infants at US children's hospitals,[9, 10, 11] and defining optimal management strategies remains an important issue in pediatric healthcare.[12] Administrative datasets are convenient and inexpensive, and can be used to evaluate practice variation, trends, and outcomes of a large, diverse group of patients within and across institutions.[9, 10] Accurately identifying febrile infants evaluated for suspected SBI in administrative databases would facilitate comparative effectiveness research, quality improvement initiatives, and institutional benchmarking.

Prior studies have validated the accuracy of administrative billing codes for identification of other common childhood illnesses, including urinary tract infection (UTI)[13] and pneumonia.[14] The accuracy of International Classification of Diseases, Ninth Revision (ICD‐9) diagnosis codes in identifying febrile young infants evaluated for SBI is not known. Reliance on administrative ICD‐9 diagnosis codes for patient identification can lead to misclassification of patients due to variable database quality, the validity of the diagnosis codes being utilized, and hospital coding practices.[15] Additionally, fever is a symptom and not a specific diagnosis. If a particular bacterial or viral diagnosis is established (eg, enterovirus meningitis), a discharge diagnosis of fever may not be attributed to the patient encounter. Thus, evaluating the performance characteristics and capture of clinical outcomes of different combinations of ICD‐9 diagnosis codes for identifying febrile infants is necessary for both the conduct and interpretation of studies that utilize administrative databases. The primary objective of this investigation was to identify the most accurate ICD‐9 coding strategies for the identification of febrile infants aged <90 days using administrative data. We also sought to evaluate capture of clinically important outcomes across identification strategies.

METHODS

Study Design and Setting

For this multicenter retrospective study, we used the Pediatric Health Information System (PHIS) database to identify infants <90 days of age[16] who presented between July 1, 2012 and June 30, 2013 to 1 of 8 EDs. We assessed performance characteristics of ICD‐9 diagnosis code case‐identification algorithms by comparing ICD‐9 code combinations to a fever reference standard determined by medical record review. The institutional review board at each participating site approved the study protocol.

Data Source

Data were obtained from 2 sources: the PHIS database and medical record review. We used the PHIS database to identify eligible patients by ICD‐9 diagnosis codes; patient encounters were randomly selected using a random number generator. The PHIS database contains demographic, diagnosis, and billing data from 44 hospitals affiliated with the Children's Hospital Association (Overland Park, Kansas) and represents 85% of freestanding children's hospitals in the United States.[17] Data are deidentified; encrypted unique patient identifiers permit tracking of patients across visits within a site.[18] The Children's Hospital Association and participating hospitals jointly assure the quality and integrity of the data.[19]

For each patient encounter identified in the PHIS database, detailed medical record review was performed by trained investigators at each of the 8 study sites (see Supporting Information, Appendix, in the online version of this article). A standardized data collection instrument was pilot tested by all investigators prior to use. Data were collected and managed using the Research Electronic Data Capture (REDCap) tool hosted at Boston Children's Hospital.[20]

Exclusions

Using PHIS data, prior to medical record review we excluded infants with a complex chronic condition as defined previously[21] and those transferred from another institution, as these infants may warrant a nonstandard evaluation and/or may have incomplete data.

ICD‐9 Diagnosis Code Groups

In the PHIS database, all patients discharged from the hospital (including hospitalized patients as well as patients discharged from the ED) receive 1 or more ICD‐9 discharge diagnosis codes. These diagnosis codes are ascribed after discharge from the hospital, or for ED patients, after ED discharge. Additionally, patients may receive an admission diagnosis, which reflects the diagnosis ascribed at the time of ED discharge or transfer to the inpatient unit.

We reviewed medical records of infants selected from the following ICD‐9 diagnosis code groups (Figure 1): (1) discharge diagnosis code of fever (780.6 [fever and other physiologic disturbances of temperature regulation], 778.4 [other disturbances of temperature regulation of newborn], 780.60 [fever, unspecified], or 780.61 [fever presenting with conditions classified elsewhere])[9, 10] regardless of the presence of admission diagnosis of fever or diagnosis of serious infection, (2) admission diagnosis code of fever without associated discharge diagnosis code of fever,[10] (3) discharge diagnosis code of serious infection determined a priori (see Supporting Information, Appendix, in the online version of this article) without discharge or admission diagnosis code of fever, and (4) infants without any diagnosis code of fever or serious infection.

jhm2441-fig-0001-m.png
Study population. 1Two of 584 medical records were unavailable for review. 2Five of 904 medical records were unavailable for review. Abbreviations: CCC, complex chronic condition; ED, emergency department.

Medical records reviewed in each of the 4 ICD‐9 diagnosis code groups were randomly selected from the overall set of ED encounters in the population of infants <90 days of age evaluated during the study period. Twenty‐five percent population sampling was used for 3 of the ICD‐9 diagnosis code groups, whereas 5% sampling was used for the no fever/no serious infection code group. The number of medical records reviewed in each ICD‐9 diagnosis code group was proportional to the distribution of ICD‐9 codes across the entire population of infants <90 days of age. These records were distributed equally across sites (228 records per site), except for 1 site that does not assign admission diagnoses (201 records).

Investigators were blinded to ICD‐9 diagnosis code groups during medical record review. Infants with multiple visits during the study period were eligible to be included more than once if the visits occurred more than 3 days apart. For infants with more than 1 ED visit on a particular calendar day, investigators were instructed to review the initial visit.

For each encounter, we also abstracted demographic characteristics (gender, race/ethnicity), insurance status, hospital region (using US Census categories[22]), and season from the PHIS database.

Reference Standard

The presence of fever was determined by medical record review. We defined fever as any documented temperature 100.4F (38.0C) at home or in the ED.[16]

ICD‐9 Code Case‐Identification Algorithms

Using the aforementioned ICD‐9 diagnosis code groups individually and in combination, the following 4 case‐identification algorithms, determined from prior study or group consensus, were compared to the reference standard: (1) ICD‐9 discharge diagnosis code of fever,[9] (2) ICD‐9 admission or discharge diagnosis code of fever,[10, 11] (3) ICD‐9 discharge diagnosis code of fever or serious infection, and (4) ICD‐9 discharge or admission diagnosis code of fever or serious infection. Algorithms were compared overall, separately for discharged and hospitalized infants, and across 3 distinct age groups (28 days, 2956 days, and 5789 days).

Patient‐Level Outcomes

To compare differences in outcomes by case‐identification algorithm, from the PHIS database we abstracted hospitalization rates, rates of UTI/pyelonephritis,[13] bacteremia/sepsis, and bacterial meningitis.[19] Severe outcomes were defined as intensive care unit admission, mechanical ventilation, central line placement, receipt of extracorporeal membrane oxygenation, or death. We assessed hospital length of stay for admitted infants and 3‐day revisits,[23, 24] and revisits resulting in hospitalization for infants discharged from the ED at the index visit. Patients billed for observation care were classified as being hospitalized.[25, 26]

Data Analysis

Accuracy of the 4 case‐identification algorithms (compared with the reference standard) was calculated using sensitivity, specificity, negative predictive value (NPV), and positive predictive value (PPV), along with 95% confidence interval (CI). Prior to analysis, a 5‐fold weighting factor was applied to the no fever/no serious infection group to account for the differential sampling used for this group (5% vs 25% for the other 3 ICD‐9 diagnosis code groups). This weighting was done to approximate the true prevalence of each ICD‐9 code group within the larger population, so that an accurate rate of false negatives (infants with fever who had neither a diagnosis of fever nor serious infection) could be calculated.

We described continuous variables using median and interquartile range or range values and categorical variables using frequencies with 95% CIs. We compared categorical variables using a 2 test. We determined statistical significance as a 2‐tailed P value <0.05. Statistical analyses were performed using SAS version 9.3 (SAS Institute, Cary, NC).

RESULTS

Study Patients

During the 1‐year study period, 23,753 ED encounters for infants <90 days of age were identified in the PHIS database at the 8 participating sites. Of these infant encounters, 2166 (9.2%) were excluded (1658 infants who had a complex chronic condition and 508 transferred into the ED), leaving 21,587 infants available for selection. After applying our sampling strategy, we identified 1797 encounters for medical record review. Seven encounters from 3 hospitals with missing medical records were excluded, resulting in a final cohort of 1790 encounters (Figure 1). Among included infants, 552 (30.8%) were 28 days, 743 (41.5%) were 29 to 56 days, and 495 (27.8%) were 57 to 89 days of age; 737 (41.2%) infants were hospitalized. Patients differed in age, race, payer, and season across ICD‐9 diagnosis code groups (see Supporting Information, Table 1, in the online version of this article).

Performance Characteristics of ICD‐9 Diagnosis Code Case‐Identification Algorithms According to Reference Standard (Overall, Hospitalized, and Discharged).*
ICD‐9 Diagnosis Code AlgorithmOverall
Sensitivity, % (95% CI)Specificity, % (95% CI)Negative Predictive Value, % (95% CI)Positive Predictive Value, % (95% CI)
  • NOTE: Abbreviations: CI, confidence interval; ED, emergency department; ICD‐9, International Classification of Diseases, Ninth Revision. *Reference standard of fever was defined by documented temperature 100.4 F (38.0 C) on review of electronic medical record.

Discharge diagnosis of fever53.2 (50.056.4)98.2 (97.898.6)90.8 (90.091.6)86.1 (83.388.9)
Hospitalized47.3 (43.151.5)97.7 (96.998.5)80.6 (78.682.6)90.2 (86.893.6)
Discharged from ED61.4 (56.666.2)98.4 (98.098.8)95.4 (94.796.1)82.1 (77.786.5)
Discharge or admission diagnosis of Fever71.1 (68.274.0)97.7 (97.398.1)94.1 (93.494.8)86.9 (84.589.3)
Hospitalized72.5 (68.876.2)97.1 (96.298.0)88.8 (87.190.5)91.7 (89.194.3)
Discharged from ED69.2 (64.773.7)98.0 (97.598.5)96.3 (95.796.9)80.8 (76.685.0)
Discharge diagnosis of fever or serious infection63.7 (60.666.8)96.5 (96.097.0)92.6 (91.893.4)79.6 (76.782.5)
Hospitalized63.9 (59.967.9)92.5 (91.094.0)85.1 (83.287.0)79.1 (75.382.9)
Discharged from ED63.4 (58.768.1)98.1 (97.698.6)95.6 (94.996.3)80.2 (75.884.6)
Discharge or admission diagnosis of fever or serious infection76.6 (73.979.3)96.2 (95.696.8)95.1 (94.595.7)81.0 (78.483.6)
Hospitalized80.8 (77.584.1)92.1 (90.693.6)91.5 (89.993.1)82.1 (78.985.3)
Discharged from ED71.0 (66.575.5)97.7 (97.298.2)96.5 (95.997.1)79.4 (75.283.6)

Among the 1790 patient encounters reviewed, a total of 766 infants (42.8%) met the reference standard definition for fever in the cohort. An additional 47 infants had abnormal temperature reported (documentation of tactile fever, history of fever without a specific temperature described, or hypothermia) but were classified as having no fever by the reference standard.

ICD‐9 Code Case‐Identification Algorithm Performance

Compared with the reference standard, the 4 case‐identification algorithms demonstrated specificity of 96.2% to 98.2% but lower sensitivity overall (Figure 2). Discharge diagnosis of fever alone demonstrated the lowest sensitivity. The algorithm of discharge or admission diagnosis of fever resulted in increased sensitivity and the highest PPV of all 4 algorithms (86.9%, 95% CI: 84.5‐89.3). Addition of serious infection codes to this algorithm resulted in a marginal increase in sensitivity and a similar decrease in PPV (Table 1). When limited to hospitalized infants, specificity was highest for the case‐identification algorithm of discharge diagnosis of fever and similarly high for discharge or admission diagnosis of fever; sensitivity was highest for the algorithm of discharge or admission diagnosis of fever or diagnosis of serious infection. For infants discharged from the ED, algorithm specificity was 97.7% to 98.4%, with lower sensitivity for all 4 algorithms (Table 1). Inclusion of the 47 infants with abnormal temperature as fever did not materially change algorithm performance (data not shown).

jhm2441-fig-0002-m.png
Algorithm sensitivity and false positive rate (1‐specificity) for identification of febrile infants aged ≤28 days, 29 to 56 days, 57 to 89 days, and overall. Horizontal and vertical bars represent 95% confidence intervals. Reference standard of fever was defined by documented temperature ≥100.4°F (38.0°C) on review of electronic medical record.

Across all 3 age groups (28 days, 2956 days, and 5789 days), the 4 case‐identification algorithms demonstrated specificity >96%, whereas algorithm sensitivity was highest in the 29‐ to 56‐days‐old age group and lowest among infants 57 to 89 days old across all 4 algorithms (Figure 2). Similar to the overall cohort, an algorithm of discharge or admission diagnosis of fever demonstrated specificity of nearly 98% in all age groups; addition of serious infection codes to this algorithm increased sensitivity, highest in the 29‐ to 56‐days‐old age group (Figure 2; see also Supporting Information, Table 2, in the online version of this article).

Performance Characteristics of ICD‐9 Diagnosis Code Case‐Identification Algorithms Across the Eight Sites According to Reference Standard.*
ICD‐9 Diagnosis Code AlgorithmSensitivity, Median % (Range)Specificity, Median % (Range)Negative Predictive Value, Median % (Range)Positive Predictive Value, Median % (Range)
  • NOTE: Abbreviations: ICD‐9, International Classification of Diseases, Ninth Revision. *Reference standard of fever was defined by documented temperature 100.4F (38.0 C) on review of electronic medical record.

Discharge diagnosis of fever56.2 (34.681.0)98.3 (96.499.1)92.1 (83.297.4)87.7 (74.093.2)
Discharge or Admission diagnosis of Fever76.7 (51.385.0)97.8 (96.298.7)95.6 (86.997.4)87.4 (80.092.9)
Discharge diagnosis of fever or serious infection68.3 (44.287.3)96.5 (95.498.0)93.6 (85.298.2)78.3 (74.289.0)
Discharge or admission diagnosis of fever or serious infection83.1 (58.390.7)95.8 (95.498.0)96.5 (88.598.2)79.1 (77.490.4)

Across the 8 study sites, median specificity was 95.8% to 98.3% for the 4 algorithms, with little interhospital variability; however, algorithm sensitivity varied widely by site. Median PPV was highest for discharge diagnosis of fever alone at 87.7% but ranged from 74.0% to 93.2% across sites. Median PPV for an algorithm of discharge or admission diagnosis of fever was similar (87.4%) but with less variation by site (range 80.0%92.9%) (Table 2).

Outcomes by ICD‐9 Diagnosis Code Group and Case‐Identification Algorithm

When compared with discharge diagnosis of fever, adding admission diagnosis of fever captured a higher proportion of hospitalized infants with SBIs (UTI/pyelonephritis, bacteremia/sepsis, or bacterial meningitis). However, median hospital length of stay, severe outcomes, and 3‐day revisits and revisits with hospitalization did not materially differ when including infants with admission diagnosis of fever in addition to discharge diagnosis of fever. Addition of infants with a diagnosis code for serious infection substantially increased the number of infants with SBIs and severe outcomes but did not capture additional 3‐day revisits (Table 3). There were no additional cases of SBI in the no fever/no serious illness diagnosis code group.

Outcomes by ICD‐9 Diagnosis Code Case‐Identification Algorithm
ICD‐9 Diagnosis Code AlgorithmOutcome3‐Day Revisit, % (95% CI)3‐Day Revisit With Hospitalization, % (95% CI)
Hospitalized, % (95% CI)UTI/Pyelonephritis, Bacteremia/Sepsis, or Bacterial Meningitis, % (95% CI)Severe Outcome, % (95% CI)*Length of Stay in Days, Median (IQR)
  • NOTE: Abbreviations: CI, confidence interval; ICD‐9, International Classification of Diseases, Ninth Revision; IQR, interquartile range; UTI, urinary tract infection. *Severe outcome was defined as intensive care unit admission, mechanical ventilation, central line placement, extracorporeal membrane oxygenation, or death. Length of stay for hospitalized infants. Percent of those discharged from the emergency department at the index visit.

Discharge diagnosis of fever44.3 (40.348.4)3.3 (1.84.7)1.4 (0.42.3)3 (23)11.7 (8.215.2)5.9 (3.38.4)
Discharge or admission diagnosis of fever52.4 (48.955.9)6.1 (4.47.8)1.9 (1.02.9)3 (23)10.9 (7.714.1)5.4 (3.17.8)
Discharge diagnosis of fever or serious infection54.0 (50.457.5)15.3 (12.717.8)3.8 (2.55.2)3 (24)11.0 (7.714.2)5.5 (3.17.9)
Discharge or admission diagnosis of fever or serious infection56.5 (53.259.7)12.9 (10.715.1)3.6 (2.44.8)3 (24)10.3 (7.313.3)5.2 (3.07.4)

Among infants who met the reference standard for fever but did not have a discharge or admission diagnosis of fever (false negatives), 11.8% had a diagnosis of SBI. Overall, 43.2% of febrile infants (and 84.4% of hospitalized infants) with SBI did not have an ICD‐9 discharge or admission diagnosis of fever. Addition of ICD‐9 diagnosis codes of serious infection to the algorithm of discharge or admission diagnosis of fever captured all additional SBIs, and no false negativeinfants missed with this algorithm had an SBI.

DISCUSSION

We described the performance of 4 ICD‐9 diagnosis code case‐identification algorithms for the identification of febrile young infants <90 days of age at US children's hospitals. Although the specificity was high across algorithms and institutions, the sensitivity was relatively low, particularly for discharge diagnosis of fever, and varied by institution. Given the high specificity, ICD‐9 diagnosis code case‐identification algorithms for fever reliably identify febrile infants using administrative data with low rates of inclusion of infants without fever. However, underidentification of patients, particularly those more prone to SBIs and severe outcomes depending on the algorithm utilized, can impact interpretation of comparative effectiveness studies or the quality of care delivered by an institution.

ICD‐9 discharge diagnosis codes are frequently used to identify pediatric patients across a variety of administrative databases, diseases, and symptoms.[19, 27, 28, 29, 30, 31] Although discharge diagnosis of fever is highly specific, sensitivity is substantially lower than other case‐identification algorithms we studied, particularly for hospitalized infants. This may be due to a fever code sometimes being omitted in favor of a more specific diagnosis (eg, bacteremia) prior to hospital discharge. Therefore, case identification relying only on ICD‐9 discharge diagnosis codes for fever may under‐report clinically important SBI or severe outcomes as demonstrated in our study. This is in contrast to ICD‐9 diagnosis code identification strategies for childhood UTI and pneumonia, which largely have higher sensitivity but lower specificity than fever codes.[13, 14]

Admission diagnosis of fever is important for febrile infants as they may not have an explicit diagnosis at the time of disposition from the ED. Addition of admission diagnosis of fever to an algorithm relying on discharge diagnosis code alone increased sensitivity without a demonstrable reduction in specificity and PPV, likely due to capture of infants with a fever diagnosis at presentation before a specific infection was identified. Although using an algorithm of discharge or admission diagnosis of fever captured a higher percentage of hospitalized febrile infants with SBIs, sensitivity was only 71% overall with this algorithm, and 43% of febrile infants with SBI would still have been missed. Importantly, though, addition of various ICD‐9 codes for serious infection to this algorithm resulted in capture of all febrile infants with SBI and should be used as a sensitivity analysis.

The test characteristics of diagnosis codes were highest in the 29‐ to 56‐days‐old age group. Given the differing low‐risk criteria[6, 7, 8] and lack of best practice guidelines[16] in this age group, the use of administrative data may allow for the comparison of testing and treatment strategies across a large cohort of febrile infants aged 29 to 56 days. However, individual hospital coding practices may affect algorithm performance, in particular sensitivity, which varied substantially by hospital. This variation in algorithm sensitivity may impact comparisons of outcomes across institutions. Therefore, when conducting studies of febrile infants using administrative data, sensitivity analyses or use of chart review should be considered to augment the use of ICD‐9 code‐based identification strategies, particularly for comparative benchmarking and outcomes studies. These additional analyses are particularly important for studies of febrile infants >56 days of age, in whom the sensitivity of diagnosis codes is particularly low. We speculate that the lower sensitivity in older febrile infants may relate to a lack of consensus on the clinical significance of fever in this age group and the varying management strategies employed.[10]

Strengths of this study include the assessment of ICD‐9 code algorithms across multiple institutions for identification of fever in young infants, and the patterns of our findings remained robust when comparing median performance characteristics of the algorithms across hospitals to our overall findings. We were also able to accurately estimate PPV and NPV using a case‐identification strategy weighted to the actual population sizes. Although sensitivity and specificity are the primary measures of test performance, predictive values are highly informative for investigators using administrative data. Additionally, our findings may inform public health efforts including disease surveillance, assessment of seasonal variation, and identification and monitoring of healthcare‐associated infections among febrile infants.

Our study has limitations. We did not review all identified records, which raises the possibility that our evaluated cohort may not be representative of the entire febrile infant population. We attempted to mitigate this possibility by using a random sampling strategy for our population selection that was weighted to the actual population sizes. Second, we identified serious infections using ICD‐9 diagnosis codes determined by group consensus, which may not capture all serious infection codes that identify febrile infants whose fever code was omitted. Third, 47 infants had abnormal temperature that did not meet our reference standard criteria for fever and were included in the no fever group. Although there may be disagreement regarding what constitutes a fever, we used a widely accepted reference standard to define fever.[16] Further, inclusion of these 47 infants as fever did not materially change algorithm performance. Last, our study was conducted at 8 large tertiary‐care children's hospitals, and our results may not be generalizable to other children's hospitals and community‐based hospitals.

CONCLUSIONS

Studies of febrile young infants that rely on ICD‐9 discharge diagnosis code of fever for case ascertainment have high specificity but low sensitivity for the identification of febrile infants, particularly among hospitalized patients. A case‐identification strategy that includes discharge or admission diagnosis of fever demonstrated higher sensitivity, and should be considered for studies of febrile infants using administrative data. However, additional strategies such as incorporation of ICD‐9 codes for serious infection should be used when comparing outcomes across institutions.

Acknowledgements

The Febrile Young Infant Research Collaborative includes the following additional collaborators who are acknowledged for their work on this study: Erica DiLeo, MA, Department of Medical Education and Research, Danbury Hospital, Danbury, Connecticut; Janet Flores, BS, Division of Emergency Medicine, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois.

Disclosures: This project funded in part by The Gerber Foundation Novice Researcher Award, (Ref No. 1827‐3835). Dr. Fran Balamuth received career development support from the National Institutes of Health (NHLBI K12‐HL109009). Funders were not involved in the design or conduct of the study; collection, management, analysis, or interpretation of the data; or preparation, review, or approval of the manuscript. The authors have no conflicts of interest relevant to this article to disclose.

Files
References
  1. Baskin MN. The prevalence of serious bacterial infections by age in febrile infants during the first 3 months of life. Pediatr Ann. 1993;22:462466.
  2. Huppler AR, Eickhoff JC, Wald ER. Performance of low‐risk criteria in the evaluation of young infants with fever: review of the literature. Pediatrics. 2010;125:228233.
  3. Schwartz S, Raveh D, Toker O, Segal G, Godovitch N, Schlesinger Y. A week‐by‐week analysis of the low‐risk criteria for serious bacterial infection in febrile neonates. Arch Dis Child. 2009;94:287292.
  4. Garcia S, Mintegi S, Gomez B, et al. Is 15 days an appropriate cut‐off age for considering serious bacterial infection in the management of febrile infants? Pediatr Infect Dis J. 2012;31:455458.
  5. Baker MD, Avner JR, Bell LM. Failure of infant observation scales in detecting serious illness in febrile, 4‐ to 8‐week‐old infants. Pediatrics. 1990;85:10401043.
  6. Baker MD, Bell LM, Avner JR. Outpatient management without antibiotics of fever in selected infants. N Engl J Med. 1993;329:14371441.
  7. Baskin MN, Fleisher GR, O'Rourke EJ. Identifying febrile infants at risk for a serious bacterial infection. J Pediatr. 1993;123:489490.
  8. Jaskiewicz JA, McCarthy CA, Richardson AC, et al. Febrile infants at low risk for serious bacterial infection—an appraisal of the Rochester criteria and implications for management. Febrile Infant Collaborative Study Group. Pediatrics. 1994;94:390396.
  9. Jain S, Cheng J, Alpern ER, et al. Management of febrile neonates in US pediatric emergency departments. Pediatrics. 2014;133:187195.
  10. Aronson PL, Thurm C, Alpern ER, et al. Variation in care of the febrile young infant <90 days in US pediatric emergency departments. Pediatrics. 2014;134:667677.
  11. Aronson PL, Thurm C, Williams DJ, et al. Association of clinical practice guidelines with emergency department management of febrile infants ≤56 days of age. J Hosp Med. 2015;10:358365.
  12. Hui C, Neto G, Tsertsvadze A, et al. Diagnosis and management of febrile infants (0‐3 months). Evid Rep Technol Assess (Full Rep). 2012;(205):1297.
  13. Tieder JS, Hall M, Auger KA, et al. Accuracy of administrative billing codes to detect urinary tract infection hospitalizations. Pediatrics. 2011;128:323330.
  14. Williams DJ, Shah SS, Myers A, et al. Identifying pediatric community‐acquired pneumonia hospitalizations: accuracy of administrative billing codes. JAMA Pediatr. 2013;167:851858.
  15. Benchimol EI, Manuel DG, To T, Griffiths AM, Rabeneck L, Guttmann A. Development and use of reporting guidelines for assessing the quality of validation studies of health administrative data. J Clin Epidemiol. 2011;64:821829.
  16. American College of Emergency Physicians Clinical Policies Committee; American College of Emergency Physicians Clinical Policies Subcommittee on Pediatric Fever. Clinical policy for children younger than three years presenting to the emergency department with fever. Ann Emerg Med. 2003;42:530545.
  17. Wood JN, Feudtner C, Medina SP, Luan X, Localio R, Rubin DM. Variation in occult injury screening for children with suspected abuse in selected US children's hospitals. Pediatrics. 2012;130:853860.
  18. Fletcher DM. Achieving data quality. How data from a pediatric health information system earns the trust of its users. J AHIMA. 2004;75:2226.
  19. Mongelluzzo J, Mohamad Z, Have TR, Shah SS. Corticosteroids and mortality in children with bacterial meningitis. JAMA. 2008;299:20482055.
  20. Harris PA, Taylor R, Thielke R, Payne J, Gonzalez N, Conde JG. Research electronic data capture (REDCap)—a metadata‐driven methodology and workflow process for providing translational research informatics support. J Biomed Inform. 2009;42:377381.
  21. Feudtner C, Hays RM, Haynes G, Geyer JR, Neff JM, Koepsell TD. Deaths attributed to pediatric complex chronic conditions: national trends and implications for supportive care services. Pediatrics. 2001;107:E99.
  22. US Census Bureau. Geographic terms and concepts—census divisions and census regions. Available at: https://www.census.gov/geo/reference/gtc/gtc_census_divreg.html. Accessed October 20, 2014.
  23. Gordon JA, An LC, Hayward RA, Williams BC. Initial emergency department diagnosis and return visits: risk versus perception. Ann Emerg Med. 1998;32:569573.
  24. Cho CS, Shapiro DJ, Cabana MD, Maselli JH, Hersh AL. A national depiction of children with return visits to the emergency department within 72 hours, 2001–2007. Pediatr Emerg Care. 2012;28:606610.
  25. Macy ML, Hall M, Shah SS, et al. Pediatric observation status: are we overlooking a growing population in children's hospitals? J Hosp Med. 2012;7:530536.
  26. Macy ML, Hall M, Shah SS, et al. Differences in designations of observation care in US freestanding children's hospitals: are they virtual or real? J Hosp Med. 2012;7:287293.
  27. Nigrovic LE, Fine AM, Monuteaux MC, Shah SS, Neuman MI. Trends in the management of viral meningitis at United States children's hospitals. Pediatrics. 2013;131:670676.
  28. Freedman SB, Hall M, Shah SS, et al. Impact of increasing ondansetron use on clinical outcomes in children with gastroenteritis. JAMA Pediatr. 2014;168:321329.
  29. Fleming‐Dutra KE, Shapiro DJ, Hicks LA, Gerber JS, Hersh AL. Race, otitis media, and antibiotic selection. Pediatrics. 2014;134:10591066.
  30. Parikh K, Hall M, Mittal V, et al. Establishing benchmarks for the hospitalized care of children with asthma, bronchiolitis, and pneumonia. Pediatrics. 2014;134:555562.
  31. Sheridan DC, Meckler GD, Spiro DM, Koch TK, Hansen ML. Diagnostic testing and treatment of pediatric headache in the emergency department. J Pediatr. 2013;163:16341637.
Article PDF
Issue
Journal of Hospital Medicine - 10(12)
Publications
Page Number
787-793
Sections
Files
Files
Article PDF
Article PDF

Fever is one of the most common reasons for emergency department (ED) evaluation of infants under 90 days of age.[1] Up to 10% to 20% of febrile young infants will have a serious bacterial infection (SBI),[2, 3, 4] but infants with SBI are difficult to distinguish from those without SBI based upon symptoms and physical examination findings alone.[5] Previously developed clinical prediction algorithms can help to identify febrile infants at low risk for SBI, but differ in age range as well as recommendations for testing and empiric treatment.[6, 7, 8] Consequently, there is widespread variation in management of febrile young infants at US children's hospitals,[9, 10, 11] and defining optimal management strategies remains an important issue in pediatric healthcare.[12] Administrative datasets are convenient and inexpensive, and can be used to evaluate practice variation, trends, and outcomes of a large, diverse group of patients within and across institutions.[9, 10] Accurately identifying febrile infants evaluated for suspected SBI in administrative databases would facilitate comparative effectiveness research, quality improvement initiatives, and institutional benchmarking.

Prior studies have validated the accuracy of administrative billing codes for identification of other common childhood illnesses, including urinary tract infection (UTI)[13] and pneumonia.[14] The accuracy of International Classification of Diseases, Ninth Revision (ICD‐9) diagnosis codes in identifying febrile young infants evaluated for SBI is not known. Reliance on administrative ICD‐9 diagnosis codes for patient identification can lead to misclassification of patients due to variable database quality, the validity of the diagnosis codes being utilized, and hospital coding practices.[15] Additionally, fever is a symptom and not a specific diagnosis. If a particular bacterial or viral diagnosis is established (eg, enterovirus meningitis), a discharge diagnosis of fever may not be attributed to the patient encounter. Thus, evaluating the performance characteristics and capture of clinical outcomes of different combinations of ICD‐9 diagnosis codes for identifying febrile infants is necessary for both the conduct and interpretation of studies that utilize administrative databases. The primary objective of this investigation was to identify the most accurate ICD‐9 coding strategies for the identification of febrile infants aged <90 days using administrative data. We also sought to evaluate capture of clinically important outcomes across identification strategies.

METHODS

Study Design and Setting

For this multicenter retrospective study, we used the Pediatric Health Information System (PHIS) database to identify infants <90 days of age[16] who presented between July 1, 2012 and June 30, 2013 to 1 of 8 EDs. We assessed performance characteristics of ICD‐9 diagnosis code case‐identification algorithms by comparing ICD‐9 code combinations to a fever reference standard determined by medical record review. The institutional review board at each participating site approved the study protocol.

Data Source

Data were obtained from 2 sources: the PHIS database and medical record review. We used the PHIS database to identify eligible patients by ICD‐9 diagnosis codes; patient encounters were randomly selected using a random number generator. The PHIS database contains demographic, diagnosis, and billing data from 44 hospitals affiliated with the Children's Hospital Association (Overland Park, Kansas) and represents 85% of freestanding children's hospitals in the United States.[17] Data are deidentified; encrypted unique patient identifiers permit tracking of patients across visits within a site.[18] The Children's Hospital Association and participating hospitals jointly assure the quality and integrity of the data.[19]

For each patient encounter identified in the PHIS database, detailed medical record review was performed by trained investigators at each of the 8 study sites (see Supporting Information, Appendix, in the online version of this article). A standardized data collection instrument was pilot tested by all investigators prior to use. Data were collected and managed using the Research Electronic Data Capture (REDCap) tool hosted at Boston Children's Hospital.[20]

Exclusions

Using PHIS data, prior to medical record review we excluded infants with a complex chronic condition as defined previously[21] and those transferred from another institution, as these infants may warrant a nonstandard evaluation and/or may have incomplete data.

ICD‐9 Diagnosis Code Groups

In the PHIS database, all patients discharged from the hospital (including hospitalized patients as well as patients discharged from the ED) receive 1 or more ICD‐9 discharge diagnosis codes. These diagnosis codes are ascribed after discharge from the hospital, or for ED patients, after ED discharge. Additionally, patients may receive an admission diagnosis, which reflects the diagnosis ascribed at the time of ED discharge or transfer to the inpatient unit.

We reviewed medical records of infants selected from the following ICD‐9 diagnosis code groups (Figure 1): (1) discharge diagnosis code of fever (780.6 [fever and other physiologic disturbances of temperature regulation], 778.4 [other disturbances of temperature regulation of newborn], 780.60 [fever, unspecified], or 780.61 [fever presenting with conditions classified elsewhere])[9, 10] regardless of the presence of admission diagnosis of fever or diagnosis of serious infection, (2) admission diagnosis code of fever without associated discharge diagnosis code of fever,[10] (3) discharge diagnosis code of serious infection determined a priori (see Supporting Information, Appendix, in the online version of this article) without discharge or admission diagnosis code of fever, and (4) infants without any diagnosis code of fever or serious infection.

jhm2441-fig-0001-m.png
Study population. 1Two of 584 medical records were unavailable for review. 2Five of 904 medical records were unavailable for review. Abbreviations: CCC, complex chronic condition; ED, emergency department.

Medical records reviewed in each of the 4 ICD‐9 diagnosis code groups were randomly selected from the overall set of ED encounters in the population of infants <90 days of age evaluated during the study period. Twenty‐five percent population sampling was used for 3 of the ICD‐9 diagnosis code groups, whereas 5% sampling was used for the no fever/no serious infection code group. The number of medical records reviewed in each ICD‐9 diagnosis code group was proportional to the distribution of ICD‐9 codes across the entire population of infants <90 days of age. These records were distributed equally across sites (228 records per site), except for 1 site that does not assign admission diagnoses (201 records).

Investigators were blinded to ICD‐9 diagnosis code groups during medical record review. Infants with multiple visits during the study period were eligible to be included more than once if the visits occurred more than 3 days apart. For infants with more than 1 ED visit on a particular calendar day, investigators were instructed to review the initial visit.

For each encounter, we also abstracted demographic characteristics (gender, race/ethnicity), insurance status, hospital region (using US Census categories[22]), and season from the PHIS database.

Reference Standard

The presence of fever was determined by medical record review. We defined fever as any documented temperature 100.4F (38.0C) at home or in the ED.[16]

ICD‐9 Code Case‐Identification Algorithms

Using the aforementioned ICD‐9 diagnosis code groups individually and in combination, the following 4 case‐identification algorithms, determined from prior study or group consensus, were compared to the reference standard: (1) ICD‐9 discharge diagnosis code of fever,[9] (2) ICD‐9 admission or discharge diagnosis code of fever,[10, 11] (3) ICD‐9 discharge diagnosis code of fever or serious infection, and (4) ICD‐9 discharge or admission diagnosis code of fever or serious infection. Algorithms were compared overall, separately for discharged and hospitalized infants, and across 3 distinct age groups (28 days, 2956 days, and 5789 days).

Patient‐Level Outcomes

To compare differences in outcomes by case‐identification algorithm, from the PHIS database we abstracted hospitalization rates, rates of UTI/pyelonephritis,[13] bacteremia/sepsis, and bacterial meningitis.[19] Severe outcomes were defined as intensive care unit admission, mechanical ventilation, central line placement, receipt of extracorporeal membrane oxygenation, or death. We assessed hospital length of stay for admitted infants and 3‐day revisits,[23, 24] and revisits resulting in hospitalization for infants discharged from the ED at the index visit. Patients billed for observation care were classified as being hospitalized.[25, 26]

Data Analysis

Accuracy of the 4 case‐identification algorithms (compared with the reference standard) was calculated using sensitivity, specificity, negative predictive value (NPV), and positive predictive value (PPV), along with 95% confidence interval (CI). Prior to analysis, a 5‐fold weighting factor was applied to the no fever/no serious infection group to account for the differential sampling used for this group (5% vs 25% for the other 3 ICD‐9 diagnosis code groups). This weighting was done to approximate the true prevalence of each ICD‐9 code group within the larger population, so that an accurate rate of false negatives (infants with fever who had neither a diagnosis of fever nor serious infection) could be calculated.

We described continuous variables using median and interquartile range or range values and categorical variables using frequencies with 95% CIs. We compared categorical variables using a 2 test. We determined statistical significance as a 2‐tailed P value <0.05. Statistical analyses were performed using SAS version 9.3 (SAS Institute, Cary, NC).

RESULTS

Study Patients

During the 1‐year study period, 23,753 ED encounters for infants <90 days of age were identified in the PHIS database at the 8 participating sites. Of these infant encounters, 2166 (9.2%) were excluded (1658 infants who had a complex chronic condition and 508 transferred into the ED), leaving 21,587 infants available for selection. After applying our sampling strategy, we identified 1797 encounters for medical record review. Seven encounters from 3 hospitals with missing medical records were excluded, resulting in a final cohort of 1790 encounters (Figure 1). Among included infants, 552 (30.8%) were 28 days, 743 (41.5%) were 29 to 56 days, and 495 (27.8%) were 57 to 89 days of age; 737 (41.2%) infants were hospitalized. Patients differed in age, race, payer, and season across ICD‐9 diagnosis code groups (see Supporting Information, Table 1, in the online version of this article).

Performance Characteristics of ICD‐9 Diagnosis Code Case‐Identification Algorithms According to Reference Standard (Overall, Hospitalized, and Discharged).*
ICD‐9 Diagnosis Code AlgorithmOverall
Sensitivity, % (95% CI)Specificity, % (95% CI)Negative Predictive Value, % (95% CI)Positive Predictive Value, % (95% CI)
  • NOTE: Abbreviations: CI, confidence interval; ED, emergency department; ICD‐9, International Classification of Diseases, Ninth Revision. *Reference standard of fever was defined by documented temperature 100.4 F (38.0 C) on review of electronic medical record.

Discharge diagnosis of fever53.2 (50.056.4)98.2 (97.898.6)90.8 (90.091.6)86.1 (83.388.9)
Hospitalized47.3 (43.151.5)97.7 (96.998.5)80.6 (78.682.6)90.2 (86.893.6)
Discharged from ED61.4 (56.666.2)98.4 (98.098.8)95.4 (94.796.1)82.1 (77.786.5)
Discharge or admission diagnosis of Fever71.1 (68.274.0)97.7 (97.398.1)94.1 (93.494.8)86.9 (84.589.3)
Hospitalized72.5 (68.876.2)97.1 (96.298.0)88.8 (87.190.5)91.7 (89.194.3)
Discharged from ED69.2 (64.773.7)98.0 (97.598.5)96.3 (95.796.9)80.8 (76.685.0)
Discharge diagnosis of fever or serious infection63.7 (60.666.8)96.5 (96.097.0)92.6 (91.893.4)79.6 (76.782.5)
Hospitalized63.9 (59.967.9)92.5 (91.094.0)85.1 (83.287.0)79.1 (75.382.9)
Discharged from ED63.4 (58.768.1)98.1 (97.698.6)95.6 (94.996.3)80.2 (75.884.6)
Discharge or admission diagnosis of fever or serious infection76.6 (73.979.3)96.2 (95.696.8)95.1 (94.595.7)81.0 (78.483.6)
Hospitalized80.8 (77.584.1)92.1 (90.693.6)91.5 (89.993.1)82.1 (78.985.3)
Discharged from ED71.0 (66.575.5)97.7 (97.298.2)96.5 (95.997.1)79.4 (75.283.6)

Among the 1790 patient encounters reviewed, a total of 766 infants (42.8%) met the reference standard definition for fever in the cohort. An additional 47 infants had abnormal temperature reported (documentation of tactile fever, history of fever without a specific temperature described, or hypothermia) but were classified as having no fever by the reference standard.

ICD‐9 Code Case‐Identification Algorithm Performance

Compared with the reference standard, the 4 case‐identification algorithms demonstrated specificity of 96.2% to 98.2% but lower sensitivity overall (Figure 2). Discharge diagnosis of fever alone demonstrated the lowest sensitivity. The algorithm of discharge or admission diagnosis of fever resulted in increased sensitivity and the highest PPV of all 4 algorithms (86.9%, 95% CI: 84.5‐89.3). Addition of serious infection codes to this algorithm resulted in a marginal increase in sensitivity and a similar decrease in PPV (Table 1). When limited to hospitalized infants, specificity was highest for the case‐identification algorithm of discharge diagnosis of fever and similarly high for discharge or admission diagnosis of fever; sensitivity was highest for the algorithm of discharge or admission diagnosis of fever or diagnosis of serious infection. For infants discharged from the ED, algorithm specificity was 97.7% to 98.4%, with lower sensitivity for all 4 algorithms (Table 1). Inclusion of the 47 infants with abnormal temperature as fever did not materially change algorithm performance (data not shown).

jhm2441-fig-0002-m.png
Algorithm sensitivity and false positive rate (1‐specificity) for identification of febrile infants aged ≤28 days, 29 to 56 days, 57 to 89 days, and overall. Horizontal and vertical bars represent 95% confidence intervals. Reference standard of fever was defined by documented temperature ≥100.4°F (38.0°C) on review of electronic medical record.

Across all 3 age groups (28 days, 2956 days, and 5789 days), the 4 case‐identification algorithms demonstrated specificity >96%, whereas algorithm sensitivity was highest in the 29‐ to 56‐days‐old age group and lowest among infants 57 to 89 days old across all 4 algorithms (Figure 2). Similar to the overall cohort, an algorithm of discharge or admission diagnosis of fever demonstrated specificity of nearly 98% in all age groups; addition of serious infection codes to this algorithm increased sensitivity, highest in the 29‐ to 56‐days‐old age group (Figure 2; see also Supporting Information, Table 2, in the online version of this article).

Performance Characteristics of ICD‐9 Diagnosis Code Case‐Identification Algorithms Across the Eight Sites According to Reference Standard.*
ICD‐9 Diagnosis Code AlgorithmSensitivity, Median % (Range)Specificity, Median % (Range)Negative Predictive Value, Median % (Range)Positive Predictive Value, Median % (Range)
  • NOTE: Abbreviations: ICD‐9, International Classification of Diseases, Ninth Revision. *Reference standard of fever was defined by documented temperature 100.4F (38.0 C) on review of electronic medical record.

Discharge diagnosis of fever56.2 (34.681.0)98.3 (96.499.1)92.1 (83.297.4)87.7 (74.093.2)
Discharge or Admission diagnosis of Fever76.7 (51.385.0)97.8 (96.298.7)95.6 (86.997.4)87.4 (80.092.9)
Discharge diagnosis of fever or serious infection68.3 (44.287.3)96.5 (95.498.0)93.6 (85.298.2)78.3 (74.289.0)
Discharge or admission diagnosis of fever or serious infection83.1 (58.390.7)95.8 (95.498.0)96.5 (88.598.2)79.1 (77.490.4)

Across the 8 study sites, median specificity was 95.8% to 98.3% for the 4 algorithms, with little interhospital variability; however, algorithm sensitivity varied widely by site. Median PPV was highest for discharge diagnosis of fever alone at 87.7% but ranged from 74.0% to 93.2% across sites. Median PPV for an algorithm of discharge or admission diagnosis of fever was similar (87.4%) but with less variation by site (range 80.0%92.9%) (Table 2).

Outcomes by ICD‐9 Diagnosis Code Group and Case‐Identification Algorithm

When compared with discharge diagnosis of fever, adding admission diagnosis of fever captured a higher proportion of hospitalized infants with SBIs (UTI/pyelonephritis, bacteremia/sepsis, or bacterial meningitis). However, median hospital length of stay, severe outcomes, and 3‐day revisits and revisits with hospitalization did not materially differ when including infants with admission diagnosis of fever in addition to discharge diagnosis of fever. Addition of infants with a diagnosis code for serious infection substantially increased the number of infants with SBIs and severe outcomes but did not capture additional 3‐day revisits (Table 3). There were no additional cases of SBI in the no fever/no serious illness diagnosis code group.

Outcomes by ICD‐9 Diagnosis Code Case‐Identification Algorithm
ICD‐9 Diagnosis Code AlgorithmOutcome3‐Day Revisit, % (95% CI)3‐Day Revisit With Hospitalization, % (95% CI)
Hospitalized, % (95% CI)UTI/Pyelonephritis, Bacteremia/Sepsis, or Bacterial Meningitis, % (95% CI)Severe Outcome, % (95% CI)*Length of Stay in Days, Median (IQR)
  • NOTE: Abbreviations: CI, confidence interval; ICD‐9, International Classification of Diseases, Ninth Revision; IQR, interquartile range; UTI, urinary tract infection. *Severe outcome was defined as intensive care unit admission, mechanical ventilation, central line placement, extracorporeal membrane oxygenation, or death. Length of stay for hospitalized infants. Percent of those discharged from the emergency department at the index visit.

Discharge diagnosis of fever44.3 (40.348.4)3.3 (1.84.7)1.4 (0.42.3)3 (23)11.7 (8.215.2)5.9 (3.38.4)
Discharge or admission diagnosis of fever52.4 (48.955.9)6.1 (4.47.8)1.9 (1.02.9)3 (23)10.9 (7.714.1)5.4 (3.17.8)
Discharge diagnosis of fever or serious infection54.0 (50.457.5)15.3 (12.717.8)3.8 (2.55.2)3 (24)11.0 (7.714.2)5.5 (3.17.9)
Discharge or admission diagnosis of fever or serious infection56.5 (53.259.7)12.9 (10.715.1)3.6 (2.44.8)3 (24)10.3 (7.313.3)5.2 (3.07.4)

Among infants who met the reference standard for fever but did not have a discharge or admission diagnosis of fever (false negatives), 11.8% had a diagnosis of SBI. Overall, 43.2% of febrile infants (and 84.4% of hospitalized infants) with SBI did not have an ICD‐9 discharge or admission diagnosis of fever. Addition of ICD‐9 diagnosis codes of serious infection to the algorithm of discharge or admission diagnosis of fever captured all additional SBIs, and no false negativeinfants missed with this algorithm had an SBI.

DISCUSSION

We described the performance of 4 ICD‐9 diagnosis code case‐identification algorithms for the identification of febrile young infants <90 days of age at US children's hospitals. Although the specificity was high across algorithms and institutions, the sensitivity was relatively low, particularly for discharge diagnosis of fever, and varied by institution. Given the high specificity, ICD‐9 diagnosis code case‐identification algorithms for fever reliably identify febrile infants using administrative data with low rates of inclusion of infants without fever. However, underidentification of patients, particularly those more prone to SBIs and severe outcomes depending on the algorithm utilized, can impact interpretation of comparative effectiveness studies or the quality of care delivered by an institution.

ICD‐9 discharge diagnosis codes are frequently used to identify pediatric patients across a variety of administrative databases, diseases, and symptoms.[19, 27, 28, 29, 30, 31] Although discharge diagnosis of fever is highly specific, sensitivity is substantially lower than other case‐identification algorithms we studied, particularly for hospitalized infants. This may be due to a fever code sometimes being omitted in favor of a more specific diagnosis (eg, bacteremia) prior to hospital discharge. Therefore, case identification relying only on ICD‐9 discharge diagnosis codes for fever may under‐report clinically important SBI or severe outcomes as demonstrated in our study. This is in contrast to ICD‐9 diagnosis code identification strategies for childhood UTI and pneumonia, which largely have higher sensitivity but lower specificity than fever codes.[13, 14]

Admission diagnosis of fever is important for febrile infants as they may not have an explicit diagnosis at the time of disposition from the ED. Addition of admission diagnosis of fever to an algorithm relying on discharge diagnosis code alone increased sensitivity without a demonstrable reduction in specificity and PPV, likely due to capture of infants with a fever diagnosis at presentation before a specific infection was identified. Although using an algorithm of discharge or admission diagnosis of fever captured a higher percentage of hospitalized febrile infants with SBIs, sensitivity was only 71% overall with this algorithm, and 43% of febrile infants with SBI would still have been missed. Importantly, though, addition of various ICD‐9 codes for serious infection to this algorithm resulted in capture of all febrile infants with SBI and should be used as a sensitivity analysis.

The test characteristics of diagnosis codes were highest in the 29‐ to 56‐days‐old age group. Given the differing low‐risk criteria[6, 7, 8] and lack of best practice guidelines[16] in this age group, the use of administrative data may allow for the comparison of testing and treatment strategies across a large cohort of febrile infants aged 29 to 56 days. However, individual hospital coding practices may affect algorithm performance, in particular sensitivity, which varied substantially by hospital. This variation in algorithm sensitivity may impact comparisons of outcomes across institutions. Therefore, when conducting studies of febrile infants using administrative data, sensitivity analyses or use of chart review should be considered to augment the use of ICD‐9 code‐based identification strategies, particularly for comparative benchmarking and outcomes studies. These additional analyses are particularly important for studies of febrile infants >56 days of age, in whom the sensitivity of diagnosis codes is particularly low. We speculate that the lower sensitivity in older febrile infants may relate to a lack of consensus on the clinical significance of fever in this age group and the varying management strategies employed.[10]

Strengths of this study include the assessment of ICD‐9 code algorithms across multiple institutions for identification of fever in young infants, and the patterns of our findings remained robust when comparing median performance characteristics of the algorithms across hospitals to our overall findings. We were also able to accurately estimate PPV and NPV using a case‐identification strategy weighted to the actual population sizes. Although sensitivity and specificity are the primary measures of test performance, predictive values are highly informative for investigators using administrative data. Additionally, our findings may inform public health efforts including disease surveillance, assessment of seasonal variation, and identification and monitoring of healthcare‐associated infections among febrile infants.

Our study has limitations. We did not review all identified records, which raises the possibility that our evaluated cohort may not be representative of the entire febrile infant population. We attempted to mitigate this possibility by using a random sampling strategy for our population selection that was weighted to the actual population sizes. Second, we identified serious infections using ICD‐9 diagnosis codes determined by group consensus, which may not capture all serious infection codes that identify febrile infants whose fever code was omitted. Third, 47 infants had abnormal temperature that did not meet our reference standard criteria for fever and were included in the no fever group. Although there may be disagreement regarding what constitutes a fever, we used a widely accepted reference standard to define fever.[16] Further, inclusion of these 47 infants as fever did not materially change algorithm performance. Last, our study was conducted at 8 large tertiary‐care children's hospitals, and our results may not be generalizable to other children's hospitals and community‐based hospitals.

CONCLUSIONS

Studies of febrile young infants that rely on ICD‐9 discharge diagnosis code of fever for case ascertainment have high specificity but low sensitivity for the identification of febrile infants, particularly among hospitalized patients. A case‐identification strategy that includes discharge or admission diagnosis of fever demonstrated higher sensitivity, and should be considered for studies of febrile infants using administrative data. However, additional strategies such as incorporation of ICD‐9 codes for serious infection should be used when comparing outcomes across institutions.

Acknowledgements

The Febrile Young Infant Research Collaborative includes the following additional collaborators who are acknowledged for their work on this study: Erica DiLeo, MA, Department of Medical Education and Research, Danbury Hospital, Danbury, Connecticut; Janet Flores, BS, Division of Emergency Medicine, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois.

Disclosures: This project funded in part by The Gerber Foundation Novice Researcher Award, (Ref No. 1827‐3835). Dr. Fran Balamuth received career development support from the National Institutes of Health (NHLBI K12‐HL109009). Funders were not involved in the design or conduct of the study; collection, management, analysis, or interpretation of the data; or preparation, review, or approval of the manuscript. The authors have no conflicts of interest relevant to this article to disclose.

Fever is one of the most common reasons for emergency department (ED) evaluation of infants under 90 days of age.[1] Up to 10% to 20% of febrile young infants will have a serious bacterial infection (SBI),[2, 3, 4] but infants with SBI are difficult to distinguish from those without SBI based upon symptoms and physical examination findings alone.[5] Previously developed clinical prediction algorithms can help to identify febrile infants at low risk for SBI, but differ in age range as well as recommendations for testing and empiric treatment.[6, 7, 8] Consequently, there is widespread variation in management of febrile young infants at US children's hospitals,[9, 10, 11] and defining optimal management strategies remains an important issue in pediatric healthcare.[12] Administrative datasets are convenient and inexpensive, and can be used to evaluate practice variation, trends, and outcomes of a large, diverse group of patients within and across institutions.[9, 10] Accurately identifying febrile infants evaluated for suspected SBI in administrative databases would facilitate comparative effectiveness research, quality improvement initiatives, and institutional benchmarking.

Prior studies have validated the accuracy of administrative billing codes for identification of other common childhood illnesses, including urinary tract infection (UTI)[13] and pneumonia.[14] The accuracy of International Classification of Diseases, Ninth Revision (ICD‐9) diagnosis codes in identifying febrile young infants evaluated for SBI is not known. Reliance on administrative ICD‐9 diagnosis codes for patient identification can lead to misclassification of patients due to variable database quality, the validity of the diagnosis codes being utilized, and hospital coding practices.[15] Additionally, fever is a symptom and not a specific diagnosis. If a particular bacterial or viral diagnosis is established (eg, enterovirus meningitis), a discharge diagnosis of fever may not be attributed to the patient encounter. Thus, evaluating the performance characteristics and capture of clinical outcomes of different combinations of ICD‐9 diagnosis codes for identifying febrile infants is necessary for both the conduct and interpretation of studies that utilize administrative databases. The primary objective of this investigation was to identify the most accurate ICD‐9 coding strategies for the identification of febrile infants aged <90 days using administrative data. We also sought to evaluate capture of clinically important outcomes across identification strategies.

METHODS

Study Design and Setting

For this multicenter retrospective study, we used the Pediatric Health Information System (PHIS) database to identify infants <90 days of age[16] who presented between July 1, 2012 and June 30, 2013 to 1 of 8 EDs. We assessed performance characteristics of ICD‐9 diagnosis code case‐identification algorithms by comparing ICD‐9 code combinations to a fever reference standard determined by medical record review. The institutional review board at each participating site approved the study protocol.

Data Source

Data were obtained from 2 sources: the PHIS database and medical record review. We used the PHIS database to identify eligible patients by ICD‐9 diagnosis codes; patient encounters were randomly selected using a random number generator. The PHIS database contains demographic, diagnosis, and billing data from 44 hospitals affiliated with the Children's Hospital Association (Overland Park, Kansas) and represents 85% of freestanding children's hospitals in the United States.[17] Data are deidentified; encrypted unique patient identifiers permit tracking of patients across visits within a site.[18] The Children's Hospital Association and participating hospitals jointly assure the quality and integrity of the data.[19]

For each patient encounter identified in the PHIS database, detailed medical record review was performed by trained investigators at each of the 8 study sites (see Supporting Information, Appendix, in the online version of this article). A standardized data collection instrument was pilot tested by all investigators prior to use. Data were collected and managed using the Research Electronic Data Capture (REDCap) tool hosted at Boston Children's Hospital.[20]

Exclusions

Using PHIS data, prior to medical record review we excluded infants with a complex chronic condition as defined previously[21] and those transferred from another institution, as these infants may warrant a nonstandard evaluation and/or may have incomplete data.

ICD‐9 Diagnosis Code Groups

In the PHIS database, all patients discharged from the hospital (including hospitalized patients as well as patients discharged from the ED) receive 1 or more ICD‐9 discharge diagnosis codes. These diagnosis codes are ascribed after discharge from the hospital, or for ED patients, after ED discharge. Additionally, patients may receive an admission diagnosis, which reflects the diagnosis ascribed at the time of ED discharge or transfer to the inpatient unit.

We reviewed medical records of infants selected from the following ICD‐9 diagnosis code groups (Figure 1): (1) discharge diagnosis code of fever (780.6 [fever and other physiologic disturbances of temperature regulation], 778.4 [other disturbances of temperature regulation of newborn], 780.60 [fever, unspecified], or 780.61 [fever presenting with conditions classified elsewhere])[9, 10] regardless of the presence of admission diagnosis of fever or diagnosis of serious infection, (2) admission diagnosis code of fever without associated discharge diagnosis code of fever,[10] (3) discharge diagnosis code of serious infection determined a priori (see Supporting Information, Appendix, in the online version of this article) without discharge or admission diagnosis code of fever, and (4) infants without any diagnosis code of fever or serious infection.

jhm2441-fig-0001-m.png
Study population. 1Two of 584 medical records were unavailable for review. 2Five of 904 medical records were unavailable for review. Abbreviations: CCC, complex chronic condition; ED, emergency department.

Medical records reviewed in each of the 4 ICD‐9 diagnosis code groups were randomly selected from the overall set of ED encounters in the population of infants <90 days of age evaluated during the study period. Twenty‐five percent population sampling was used for 3 of the ICD‐9 diagnosis code groups, whereas 5% sampling was used for the no fever/no serious infection code group. The number of medical records reviewed in each ICD‐9 diagnosis code group was proportional to the distribution of ICD‐9 codes across the entire population of infants <90 days of age. These records were distributed equally across sites (228 records per site), except for 1 site that does not assign admission diagnoses (201 records).

Investigators were blinded to ICD‐9 diagnosis code groups during medical record review. Infants with multiple visits during the study period were eligible to be included more than once if the visits occurred more than 3 days apart. For infants with more than 1 ED visit on a particular calendar day, investigators were instructed to review the initial visit.

For each encounter, we also abstracted demographic characteristics (gender, race/ethnicity), insurance status, hospital region (using US Census categories[22]), and season from the PHIS database.

Reference Standard

The presence of fever was determined by medical record review. We defined fever as any documented temperature 100.4F (38.0C) at home or in the ED.[16]

ICD‐9 Code Case‐Identification Algorithms

Using the aforementioned ICD‐9 diagnosis code groups individually and in combination, the following 4 case‐identification algorithms, determined from prior study or group consensus, were compared to the reference standard: (1) ICD‐9 discharge diagnosis code of fever,[9] (2) ICD‐9 admission or discharge diagnosis code of fever,[10, 11] (3) ICD‐9 discharge diagnosis code of fever or serious infection, and (4) ICD‐9 discharge or admission diagnosis code of fever or serious infection. Algorithms were compared overall, separately for discharged and hospitalized infants, and across 3 distinct age groups (28 days, 2956 days, and 5789 days).

Patient‐Level Outcomes

To compare differences in outcomes by case‐identification algorithm, from the PHIS database we abstracted hospitalization rates, rates of UTI/pyelonephritis,[13] bacteremia/sepsis, and bacterial meningitis.[19] Severe outcomes were defined as intensive care unit admission, mechanical ventilation, central line placement, receipt of extracorporeal membrane oxygenation, or death. We assessed hospital length of stay for admitted infants and 3‐day revisits,[23, 24] and revisits resulting in hospitalization for infants discharged from the ED at the index visit. Patients billed for observation care were classified as being hospitalized.[25, 26]

Data Analysis

Accuracy of the 4 case‐identification algorithms (compared with the reference standard) was calculated using sensitivity, specificity, negative predictive value (NPV), and positive predictive value (PPV), along with 95% confidence interval (CI). Prior to analysis, a 5‐fold weighting factor was applied to the no fever/no serious infection group to account for the differential sampling used for this group (5% vs 25% for the other 3 ICD‐9 diagnosis code groups). This weighting was done to approximate the true prevalence of each ICD‐9 code group within the larger population, so that an accurate rate of false negatives (infants with fever who had neither a diagnosis of fever nor serious infection) could be calculated.

We described continuous variables using median and interquartile range or range values and categorical variables using frequencies with 95% CIs. We compared categorical variables using a 2 test. We determined statistical significance as a 2‐tailed P value <0.05. Statistical analyses were performed using SAS version 9.3 (SAS Institute, Cary, NC).

RESULTS

Study Patients

During the 1‐year study period, 23,753 ED encounters for infants <90 days of age were identified in the PHIS database at the 8 participating sites. Of these infant encounters, 2166 (9.2%) were excluded (1658 infants who had a complex chronic condition and 508 transferred into the ED), leaving 21,587 infants available for selection. After applying our sampling strategy, we identified 1797 encounters for medical record review. Seven encounters from 3 hospitals with missing medical records were excluded, resulting in a final cohort of 1790 encounters (Figure 1). Among included infants, 552 (30.8%) were 28 days, 743 (41.5%) were 29 to 56 days, and 495 (27.8%) were 57 to 89 days of age; 737 (41.2%) infants were hospitalized. Patients differed in age, race, payer, and season across ICD‐9 diagnosis code groups (see Supporting Information, Table 1, in the online version of this article).

Performance Characteristics of ICD‐9 Diagnosis Code Case‐Identification Algorithms According to Reference Standard (Overall, Hospitalized, and Discharged).*
ICD‐9 Diagnosis Code AlgorithmOverall
Sensitivity, % (95% CI)Specificity, % (95% CI)Negative Predictive Value, % (95% CI)Positive Predictive Value, % (95% CI)
  • NOTE: Abbreviations: CI, confidence interval; ED, emergency department; ICD‐9, International Classification of Diseases, Ninth Revision. *Reference standard of fever was defined by documented temperature 100.4 F (38.0 C) on review of electronic medical record.

Discharge diagnosis of fever53.2 (50.056.4)98.2 (97.898.6)90.8 (90.091.6)86.1 (83.388.9)
Hospitalized47.3 (43.151.5)97.7 (96.998.5)80.6 (78.682.6)90.2 (86.893.6)
Discharged from ED61.4 (56.666.2)98.4 (98.098.8)95.4 (94.796.1)82.1 (77.786.5)
Discharge or admission diagnosis of Fever71.1 (68.274.0)97.7 (97.398.1)94.1 (93.494.8)86.9 (84.589.3)
Hospitalized72.5 (68.876.2)97.1 (96.298.0)88.8 (87.190.5)91.7 (89.194.3)
Discharged from ED69.2 (64.773.7)98.0 (97.598.5)96.3 (95.796.9)80.8 (76.685.0)
Discharge diagnosis of fever or serious infection63.7 (60.666.8)96.5 (96.097.0)92.6 (91.893.4)79.6 (76.782.5)
Hospitalized63.9 (59.967.9)92.5 (91.094.0)85.1 (83.287.0)79.1 (75.382.9)
Discharged from ED63.4 (58.768.1)98.1 (97.698.6)95.6 (94.996.3)80.2 (75.884.6)
Discharge or admission diagnosis of fever or serious infection76.6 (73.979.3)96.2 (95.696.8)95.1 (94.595.7)81.0 (78.483.6)
Hospitalized80.8 (77.584.1)92.1 (90.693.6)91.5 (89.993.1)82.1 (78.985.3)
Discharged from ED71.0 (66.575.5)97.7 (97.298.2)96.5 (95.997.1)79.4 (75.283.6)

Among the 1790 patient encounters reviewed, a total of 766 infants (42.8%) met the reference standard definition for fever in the cohort. An additional 47 infants had abnormal temperature reported (documentation of tactile fever, history of fever without a specific temperature described, or hypothermia) but were classified as having no fever by the reference standard.

ICD‐9 Code Case‐Identification Algorithm Performance

Compared with the reference standard, the 4 case‐identification algorithms demonstrated specificity of 96.2% to 98.2% but lower sensitivity overall (Figure 2). Discharge diagnosis of fever alone demonstrated the lowest sensitivity. The algorithm of discharge or admission diagnosis of fever resulted in increased sensitivity and the highest PPV of all 4 algorithms (86.9%, 95% CI: 84.5‐89.3). Addition of serious infection codes to this algorithm resulted in a marginal increase in sensitivity and a similar decrease in PPV (Table 1). When limited to hospitalized infants, specificity was highest for the case‐identification algorithm of discharge diagnosis of fever and similarly high for discharge or admission diagnosis of fever; sensitivity was highest for the algorithm of discharge or admission diagnosis of fever or diagnosis of serious infection. For infants discharged from the ED, algorithm specificity was 97.7% to 98.4%, with lower sensitivity for all 4 algorithms (Table 1). Inclusion of the 47 infants with abnormal temperature as fever did not materially change algorithm performance (data not shown).

jhm2441-fig-0002-m.png
Algorithm sensitivity and false positive rate (1‐specificity) for identification of febrile infants aged ≤28 days, 29 to 56 days, 57 to 89 days, and overall. Horizontal and vertical bars represent 95% confidence intervals. Reference standard of fever was defined by documented temperature ≥100.4°F (38.0°C) on review of electronic medical record.

Across all 3 age groups (28 days, 2956 days, and 5789 days), the 4 case‐identification algorithms demonstrated specificity >96%, whereas algorithm sensitivity was highest in the 29‐ to 56‐days‐old age group and lowest among infants 57 to 89 days old across all 4 algorithms (Figure 2). Similar to the overall cohort, an algorithm of discharge or admission diagnosis of fever demonstrated specificity of nearly 98% in all age groups; addition of serious infection codes to this algorithm increased sensitivity, highest in the 29‐ to 56‐days‐old age group (Figure 2; see also Supporting Information, Table 2, in the online version of this article).

Performance Characteristics of ICD‐9 Diagnosis Code Case‐Identification Algorithms Across the Eight Sites According to Reference Standard.*
ICD‐9 Diagnosis Code AlgorithmSensitivity, Median % (Range)Specificity, Median % (Range)Negative Predictive Value, Median % (Range)Positive Predictive Value, Median % (Range)
  • NOTE: Abbreviations: ICD‐9, International Classification of Diseases, Ninth Revision. *Reference standard of fever was defined by documented temperature 100.4F (38.0 C) on review of electronic medical record.

Discharge diagnosis of fever56.2 (34.681.0)98.3 (96.499.1)92.1 (83.297.4)87.7 (74.093.2)
Discharge or Admission diagnosis of Fever76.7 (51.385.0)97.8 (96.298.7)95.6 (86.997.4)87.4 (80.092.9)
Discharge diagnosis of fever or serious infection68.3 (44.287.3)96.5 (95.498.0)93.6 (85.298.2)78.3 (74.289.0)
Discharge or admission diagnosis of fever or serious infection83.1 (58.390.7)95.8 (95.498.0)96.5 (88.598.2)79.1 (77.490.4)

Across the 8 study sites, median specificity was 95.8% to 98.3% for the 4 algorithms, with little interhospital variability; however, algorithm sensitivity varied widely by site. Median PPV was highest for discharge diagnosis of fever alone at 87.7% but ranged from 74.0% to 93.2% across sites. Median PPV for an algorithm of discharge or admission diagnosis of fever was similar (87.4%) but with less variation by site (range 80.0%92.9%) (Table 2).

Outcomes by ICD‐9 Diagnosis Code Group and Case‐Identification Algorithm

When compared with discharge diagnosis of fever, adding admission diagnosis of fever captured a higher proportion of hospitalized infants with SBIs (UTI/pyelonephritis, bacteremia/sepsis, or bacterial meningitis). However, median hospital length of stay, severe outcomes, and 3‐day revisits and revisits with hospitalization did not materially differ when including infants with admission diagnosis of fever in addition to discharge diagnosis of fever. Addition of infants with a diagnosis code for serious infection substantially increased the number of infants with SBIs and severe outcomes but did not capture additional 3‐day revisits (Table 3). There were no additional cases of SBI in the no fever/no serious illness diagnosis code group.

Outcomes by ICD‐9 Diagnosis Code Case‐Identification Algorithm
ICD‐9 Diagnosis Code AlgorithmOutcome3‐Day Revisit, % (95% CI)3‐Day Revisit With Hospitalization, % (95% CI)
Hospitalized, % (95% CI)UTI/Pyelonephritis, Bacteremia/Sepsis, or Bacterial Meningitis, % (95% CI)Severe Outcome, % (95% CI)*Length of Stay in Days, Median (IQR)
  • NOTE: Abbreviations: CI, confidence interval; ICD‐9, International Classification of Diseases, Ninth Revision; IQR, interquartile range; UTI, urinary tract infection. *Severe outcome was defined as intensive care unit admission, mechanical ventilation, central line placement, extracorporeal membrane oxygenation, or death. Length of stay for hospitalized infants. Percent of those discharged from the emergency department at the index visit.

Discharge diagnosis of fever44.3 (40.348.4)3.3 (1.84.7)1.4 (0.42.3)3 (23)11.7 (8.215.2)5.9 (3.38.4)
Discharge or admission diagnosis of fever52.4 (48.955.9)6.1 (4.47.8)1.9 (1.02.9)3 (23)10.9 (7.714.1)5.4 (3.17.8)
Discharge diagnosis of fever or serious infection54.0 (50.457.5)15.3 (12.717.8)3.8 (2.55.2)3 (24)11.0 (7.714.2)5.5 (3.17.9)
Discharge or admission diagnosis of fever or serious infection56.5 (53.259.7)12.9 (10.715.1)3.6 (2.44.8)3 (24)10.3 (7.313.3)5.2 (3.07.4)

Among infants who met the reference standard for fever but did not have a discharge or admission diagnosis of fever (false negatives), 11.8% had a diagnosis of SBI. Overall, 43.2% of febrile infants (and 84.4% of hospitalized infants) with SBI did not have an ICD‐9 discharge or admission diagnosis of fever. Addition of ICD‐9 diagnosis codes of serious infection to the algorithm of discharge or admission diagnosis of fever captured all additional SBIs, and no false negativeinfants missed with this algorithm had an SBI.

DISCUSSION

We described the performance of 4 ICD‐9 diagnosis code case‐identification algorithms for the identification of febrile young infants <90 days of age at US children's hospitals. Although the specificity was high across algorithms and institutions, the sensitivity was relatively low, particularly for discharge diagnosis of fever, and varied by institution. Given the high specificity, ICD‐9 diagnosis code case‐identification algorithms for fever reliably identify febrile infants using administrative data with low rates of inclusion of infants without fever. However, underidentification of patients, particularly those more prone to SBIs and severe outcomes depending on the algorithm utilized, can impact interpretation of comparative effectiveness studies or the quality of care delivered by an institution.

ICD‐9 discharge diagnosis codes are frequently used to identify pediatric patients across a variety of administrative databases, diseases, and symptoms.[19, 27, 28, 29, 30, 31] Although discharge diagnosis of fever is highly specific, sensitivity is substantially lower than other case‐identification algorithms we studied, particularly for hospitalized infants. This may be due to a fever code sometimes being omitted in favor of a more specific diagnosis (eg, bacteremia) prior to hospital discharge. Therefore, case identification relying only on ICD‐9 discharge diagnosis codes for fever may under‐report clinically important SBI or severe outcomes as demonstrated in our study. This is in contrast to ICD‐9 diagnosis code identification strategies for childhood UTI and pneumonia, which largely have higher sensitivity but lower specificity than fever codes.[13, 14]

Admission diagnosis of fever is important for febrile infants as they may not have an explicit diagnosis at the time of disposition from the ED. Addition of admission diagnosis of fever to an algorithm relying on discharge diagnosis code alone increased sensitivity without a demonstrable reduction in specificity and PPV, likely due to capture of infants with a fever diagnosis at presentation before a specific infection was identified. Although using an algorithm of discharge or admission diagnosis of fever captured a higher percentage of hospitalized febrile infants with SBIs, sensitivity was only 71% overall with this algorithm, and 43% of febrile infants with SBI would still have been missed. Importantly, though, addition of various ICD‐9 codes for serious infection to this algorithm resulted in capture of all febrile infants with SBI and should be used as a sensitivity analysis.

The test characteristics of diagnosis codes were highest in the 29‐ to 56‐days‐old age group. Given the differing low‐risk criteria[6, 7, 8] and lack of best practice guidelines[16] in this age group, the use of administrative data may allow for the comparison of testing and treatment strategies across a large cohort of febrile infants aged 29 to 56 days. However, individual hospital coding practices may affect algorithm performance, in particular sensitivity, which varied substantially by hospital. This variation in algorithm sensitivity may impact comparisons of outcomes across institutions. Therefore, when conducting studies of febrile infants using administrative data, sensitivity analyses or use of chart review should be considered to augment the use of ICD‐9 code‐based identification strategies, particularly for comparative benchmarking and outcomes studies. These additional analyses are particularly important for studies of febrile infants >56 days of age, in whom the sensitivity of diagnosis codes is particularly low. We speculate that the lower sensitivity in older febrile infants may relate to a lack of consensus on the clinical significance of fever in this age group and the varying management strategies employed.[10]

Strengths of this study include the assessment of ICD‐9 code algorithms across multiple institutions for identification of fever in young infants, and the patterns of our findings remained robust when comparing median performance characteristics of the algorithms across hospitals to our overall findings. We were also able to accurately estimate PPV and NPV using a case‐identification strategy weighted to the actual population sizes. Although sensitivity and specificity are the primary measures of test performance, predictive values are highly informative for investigators using administrative data. Additionally, our findings may inform public health efforts including disease surveillance, assessment of seasonal variation, and identification and monitoring of healthcare‐associated infections among febrile infants.

Our study has limitations. We did not review all identified records, which raises the possibility that our evaluated cohort may not be representative of the entire febrile infant population. We attempted to mitigate this possibility by using a random sampling strategy for our population selection that was weighted to the actual population sizes. Second, we identified serious infections using ICD‐9 diagnosis codes determined by group consensus, which may not capture all serious infection codes that identify febrile infants whose fever code was omitted. Third, 47 infants had abnormal temperature that did not meet our reference standard criteria for fever and were included in the no fever group. Although there may be disagreement regarding what constitutes a fever, we used a widely accepted reference standard to define fever.[16] Further, inclusion of these 47 infants as fever did not materially change algorithm performance. Last, our study was conducted at 8 large tertiary‐care children's hospitals, and our results may not be generalizable to other children's hospitals and community‐based hospitals.

CONCLUSIONS

Studies of febrile young infants that rely on ICD‐9 discharge diagnosis code of fever for case ascertainment have high specificity but low sensitivity for the identification of febrile infants, particularly among hospitalized patients. A case‐identification strategy that includes discharge or admission diagnosis of fever demonstrated higher sensitivity, and should be considered for studies of febrile infants using administrative data. However, additional strategies such as incorporation of ICD‐9 codes for serious infection should be used when comparing outcomes across institutions.

Acknowledgements

The Febrile Young Infant Research Collaborative includes the following additional collaborators who are acknowledged for their work on this study: Erica DiLeo, MA, Department of Medical Education and Research, Danbury Hospital, Danbury, Connecticut; Janet Flores, BS, Division of Emergency Medicine, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois.

Disclosures: This project funded in part by The Gerber Foundation Novice Researcher Award, (Ref No. 1827‐3835). Dr. Fran Balamuth received career development support from the National Institutes of Health (NHLBI K12‐HL109009). Funders were not involved in the design or conduct of the study; collection, management, analysis, or interpretation of the data; or preparation, review, or approval of the manuscript. The authors have no conflicts of interest relevant to this article to disclose.

References
  1. Baskin MN. The prevalence of serious bacterial infections by age in febrile infants during the first 3 months of life. Pediatr Ann. 1993;22:462466.
  2. Huppler AR, Eickhoff JC, Wald ER. Performance of low‐risk criteria in the evaluation of young infants with fever: review of the literature. Pediatrics. 2010;125:228233.
  3. Schwartz S, Raveh D, Toker O, Segal G, Godovitch N, Schlesinger Y. A week‐by‐week analysis of the low‐risk criteria for serious bacterial infection in febrile neonates. Arch Dis Child. 2009;94:287292.
  4. Garcia S, Mintegi S, Gomez B, et al. Is 15 days an appropriate cut‐off age for considering serious bacterial infection in the management of febrile infants? Pediatr Infect Dis J. 2012;31:455458.
  5. Baker MD, Avner JR, Bell LM. Failure of infant observation scales in detecting serious illness in febrile, 4‐ to 8‐week‐old infants. Pediatrics. 1990;85:10401043.
  6. Baker MD, Bell LM, Avner JR. Outpatient management without antibiotics of fever in selected infants. N Engl J Med. 1993;329:14371441.
  7. Baskin MN, Fleisher GR, O'Rourke EJ. Identifying febrile infants at risk for a serious bacterial infection. J Pediatr. 1993;123:489490.
  8. Jaskiewicz JA, McCarthy CA, Richardson AC, et al. Febrile infants at low risk for serious bacterial infection—an appraisal of the Rochester criteria and implications for management. Febrile Infant Collaborative Study Group. Pediatrics. 1994;94:390396.
  9. Jain S, Cheng J, Alpern ER, et al. Management of febrile neonates in US pediatric emergency departments. Pediatrics. 2014;133:187195.
  10. Aronson PL, Thurm C, Alpern ER, et al. Variation in care of the febrile young infant <90 days in US pediatric emergency departments. Pediatrics. 2014;134:667677.
  11. Aronson PL, Thurm C, Williams DJ, et al. Association of clinical practice guidelines with emergency department management of febrile infants ≤56 days of age. J Hosp Med. 2015;10:358365.
  12. Hui C, Neto G, Tsertsvadze A, et al. Diagnosis and management of febrile infants (0‐3 months). Evid Rep Technol Assess (Full Rep). 2012;(205):1297.
  13. Tieder JS, Hall M, Auger KA, et al. Accuracy of administrative billing codes to detect urinary tract infection hospitalizations. Pediatrics. 2011;128:323330.
  14. Williams DJ, Shah SS, Myers A, et al. Identifying pediatric community‐acquired pneumonia hospitalizations: accuracy of administrative billing codes. JAMA Pediatr. 2013;167:851858.
  15. Benchimol EI, Manuel DG, To T, Griffiths AM, Rabeneck L, Guttmann A. Development and use of reporting guidelines for assessing the quality of validation studies of health administrative data. J Clin Epidemiol. 2011;64:821829.
  16. American College of Emergency Physicians Clinical Policies Committee; American College of Emergency Physicians Clinical Policies Subcommittee on Pediatric Fever. Clinical policy for children younger than three years presenting to the emergency department with fever. Ann Emerg Med. 2003;42:530545.
  17. Wood JN, Feudtner C, Medina SP, Luan X, Localio R, Rubin DM. Variation in occult injury screening for children with suspected abuse in selected US children's hospitals. Pediatrics. 2012;130:853860.
  18. Fletcher DM. Achieving data quality. How data from a pediatric health information system earns the trust of its users. J AHIMA. 2004;75:2226.
  19. Mongelluzzo J, Mohamad Z, Have TR, Shah SS. Corticosteroids and mortality in children with bacterial meningitis. JAMA. 2008;299:20482055.
  20. Harris PA, Taylor R, Thielke R, Payne J, Gonzalez N, Conde JG. Research electronic data capture (REDCap)—a metadata‐driven methodology and workflow process for providing translational research informatics support. J Biomed Inform. 2009;42:377381.
  21. Feudtner C, Hays RM, Haynes G, Geyer JR, Neff JM, Koepsell TD. Deaths attributed to pediatric complex chronic conditions: national trends and implications for supportive care services. Pediatrics. 2001;107:E99.
  22. US Census Bureau. Geographic terms and concepts—census divisions and census regions. Available at: https://www.census.gov/geo/reference/gtc/gtc_census_divreg.html. Accessed October 20, 2014.
  23. Gordon JA, An LC, Hayward RA, Williams BC. Initial emergency department diagnosis and return visits: risk versus perception. Ann Emerg Med. 1998;32:569573.
  24. Cho CS, Shapiro DJ, Cabana MD, Maselli JH, Hersh AL. A national depiction of children with return visits to the emergency department within 72 hours, 2001–2007. Pediatr Emerg Care. 2012;28:606610.
  25. Macy ML, Hall M, Shah SS, et al. Pediatric observation status: are we overlooking a growing population in children's hospitals? J Hosp Med. 2012;7:530536.
  26. Macy ML, Hall M, Shah SS, et al. Differences in designations of observation care in US freestanding children's hospitals: are they virtual or real? J Hosp Med. 2012;7:287293.
  27. Nigrovic LE, Fine AM, Monuteaux MC, Shah SS, Neuman MI. Trends in the management of viral meningitis at United States children's hospitals. Pediatrics. 2013;131:670676.
  28. Freedman SB, Hall M, Shah SS, et al. Impact of increasing ondansetron use on clinical outcomes in children with gastroenteritis. JAMA Pediatr. 2014;168:321329.
  29. Fleming‐Dutra KE, Shapiro DJ, Hicks LA, Gerber JS, Hersh AL. Race, otitis media, and antibiotic selection. Pediatrics. 2014;134:10591066.
  30. Parikh K, Hall M, Mittal V, et al. Establishing benchmarks for the hospitalized care of children with asthma, bronchiolitis, and pneumonia. Pediatrics. 2014;134:555562.
  31. Sheridan DC, Meckler GD, Spiro DM, Koch TK, Hansen ML. Diagnostic testing and treatment of pediatric headache in the emergency department. J Pediatr. 2013;163:16341637.
References
  1. Baskin MN. The prevalence of serious bacterial infections by age in febrile infants during the first 3 months of life. Pediatr Ann. 1993;22:462466.
  2. Huppler AR, Eickhoff JC, Wald ER. Performance of low‐risk criteria in the evaluation of young infants with fever: review of the literature. Pediatrics. 2010;125:228233.
  3. Schwartz S, Raveh D, Toker O, Segal G, Godovitch N, Schlesinger Y. A week‐by‐week analysis of the low‐risk criteria for serious bacterial infection in febrile neonates. Arch Dis Child. 2009;94:287292.
  4. Garcia S, Mintegi S, Gomez B, et al. Is 15 days an appropriate cut‐off age for considering serious bacterial infection in the management of febrile infants? Pediatr Infect Dis J. 2012;31:455458.
  5. Baker MD, Avner JR, Bell LM. Failure of infant observation scales in detecting serious illness in febrile, 4‐ to 8‐week‐old infants. Pediatrics. 1990;85:10401043.
  6. Baker MD, Bell LM, Avner JR. Outpatient management without antibiotics of fever in selected infants. N Engl J Med. 1993;329:14371441.
  7. Baskin MN, Fleisher GR, O'Rourke EJ. Identifying febrile infants at risk for a serious bacterial infection. J Pediatr. 1993;123:489490.
  8. Jaskiewicz JA, McCarthy CA, Richardson AC, et al. Febrile infants at low risk for serious bacterial infection—an appraisal of the Rochester criteria and implications for management. Febrile Infant Collaborative Study Group. Pediatrics. 1994;94:390396.
  9. Jain S, Cheng J, Alpern ER, et al. Management of febrile neonates in US pediatric emergency departments. Pediatrics. 2014;133:187195.
  10. Aronson PL, Thurm C, Alpern ER, et al. Variation in care of the febrile young infant <90 days in US pediatric emergency departments. Pediatrics. 2014;134:667677.
  11. Aronson PL, Thurm C, Williams DJ, et al. Association of clinical practice guidelines with emergency department management of febrile infants ≤56 days of age. J Hosp Med. 2015;10:358365.
  12. Hui C, Neto G, Tsertsvadze A, et al. Diagnosis and management of febrile infants (0‐3 months). Evid Rep Technol Assess (Full Rep). 2012;(205):1297.
  13. Tieder JS, Hall M, Auger KA, et al. Accuracy of administrative billing codes to detect urinary tract infection hospitalizations. Pediatrics. 2011;128:323330.
  14. Williams DJ, Shah SS, Myers A, et al. Identifying pediatric community‐acquired pneumonia hospitalizations: accuracy of administrative billing codes. JAMA Pediatr. 2013;167:851858.
  15. Benchimol EI, Manuel DG, To T, Griffiths AM, Rabeneck L, Guttmann A. Development and use of reporting guidelines for assessing the quality of validation studies of health administrative data. J Clin Epidemiol. 2011;64:821829.
  16. American College of Emergency Physicians Clinical Policies Committee; American College of Emergency Physicians Clinical Policies Subcommittee on Pediatric Fever. Clinical policy for children younger than three years presenting to the emergency department with fever. Ann Emerg Med. 2003;42:530545.
  17. Wood JN, Feudtner C, Medina SP, Luan X, Localio R, Rubin DM. Variation in occult injury screening for children with suspected abuse in selected US children's hospitals. Pediatrics. 2012;130:853860.
  18. Fletcher DM. Achieving data quality. How data from a pediatric health information system earns the trust of its users. J AHIMA. 2004;75:2226.
  19. Mongelluzzo J, Mohamad Z, Have TR, Shah SS. Corticosteroids and mortality in children with bacterial meningitis. JAMA. 2008;299:20482055.
  20. Harris PA, Taylor R, Thielke R, Payne J, Gonzalez N, Conde JG. Research electronic data capture (REDCap)—a metadata‐driven methodology and workflow process for providing translational research informatics support. J Biomed Inform. 2009;42:377381.
  21. Feudtner C, Hays RM, Haynes G, Geyer JR, Neff JM, Koepsell TD. Deaths attributed to pediatric complex chronic conditions: national trends and implications for supportive care services. Pediatrics. 2001;107:E99.
  22. US Census Bureau. Geographic terms and concepts—census divisions and census regions. Available at: https://www.census.gov/geo/reference/gtc/gtc_census_divreg.html. Accessed October 20, 2014.
  23. Gordon JA, An LC, Hayward RA, Williams BC. Initial emergency department diagnosis and return visits: risk versus perception. Ann Emerg Med. 1998;32:569573.
  24. Cho CS, Shapiro DJ, Cabana MD, Maselli JH, Hersh AL. A national depiction of children with return visits to the emergency department within 72 hours, 2001–2007. Pediatr Emerg Care. 2012;28:606610.
  25. Macy ML, Hall M, Shah SS, et al. Pediatric observation status: are we overlooking a growing population in children's hospitals? J Hosp Med. 2012;7:530536.
  26. Macy ML, Hall M, Shah SS, et al. Differences in designations of observation care in US freestanding children's hospitals: are they virtual or real? J Hosp Med. 2012;7:287293.
  27. Nigrovic LE, Fine AM, Monuteaux MC, Shah SS, Neuman MI. Trends in the management of viral meningitis at United States children's hospitals. Pediatrics. 2013;131:670676.
  28. Freedman SB, Hall M, Shah SS, et al. Impact of increasing ondansetron use on clinical outcomes in children with gastroenteritis. JAMA Pediatr. 2014;168:321329.
  29. Fleming‐Dutra KE, Shapiro DJ, Hicks LA, Gerber JS, Hersh AL. Race, otitis media, and antibiotic selection. Pediatrics. 2014;134:10591066.
  30. Parikh K, Hall M, Mittal V, et al. Establishing benchmarks for the hospitalized care of children with asthma, bronchiolitis, and pneumonia. Pediatrics. 2014;134:555562.
  31. Sheridan DC, Meckler GD, Spiro DM, Koch TK, Hansen ML. Diagnostic testing and treatment of pediatric headache in the emergency department. J Pediatr. 2013;163:16341637.
Issue
Journal of Hospital Medicine - 10(12)
Issue
Journal of Hospital Medicine - 10(12)
Page Number
787-793
Page Number
787-793
Publications
Publications
Article Type
Display Headline
Accuracy of diagnosis codes to identify febrile young infants using administrative data
Display Headline
Accuracy of diagnosis codes to identify febrile young infants using administrative data
Sections
Article Source

© 2015 Society of Hospital Medicine

Disallow All Ads
Correspondence Location
Address for correspondence and reprint requests: Paul L. Aronson, MD, Section of Pediatric Emergency Medicine, Yale School of Medicine, 100 York Street, Suite 1F, New Haven, CT, 06511; Telephone: 203‐737‐7443; Fax: 203‐737‐7447; E‐mail: paul.aronson@yale.edu
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Article PDF Media
Image
Disable zoom
Off
Media Files
Image
Disable zoom
Off
Image
Disable zoom
Off

OUs and Patient Outcomes

Article Type
Changed
Sun, 05/21/2017 - 13:05
Display Headline
Observation‐status patients in children's hospitals with and without dedicated observation units in 2011

Many pediatric hospitalizations are of short duration, and more than half of short‐stay hospitalizations are designated as observation status.[1, 2] Observation status is an administrative label assigned to patients who do not meet hospital or payer criteria for inpatient‐status care. Short‐stay observation‐status patients do not fit in traditional models of emergency department (ED) or inpatient care. EDs often focus on discharging or admitting patients within a matter of hours, whereas inpatient units tend to measure length of stay (LOS) in terms of days[3] and may not have systems in place to facilitate rapid discharge of short‐stay patients.[4] Observation units (OUs) have been established in some hospitals to address the unique care needs of short‐stay patients.[5, 6, 7]

Single‐site reports from children's hospitals with successful OUs have demonstrated shorter LOS and lower costs compared with inpatient settings.[6, 8, 9, 10, 11, 12, 13, 14] No prior study has examined hospital‐level effects of an OU on observation‐status patient outcomes. The Pediatric Health Information System (PHIS) database provides a unique opportunity to explore this question, because unlike other national hospital administrative databases,[15, 16] the PHIS dataset contains information about children under observation status. In addition, we know which PHIS hospitals had a dedicated OU in 2011.7

We hypothesized that overall observation‐status stays in hospitals with a dedicated OU would be of shorter duration with earlier discharges at lower cost than observation‐status stays in hospitals without a dedicated OU. We compared hospitals with and without a dedicated OU on secondary outcomes including rates of conversion to inpatient status and return care for any reason.

METHODS

We conducted a cross‐sectional analysis of hospital administrative data using the 2011 PHIS databasea national administrative database that contains resource utilization data from 43 participating hospitals located in 26 states plus the District of Columbia. These hospitals account for approximately 20% of pediatric hospitalizations in the United States.

For each hospital encounter, PHIS includes patient demographics, up to 41 International Classification of Diseases, Ninth Revision, Clinical Modification (ICD‐9‐CM) diagnoses, up to 41 ICD‐9‐CM procedures, and hospital charges for services. Data are deidentified prior to inclusion, but unique identifiers allow for determination of return visits and readmissions following an index visit for an individual patient. Data quality and reliability are assured jointly by the Children's Hospital Association (formerly Child Health Corporation of America, Overland Park, KS), participating hospitals, and Truven Health Analytics (New York, NY). This study, using administrative data, was not considered human subjects research by the policies of the Cincinnati Children's Hospital Medical Center Institutional Review Board.

Hospital Selection and Hospital Characteristics

The study sample was drawn from the 31 hospitals that reported observation‐status patient data to PHIS in 2011. Analyses were conducted in 2013, at which time 2011 was the most recent year of data. We categorized 14 hospitals as having a dedicated OU during 2011 based on information collected in 2013.7 To summarize briefly, we interviewed by telephone representatives of hospitals responding to an email query as to the presence of a geographically distinct OU for the care of unscheduled patients from the ED. Three of the 14 representatives reported their hospital had 2 OUs, 1 of which was a separate surgical OU. Ten OUs cared for both ED patients and patients with scheduled procedures; 8 units received patients from non‐ED sources. Hospitalists provided staffing in more than half of the OUs.

We attempted to identify administrative data that would signal care delivered in a dedicated OU using hospital charge codes reported to PHIS, but learned this was not possible due to between‐hospital variation in the specificity of the charge codes. Therefore, we were unable to determine if patient care was delivered in a dedicated OU or another setting, such as a general inpatient unit or the ED. Other hospital characteristics available from the PHIS dataset included the number of inpatient beds, ED visits, inpatient admissions, observation‐status stays, and payer mix. We calculated the percentage of ED visits resulting in admission by dividing the number of ED visits with associated inpatient or observation status by the total number of ED visits and the percentage of admissions under observation status by dividing the number of observation‐status stays by the total number of admissions under observation or inpatient status.

Visit Selection and Patient Characteristics

All observation‐status stays regardless of the point of entry into the hospital were eligible for this study. We excluded stays that were birth‐related, included intensive care, or resulted in transfer or death. Patient demographic characteristics used to describe the cohort included age, gender, race/ethnicity, and primary payer. Stays that began in the ED were identified by an emergency room charge within PHIS. Eligible stays were categorized using All Patient Refined Diagnosis Related Groups (APR‐DRGs) version 24 using the ICD‐9‐CM code‐based proprietary 3M software (3M Health Information Systems, St. Paul, MN). We determined the 15 top‐ranking APR‐DRGs among observation‐status stays in hospitals with a dedicated OU and hospitals without. Procedural stays were identified based on procedural APR‐DRGs (eg, tonsil and adenoid procedures) or the presence of an ICD‐9‐CM procedure code (eg, 331 spinal tap).

Measured Outcomes

Outcomes of observation‐status stays were determined within 4 categories: (1) LOS, (2) standardized costs, (3) conversion to inpatient status, and (4) return visits and readmissions. LOS was calculated in terms of nights spent in hospital for all stays by subtracting the discharge date from the admission date and in terms of hours for stays in the 28 hospitals that report admission and discharge hour to the PHIS database. Discharge timing was examined in 4, 6‐hour blocks starting at midnight. Standardized costs were derived from a charge master index that was created by taking the median costs from all PHIS hospitals for each charged service.[17] Standardized costs represent the estimated cost of providing any particular clinical activity but are not the cost to patients, nor do they represent the actual cost to any given hospital. This approach allows for cost comparisons across hospitals, without biases arising from using charges or from deriving costs using hospitals' ratios of costs to charges.[18] Conversion from observation to inpatient status was calculated by dividing the number of inpatient‐status stays with observation codes by the number of observation‐statusonly stays plus the number of inpatient‐status stays with observation codes. All‐cause 3‐day ED return visits and 30‐day readmissions to the same hospital were assessed using patient‐specific identifiers that allowed for tracking of ED return visits and readmissions following the index observation stay.

Data Analysis

Descriptive statistics were calculated for hospital and patient characteristics using medians and interquartile ranges (IQRs) for continuous factors and frequencies with percentages for categorical factors. Comparisons of these factors between hospitals with dedicated OUs and without were made using [2] and Wilcoxon rank sum tests as appropriate. Multivariable regression was performed using generalized linear mixed models treating hospital as a random effect and used patient age, the case‐mix index based on the APR‐DRG severity of illness, ED visit, and procedures associated with the index observation‐status stay. For continuous outcomes, we performed a log transformation on the outcome, confirmed the normality assumption, and back transformed the results. Sensitivity analyses were conducted to compare LOS, standardized costs, and conversation rates by hospital type for 10 of the 15 top‐ranking APR‐DRGs commonly cared for by pediatric hospitalists and to compare hospitals that reported the presence of an OU that was consistently open (24 hours per day, 7 days per week) and operating during the entire 2011 calendar year, and those without. Based on information gathered from the telephone interviews, hospitals with partially open OUs were similar to hospitals with continuously open OUs, such that they were included in our main analyses. All statistical analyses were performed using SAS version 9.3 (SAS Institute, Cary, NC). P values <0.05 were considered statistically significant.

RESULTS

Hospital Characteristics

Dedicated OUs were present in 14 of the 31 hospitals that reported observation‐status patient data to PHIS (Figure 1). Three of these hospitals had OUs that were open for 5 months or less in 2011; 1 unit opened, 1 unit closed, and 1 hospital operated a seasonal unit. The remaining 17 hospitals reported no OU that admitted unscheduled patients from the ED during 2011. Hospitals with a dedicated OU had more inpatient beds and higher median number of inpatient admissions than those without (Table 1). Hospitals were statistically similar in terms of total volume of ED visits, percentage of ED visits resulting in admission, total number of observation‐status stays, percentage of admissions under observation status, and payer mix.

jhm2339-fig-0001-m.png
Study Hospital Cohort Selection
Hospitals* With and Without Dedicated Observation Units
 Overall, Median (IQR)Hospitals With a Dedicated Observation Unit, Median (IQR)Hospitals Without a Dedicated Observation Unit, Median (IQR)P Value
  • NOTE: Abbreviations: ED, emergency department; IQR, interquartile range. *Among hospitals that reported observation‐status patient data to the Pediatric Health Information System database in 2011. Hospitals reporting the presence of at least 1 dedicated observation unit that admitted unscheduled patients from the ED in 2011. Percent of ED visits resulting in admission=number of ED visits admitted to inpatient or observation status divided by total number of ED visits in 2011. Percent of admissions under observation status=number of observation‐status stays divided by the total number of admissions (observation and inpatient status) in 2011.

No. of hospitals311417 
Total no. of inpatient beds273 (213311)304 (269425)246 (175293)0.006
Total no. ED visits62971 (47,50497,723)87,892 (55,102117,119)53,151 (4750470,882)0.21
ED visits resulting in admission, %13.1 (9.715.0)13.8 (10.5, 19.1)12.5 (9.714.5)0.31
Total no. of inpatient admissions11,537 (9,26814,568)13,206 (11,32517,869)10,207 (8,64013,363)0.04
Admissions under observation status, %25.7 (19.733.8)25.5 (21.431.4)26.0 (16.935.1)0.98
Total no. of observation stays3,820 (27935672)4,850 (3,309 6,196)3,141 (2,3654,616)0.07
Government payer, %60.2 (53.371.2)62.1 (54.9, 65.9)59.2 (53.373.7)0.89

Observation‐Status Patients by Hospital Type

In 2011, there were a total of 136,239 observation‐status stays69,983 (51.4%) within the 14 hospitals with a dedicated OU and 66,256 (48.6%) within the 17 hospitals without. Patient care originated in the ED for 57.8% observation‐status stays in hospitals with an OU compared with 53.0% of observation‐status stays in hospitals without (P<0.001). Compared with hospitals with a dedicated OU, those without a dedicated OU had higher percentages of observation‐status patients older than 12 years and non‐Hispanic and a higher percentage of observation‐status patients with private payer type (Table 2). The 15 top‐ranking APR‐DRGs accounted for roughly half of all observation‐status stays and were relatively consistent between hospitals with and without a dedicated OU (Table 3). Procedural care was frequently associated with observation‐status stays.

Observation‐Status Patients by Hospital Type
 Overall, No. (%)Hospitals With a Dedicated Observation Unit, No. (%)*Hospitals Without a Dedicated Observation Unit, No. (%)P Value
  • NOTE: *Hospitals reporting the presence of at least 1 dedicated observation unit that admitted unscheduled patients from the emergency department in 2011.

Age    
<1 year23,845 (17.5)12,101 (17.3)11,744 (17.7)<0.001
15 years53,405 (38.5)28,052 (40.1)24,353 (36.8) 
612 years33,674 (24.7)17,215 (24.6)16,459 (24.8) 
1318 years23,607 (17.3)11,472 (16.4)12,135 (18.3) 
>18 years2,708 (2)1,143 (1.6)1,565 (2.4) 
Gender    
Male76,142 (55.9)39,178 (56)36,964 (55.8)0.43
Female60,025 (44.1)30,756 (44)29,269 (44.2) 
Race/ethnicity    
Non‐Hispanic white72,183 (53.0)30,653 (43.8)41,530 (62.7)<0.001
Non‐Hispanic black30,995 (22.8)16,314 (23.3)14,681 (22.2) 
Hispanic21,255 (15.6)16,583 (23.7)4,672 (7.1) 
Asian2,075 (1.5)1,313 (1.9)762 (1.2) 
Non‐Hispanic other9,731 (7.1)5,120 (7.3)4,611 (7.0) 
Payer    
Government68,725 (50.4)36,967 (52.8)31,758 (47.9)<0.001
Private48,416 (35.5)21,112 (30.2)27,304 (41.2) 
Other19,098 (14.0)11,904 (17)7,194 (10.9) 
Fifteen Most Common APR‐DRGs for Observation‐Status Patients by Hospital Type
Observation‐Status Patients in Hospitals With a Dedicated Observation Unit*Observation‐Status Patients in Hospitals Without a Dedicated Observation Unit
RankAPR‐DRGNo.% of All Observation Status Stays% Began in EDRankAPR‐DRGNo.% of All Observation Status Stays% Began in ED
  • NOTE: Abbreviations: APR‐DRG, All Patient Refined Diagnosis Related Group; ED, emergency department; ENT, ear, nose, and throat; NEC, not elsewhere classified; RSV, respiratory syncytial virus. *Hospitals reporting the presence of at least 1 dedicated observation unit that admitted unscheduled patients from the ED in 2011. Within the APR‐DRG. Procedure codes associated with 99% to 100% of observation stays within the APR‐DRG. Procedure codes associated with 20% 45% of observation stays within APR‐DRG; procedure codes were associated with <20% of observation stays within the APR‐DRG that are not indicated otherwise.

1Tonsil and adenoid procedures4,6216.61.31Tonsil and adenoid procedures3,8065.71.6
2Asthma4,2466.185.32Asthma3,7565.779.0
3Seizure3,5165.052.03Seizure2,8464.354.9
4Nonbacterial gastroenteritis3,2864.785.84Upper respiratory infections2,7334.169.6
5Bronchiolitis, RSV pneumonia3,0934.478.55Nonbacterial gastroenteritis2,6824.074.5
6Upper respiratory infections2,9234.280.06Other digestive system diagnoses2,5453.866.3
7Other digestive system diagnoses2,0642.974.07Bronchiolitis, RSV pneumonia2,5443.869.2
8Respiratory signs, symptoms, diagnoses2,0522.981.68Shoulder and arm procedures1,8622.872.6
9Other ENT/cranial/facial diagnoses1,6842.443.69Appendectomy1,7852.779.2
10Shoulder and arm procedures1,6242.379.110Other ENT/cranial/facial diagnoses1,6242.529.9
11Abdominal pain1,6122.386.211Abdominal pain1,4612.282.3
12Fever1,4942.185.112Other factors influencing health status1,4612.266.3
13Appendectomy1,4652.166.413Cellulitis/other bacterial skin infections1,3832.184.2
14Cellulitis/other bacterial skin infections1,3932.086.414Respiratory signs, symptoms, diagnoses1,3082.039.1
15Pneumonia NEC1,3561.979.115Pneumonia NEC1,2451.973.1
 Total36,42952.057.8 Total33,04149.8753.0

Outcomes of Observation‐Status Stays

A greater percentage of observation‐status stays in hospitals with a dedicated OU experienced a same‐day discharge (Table 4). In addition, a higher percentage of discharges occurred between midnight and 11 am in hospitals with a dedicated OU. However, overall risk‐adjusted LOS in hours (12.8 vs 12.2 hours, P=0.90) and risk‐adjusted total standardized costs ($2551 vs $2433, P=0.75) were similar between hospital types. These findings were consistent within the 1 APR‐DRGs commonly cared for by pediatric hospitalists (see Supporting Information, Appendix 1, in the online version of this article). Overall, conversion from observation to inpatient status was significantly higher in hospitals with a dedicated OU compared with hospitals without; however, this pattern was not consistent across the 10 APR‐DRGs commonly cared for by pediatric hospitalists (see Supporting Information, Appendix 1, in the online version of this article). Adjusted odds of 3‐day ED return visits and 30‐day readmissions were comparable between hospital groups.

Risk‐Adjusted* Outcomes for Observation‐Status Stays in Hospitals With and Without a Dedicated Observation Unit
 Observation‐Status Patients in Hospitals With a Dedicated Observation UnitObservation‐Status Patients in Hospitals Without a Dedicated Observation UnitP Value
  • NOTE: Abbreviations: AOR, adjusted odds ratio; APR‐DRG, All Patient Refined Diagnosis Related Group; ED, emergency department; IQR, interquartile range. *Risk‐adjusted using generalized linear mixed models treating hospital as a random effect and used patient age, the case‐mix index based on the APR‐DRG severity of illness, ED visit, and procedures associated with the index observation‐status stay. Hospitals reporting the presence of at least 1 dedicated observation unit that admitted unscheduled patients from the ED in 2011. Three hospitals excluded from the analysis for poor data quality for admission/discharge hour; hospitals report admission and discharge in terms of whole hours.

No. of hospitals1417 
Length of stay, h, median (IQR)12.8 (6.923.7)12.2 (721.3)0.90
0 midnights, no. (%)16,678 (23.8)14,648 (22.1)<.001
1 midnight, no. (%)46,144 (65.9)44,559 (67.3) 
2 midnights or more, no. (%)7,161 (10.2)7,049 (10.6) 
Discharge timing, no. (%)   
Midnight5 am1,223 (1.9)408 (0.7)<0.001
6 am11 am18,916 (29.3)15,914 (27.1) 
Noon5 pm32,699 (50.7)31,619 (53.9) 
6 pm11 pm11,718 (18.2)10,718 (18.3) 
Total standardized costs, $, median (IQR)2,551.3 (2,053.93,169.1)2,433.4 (1,998.42,963)0.75
Conversion to inpatient status11.06%9.63%<0.01
Return care, AOR (95% CI)   
3‐day ED return visit0.93 (0.77‐1.12)Referent0.46
30‐day readmission0.88 (0.67‐1.15)Referent0.36

We found similar results in sensitivity analyses comparing observation‐status stays in hospitals with a continuously open OU (open 24 hours per day, 7 days per week, for all of 2011 [n=10 hospitals]) to those without(see Supporting Information, Appendix 2, in the online version of this article). However, there were, on average, more observation‐status stays in hospitals with a continuously open OU (median 5605, IQR 42077089) than hospitals without (median 3309, IQR 26784616) (P=0.04). In contrast to our main results, conversion to inpatient status was lower in hospitals with a continuously open OU compared with hospitals without (8.52% vs 11.57%, P<0.01).

DISCUSSION

Counter to our hypothesis, we did not find hospital‐level differences in length of stay or costs for observation‐status patients cared for in hospitals with and without a dedicated OU, though hospitals with dedicated OUs did have more same‐day discharges and more morning discharges. The lack of observed differences in LOS and costs may reflect the fact that many children under observation status are treated throughout the hospital, even in facilities with a dedicated OU. Access to a dedicated OU is limited by factors including small numbers of OU beds and specific low acuity/low complexity OU admission criteria.[7] The inclusion of all children admitted under observation status in our analyses may have diluted any effect of dedicated OUs at the hospital level, but was necessary due to the inability to identify location of care for children admitted under observation status. Location of care is an important variable that should be incorporated into administrative databases to allow for comparative effectiveness research designs. Until such data are available, chart review at individual hospitals would be necessary to determine which patients received care in an OU.

We did find that discharges for observation‐status patients occurred earlier in the day in hospitals with a dedicated OU when compared with observation‐status patients in hospitals without a dedicated OU. In addition, the percentage of same‐day discharges was higher among observation‐status patients treated in hospitals with a dedicated OU. These differences may stem from policies and procedures that encourage rapid discharge in dedicated OUs, and those practices may affect other care areas. For example, OUs may enforce policies requiring family presence at the bedside or utilize staffing models where doctors and nurses are in frequent communication, both of which would facilitate discharge as soon as a patient no longer required hospital‐based care.[7] A retrospective chart review study design could be used to identify discharge processes and other key characteristics of highly performing OUs.

We found conflicting results in our main and sensitivity analyses related to conversion to inpatient status. Lower percentages of observation‐status patients converting to inpatient status indicates greater success in the delivery of observation care based on established performance metrics.[19] Lower rates of conversion to inpatient status may be the result of stricter admission criteria for some diagnosis and in hospitals with a continuously open dedicate OU, more refined processes for utilization review that allow for patients to be placed into the correct status (observation vs inpatient) at the time of admission, or efforts to educate providers about the designation of observation status.[7] It is also possible that fewer observation‐status patients convert to inpatient status in hospitals with a continuously open dedicated OU because such a change would require movement of the patient to an inpatient bed.

These analyses were more comprehensive than our prior studies[2, 20] in that we included both patients who were treated first in the ED and those who were not. In addition to the APR‐DRGs representative of conditions that have been successfully treated in ED‐based pediatric OUs (eg, asthma, seizures, gastroenteritis, cellulitis),[8, 9, 21, 22] we found observation‐status was commonly associated with procedural care. This population of patients may be relevant to hospitalists who staff OUs that provide both unscheduled and postprocedural care. The colocation of medical and postprocedural patients has been described by others[8, 23] and was reported to occur in over half of the OUs included in this study.[7] The extent to which postprocedure observation care is provided in general OUs staffed by hospitalists represents another opportunity for further study.

Hospitals face many considerations when determining if and how they will provide observation services to patients expected to experience short stays.[7] Some hospitals may be unable to justify an OU for all or part of the year based on the volume of admissions or the costs to staff an OU.[24, 25] Other hospitals may open an OU to promote patient flow and reduce ED crowding.[26] Hospitals may also be influenced by reimbursement policies related to observation‐status stays. Although we did not observe differences in overall payer mix, we did find higher percentages of observation‐status patients in hospitals with dedicated OUs to have public insurance. Although hospital contracts with payers around observation status patients are complex and beyond the scope of this analysis, it is possible that hospitals have established OUs because of increasingly stringent rules or criteria to meet inpatient status or experiences with high volumes of observation‐status patients covered by a particular payer. Nevertheless, the brief nature of many pediatric hospitalizations and the scarcity of pediatric OU beds must be considered in policy changes that result from national discussions about the appropriateness of inpatient stays shorter than 2 nights in duration.[27]

Limitations

The primary limitation to our analyses is the lack of ability to identify patients who were treated in a dedicated OU because few hospitals provided data to PHIS that allowed for the identification of the unit or location of care. Second, it is possible that some hospitals were misclassified as not having a dedicated OU based on our survey, which initially inquired about OUs that provided care to patients first treated in the ED. Therefore, OUs that exclusively care for postoperative patients or patients with scheduled treatments may be present in hospitals that we have labeled as not having a dedicated OU. This potential misclassification would bias our results toward finding no differences. Third, in any study of administrative data there is potential that diagnosis codes are incomplete or inaccurately capture the underlying reason for the episode of care. Fourth, the experiences of the free‐standing children's hospitals that contribute data to PHIS may not be generalizable to other hospitals that provide observation care to children. Finally, return care may be underestimated, as children could receive treatment at another hospital following discharge from a PHIS hospital. Care outside of PHIS hospitals would not be captured, but we do not expect this to differ for hospitals with and without dedicated OUs. It is possible that health information exchanges will permit more comprehensive analyses of care across different hospitals in the future.

CONCLUSION

Observation status patients are similar in hospitals with and without dedicated observation units that admit children from the ED. The presence of a dedicated OU appears to have an influence on same‐day and morning discharges across all observation‐status stays without impacting other hospital‐level outcomes. Inclusion of location of care (eg, geographically distinct dedicated OU vs general inpatient unit vs ED) in hospital administrative datasets would allow for meaningful comparisons of different models of care for short‐stay observation‐status patients.

Acknowledgements

The authors thank John P. Harding, MBA, FACHE, Children's Hospital of the King's Daughters, Norfolk, Virginia for his input on the study design.

Disclosures: Dr. Hall had full access to the data and takes responsibility for the integrity of the data and the accuracy of the data analysis. Internal funds from the Children's Hospital Association supported the conduct of this work. The authors have no financial relationships or conflicts of interest to disclose.

Files
Article PDF
Issue
Journal of Hospital Medicine - 10(6)
Publications
Page Number
366-372
Sections
Files
Files
Article PDF
Article PDF

Many pediatric hospitalizations are of short duration, and more than half of short‐stay hospitalizations are designated as observation status.[1, 2] Observation status is an administrative label assigned to patients who do not meet hospital or payer criteria for inpatient‐status care. Short‐stay observation‐status patients do not fit in traditional models of emergency department (ED) or inpatient care. EDs often focus on discharging or admitting patients within a matter of hours, whereas inpatient units tend to measure length of stay (LOS) in terms of days[3] and may not have systems in place to facilitate rapid discharge of short‐stay patients.[4] Observation units (OUs) have been established in some hospitals to address the unique care needs of short‐stay patients.[5, 6, 7]

Single‐site reports from children's hospitals with successful OUs have demonstrated shorter LOS and lower costs compared with inpatient settings.[6, 8, 9, 10, 11, 12, 13, 14] No prior study has examined hospital‐level effects of an OU on observation‐status patient outcomes. The Pediatric Health Information System (PHIS) database provides a unique opportunity to explore this question, because unlike other national hospital administrative databases,[15, 16] the PHIS dataset contains information about children under observation status. In addition, we know which PHIS hospitals had a dedicated OU in 2011.7

We hypothesized that overall observation‐status stays in hospitals with a dedicated OU would be of shorter duration with earlier discharges at lower cost than observation‐status stays in hospitals without a dedicated OU. We compared hospitals with and without a dedicated OU on secondary outcomes including rates of conversion to inpatient status and return care for any reason.

METHODS

We conducted a cross‐sectional analysis of hospital administrative data using the 2011 PHIS databasea national administrative database that contains resource utilization data from 43 participating hospitals located in 26 states plus the District of Columbia. These hospitals account for approximately 20% of pediatric hospitalizations in the United States.

For each hospital encounter, PHIS includes patient demographics, up to 41 International Classification of Diseases, Ninth Revision, Clinical Modification (ICD‐9‐CM) diagnoses, up to 41 ICD‐9‐CM procedures, and hospital charges for services. Data are deidentified prior to inclusion, but unique identifiers allow for determination of return visits and readmissions following an index visit for an individual patient. Data quality and reliability are assured jointly by the Children's Hospital Association (formerly Child Health Corporation of America, Overland Park, KS), participating hospitals, and Truven Health Analytics (New York, NY). This study, using administrative data, was not considered human subjects research by the policies of the Cincinnati Children's Hospital Medical Center Institutional Review Board.

Hospital Selection and Hospital Characteristics

The study sample was drawn from the 31 hospitals that reported observation‐status patient data to PHIS in 2011. Analyses were conducted in 2013, at which time 2011 was the most recent year of data. We categorized 14 hospitals as having a dedicated OU during 2011 based on information collected in 2013.7 To summarize briefly, we interviewed by telephone representatives of hospitals responding to an email query as to the presence of a geographically distinct OU for the care of unscheduled patients from the ED. Three of the 14 representatives reported their hospital had 2 OUs, 1 of which was a separate surgical OU. Ten OUs cared for both ED patients and patients with scheduled procedures; 8 units received patients from non‐ED sources. Hospitalists provided staffing in more than half of the OUs.

We attempted to identify administrative data that would signal care delivered in a dedicated OU using hospital charge codes reported to PHIS, but learned this was not possible due to between‐hospital variation in the specificity of the charge codes. Therefore, we were unable to determine if patient care was delivered in a dedicated OU or another setting, such as a general inpatient unit or the ED. Other hospital characteristics available from the PHIS dataset included the number of inpatient beds, ED visits, inpatient admissions, observation‐status stays, and payer mix. We calculated the percentage of ED visits resulting in admission by dividing the number of ED visits with associated inpatient or observation status by the total number of ED visits and the percentage of admissions under observation status by dividing the number of observation‐status stays by the total number of admissions under observation or inpatient status.

Visit Selection and Patient Characteristics

All observation‐status stays regardless of the point of entry into the hospital were eligible for this study. We excluded stays that were birth‐related, included intensive care, or resulted in transfer or death. Patient demographic characteristics used to describe the cohort included age, gender, race/ethnicity, and primary payer. Stays that began in the ED were identified by an emergency room charge within PHIS. Eligible stays were categorized using All Patient Refined Diagnosis Related Groups (APR‐DRGs) version 24 using the ICD‐9‐CM code‐based proprietary 3M software (3M Health Information Systems, St. Paul, MN). We determined the 15 top‐ranking APR‐DRGs among observation‐status stays in hospitals with a dedicated OU and hospitals without. Procedural stays were identified based on procedural APR‐DRGs (eg, tonsil and adenoid procedures) or the presence of an ICD‐9‐CM procedure code (eg, 331 spinal tap).

Measured Outcomes

Outcomes of observation‐status stays were determined within 4 categories: (1) LOS, (2) standardized costs, (3) conversion to inpatient status, and (4) return visits and readmissions. LOS was calculated in terms of nights spent in hospital for all stays by subtracting the discharge date from the admission date and in terms of hours for stays in the 28 hospitals that report admission and discharge hour to the PHIS database. Discharge timing was examined in 4, 6‐hour blocks starting at midnight. Standardized costs were derived from a charge master index that was created by taking the median costs from all PHIS hospitals for each charged service.[17] Standardized costs represent the estimated cost of providing any particular clinical activity but are not the cost to patients, nor do they represent the actual cost to any given hospital. This approach allows for cost comparisons across hospitals, without biases arising from using charges or from deriving costs using hospitals' ratios of costs to charges.[18] Conversion from observation to inpatient status was calculated by dividing the number of inpatient‐status stays with observation codes by the number of observation‐statusonly stays plus the number of inpatient‐status stays with observation codes. All‐cause 3‐day ED return visits and 30‐day readmissions to the same hospital were assessed using patient‐specific identifiers that allowed for tracking of ED return visits and readmissions following the index observation stay.

Data Analysis

Descriptive statistics were calculated for hospital and patient characteristics using medians and interquartile ranges (IQRs) for continuous factors and frequencies with percentages for categorical factors. Comparisons of these factors between hospitals with dedicated OUs and without were made using [2] and Wilcoxon rank sum tests as appropriate. Multivariable regression was performed using generalized linear mixed models treating hospital as a random effect and used patient age, the case‐mix index based on the APR‐DRG severity of illness, ED visit, and procedures associated with the index observation‐status stay. For continuous outcomes, we performed a log transformation on the outcome, confirmed the normality assumption, and back transformed the results. Sensitivity analyses were conducted to compare LOS, standardized costs, and conversation rates by hospital type for 10 of the 15 top‐ranking APR‐DRGs commonly cared for by pediatric hospitalists and to compare hospitals that reported the presence of an OU that was consistently open (24 hours per day, 7 days per week) and operating during the entire 2011 calendar year, and those without. Based on information gathered from the telephone interviews, hospitals with partially open OUs were similar to hospitals with continuously open OUs, such that they were included in our main analyses. All statistical analyses were performed using SAS version 9.3 (SAS Institute, Cary, NC). P values <0.05 were considered statistically significant.

RESULTS

Hospital Characteristics

Dedicated OUs were present in 14 of the 31 hospitals that reported observation‐status patient data to PHIS (Figure 1). Three of these hospitals had OUs that were open for 5 months or less in 2011; 1 unit opened, 1 unit closed, and 1 hospital operated a seasonal unit. The remaining 17 hospitals reported no OU that admitted unscheduled patients from the ED during 2011. Hospitals with a dedicated OU had more inpatient beds and higher median number of inpatient admissions than those without (Table 1). Hospitals were statistically similar in terms of total volume of ED visits, percentage of ED visits resulting in admission, total number of observation‐status stays, percentage of admissions under observation status, and payer mix.

jhm2339-fig-0001-m.png
Study Hospital Cohort Selection
Hospitals* With and Without Dedicated Observation Units
 Overall, Median (IQR)Hospitals With a Dedicated Observation Unit, Median (IQR)Hospitals Without a Dedicated Observation Unit, Median (IQR)P Value
  • NOTE: Abbreviations: ED, emergency department; IQR, interquartile range. *Among hospitals that reported observation‐status patient data to the Pediatric Health Information System database in 2011. Hospitals reporting the presence of at least 1 dedicated observation unit that admitted unscheduled patients from the ED in 2011. Percent of ED visits resulting in admission=number of ED visits admitted to inpatient or observation status divided by total number of ED visits in 2011. Percent of admissions under observation status=number of observation‐status stays divided by the total number of admissions (observation and inpatient status) in 2011.

No. of hospitals311417 
Total no. of inpatient beds273 (213311)304 (269425)246 (175293)0.006
Total no. ED visits62971 (47,50497,723)87,892 (55,102117,119)53,151 (4750470,882)0.21
ED visits resulting in admission, %13.1 (9.715.0)13.8 (10.5, 19.1)12.5 (9.714.5)0.31
Total no. of inpatient admissions11,537 (9,26814,568)13,206 (11,32517,869)10,207 (8,64013,363)0.04
Admissions under observation status, %25.7 (19.733.8)25.5 (21.431.4)26.0 (16.935.1)0.98
Total no. of observation stays3,820 (27935672)4,850 (3,309 6,196)3,141 (2,3654,616)0.07
Government payer, %60.2 (53.371.2)62.1 (54.9, 65.9)59.2 (53.373.7)0.89

Observation‐Status Patients by Hospital Type

In 2011, there were a total of 136,239 observation‐status stays69,983 (51.4%) within the 14 hospitals with a dedicated OU and 66,256 (48.6%) within the 17 hospitals without. Patient care originated in the ED for 57.8% observation‐status stays in hospitals with an OU compared with 53.0% of observation‐status stays in hospitals without (P<0.001). Compared with hospitals with a dedicated OU, those without a dedicated OU had higher percentages of observation‐status patients older than 12 years and non‐Hispanic and a higher percentage of observation‐status patients with private payer type (Table 2). The 15 top‐ranking APR‐DRGs accounted for roughly half of all observation‐status stays and were relatively consistent between hospitals with and without a dedicated OU (Table 3). Procedural care was frequently associated with observation‐status stays.

Observation‐Status Patients by Hospital Type
 Overall, No. (%)Hospitals With a Dedicated Observation Unit, No. (%)*Hospitals Without a Dedicated Observation Unit, No. (%)P Value
  • NOTE: *Hospitals reporting the presence of at least 1 dedicated observation unit that admitted unscheduled patients from the emergency department in 2011.

Age    
<1 year23,845 (17.5)12,101 (17.3)11,744 (17.7)<0.001
15 years53,405 (38.5)28,052 (40.1)24,353 (36.8) 
612 years33,674 (24.7)17,215 (24.6)16,459 (24.8) 
1318 years23,607 (17.3)11,472 (16.4)12,135 (18.3) 
>18 years2,708 (2)1,143 (1.6)1,565 (2.4) 
Gender    
Male76,142 (55.9)39,178 (56)36,964 (55.8)0.43
Female60,025 (44.1)30,756 (44)29,269 (44.2) 
Race/ethnicity    
Non‐Hispanic white72,183 (53.0)30,653 (43.8)41,530 (62.7)<0.001
Non‐Hispanic black30,995 (22.8)16,314 (23.3)14,681 (22.2) 
Hispanic21,255 (15.6)16,583 (23.7)4,672 (7.1) 
Asian2,075 (1.5)1,313 (1.9)762 (1.2) 
Non‐Hispanic other9,731 (7.1)5,120 (7.3)4,611 (7.0) 
Payer    
Government68,725 (50.4)36,967 (52.8)31,758 (47.9)<0.001
Private48,416 (35.5)21,112 (30.2)27,304 (41.2) 
Other19,098 (14.0)11,904 (17)7,194 (10.9) 
Fifteen Most Common APR‐DRGs for Observation‐Status Patients by Hospital Type
Observation‐Status Patients in Hospitals With a Dedicated Observation Unit*Observation‐Status Patients in Hospitals Without a Dedicated Observation Unit
RankAPR‐DRGNo.% of All Observation Status Stays% Began in EDRankAPR‐DRGNo.% of All Observation Status Stays% Began in ED
  • NOTE: Abbreviations: APR‐DRG, All Patient Refined Diagnosis Related Group; ED, emergency department; ENT, ear, nose, and throat; NEC, not elsewhere classified; RSV, respiratory syncytial virus. *Hospitals reporting the presence of at least 1 dedicated observation unit that admitted unscheduled patients from the ED in 2011. Within the APR‐DRG. Procedure codes associated with 99% to 100% of observation stays within the APR‐DRG. Procedure codes associated with 20% 45% of observation stays within APR‐DRG; procedure codes were associated with <20% of observation stays within the APR‐DRG that are not indicated otherwise.

1Tonsil and adenoid procedures4,6216.61.31Tonsil and adenoid procedures3,8065.71.6
2Asthma4,2466.185.32Asthma3,7565.779.0
3Seizure3,5165.052.03Seizure2,8464.354.9
4Nonbacterial gastroenteritis3,2864.785.84Upper respiratory infections2,7334.169.6
5Bronchiolitis, RSV pneumonia3,0934.478.55Nonbacterial gastroenteritis2,6824.074.5
6Upper respiratory infections2,9234.280.06Other digestive system diagnoses2,5453.866.3
7Other digestive system diagnoses2,0642.974.07Bronchiolitis, RSV pneumonia2,5443.869.2
8Respiratory signs, symptoms, diagnoses2,0522.981.68Shoulder and arm procedures1,8622.872.6
9Other ENT/cranial/facial diagnoses1,6842.443.69Appendectomy1,7852.779.2
10Shoulder and arm procedures1,6242.379.110Other ENT/cranial/facial diagnoses1,6242.529.9
11Abdominal pain1,6122.386.211Abdominal pain1,4612.282.3
12Fever1,4942.185.112Other factors influencing health status1,4612.266.3
13Appendectomy1,4652.166.413Cellulitis/other bacterial skin infections1,3832.184.2
14Cellulitis/other bacterial skin infections1,3932.086.414Respiratory signs, symptoms, diagnoses1,3082.039.1
15Pneumonia NEC1,3561.979.115Pneumonia NEC1,2451.973.1
 Total36,42952.057.8 Total33,04149.8753.0

Outcomes of Observation‐Status Stays

A greater percentage of observation‐status stays in hospitals with a dedicated OU experienced a same‐day discharge (Table 4). In addition, a higher percentage of discharges occurred between midnight and 11 am in hospitals with a dedicated OU. However, overall risk‐adjusted LOS in hours (12.8 vs 12.2 hours, P=0.90) and risk‐adjusted total standardized costs ($2551 vs $2433, P=0.75) were similar between hospital types. These findings were consistent within the 1 APR‐DRGs commonly cared for by pediatric hospitalists (see Supporting Information, Appendix 1, in the online version of this article). Overall, conversion from observation to inpatient status was significantly higher in hospitals with a dedicated OU compared with hospitals without; however, this pattern was not consistent across the 10 APR‐DRGs commonly cared for by pediatric hospitalists (see Supporting Information, Appendix 1, in the online version of this article). Adjusted odds of 3‐day ED return visits and 30‐day readmissions were comparable between hospital groups.

Risk‐Adjusted* Outcomes for Observation‐Status Stays in Hospitals With and Without a Dedicated Observation Unit
 Observation‐Status Patients in Hospitals With a Dedicated Observation UnitObservation‐Status Patients in Hospitals Without a Dedicated Observation UnitP Value
  • NOTE: Abbreviations: AOR, adjusted odds ratio; APR‐DRG, All Patient Refined Diagnosis Related Group; ED, emergency department; IQR, interquartile range. *Risk‐adjusted using generalized linear mixed models treating hospital as a random effect and used patient age, the case‐mix index based on the APR‐DRG severity of illness, ED visit, and procedures associated with the index observation‐status stay. Hospitals reporting the presence of at least 1 dedicated observation unit that admitted unscheduled patients from the ED in 2011. Three hospitals excluded from the analysis for poor data quality for admission/discharge hour; hospitals report admission and discharge in terms of whole hours.

No. of hospitals1417 
Length of stay, h, median (IQR)12.8 (6.923.7)12.2 (721.3)0.90
0 midnights, no. (%)16,678 (23.8)14,648 (22.1)<.001
1 midnight, no. (%)46,144 (65.9)44,559 (67.3) 
2 midnights or more, no. (%)7,161 (10.2)7,049 (10.6) 
Discharge timing, no. (%)   
Midnight5 am1,223 (1.9)408 (0.7)<0.001
6 am11 am18,916 (29.3)15,914 (27.1) 
Noon5 pm32,699 (50.7)31,619 (53.9) 
6 pm11 pm11,718 (18.2)10,718 (18.3) 
Total standardized costs, $, median (IQR)2,551.3 (2,053.93,169.1)2,433.4 (1,998.42,963)0.75
Conversion to inpatient status11.06%9.63%<0.01
Return care, AOR (95% CI)   
3‐day ED return visit0.93 (0.77‐1.12)Referent0.46
30‐day readmission0.88 (0.67‐1.15)Referent0.36

We found similar results in sensitivity analyses comparing observation‐status stays in hospitals with a continuously open OU (open 24 hours per day, 7 days per week, for all of 2011 [n=10 hospitals]) to those without(see Supporting Information, Appendix 2, in the online version of this article). However, there were, on average, more observation‐status stays in hospitals with a continuously open OU (median 5605, IQR 42077089) than hospitals without (median 3309, IQR 26784616) (P=0.04). In contrast to our main results, conversion to inpatient status was lower in hospitals with a continuously open OU compared with hospitals without (8.52% vs 11.57%, P<0.01).

DISCUSSION

Counter to our hypothesis, we did not find hospital‐level differences in length of stay or costs for observation‐status patients cared for in hospitals with and without a dedicated OU, though hospitals with dedicated OUs did have more same‐day discharges and more morning discharges. The lack of observed differences in LOS and costs may reflect the fact that many children under observation status are treated throughout the hospital, even in facilities with a dedicated OU. Access to a dedicated OU is limited by factors including small numbers of OU beds and specific low acuity/low complexity OU admission criteria.[7] The inclusion of all children admitted under observation status in our analyses may have diluted any effect of dedicated OUs at the hospital level, but was necessary due to the inability to identify location of care for children admitted under observation status. Location of care is an important variable that should be incorporated into administrative databases to allow for comparative effectiveness research designs. Until such data are available, chart review at individual hospitals would be necessary to determine which patients received care in an OU.

We did find that discharges for observation‐status patients occurred earlier in the day in hospitals with a dedicated OU when compared with observation‐status patients in hospitals without a dedicated OU. In addition, the percentage of same‐day discharges was higher among observation‐status patients treated in hospitals with a dedicated OU. These differences may stem from policies and procedures that encourage rapid discharge in dedicated OUs, and those practices may affect other care areas. For example, OUs may enforce policies requiring family presence at the bedside or utilize staffing models where doctors and nurses are in frequent communication, both of which would facilitate discharge as soon as a patient no longer required hospital‐based care.[7] A retrospective chart review study design could be used to identify discharge processes and other key characteristics of highly performing OUs.

We found conflicting results in our main and sensitivity analyses related to conversion to inpatient status. Lower percentages of observation‐status patients converting to inpatient status indicates greater success in the delivery of observation care based on established performance metrics.[19] Lower rates of conversion to inpatient status may be the result of stricter admission criteria for some diagnosis and in hospitals with a continuously open dedicate OU, more refined processes for utilization review that allow for patients to be placed into the correct status (observation vs inpatient) at the time of admission, or efforts to educate providers about the designation of observation status.[7] It is also possible that fewer observation‐status patients convert to inpatient status in hospitals with a continuously open dedicated OU because such a change would require movement of the patient to an inpatient bed.

These analyses were more comprehensive than our prior studies[2, 20] in that we included both patients who were treated first in the ED and those who were not. In addition to the APR‐DRGs representative of conditions that have been successfully treated in ED‐based pediatric OUs (eg, asthma, seizures, gastroenteritis, cellulitis),[8, 9, 21, 22] we found observation‐status was commonly associated with procedural care. This population of patients may be relevant to hospitalists who staff OUs that provide both unscheduled and postprocedural care. The colocation of medical and postprocedural patients has been described by others[8, 23] and was reported to occur in over half of the OUs included in this study.[7] The extent to which postprocedure observation care is provided in general OUs staffed by hospitalists represents another opportunity for further study.

Hospitals face many considerations when determining if and how they will provide observation services to patients expected to experience short stays.[7] Some hospitals may be unable to justify an OU for all or part of the year based on the volume of admissions or the costs to staff an OU.[24, 25] Other hospitals may open an OU to promote patient flow and reduce ED crowding.[26] Hospitals may also be influenced by reimbursement policies related to observation‐status stays. Although we did not observe differences in overall payer mix, we did find higher percentages of observation‐status patients in hospitals with dedicated OUs to have public insurance. Although hospital contracts with payers around observation status patients are complex and beyond the scope of this analysis, it is possible that hospitals have established OUs because of increasingly stringent rules or criteria to meet inpatient status or experiences with high volumes of observation‐status patients covered by a particular payer. Nevertheless, the brief nature of many pediatric hospitalizations and the scarcity of pediatric OU beds must be considered in policy changes that result from national discussions about the appropriateness of inpatient stays shorter than 2 nights in duration.[27]

Limitations

The primary limitation to our analyses is the lack of ability to identify patients who were treated in a dedicated OU because few hospitals provided data to PHIS that allowed for the identification of the unit or location of care. Second, it is possible that some hospitals were misclassified as not having a dedicated OU based on our survey, which initially inquired about OUs that provided care to patients first treated in the ED. Therefore, OUs that exclusively care for postoperative patients or patients with scheduled treatments may be present in hospitals that we have labeled as not having a dedicated OU. This potential misclassification would bias our results toward finding no differences. Third, in any study of administrative data there is potential that diagnosis codes are incomplete or inaccurately capture the underlying reason for the episode of care. Fourth, the experiences of the free‐standing children's hospitals that contribute data to PHIS may not be generalizable to other hospitals that provide observation care to children. Finally, return care may be underestimated, as children could receive treatment at another hospital following discharge from a PHIS hospital. Care outside of PHIS hospitals would not be captured, but we do not expect this to differ for hospitals with and without dedicated OUs. It is possible that health information exchanges will permit more comprehensive analyses of care across different hospitals in the future.

CONCLUSION

Observation status patients are similar in hospitals with and without dedicated observation units that admit children from the ED. The presence of a dedicated OU appears to have an influence on same‐day and morning discharges across all observation‐status stays without impacting other hospital‐level outcomes. Inclusion of location of care (eg, geographically distinct dedicated OU vs general inpatient unit vs ED) in hospital administrative datasets would allow for meaningful comparisons of different models of care for short‐stay observation‐status patients.

Acknowledgements

The authors thank John P. Harding, MBA, FACHE, Children's Hospital of the King's Daughters, Norfolk, Virginia for his input on the study design.

Disclosures: Dr. Hall had full access to the data and takes responsibility for the integrity of the data and the accuracy of the data analysis. Internal funds from the Children's Hospital Association supported the conduct of this work. The authors have no financial relationships or conflicts of interest to disclose.

Many pediatric hospitalizations are of short duration, and more than half of short‐stay hospitalizations are designated as observation status.[1, 2] Observation status is an administrative label assigned to patients who do not meet hospital or payer criteria for inpatient‐status care. Short‐stay observation‐status patients do not fit in traditional models of emergency department (ED) or inpatient care. EDs often focus on discharging or admitting patients within a matter of hours, whereas inpatient units tend to measure length of stay (LOS) in terms of days[3] and may not have systems in place to facilitate rapid discharge of short‐stay patients.[4] Observation units (OUs) have been established in some hospitals to address the unique care needs of short‐stay patients.[5, 6, 7]

Single‐site reports from children's hospitals with successful OUs have demonstrated shorter LOS and lower costs compared with inpatient settings.[6, 8, 9, 10, 11, 12, 13, 14] No prior study has examined hospital‐level effects of an OU on observation‐status patient outcomes. The Pediatric Health Information System (PHIS) database provides a unique opportunity to explore this question, because unlike other national hospital administrative databases,[15, 16] the PHIS dataset contains information about children under observation status. In addition, we know which PHIS hospitals had a dedicated OU in 2011.7

We hypothesized that overall observation‐status stays in hospitals with a dedicated OU would be of shorter duration with earlier discharges at lower cost than observation‐status stays in hospitals without a dedicated OU. We compared hospitals with and without a dedicated OU on secondary outcomes including rates of conversion to inpatient status and return care for any reason.

METHODS

We conducted a cross‐sectional analysis of hospital administrative data using the 2011 PHIS databasea national administrative database that contains resource utilization data from 43 participating hospitals located in 26 states plus the District of Columbia. These hospitals account for approximately 20% of pediatric hospitalizations in the United States.

For each hospital encounter, PHIS includes patient demographics, up to 41 International Classification of Diseases, Ninth Revision, Clinical Modification (ICD‐9‐CM) diagnoses, up to 41 ICD‐9‐CM procedures, and hospital charges for services. Data are deidentified prior to inclusion, but unique identifiers allow for determination of return visits and readmissions following an index visit for an individual patient. Data quality and reliability are assured jointly by the Children's Hospital Association (formerly Child Health Corporation of America, Overland Park, KS), participating hospitals, and Truven Health Analytics (New York, NY). This study, using administrative data, was not considered human subjects research by the policies of the Cincinnati Children's Hospital Medical Center Institutional Review Board.

Hospital Selection and Hospital Characteristics

The study sample was drawn from the 31 hospitals that reported observation‐status patient data to PHIS in 2011. Analyses were conducted in 2013, at which time 2011 was the most recent year of data. We categorized 14 hospitals as having a dedicated OU during 2011 based on information collected in 2013.7 To summarize briefly, we interviewed by telephone representatives of hospitals responding to an email query as to the presence of a geographically distinct OU for the care of unscheduled patients from the ED. Three of the 14 representatives reported their hospital had 2 OUs, 1 of which was a separate surgical OU. Ten OUs cared for both ED patients and patients with scheduled procedures; 8 units received patients from non‐ED sources. Hospitalists provided staffing in more than half of the OUs.

We attempted to identify administrative data that would signal care delivered in a dedicated OU using hospital charge codes reported to PHIS, but learned this was not possible due to between‐hospital variation in the specificity of the charge codes. Therefore, we were unable to determine if patient care was delivered in a dedicated OU or another setting, such as a general inpatient unit or the ED. Other hospital characteristics available from the PHIS dataset included the number of inpatient beds, ED visits, inpatient admissions, observation‐status stays, and payer mix. We calculated the percentage of ED visits resulting in admission by dividing the number of ED visits with associated inpatient or observation status by the total number of ED visits and the percentage of admissions under observation status by dividing the number of observation‐status stays by the total number of admissions under observation or inpatient status.

Visit Selection and Patient Characteristics

All observation‐status stays regardless of the point of entry into the hospital were eligible for this study. We excluded stays that were birth‐related, included intensive care, or resulted in transfer or death. Patient demographic characteristics used to describe the cohort included age, gender, race/ethnicity, and primary payer. Stays that began in the ED were identified by an emergency room charge within PHIS. Eligible stays were categorized using All Patient Refined Diagnosis Related Groups (APR‐DRGs) version 24 using the ICD‐9‐CM code‐based proprietary 3M software (3M Health Information Systems, St. Paul, MN). We determined the 15 top‐ranking APR‐DRGs among observation‐status stays in hospitals with a dedicated OU and hospitals without. Procedural stays were identified based on procedural APR‐DRGs (eg, tonsil and adenoid procedures) or the presence of an ICD‐9‐CM procedure code (eg, 331 spinal tap).

Measured Outcomes

Outcomes of observation‐status stays were determined within 4 categories: (1) LOS, (2) standardized costs, (3) conversion to inpatient status, and (4) return visits and readmissions. LOS was calculated in terms of nights spent in hospital for all stays by subtracting the discharge date from the admission date and in terms of hours for stays in the 28 hospitals that report admission and discharge hour to the PHIS database. Discharge timing was examined in 4, 6‐hour blocks starting at midnight. Standardized costs were derived from a charge master index that was created by taking the median costs from all PHIS hospitals for each charged service.[17] Standardized costs represent the estimated cost of providing any particular clinical activity but are not the cost to patients, nor do they represent the actual cost to any given hospital. This approach allows for cost comparisons across hospitals, without biases arising from using charges or from deriving costs using hospitals' ratios of costs to charges.[18] Conversion from observation to inpatient status was calculated by dividing the number of inpatient‐status stays with observation codes by the number of observation‐statusonly stays plus the number of inpatient‐status stays with observation codes. All‐cause 3‐day ED return visits and 30‐day readmissions to the same hospital were assessed using patient‐specific identifiers that allowed for tracking of ED return visits and readmissions following the index observation stay.

Data Analysis

Descriptive statistics were calculated for hospital and patient characteristics using medians and interquartile ranges (IQRs) for continuous factors and frequencies with percentages for categorical factors. Comparisons of these factors between hospitals with dedicated OUs and without were made using [2] and Wilcoxon rank sum tests as appropriate. Multivariable regression was performed using generalized linear mixed models treating hospital as a random effect and used patient age, the case‐mix index based on the APR‐DRG severity of illness, ED visit, and procedures associated with the index observation‐status stay. For continuous outcomes, we performed a log transformation on the outcome, confirmed the normality assumption, and back transformed the results. Sensitivity analyses were conducted to compare LOS, standardized costs, and conversation rates by hospital type for 10 of the 15 top‐ranking APR‐DRGs commonly cared for by pediatric hospitalists and to compare hospitals that reported the presence of an OU that was consistently open (24 hours per day, 7 days per week) and operating during the entire 2011 calendar year, and those without. Based on information gathered from the telephone interviews, hospitals with partially open OUs were similar to hospitals with continuously open OUs, such that they were included in our main analyses. All statistical analyses were performed using SAS version 9.3 (SAS Institute, Cary, NC). P values <0.05 were considered statistically significant.

RESULTS

Hospital Characteristics

Dedicated OUs were present in 14 of the 31 hospitals that reported observation‐status patient data to PHIS (Figure 1). Three of these hospitals had OUs that were open for 5 months or less in 2011; 1 unit opened, 1 unit closed, and 1 hospital operated a seasonal unit. The remaining 17 hospitals reported no OU that admitted unscheduled patients from the ED during 2011. Hospitals with a dedicated OU had more inpatient beds and higher median number of inpatient admissions than those without (Table 1). Hospitals were statistically similar in terms of total volume of ED visits, percentage of ED visits resulting in admission, total number of observation‐status stays, percentage of admissions under observation status, and payer mix.

jhm2339-fig-0001-m.png
Study Hospital Cohort Selection
Hospitals* With and Without Dedicated Observation Units
 Overall, Median (IQR)Hospitals With a Dedicated Observation Unit, Median (IQR)Hospitals Without a Dedicated Observation Unit, Median (IQR)P Value
  • NOTE: Abbreviations: ED, emergency department; IQR, interquartile range. *Among hospitals that reported observation‐status patient data to the Pediatric Health Information System database in 2011. Hospitals reporting the presence of at least 1 dedicated observation unit that admitted unscheduled patients from the ED in 2011. Percent of ED visits resulting in admission=number of ED visits admitted to inpatient or observation status divided by total number of ED visits in 2011. Percent of admissions under observation status=number of observation‐status stays divided by the total number of admissions (observation and inpatient status) in 2011.

No. of hospitals311417 
Total no. of inpatient beds273 (213311)304 (269425)246 (175293)0.006
Total no. ED visits62971 (47,50497,723)87,892 (55,102117,119)53,151 (4750470,882)0.21
ED visits resulting in admission, %13.1 (9.715.0)13.8 (10.5, 19.1)12.5 (9.714.5)0.31
Total no. of inpatient admissions11,537 (9,26814,568)13,206 (11,32517,869)10,207 (8,64013,363)0.04
Admissions under observation status, %25.7 (19.733.8)25.5 (21.431.4)26.0 (16.935.1)0.98
Total no. of observation stays3,820 (27935672)4,850 (3,309 6,196)3,141 (2,3654,616)0.07
Government payer, %60.2 (53.371.2)62.1 (54.9, 65.9)59.2 (53.373.7)0.89

Observation‐Status Patients by Hospital Type

In 2011, there were a total of 136,239 observation‐status stays69,983 (51.4%) within the 14 hospitals with a dedicated OU and 66,256 (48.6%) within the 17 hospitals without. Patient care originated in the ED for 57.8% observation‐status stays in hospitals with an OU compared with 53.0% of observation‐status stays in hospitals without (P<0.001). Compared with hospitals with a dedicated OU, those without a dedicated OU had higher percentages of observation‐status patients older than 12 years and non‐Hispanic and a higher percentage of observation‐status patients with private payer type (Table 2). The 15 top‐ranking APR‐DRGs accounted for roughly half of all observation‐status stays and were relatively consistent between hospitals with and without a dedicated OU (Table 3). Procedural care was frequently associated with observation‐status stays.

Observation‐Status Patients by Hospital Type
 Overall, No. (%)Hospitals With a Dedicated Observation Unit, No. (%)*Hospitals Without a Dedicated Observation Unit, No. (%)P Value
  • NOTE: *Hospitals reporting the presence of at least 1 dedicated observation unit that admitted unscheduled patients from the emergency department in 2011.

Age    
<1 year23,845 (17.5)12,101 (17.3)11,744 (17.7)<0.001
15 years53,405 (38.5)28,052 (40.1)24,353 (36.8) 
612 years33,674 (24.7)17,215 (24.6)16,459 (24.8) 
1318 years23,607 (17.3)11,472 (16.4)12,135 (18.3) 
>18 years2,708 (2)1,143 (1.6)1,565 (2.4) 
Gender    
Male76,142 (55.9)39,178 (56)36,964 (55.8)0.43
Female60,025 (44.1)30,756 (44)29,269 (44.2) 
Race/ethnicity    
Non‐Hispanic white72,183 (53.0)30,653 (43.8)41,530 (62.7)<0.001
Non‐Hispanic black30,995 (22.8)16,314 (23.3)14,681 (22.2) 
Hispanic21,255 (15.6)16,583 (23.7)4,672 (7.1) 
Asian2,075 (1.5)1,313 (1.9)762 (1.2) 
Non‐Hispanic other9,731 (7.1)5,120 (7.3)4,611 (7.0) 
Payer    
Government68,725 (50.4)36,967 (52.8)31,758 (47.9)<0.001
Private48,416 (35.5)21,112 (30.2)27,304 (41.2) 
Other19,098 (14.0)11,904 (17)7,194 (10.9) 
Fifteen Most Common APR‐DRGs for Observation‐Status Patients by Hospital Type
Observation‐Status Patients in Hospitals With a Dedicated Observation Unit*Observation‐Status Patients in Hospitals Without a Dedicated Observation Unit
RankAPR‐DRGNo.% of All Observation Status Stays% Began in EDRankAPR‐DRGNo.% of All Observation Status Stays% Began in ED
  • NOTE: Abbreviations: APR‐DRG, All Patient Refined Diagnosis Related Group; ED, emergency department; ENT, ear, nose, and throat; NEC, not elsewhere classified; RSV, respiratory syncytial virus. *Hospitals reporting the presence of at least 1 dedicated observation unit that admitted unscheduled patients from the ED in 2011. Within the APR‐DRG. Procedure codes associated with 99% to 100% of observation stays within the APR‐DRG. Procedure codes associated with 20% 45% of observation stays within APR‐DRG; procedure codes were associated with <20% of observation stays within the APR‐DRG that are not indicated otherwise.

1Tonsil and adenoid procedures4,6216.61.31Tonsil and adenoid procedures3,8065.71.6
2Asthma4,2466.185.32Asthma3,7565.779.0
3Seizure3,5165.052.03Seizure2,8464.354.9
4Nonbacterial gastroenteritis3,2864.785.84Upper respiratory infections2,7334.169.6
5Bronchiolitis, RSV pneumonia3,0934.478.55Nonbacterial gastroenteritis2,6824.074.5
6Upper respiratory infections2,9234.280.06Other digestive system diagnoses2,5453.866.3
7Other digestive system diagnoses2,0642.974.07Bronchiolitis, RSV pneumonia2,5443.869.2
8Respiratory signs, symptoms, diagnoses2,0522.981.68Shoulder and arm procedures1,8622.872.6
9Other ENT/cranial/facial diagnoses1,6842.443.69Appendectomy1,7852.779.2
10Shoulder and arm procedures1,6242.379.110Other ENT/cranial/facial diagnoses1,6242.529.9
11Abdominal pain1,6122.386.211Abdominal pain1,4612.282.3
12Fever1,4942.185.112Other factors influencing health status1,4612.266.3
13Appendectomy1,4652.166.413Cellulitis/other bacterial skin infections1,3832.184.2
14Cellulitis/other bacterial skin infections1,3932.086.414Respiratory signs, symptoms, diagnoses1,3082.039.1
15Pneumonia NEC1,3561.979.115Pneumonia NEC1,2451.973.1
 Total36,42952.057.8 Total33,04149.8753.0

Outcomes of Observation‐Status Stays

A greater percentage of observation‐status stays in hospitals with a dedicated OU experienced a same‐day discharge (Table 4). In addition, a higher percentage of discharges occurred between midnight and 11 am in hospitals with a dedicated OU. However, overall risk‐adjusted LOS in hours (12.8 vs 12.2 hours, P=0.90) and risk‐adjusted total standardized costs ($2551 vs $2433, P=0.75) were similar between hospital types. These findings were consistent within the 1 APR‐DRGs commonly cared for by pediatric hospitalists (see Supporting Information, Appendix 1, in the online version of this article). Overall, conversion from observation to inpatient status was significantly higher in hospitals with a dedicated OU compared with hospitals without; however, this pattern was not consistent across the 10 APR‐DRGs commonly cared for by pediatric hospitalists (see Supporting Information, Appendix 1, in the online version of this article). Adjusted odds of 3‐day ED return visits and 30‐day readmissions were comparable between hospital groups.

Risk‐Adjusted* Outcomes for Observation‐Status Stays in Hospitals With and Without a Dedicated Observation Unit
 Observation‐Status Patients in Hospitals With a Dedicated Observation UnitObservation‐Status Patients in Hospitals Without a Dedicated Observation UnitP Value
  • NOTE: Abbreviations: AOR, adjusted odds ratio; APR‐DRG, All Patient Refined Diagnosis Related Group; ED, emergency department; IQR, interquartile range. *Risk‐adjusted using generalized linear mixed models treating hospital as a random effect and used patient age, the case‐mix index based on the APR‐DRG severity of illness, ED visit, and procedures associated with the index observation‐status stay. Hospitals reporting the presence of at least 1 dedicated observation unit that admitted unscheduled patients from the ED in 2011. Three hospitals excluded from the analysis for poor data quality for admission/discharge hour; hospitals report admission and discharge in terms of whole hours.

No. of hospitals1417 
Length of stay, h, median (IQR)12.8 (6.923.7)12.2 (721.3)0.90
0 midnights, no. (%)16,678 (23.8)14,648 (22.1)<.001
1 midnight, no. (%)46,144 (65.9)44,559 (67.3) 
2 midnights or more, no. (%)7,161 (10.2)7,049 (10.6) 
Discharge timing, no. (%)   
Midnight5 am1,223 (1.9)408 (0.7)<0.001
6 am11 am18,916 (29.3)15,914 (27.1) 
Noon5 pm32,699 (50.7)31,619 (53.9) 
6 pm11 pm11,718 (18.2)10,718 (18.3) 
Total standardized costs, $, median (IQR)2,551.3 (2,053.93,169.1)2,433.4 (1,998.42,963)0.75
Conversion to inpatient status11.06%9.63%<0.01
Return care, AOR (95% CI)   
3‐day ED return visit0.93 (0.77‐1.12)Referent0.46
30‐day readmission0.88 (0.67‐1.15)Referent0.36

We found similar results in sensitivity analyses comparing observation‐status stays in hospitals with a continuously open OU (open 24 hours per day, 7 days per week, for all of 2011 [n=10 hospitals]) to those without(see Supporting Information, Appendix 2, in the online version of this article). However, there were, on average, more observation‐status stays in hospitals with a continuously open OU (median 5605, IQR 42077089) than hospitals without (median 3309, IQR 26784616) (P=0.04). In contrast to our main results, conversion to inpatient status was lower in hospitals with a continuously open OU compared with hospitals without (8.52% vs 11.57%, P<0.01).

DISCUSSION

Counter to our hypothesis, we did not find hospital‐level differences in length of stay or costs for observation‐status patients cared for in hospitals with and without a dedicated OU, though hospitals with dedicated OUs did have more same‐day discharges and more morning discharges. The lack of observed differences in LOS and costs may reflect the fact that many children under observation status are treated throughout the hospital, even in facilities with a dedicated OU. Access to a dedicated OU is limited by factors including small numbers of OU beds and specific low acuity/low complexity OU admission criteria.[7] The inclusion of all children admitted under observation status in our analyses may have diluted any effect of dedicated OUs at the hospital level, but was necessary due to the inability to identify location of care for children admitted under observation status. Location of care is an important variable that should be incorporated into administrative databases to allow for comparative effectiveness research designs. Until such data are available, chart review at individual hospitals would be necessary to determine which patients received care in an OU.

We did find that discharges for observation‐status patients occurred earlier in the day in hospitals with a dedicated OU when compared with observation‐status patients in hospitals without a dedicated OU. In addition, the percentage of same‐day discharges was higher among observation‐status patients treated in hospitals with a dedicated OU. These differences may stem from policies and procedures that encourage rapid discharge in dedicated OUs, and those practices may affect other care areas. For example, OUs may enforce policies requiring family presence at the bedside or utilize staffing models where doctors and nurses are in frequent communication, both of which would facilitate discharge as soon as a patient no longer required hospital‐based care.[7] A retrospective chart review study design could be used to identify discharge processes and other key characteristics of highly performing OUs.

We found conflicting results in our main and sensitivity analyses related to conversion to inpatient status. Lower percentages of observation‐status patients converting to inpatient status indicates greater success in the delivery of observation care based on established performance metrics.[19] Lower rates of conversion to inpatient status may be the result of stricter admission criteria for some diagnosis and in hospitals with a continuously open dedicate OU, more refined processes for utilization review that allow for patients to be placed into the correct status (observation vs inpatient) at the time of admission, or efforts to educate providers about the designation of observation status.[7] It is also possible that fewer observation‐status patients convert to inpatient status in hospitals with a continuously open dedicated OU because such a change would require movement of the patient to an inpatient bed.

These analyses were more comprehensive than our prior studies[2, 20] in that we included both patients who were treated first in the ED and those who were not. In addition to the APR‐DRGs representative of conditions that have been successfully treated in ED‐based pediatric OUs (eg, asthma, seizures, gastroenteritis, cellulitis),[8, 9, 21, 22] we found observation‐status was commonly associated with procedural care. This population of patients may be relevant to hospitalists who staff OUs that provide both unscheduled and postprocedural care. The colocation of medical and postprocedural patients has been described by others[8, 23] and was reported to occur in over half of the OUs included in this study.[7] The extent to which postprocedure observation care is provided in general OUs staffed by hospitalists represents another opportunity for further study.

Hospitals face many considerations when determining if and how they will provide observation services to patients expected to experience short stays.[7] Some hospitals may be unable to justify an OU for all or part of the year based on the volume of admissions or the costs to staff an OU.[24, 25] Other hospitals may open an OU to promote patient flow and reduce ED crowding.[26] Hospitals may also be influenced by reimbursement policies related to observation‐status stays. Although we did not observe differences in overall payer mix, we did find higher percentages of observation‐status patients in hospitals with dedicated OUs to have public insurance. Although hospital contracts with payers around observation status patients are complex and beyond the scope of this analysis, it is possible that hospitals have established OUs because of increasingly stringent rules or criteria to meet inpatient status or experiences with high volumes of observation‐status patients covered by a particular payer. Nevertheless, the brief nature of many pediatric hospitalizations and the scarcity of pediatric OU beds must be considered in policy changes that result from national discussions about the appropriateness of inpatient stays shorter than 2 nights in duration.[27]

Limitations

The primary limitation to our analyses is the lack of ability to identify patients who were treated in a dedicated OU because few hospitals provided data to PHIS that allowed for the identification of the unit or location of care. Second, it is possible that some hospitals were misclassified as not having a dedicated OU based on our survey, which initially inquired about OUs that provided care to patients first treated in the ED. Therefore, OUs that exclusively care for postoperative patients or patients with scheduled treatments may be present in hospitals that we have labeled as not having a dedicated OU. This potential misclassification would bias our results toward finding no differences. Third, in any study of administrative data there is potential that diagnosis codes are incomplete or inaccurately capture the underlying reason for the episode of care. Fourth, the experiences of the free‐standing children's hospitals that contribute data to PHIS may not be generalizable to other hospitals that provide observation care to children. Finally, return care may be underestimated, as children could receive treatment at another hospital following discharge from a PHIS hospital. Care outside of PHIS hospitals would not be captured, but we do not expect this to differ for hospitals with and without dedicated OUs. It is possible that health information exchanges will permit more comprehensive analyses of care across different hospitals in the future.

CONCLUSION

Observation status patients are similar in hospitals with and without dedicated observation units that admit children from the ED. The presence of a dedicated OU appears to have an influence on same‐day and morning discharges across all observation‐status stays without impacting other hospital‐level outcomes. Inclusion of location of care (eg, geographically distinct dedicated OU vs general inpatient unit vs ED) in hospital administrative datasets would allow for meaningful comparisons of different models of care for short‐stay observation‐status patients.

Acknowledgements

The authors thank John P. Harding, MBA, FACHE, Children's Hospital of the King's Daughters, Norfolk, Virginia for his input on the study design.

Disclosures: Dr. Hall had full access to the data and takes responsibility for the integrity of the data and the accuracy of the data analysis. Internal funds from the Children's Hospital Association supported the conduct of this work. The authors have no financial relationships or conflicts of interest to disclose.

Issue
Journal of Hospital Medicine - 10(6)
Issue
Journal of Hospital Medicine - 10(6)
Page Number
366-372
Page Number
366-372
Publications
Publications
Article Type
Display Headline
Observation‐status patients in children's hospitals with and without dedicated observation units in 2011
Display Headline
Observation‐status patients in children's hospitals with and without dedicated observation units in 2011
Sections
Article Source

© 2015 Society of Hospital Medicine

Disallow All Ads
Correspondence Location
Address for correspondence and reprint requests: Michelle L. Macy, MD, Division of General Pediatrics, University of Michigan, 300 North Ingalls 6C13, Ann Arbor, MI 48109‐5456; Telephone: 734‐936‐8338; Fax: 734‐764‐2599; E‐mail: mlmacy@umich.edu
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Article PDF Media
Image
Disable zoom
Off
Media Files
Image
Disable zoom
Off
Image
Disable zoom
Off
Image
Disable zoom
Off

Febrile Infant CPGs

Article Type
Changed
Sun, 05/21/2017 - 13:08
Display Headline
Association of clinical practice guidelines with emergency department management of febrile infants ≤56 days of age

Febrile young infants are at high risk for serious bacterial infection (SBI) with reported rates of 8.5% to 12%, even higher in neonates 28 days of age.[1, 2, 3] As a result, febrile infants often undergo extensive diagnostic evaluation consisting of a combination of urine, blood, and cerebrospinal fluid (CSF) testing.[4, 5, 6] Several clinical prediction algorithms use this diagnostic testing to identify febrile infants at low risk for SBI, but they differ with respect to age range, recommended testing, antibiotic administration, and threshold for hospitalization.[4, 5, 6] Additionally, the optimal management strategy for this population has not been defined.[7] Consequently, laboratory testing, antibiotic use, and hospitalization for febrile young infants vary widely among hospitals.[8, 9, 10]

Clinical practice guidelines (CPGs) are designed to implement evidence‐based care and reduce practice variability, with the goal of improving quality of care and optimizing costs.[11] Implementation of a CPG for management of febrile young infants in the Intermountain Healthcare System was associated with greater adherence to evidence‐based care and lower costs.[12] However, when strong evidence is lacking, different interpretations of febrile infant risk classification incorporated into local CPGs may be a major driver of the across‐hospital practice variation observed in prior studies.[8, 9] Understanding sources of variability as well as determining the association of CPGs with clinicians' practice patterns can help identify quality improvement opportunities, either through national benchmarking or local efforts.

Our primary objectives were to compare (1) recommendations of pediatric emergency departmentbased institutional CPGs for febrile young infants and (2) rates of urine, blood, CSF testing, hospitalization, and ceftriaxone use at emergency department (ED) discharge based upon CPG presence and the specific CPG recommendations. Our secondary objectives were to describe the association of CPGs with healthcare costs and return visits for SBI.

METHODS

Study Design

We used the Pediatric Health Information System (PHIS) to identify febrile infants 56 days of age who presented to the ED between January 1, 2013 and December 31, 2013. We also surveyed ED providers at participating PHIS hospitals. Informed consent was obtained from survey respondents. The institutional review board at Boston Children's Hospital approved the study protocol.

Clinical Practice Guideline Survey

We sent an electronic survey to medical directors or division directors at 37 pediatric EDs to determine whether their ED utilized a CPG for the management of the febrile young infant in 2013. If no response was received after the second attempt, we queried ED fellowship directors or other ED attending physicians at nonresponding hospitals. Survey items included the presence of a febrile young infant CPG, and if present, the year of implementation, ages targeted, and CPG content. As applicable, respondents were asked to share their CPG and/or provide the specific CPG recommendations.

We collected and managed survey data using the Research Electronic Data Capture (REDCap) electronic data capture tools hosted at Boston Children's Hospital. REDCap is a secure, Web‐based application designed to support data capture for research studies.[13]

Data Source

The PHIS database contains administrative data from 44 US children's hospitals. These hospitals, affiliated with the Children's Hospital Association, represent 85% of freestanding US children's hospitals.[14] Encrypted patient identifiers permit tracking of patients across encounters.[15] Data quality and integrity are assured jointly by the Children's Hospital Association and participating hospitals.[16] For this study, 7 hospitals were excluded due to incomplete ED data or known data‐quality issues.[17]

Patients

We identified study infants using the following International Classification of Diseases, 9th Revision (ICD‐9) admission or discharge diagnosis codes for fever as defined previously[8, 9]: 780.6, 778.4, 780.60, or 780.61. We excluded infants with a complex chronic condition[18] and those transferred from another institution, as these infants may warrant a nonstandard evaluation and/or may have incomplete data. For infants with >1 ED visit for fever during the study period, repeat visits within 3 days of an index visit were considered a revisit for the same episode of illness; visits >3 days following an index visit were considered as a new index visit.

Study Definitions

From the PHIS database, we abstracted demographic characteristics (gender, race/ethnicity), insurance status, and region where the hospital was located (using US Census categories[19]). Billing codes were used to assess whether urine, blood, and CSF testing (as defined previously[9]) were performed during the ED evaluation. To account for ED visits that spanned the midnight hour, for hospitalized patients we considered any testing or treatment occurring on the initial or second hospital day to be performed in the ED; billing code data in PHIS are based upon calendar day and do not distinguish testing performed in the ED versus inpatient setting.[8, 9] Patients billed for observation care were classified as being hospitalized.[20, 21]

We identified the presence of an SBI using ICD‐9 diagnosis codes for the following infections as described previously[9]: urinary tract infection or pyelonephritis,[22] bacteremia or sepsis, bacterial meningitis,[16] pneumonia,[23] or bacterial enteritis. To assess return visits for SBI that required inpatient management, we defined an ED revisit for an SBI as a return visit within 3 days of ED discharge[24, 25] that resulted in hospitalization with an associated ICD‐9 discharge diagnosis code for an SBI.

Hospitals charges in PHIS database were adjusted for hospital location by using the Centers for Medicare and Medicaid Services price/wage index. Costs were estimated by applying hospital‐level cost‐to‐charge ratios to charge data.[26]

Measured Exposures

The primary exposure was the presence of an ED‐based CPG for management of the febrile young infant aged 28 days and 29 to 56 days; 56 days was used as the upper age limit as all of the CPGs included infants up to this age or beyond. Six institutions utilized CPGs with different thresholds to define the age categories (eg, dichotomized at 27 or 30 days); these CPGs were classified into the aforementioned age groups to permit comparisons across standardized age groups. We classified institutions based on the presence of a CPG. To assess differences in the application of low‐risk criteria, the CPGs were further classified a priori based upon specific recommendations around laboratory testing and hospitalization, as well as ceftriaxone use for infants aged 29 to 56 days discharged from the ED. CPGs were categorized based upon whether testing, hospitalization, and ceftriaxone use were: (1) recommended for all patients, (2) recommended only if patients were classified as high risk (absence of low‐risk criteria), (3) recommended against, or (4) recommended to consider at clinician discretion.

Outcome Measures

Measured outcomes were performance of urine, blood, CSF testing, and hospitalization rate, as well as rate of ceftriaxone use for discharged infants aged 29 to 56 days, 3‐day revisits for SBI, and costs per visit, which included hospitalization costs for admitted patients.

Data Analysis

We described continuous variables using median and interquartile range or range values and categorical variables using frequencies. We compared medians using Wilcoxon rank sum and categorical variables using a [2] test. We compared rates of testing, hospitalization, ceftriaxone use, and 3‐day revisits for SBI based on the presence of a CPG, and when present, the specific CPG recommendations. Costs per visit were compared between institutions with and without CPGs and assessed separately for admitted and discharged patients. To adjust for potential confounders and clustering of patients within hospitals, we used generalized estimating equations with logistic regression to generate adjusted odd ratios (aORs) and 95% confidence intervals (CIs). Models were adjusted for geographic region, payer, race, and gender. Statistical analyses were performed by using SAS version 9.3 (SAS Institute, Cary, NC). We determined statistical significance as a 2‐tailed P value <0.05.

Febrile infants with bronchiolitis or a history of prematurity may be managed differently from full‐term febrile young infants without bronchiolitis.[6, 27] Therefore, we performed a subgroup analysis after exclusion of infants with an ICD‐9 discharge diagnosis code for bronchiolitis (466.11 and 466.19)[28] or prematurity (765).

Because our study included ED encounters in 2013, we repeated our analyses after exclusion of hospitals with CPGs implemented during the 2013 calendar year.

RESULTS

CPG by Institution

Thirty‐three (89.2%) of the 37 EDs surveyed completed the questionnaire. Overall, 21 (63.6%) of the 33 EDs had a CPG; 15 (45.5%) had a CPG for all infants 56 days of age, 5 (15.2%) had a CPG for infants 28 days only, and 1 (3.0%) had a CPG for infants 29 to 56 days but not 28 days of age (Figure 1). Seventeen EDs had an established CPG prior to 2013, and 4 hospitals implemented a CPG during the 2013 calendar year, 2 with CPGs for neonates 28 days and 2 with CPGs for both 28 days and 29 to 56 days of age. Hospitals with CPGs were more likely to be located in the Northeast and West regions of the United States and provide care to a higher proportion of non‐Hispanic white patients, as well as those with commercial insurance (Table 1).

jhm2329-fig-0001-m.png
Specific clinical practice guideline (CPG) recommendations for diagnostic testing, hospitalization, and ceftriaxone use at ED discharge by institution among the 21 institutions with a CPG. Urine testing is defined as urine dipstick, urinalysis, or urine culture; blood testing as complete blood count or blood culture, and cerebrospinal fluid (CSF) testing as cell count, culture, or procedure code for lumbar puncture. Abbreviations: ED, emergency department.
Characteristics of Patients in Hospitals With and Without CPGs for the Febrile Young Infant 56 Days of Age
Characteristic28 Days2956 Days
No CPG, n=996, N (%)CPG, n=2,149, N (%)P ValueNo CPG, n=2,460, N (%)CPG, n=3,772, N (%)P Value
  • NOTE: Abbreviations: CPG, clinical practice guideline; IQR, interquartile range; UTI, urinary tract infection. *Includes UTI/pyelonephritis, bacteremia/sepsis, bacterial meningitis, pneumonia, and bacterial enteritis. Some infants had more than 1 site of infection.

Race      
Non‐Hispanic white325 (32.6)996 (46.3) 867 (35.2)1,728 (45.8) 
Non‐Hispanic black248 (24.9)381 (17.7) 593 (24.1)670 (17.8) 
Hispanic243 (24.4)531 (24.7) 655 (26.6)986 (26.1) 
Asian28 (2.8)78 (3.6) 40 (1.6)122 (3.2) 
Other Race152 (15.3)163 (7.6)<0.001305 (12.4)266 (7.1)<0.001
Gender      
Female435 (43.7)926 (43.1)0.761,067 (43.4)1,714 (45.4)0.22
Payer      
Commercial243 (24.4)738 (34.3) 554 (22.5)1,202 (31.9) 
Government664 (66.7)1,269 (59.1) 1,798 (73.1)2,342 (62.1) 
Other payer89 (8.9)142 (6.6)<0.001108 (4.4)228 (6.0)<0.001
Region      
Northeast39 (3.9)245 (11.4) 77 (3.1)572 (15.2) 
South648 (65.1)915 (42.6) 1,662 (67.6)1,462 (38.8) 
Midwest271 (27.2)462 (21.5) 506 (20.6)851 (22.6) 
West38 (3.8)527 (24.5)<0.001215 (8.7)887 (23.5)<0.001
Serious bacterial infection      
Overall*131 (13.2)242 (11.3)0.14191 (7.8)237 (6.3)0.03
UTI/pyelonephritis73 (7.3)153 (7.1) 103 (4.2)154 (4.1) 
Bacteremia/sepsis56 (5.6)91 (4.2) 78 (3.2)61 (1.6) 
Bacterial meningitis15 (1.5)15 (0.7) 4 (0.2)14 (0.4) 
Age, d, median (IQR)18 (11, 24)18 (11, 23)0.6746 (37, 53)45 (37, 53)0.11

All 20 CPGs for the febrile young infant 28 days of age recommended urine, blood, CSF testing, and hospitalization for all infants (Figure 1). Of the 16 hospitals with CPGs for febrile infants aged 29 to 56 days, all recommended urine and blood testing for all patients, except for 1 CPG, which recommended consideration of blood testing but not to obtain routinely. Hospitals varied in recommendations for CSF testing among infants aged 29 to 56 days: 8 (50%) recommended CSF testing in all patients and 8 (50%) recommended CSF testing only if the patient was high risk per defined criteria (based on history, physical examination, urine, and blood testing). In all 16 CPGs, hospitalization was recommended only for high‐risk infants. For low‐risk infants aged 2956 days being discharged from the ED, 3 hospitals recommended ceftriaxone for all, 9 recommended consideration of ceftriaxone, and 4 recommended against antibiotics (Figure 1).

Study Patients

During the study period, there were 10,415 infants 56 days old with a diagnosis of fever at the 33 participating hospitals. After exclusion of 635 (6.1%) infants with a complex chronic condition and 445 (4.3%) transferred from another institution (including 42 with a complex chronic condition), 9377 infants remained in our study cohort. Approximately one‐third of the cohort was 28 days of age and two‐thirds aged 29 to 56 days. The overall SBI rate was 8.5% but varied by age (11.9% in infants 28 days and 6.9% in infants 29 to 56 days of age) (Table 1).

CPGs and Use of Diagnostic Testing, Hospitalization Rates, Ceftriaxone Use, and Revisits for SBI

For infants 28 days of age, the presence of a CPG was not associated with urine, blood, CSF testing, or hospitalization after multivariable adjustment (Table 2). Among infants aged 29 to 56 days, urine testing did not differ based on the presence of a CPG, whereas blood testing was performed less often at the 1 hospital whose CPG recommended to consider, but not routinely obtain, testing (aOR: 0.4, 95% CI: 0.3‐0.7, P=0.001). Compared to hospitals without a CPG, CSF testing was performed less often at hospitals with CPG recommendations to only obtain CSF if high risk (aOR: 0.5, 95% CI: 0.3‐0.8, P=0.002). However, the odds of hospitalization did not differ at institutions with and without a febrile infant CPG (aOR: 0.7, 95% CI: 0.5‐1.1, P=0.10). For infants aged 29 to 56 days discharged from the ED, ceftriaxone was administered more often at hospitals with CPGs that recommended ceftriaxone for all discharged patients (aOR: 4.6, 95% CI: 2.39.3, P<0.001) and less often at hospitals whose CPGs recommended against antibiotics (aOR: 0.3, 95% CI: 0.1‐0.9, P=0.03) (Table 3). Our findings were similar in the subgroup of infants without bronchiolitis or prematurity (see Supporting Tables 1 and 2 in the online version of this article). After exclusion of hospitals with a CPG implemented during the 2013 calendar year (4 hospitals excluded in the 28 days age group and 2 hospitals excluded in the 29 to 56 days age group), infants aged 29 to 56 days cared for at a hospital with a CPG experienced a lower odds of hospitalization (aOR: 0.7, 95% CI: 0.4‐0.98, P=0.04). Otherwise, our findings in both age groups did not materially differ from the main analyses.

Variation in Testing and Hospitalization Based on CPG‐Specific Recommendations Among Infants 28 Days of Age With Diagnosis of Fever
Testing/HospitalizationNo. of HospitalsNo. of Patients% Received*aOR (95% CI)P Value
  • NOTE: Abbreviations: aOR, adjusted odds ratio; CI, confidence interval; CPG, clinical practice guideline; CSF, cerebrospinal fluid. *Percent of infants who received test or were hospitalized. Adjusted for hospital clustering, geographic region, payer, race, and gender. Urine testing defined as urine dipstick, urinalysis, or urine culture; Blood testing defined as complete blood count or blood culture. ‖CSF testing defined as cell count, culture, or procedure code for lumbar puncture

Laboratory testing     
Urine testing     
No CPG1399675.6Ref 
CPG: recommend for all202,14980.71.2 (0.9‐1.7)0.22
Blood testing     
No CPG1399676.9Ref 
CPG: recommend for all202,14981.81.2 (0.9‐1.7)0.25
CSF testing     
No CPG1399671.0Ref 
CPG: recommend for all202,14977.51.3 (1.01.7)0.08
Disposition     
Hospitalization     
No CPG1399675.4Ref 
CPG: recommend for all202,14981.61.2 (0.9‐1.8)0.26
Variation in Testing, Hospitalization, and Ceftriaxone Use Based on CPG‐Specific Recommendations Among Infants 29 to 56 Days of Age With Diagnosis of Fever
Testing/HospitalizationNo. of HospitalsNo. of Patients% Received*aOR (95% CI)P Value
  • NOTE: Abbreviations: aOR, adjusted odds ratio; CI, confidence interval; CPG, clinical practice guideline; CSF, cerebrospinal fluid. *Percent of infants who received test, were hospitalized, or received ceftriaxone. Adjusted for hospital clustering, geographic region, payer, race, and gender. Urine testing defined as urine dipstick, urinalysis, or urine culture. Blood testing defined as complete blood count or blood culture. CSF testing defined as cell count, culture, or procedure code for lumbar puncture. For low‐risk infants discharged from the emergency department.

Laboratory resting     
Urine testing     
No CPG172,46081.1Ref 
CPG: recommend for all163,77282.10.9 (0.7‐1.4)0.76
Blood testing     
No CPG172,46079.4Ref 
CPG: recommend for all153,62882.61.1 (0.7‐1.6)0.70
CPG: recommend consider114462.50.4 (0.3‐0.7)0.001
CSF testing     
No CPG172,46046.3Ref 
CPG: recommend for all81,51770.31.3 (0.9‐1.9)0.11
CPG: recommend if high‐risk82,25539.90.5 (0.3‐0.8)0.002
Disposition     
Hospitalization     
No CPG172,46047.0Ref 
CPG: recommend if high‐risk163,77242.00.7 (0.5‐1.1)0.10
Ceftriaxone if discharged     
No CPG171,30411.7Ref 
CPG: recommend against431310.90.3 (0.1‐0.9)0.03
CPG: recommend consider91,56714.41.5 (0.9‐2.4)0.09
CPG: recommend for all330664.14.6 (2.39.3)< 0.001

Three‐day revisits for SBI were similarly low at hospitals with and without CPGs among infants 28 days (1.5% vs 0.8%, P=0.44) and 29 to 56 days of age (1.4% vs 1.1%, P=0.44) and did not differ after exclusion of hospitals with a CPG implemented in 2013.

CPGs and Costs

Among infants 28 days of age, costs per visit did not differ for admitted and discharged patients based on CPG presence. The presence of an ED febrile infant CPG was associated with higher costs for both admitted and discharged infants 29 to 56 days of age (Table 4). The cost analysis did not significantly differ after exclusion of hospitals with CPGs implemented in 2013.

Costs per Visit for Febrile Young Infants 56 Days of Age at Institutions With and Without CPGs
 28 Days, Cost, Median (IQR)29 to 56 Days, Cost, Median (IQR)
No CPGCPGP ValueNo CPGCPGP Value
  • NOTE: Abbreviations: CPG, clinical practice guideline; IQR, interquartile range.

Admitted$4,979 ($3,408$6,607) [n=751]$4,715 ($3,472$6,526) [n=1,753]0.79$3,756 ($2,725$5,041) [n=1,156]$3,923 ($3,077$5,243) [n=1,586]<0.001
Discharged$298 ($166$510) [n=245]$231 ($160$464) [n=396]0.10$681($398$982) [n=1,304)]$764 ($412$1,100) [n=2,186]<0.001

DISCUSSION

We described the content and association of CPGs with management of the febrile infant 56 days of age across a large sample of children's hospitals. Nearly two‐thirds of included pediatric EDs have a CPG for the management of young febrile infants. Management of febrile infants 28 days was uniform, with a majority hospitalized after urine, blood, and CSF testing regardless of the presence of a CPG. In contrast, CPGs for infants 29 to 56 days of age varied in their recommendations for CSF testing as well as ceftriaxone use for infants discharged from the ED. Consequently, we observed considerable hospital variability in CSF testing and ceftriaxone use for discharged infants, which correlates with variation in the presence and content of CPGs. Institutional CPGs may be a source of the across‐hospital variation in care of febrile young infants observed in prior study.[9]

Febrile infants 28 days of age are at particularly high risk for SBI, with a prevalence of nearly 20% or higher.[2, 3, 29] The high prevalence of SBI, combined with the inherent difficulty in distinguishing neonates with and without SBI,[2, 30] has resulted in uniform CPG recommendations to perform the full‐sepsis workup in this young age group. Similar to prior studies,[8, 9] we observed that most febrile infants 28 days undergo the full sepsis evaluation, including CSF testing, and are hospitalized regardless of the presence of a CPG.

However, given the conflicting recommendations for febrile infants 29 to 56 days of age,[4, 5, 6] the optimal management strategy is less certain.[7] The Rochester, Philadelphia, and Boston criteria, 3 published models to identify infants at low risk for SBI, primarily differ in their recommendations for CSF testing and ceftriaxone use in this age group.[4, 5, 6] Half of the CPGs recommended CSF testing for all febrile infants, and half recommended CSF testing only if the infant was high risk. Institutional guidelines that recommended selective CSF testing for febrile infants aged 29 to 56 days were associated with lower rates of CSF testing. Furthermore, ceftriaxone use varied based on CPG recommendations for low‐risk infants discharged from the ED. Therefore, the influence of febrile infant CPGs mainly relates to the limiting of CSF testing and targeted ceftriaxone use in low‐risk infants. As the rate of return visits for SBI is low across hospitals, future study should assess outcomes at hospitals with CPGs recommending selective CSF testing. Of note, infants 29 to 56 days of age were less likely to be hospitalized when cared for at a hospital with an established CPG prior to 2013 without increase in 3‐day revisits for SBI. This finding may indicate that longer duration of CPG implementation is associated with lower rates of hospitalization for low‐risk infants; this finding merits further study.

The presence of a CPG was not associated with lower costs for febrile infants in either age group. Although individual healthcare systems have achieved lower costs with CPG implementation,[12] the mere presence of a CPG is not associated with lower costs when assessed across institutions. Higher costs for admitted and discharged infants 29 to 56 days of age in the presence of a CPG likely reflects the higher rate of CSF testing at hospitals whose CPGs recommend testing for all febrile infants, as well as inpatient management strategies for hospitalized infants not captured in our study. Future investigation should include an assessment of the cost‐effectiveness of the various testing and treatment strategies employed for the febrile young infant.

Our study has several limitations. First, the validity of ICD‐9 diagnosis codes for identifying young infants with fever is not well established, and thus our study is subject to misclassification bias. To minimize missed patients, we included infants with either an ICD‐9 admission or discharge diagnosis of fever; however, utilization of diagnosis codes for patient identification may have resulted in undercapture of infants with a measured temperature of 38.0C. It is also possible that some patients who did not undergo testing were misclassified as having a fever or had temperatures below standard thresholds to prompt diagnostic testing. This is a potential reason that testing was not performed in 100% of infants, even at hospitals with CPGs that recommended testing for all patients. Additionally, some febrile infants diagnosed with SBI may not have an associated ICD‐9 diagnosis code for fever. Although the overall SBI rate observed in our study was similar to prior studies,[4, 31] the rate in neonates 28 days of age was lower than reported in recent investigations,[2, 3] which may indicate inclusion of a higher proportion of low‐risk febrile infants. With the exception of bronchiolitis, we also did not assess diagnostic testing in the presence of other identified sources of infection such as herpes simplex virus.

Second, we were unable to assess the presence or absence of a CPG at the 4 excluded EDs that did not respond to the survey or the institutions excluded for data‐quality issues. However, included and excluded hospitals did not differ in region or annual ED volume (data not shown).

Third, although we classified hospitals based upon the presence and content of CPGs, we were unable to fully evaluate adherence to the CPG at each site.

Last, though PHIS hospitals represent 85% of freestanding children's hospitals, many febrile infants are hospitalized at non‐PHIS institutions; our results may not be generalizable to care provided at nonchildren's hospitals.

CONCLUSIONS

Management of febrile neonates 28 days of age does not vary based on CPG presence. However, CPGs for the febrile infant aged 29 to 56 days vary in recommendations for CSF testing as well as ceftriaxone use for low‐risk patients, which significantly contributes to practice variation and healthcare costs across institutions.

Acknowledgements

The Febrile Young Infant Research Collaborative includes the following additional investigators who are acknowledged for their work on this study: Kao‐Ping Chua, MD, Harvard PhD Program in Health Policy, Harvard University, Cambridge, Massachusetts, and Division of Emergency Medicine, Department of Pediatrics, Boston Children's Hospital, Boston, Massachusetts; Elana A. Feldman, BA, University of Washington School of Medicine, Seattle, Washington; and Katie L. Hayes, BS, Division of Emergency Medicine, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania.

Disclosures

This project was funded in part by The Gerber Foundation Novice Researcher Award (Ref #18273835). Dr. Fran Balamuth received career development support from the National Institutes of Health (NHLBI K12‐HL109009). Funders were not involved in design or conduct of the study; collection, management, analysis, or interpretation of the data; preparation, review, or approval of the manuscript. The authors have no financial relationships relevant to this article to disclose. No payment was received for the production of this article. The authors have no conflicts of interest relevant to this article to disclose.

Files
References
  1. Huppler AR, Eickhoff JC, Wald ER. Performance of low‐risk criteria in the evaluation of young infants with fever: review of the literature. Pediatrics. 2010;125:228233.
  2. Schwartz S, Raveh D, Toker O, Segal G, Godovitch N, Schlesinger Y. A week‐by‐week analysis of the low‐risk criteria for serious bacterial infection in febrile neonates. Arch Dis Child. 2009;94:287292.
  3. Garcia S, Mintegi S, Gomez B, et al. Is 15 days an appropriate cut‐off age for considering serious bacterial infection in the management of febrile infants? Pediatr Infect Dis J. 2012;31:455458.
  4. Baker MD, Bell LM, Avner JR. Outpatient management without antibiotics of fever in selected infants. N Engl J Med. 1993;329:14371441.
  5. Baskin MN, Fleisher GR, O'Rourke EJ. Identifying febrile infants at risk for a serious bacterial infection. J Pediatr. 1993;123:489490.
  6. Jaskiewicz JA, McCarthy CA, Richardson AC, et al. Febrile infants at low risk for serious bacterial infection—an appraisal of the Rochester criteria and implications for management. Febrile Infant Collaborative Study Group. Pediatrics. 1994;94:390396.
  7. American College of Emergency Physicians Clinical Policies Committee; American College of Emergency Physicians Clinical Policies Subcommittee on Pediatric Fever. Clinical policy for children younger than three years presenting to the emergency department with fever. Ann Emerg Med. 2003;42:530545.
  8. Jain S, Cheng J, Alpern ER, et al. Management of febrile neonates in US pediatric emergency departments. Pediatrics. 2014;133:187195.
  9. Aronson PL, Thurm C, Alpern ER, et al. Variation in care of the febrile young infant <90 days in US pediatric emergency departments. Pediatrics. 2014;134:667677.
  10. Yarden‐Bilavsky H, Ashkenazi S, Amir J, Schlesinger Y, Bilavsky E. Fever survey highlights significant variations in how infants aged ≤60 days are evaluated and underline the need for guidelines. Acta Paediatr. 2014;103:379385.
  11. Bergman DA. Evidence‐based guidelines and critical pathways for quality improvement. Pediatrics. 1999;103:225232.
  12. Byington CL, Reynolds CC, Korgenski K, et al. Costs and infant outcomes after implementation of a care process model for febrile infants. Pediatrics. 2012;130:e16e24.
  13. Harris PA, Taylor R, Thielke R, Payne J, Gonzalez N, Conde JG. Research electronic data capture (REDCap)—a metadata‐driven methodology and workflow process for providing translational research informatics support. J Biomed Inform. 2009;42:377381.
  14. Wood JN, Feudtner C, Medina SP, Luan X, Localio R, Rubin DM. Variation in occult injury screening for children with suspected abuse in selected US children's hospitals. Pediatrics. 2012;130:853860.
  15. Fletcher DM. Achieving data quality. How data from a pediatric health information system earns the trust of its users. J AHIMA. 2004;75:2226.
  16. Mongelluzzo J, Mohamad Z, Have TR, Shah SS. Corticosteroids and mortality in children with bacterial meningitis. JAMA. 2008;299:20482055.
  17. Kharbanda AB, Hall M, Shah SS, et al. Variation in resource utilization across a national sample of pediatric emergency departments. J Pediatr. 2013;163:230236.
  18. Feudtner C, Hays RM, Haynes G, Geyer JR, Neff JM, Koepsell TD. Deaths attributed to pediatric complex chronic conditions: national trends and implications for supportive care services. Pediatrics. 2001;107:E99.
  19. US Census Bureau. Geographic terms and concepts—census divisions and census regions. Available at: https://www.census.gov/geo/reference/gtc/gtc_census_divreg.html. Accessed September 10, 2014.
  20. Macy ML, Hall M, Shah SS, et al. Pediatric observation status: are we overlooking a growing population in children's hospitals? J Hosp Med. 2012;7:530536.
  21. Macy ML, Hall M, Shah SS, et al. Differences in designations of observation care in US freestanding children's hospitals: are they virtual or real? J Hosp Med. 2012;7:287293.
  22. Tieder JS, Hall M, Auger KA, et al. Accuracy of administrative billing codes to detect urinary tract infection hospitalizations. Pediatrics. 2011;128:323330.
  23. Williams DJ, Shah SS, Myers A, et al. Identifying pediatric community‐acquired pneumonia hospitalizations: accuracy of administrative billing codes. JAMA Pediatr. 2013;167:851858.
  24. Gordon JA, An LC, Hayward RA, Williams BC. Initial emergency department diagnosis and return visits: risk versus perception. Ann Emerg Med. 1998;32:569573.
  25. Cho CS, Shapiro DJ, Cabana MD, Maselli JH, Hersh AL. A national depiction of children with return visits to the emergency department within 72 hours, 2001–2007. Pediatr Emerg Care. 2012;28:606610.
  26. Healthcare Cost and Utilization Project. Cost‐to‐charge ratio files. Available at: http://www.hcup‐us.ahrq.gov/db/state/costtocharge.jsp. Accessed September 11, 2014.
  27. Levine DA, Platt SL, Dayan PS, et al. Risk of serious bacterial infection in young febrile infants with respiratory syncytial virus infections. Pediatrics. 2004;113:17281734.
  28. Parikh K, Hall M, Mittal V, et al. Establishing benchmarks for the hospitalized care of children with asthma, bronchiolitis, and pneumonia. Pediatrics. 2014;134:555562.
  29. Mintegi S, Benito J, Astobiza E, Capape S, Gomez B, Eguireun A. Well appearing young infants with fever without known source in the emergency department: are lumbar punctures always necessary? Eur J Emerg Med. 2010;17:167169.
  30. Baker MD, Bell LM. Unpredictability of serious bacterial illness in febrile infants from birth to 1 month of age. Arch Pediatr Adolesc Med. 1999;153:508511.
  31. Pantell RH, Newman TB, Bernzweig J, et al. Management and outcomes of care of fever in early infancy. JAMA. 2004;291:12031212.
Article PDF
Issue
Journal of Hospital Medicine - 10(6)
Publications
Page Number
358-365
Sections
Files
Files
Article PDF
Article PDF

Febrile young infants are at high risk for serious bacterial infection (SBI) with reported rates of 8.5% to 12%, even higher in neonates 28 days of age.[1, 2, 3] As a result, febrile infants often undergo extensive diagnostic evaluation consisting of a combination of urine, blood, and cerebrospinal fluid (CSF) testing.[4, 5, 6] Several clinical prediction algorithms use this diagnostic testing to identify febrile infants at low risk for SBI, but they differ with respect to age range, recommended testing, antibiotic administration, and threshold for hospitalization.[4, 5, 6] Additionally, the optimal management strategy for this population has not been defined.[7] Consequently, laboratory testing, antibiotic use, and hospitalization for febrile young infants vary widely among hospitals.[8, 9, 10]

Clinical practice guidelines (CPGs) are designed to implement evidence‐based care and reduce practice variability, with the goal of improving quality of care and optimizing costs.[11] Implementation of a CPG for management of febrile young infants in the Intermountain Healthcare System was associated with greater adherence to evidence‐based care and lower costs.[12] However, when strong evidence is lacking, different interpretations of febrile infant risk classification incorporated into local CPGs may be a major driver of the across‐hospital practice variation observed in prior studies.[8, 9] Understanding sources of variability as well as determining the association of CPGs with clinicians' practice patterns can help identify quality improvement opportunities, either through national benchmarking or local efforts.

Our primary objectives were to compare (1) recommendations of pediatric emergency departmentbased institutional CPGs for febrile young infants and (2) rates of urine, blood, CSF testing, hospitalization, and ceftriaxone use at emergency department (ED) discharge based upon CPG presence and the specific CPG recommendations. Our secondary objectives were to describe the association of CPGs with healthcare costs and return visits for SBI.

METHODS

Study Design

We used the Pediatric Health Information System (PHIS) to identify febrile infants 56 days of age who presented to the ED between January 1, 2013 and December 31, 2013. We also surveyed ED providers at participating PHIS hospitals. Informed consent was obtained from survey respondents. The institutional review board at Boston Children's Hospital approved the study protocol.

Clinical Practice Guideline Survey

We sent an electronic survey to medical directors or division directors at 37 pediatric EDs to determine whether their ED utilized a CPG for the management of the febrile young infant in 2013. If no response was received after the second attempt, we queried ED fellowship directors or other ED attending physicians at nonresponding hospitals. Survey items included the presence of a febrile young infant CPG, and if present, the year of implementation, ages targeted, and CPG content. As applicable, respondents were asked to share their CPG and/or provide the specific CPG recommendations.

We collected and managed survey data using the Research Electronic Data Capture (REDCap) electronic data capture tools hosted at Boston Children's Hospital. REDCap is a secure, Web‐based application designed to support data capture for research studies.[13]

Data Source

The PHIS database contains administrative data from 44 US children's hospitals. These hospitals, affiliated with the Children's Hospital Association, represent 85% of freestanding US children's hospitals.[14] Encrypted patient identifiers permit tracking of patients across encounters.[15] Data quality and integrity are assured jointly by the Children's Hospital Association and participating hospitals.[16] For this study, 7 hospitals were excluded due to incomplete ED data or known data‐quality issues.[17]

Patients

We identified study infants using the following International Classification of Diseases, 9th Revision (ICD‐9) admission or discharge diagnosis codes for fever as defined previously[8, 9]: 780.6, 778.4, 780.60, or 780.61. We excluded infants with a complex chronic condition[18] and those transferred from another institution, as these infants may warrant a nonstandard evaluation and/or may have incomplete data. For infants with >1 ED visit for fever during the study period, repeat visits within 3 days of an index visit were considered a revisit for the same episode of illness; visits >3 days following an index visit were considered as a new index visit.

Study Definitions

From the PHIS database, we abstracted demographic characteristics (gender, race/ethnicity), insurance status, and region where the hospital was located (using US Census categories[19]). Billing codes were used to assess whether urine, blood, and CSF testing (as defined previously[9]) were performed during the ED evaluation. To account for ED visits that spanned the midnight hour, for hospitalized patients we considered any testing or treatment occurring on the initial or second hospital day to be performed in the ED; billing code data in PHIS are based upon calendar day and do not distinguish testing performed in the ED versus inpatient setting.[8, 9] Patients billed for observation care were classified as being hospitalized.[20, 21]

We identified the presence of an SBI using ICD‐9 diagnosis codes for the following infections as described previously[9]: urinary tract infection or pyelonephritis,[22] bacteremia or sepsis, bacterial meningitis,[16] pneumonia,[23] or bacterial enteritis. To assess return visits for SBI that required inpatient management, we defined an ED revisit for an SBI as a return visit within 3 days of ED discharge[24, 25] that resulted in hospitalization with an associated ICD‐9 discharge diagnosis code for an SBI.

Hospitals charges in PHIS database were adjusted for hospital location by using the Centers for Medicare and Medicaid Services price/wage index. Costs were estimated by applying hospital‐level cost‐to‐charge ratios to charge data.[26]

Measured Exposures

The primary exposure was the presence of an ED‐based CPG for management of the febrile young infant aged 28 days and 29 to 56 days; 56 days was used as the upper age limit as all of the CPGs included infants up to this age or beyond. Six institutions utilized CPGs with different thresholds to define the age categories (eg, dichotomized at 27 or 30 days); these CPGs were classified into the aforementioned age groups to permit comparisons across standardized age groups. We classified institutions based on the presence of a CPG. To assess differences in the application of low‐risk criteria, the CPGs were further classified a priori based upon specific recommendations around laboratory testing and hospitalization, as well as ceftriaxone use for infants aged 29 to 56 days discharged from the ED. CPGs were categorized based upon whether testing, hospitalization, and ceftriaxone use were: (1) recommended for all patients, (2) recommended only if patients were classified as high risk (absence of low‐risk criteria), (3) recommended against, or (4) recommended to consider at clinician discretion.

Outcome Measures

Measured outcomes were performance of urine, blood, CSF testing, and hospitalization rate, as well as rate of ceftriaxone use for discharged infants aged 29 to 56 days, 3‐day revisits for SBI, and costs per visit, which included hospitalization costs for admitted patients.

Data Analysis

We described continuous variables using median and interquartile range or range values and categorical variables using frequencies. We compared medians using Wilcoxon rank sum and categorical variables using a [2] test. We compared rates of testing, hospitalization, ceftriaxone use, and 3‐day revisits for SBI based on the presence of a CPG, and when present, the specific CPG recommendations. Costs per visit were compared between institutions with and without CPGs and assessed separately for admitted and discharged patients. To adjust for potential confounders and clustering of patients within hospitals, we used generalized estimating equations with logistic regression to generate adjusted odd ratios (aORs) and 95% confidence intervals (CIs). Models were adjusted for geographic region, payer, race, and gender. Statistical analyses were performed by using SAS version 9.3 (SAS Institute, Cary, NC). We determined statistical significance as a 2‐tailed P value <0.05.

Febrile infants with bronchiolitis or a history of prematurity may be managed differently from full‐term febrile young infants without bronchiolitis.[6, 27] Therefore, we performed a subgroup analysis after exclusion of infants with an ICD‐9 discharge diagnosis code for bronchiolitis (466.11 and 466.19)[28] or prematurity (765).

Because our study included ED encounters in 2013, we repeated our analyses after exclusion of hospitals with CPGs implemented during the 2013 calendar year.

RESULTS

CPG by Institution

Thirty‐three (89.2%) of the 37 EDs surveyed completed the questionnaire. Overall, 21 (63.6%) of the 33 EDs had a CPG; 15 (45.5%) had a CPG for all infants 56 days of age, 5 (15.2%) had a CPG for infants 28 days only, and 1 (3.0%) had a CPG for infants 29 to 56 days but not 28 days of age (Figure 1). Seventeen EDs had an established CPG prior to 2013, and 4 hospitals implemented a CPG during the 2013 calendar year, 2 with CPGs for neonates 28 days and 2 with CPGs for both 28 days and 29 to 56 days of age. Hospitals with CPGs were more likely to be located in the Northeast and West regions of the United States and provide care to a higher proportion of non‐Hispanic white patients, as well as those with commercial insurance (Table 1).

jhm2329-fig-0001-m.png
Specific clinical practice guideline (CPG) recommendations for diagnostic testing, hospitalization, and ceftriaxone use at ED discharge by institution among the 21 institutions with a CPG. Urine testing is defined as urine dipstick, urinalysis, or urine culture; blood testing as complete blood count or blood culture, and cerebrospinal fluid (CSF) testing as cell count, culture, or procedure code for lumbar puncture. Abbreviations: ED, emergency department.
Characteristics of Patients in Hospitals With and Without CPGs for the Febrile Young Infant 56 Days of Age
Characteristic28 Days2956 Days
No CPG, n=996, N (%)CPG, n=2,149, N (%)P ValueNo CPG, n=2,460, N (%)CPG, n=3,772, N (%)P Value
  • NOTE: Abbreviations: CPG, clinical practice guideline; IQR, interquartile range; UTI, urinary tract infection. *Includes UTI/pyelonephritis, bacteremia/sepsis, bacterial meningitis, pneumonia, and bacterial enteritis. Some infants had more than 1 site of infection.

Race      
Non‐Hispanic white325 (32.6)996 (46.3) 867 (35.2)1,728 (45.8) 
Non‐Hispanic black248 (24.9)381 (17.7) 593 (24.1)670 (17.8) 
Hispanic243 (24.4)531 (24.7) 655 (26.6)986 (26.1) 
Asian28 (2.8)78 (3.6) 40 (1.6)122 (3.2) 
Other Race152 (15.3)163 (7.6)<0.001305 (12.4)266 (7.1)<0.001
Gender      
Female435 (43.7)926 (43.1)0.761,067 (43.4)1,714 (45.4)0.22
Payer      
Commercial243 (24.4)738 (34.3) 554 (22.5)1,202 (31.9) 
Government664 (66.7)1,269 (59.1) 1,798 (73.1)2,342 (62.1) 
Other payer89 (8.9)142 (6.6)<0.001108 (4.4)228 (6.0)<0.001
Region      
Northeast39 (3.9)245 (11.4) 77 (3.1)572 (15.2) 
South648 (65.1)915 (42.6) 1,662 (67.6)1,462 (38.8) 
Midwest271 (27.2)462 (21.5) 506 (20.6)851 (22.6) 
West38 (3.8)527 (24.5)<0.001215 (8.7)887 (23.5)<0.001
Serious bacterial infection      
Overall*131 (13.2)242 (11.3)0.14191 (7.8)237 (6.3)0.03
UTI/pyelonephritis73 (7.3)153 (7.1) 103 (4.2)154 (4.1) 
Bacteremia/sepsis56 (5.6)91 (4.2) 78 (3.2)61 (1.6) 
Bacterial meningitis15 (1.5)15 (0.7) 4 (0.2)14 (0.4) 
Age, d, median (IQR)18 (11, 24)18 (11, 23)0.6746 (37, 53)45 (37, 53)0.11

All 20 CPGs for the febrile young infant 28 days of age recommended urine, blood, CSF testing, and hospitalization for all infants (Figure 1). Of the 16 hospitals with CPGs for febrile infants aged 29 to 56 days, all recommended urine and blood testing for all patients, except for 1 CPG, which recommended consideration of blood testing but not to obtain routinely. Hospitals varied in recommendations for CSF testing among infants aged 29 to 56 days: 8 (50%) recommended CSF testing in all patients and 8 (50%) recommended CSF testing only if the patient was high risk per defined criteria (based on history, physical examination, urine, and blood testing). In all 16 CPGs, hospitalization was recommended only for high‐risk infants. For low‐risk infants aged 2956 days being discharged from the ED, 3 hospitals recommended ceftriaxone for all, 9 recommended consideration of ceftriaxone, and 4 recommended against antibiotics (Figure 1).

Study Patients

During the study period, there were 10,415 infants 56 days old with a diagnosis of fever at the 33 participating hospitals. After exclusion of 635 (6.1%) infants with a complex chronic condition and 445 (4.3%) transferred from another institution (including 42 with a complex chronic condition), 9377 infants remained in our study cohort. Approximately one‐third of the cohort was 28 days of age and two‐thirds aged 29 to 56 days. The overall SBI rate was 8.5% but varied by age (11.9% in infants 28 days and 6.9% in infants 29 to 56 days of age) (Table 1).

CPGs and Use of Diagnostic Testing, Hospitalization Rates, Ceftriaxone Use, and Revisits for SBI

For infants 28 days of age, the presence of a CPG was not associated with urine, blood, CSF testing, or hospitalization after multivariable adjustment (Table 2). Among infants aged 29 to 56 days, urine testing did not differ based on the presence of a CPG, whereas blood testing was performed less often at the 1 hospital whose CPG recommended to consider, but not routinely obtain, testing (aOR: 0.4, 95% CI: 0.3‐0.7, P=0.001). Compared to hospitals without a CPG, CSF testing was performed less often at hospitals with CPG recommendations to only obtain CSF if high risk (aOR: 0.5, 95% CI: 0.3‐0.8, P=0.002). However, the odds of hospitalization did not differ at institutions with and without a febrile infant CPG (aOR: 0.7, 95% CI: 0.5‐1.1, P=0.10). For infants aged 29 to 56 days discharged from the ED, ceftriaxone was administered more often at hospitals with CPGs that recommended ceftriaxone for all discharged patients (aOR: 4.6, 95% CI: 2.39.3, P<0.001) and less often at hospitals whose CPGs recommended against antibiotics (aOR: 0.3, 95% CI: 0.1‐0.9, P=0.03) (Table 3). Our findings were similar in the subgroup of infants without bronchiolitis or prematurity (see Supporting Tables 1 and 2 in the online version of this article). After exclusion of hospitals with a CPG implemented during the 2013 calendar year (4 hospitals excluded in the 28 days age group and 2 hospitals excluded in the 29 to 56 days age group), infants aged 29 to 56 days cared for at a hospital with a CPG experienced a lower odds of hospitalization (aOR: 0.7, 95% CI: 0.4‐0.98, P=0.04). Otherwise, our findings in both age groups did not materially differ from the main analyses.

Variation in Testing and Hospitalization Based on CPG‐Specific Recommendations Among Infants 28 Days of Age With Diagnosis of Fever
Testing/HospitalizationNo. of HospitalsNo. of Patients% Received*aOR (95% CI)P Value
  • NOTE: Abbreviations: aOR, adjusted odds ratio; CI, confidence interval; CPG, clinical practice guideline; CSF, cerebrospinal fluid. *Percent of infants who received test or were hospitalized. Adjusted for hospital clustering, geographic region, payer, race, and gender. Urine testing defined as urine dipstick, urinalysis, or urine culture; Blood testing defined as complete blood count or blood culture. ‖CSF testing defined as cell count, culture, or procedure code for lumbar puncture

Laboratory testing     
Urine testing     
No CPG1399675.6Ref 
CPG: recommend for all202,14980.71.2 (0.9‐1.7)0.22
Blood testing     
No CPG1399676.9Ref 
CPG: recommend for all202,14981.81.2 (0.9‐1.7)0.25
CSF testing     
No CPG1399671.0Ref 
CPG: recommend for all202,14977.51.3 (1.01.7)0.08
Disposition     
Hospitalization     
No CPG1399675.4Ref 
CPG: recommend for all202,14981.61.2 (0.9‐1.8)0.26
Variation in Testing, Hospitalization, and Ceftriaxone Use Based on CPG‐Specific Recommendations Among Infants 29 to 56 Days of Age With Diagnosis of Fever
Testing/HospitalizationNo. of HospitalsNo. of Patients% Received*aOR (95% CI)P Value
  • NOTE: Abbreviations: aOR, adjusted odds ratio; CI, confidence interval; CPG, clinical practice guideline; CSF, cerebrospinal fluid. *Percent of infants who received test, were hospitalized, or received ceftriaxone. Adjusted for hospital clustering, geographic region, payer, race, and gender. Urine testing defined as urine dipstick, urinalysis, or urine culture. Blood testing defined as complete blood count or blood culture. CSF testing defined as cell count, culture, or procedure code for lumbar puncture. For low‐risk infants discharged from the emergency department.

Laboratory resting     
Urine testing     
No CPG172,46081.1Ref 
CPG: recommend for all163,77282.10.9 (0.7‐1.4)0.76
Blood testing     
No CPG172,46079.4Ref 
CPG: recommend for all153,62882.61.1 (0.7‐1.6)0.70
CPG: recommend consider114462.50.4 (0.3‐0.7)0.001
CSF testing     
No CPG172,46046.3Ref 
CPG: recommend for all81,51770.31.3 (0.9‐1.9)0.11
CPG: recommend if high‐risk82,25539.90.5 (0.3‐0.8)0.002
Disposition     
Hospitalization     
No CPG172,46047.0Ref 
CPG: recommend if high‐risk163,77242.00.7 (0.5‐1.1)0.10
Ceftriaxone if discharged     
No CPG171,30411.7Ref 
CPG: recommend against431310.90.3 (0.1‐0.9)0.03
CPG: recommend consider91,56714.41.5 (0.9‐2.4)0.09
CPG: recommend for all330664.14.6 (2.39.3)< 0.001

Three‐day revisits for SBI were similarly low at hospitals with and without CPGs among infants 28 days (1.5% vs 0.8%, P=0.44) and 29 to 56 days of age (1.4% vs 1.1%, P=0.44) and did not differ after exclusion of hospitals with a CPG implemented in 2013.

CPGs and Costs

Among infants 28 days of age, costs per visit did not differ for admitted and discharged patients based on CPG presence. The presence of an ED febrile infant CPG was associated with higher costs for both admitted and discharged infants 29 to 56 days of age (Table 4). The cost analysis did not significantly differ after exclusion of hospitals with CPGs implemented in 2013.

Costs per Visit for Febrile Young Infants 56 Days of Age at Institutions With and Without CPGs
 28 Days, Cost, Median (IQR)29 to 56 Days, Cost, Median (IQR)
No CPGCPGP ValueNo CPGCPGP Value
  • NOTE: Abbreviations: CPG, clinical practice guideline; IQR, interquartile range.

Admitted$4,979 ($3,408$6,607) [n=751]$4,715 ($3,472$6,526) [n=1,753]0.79$3,756 ($2,725$5,041) [n=1,156]$3,923 ($3,077$5,243) [n=1,586]<0.001
Discharged$298 ($166$510) [n=245]$231 ($160$464) [n=396]0.10$681($398$982) [n=1,304)]$764 ($412$1,100) [n=2,186]<0.001

DISCUSSION

We described the content and association of CPGs with management of the febrile infant 56 days of age across a large sample of children's hospitals. Nearly two‐thirds of included pediatric EDs have a CPG for the management of young febrile infants. Management of febrile infants 28 days was uniform, with a majority hospitalized after urine, blood, and CSF testing regardless of the presence of a CPG. In contrast, CPGs for infants 29 to 56 days of age varied in their recommendations for CSF testing as well as ceftriaxone use for infants discharged from the ED. Consequently, we observed considerable hospital variability in CSF testing and ceftriaxone use for discharged infants, which correlates with variation in the presence and content of CPGs. Institutional CPGs may be a source of the across‐hospital variation in care of febrile young infants observed in prior study.[9]

Febrile infants 28 days of age are at particularly high risk for SBI, with a prevalence of nearly 20% or higher.[2, 3, 29] The high prevalence of SBI, combined with the inherent difficulty in distinguishing neonates with and without SBI,[2, 30] has resulted in uniform CPG recommendations to perform the full‐sepsis workup in this young age group. Similar to prior studies,[8, 9] we observed that most febrile infants 28 days undergo the full sepsis evaluation, including CSF testing, and are hospitalized regardless of the presence of a CPG.

However, given the conflicting recommendations for febrile infants 29 to 56 days of age,[4, 5, 6] the optimal management strategy is less certain.[7] The Rochester, Philadelphia, and Boston criteria, 3 published models to identify infants at low risk for SBI, primarily differ in their recommendations for CSF testing and ceftriaxone use in this age group.[4, 5, 6] Half of the CPGs recommended CSF testing for all febrile infants, and half recommended CSF testing only if the infant was high risk. Institutional guidelines that recommended selective CSF testing for febrile infants aged 29 to 56 days were associated with lower rates of CSF testing. Furthermore, ceftriaxone use varied based on CPG recommendations for low‐risk infants discharged from the ED. Therefore, the influence of febrile infant CPGs mainly relates to the limiting of CSF testing and targeted ceftriaxone use in low‐risk infants. As the rate of return visits for SBI is low across hospitals, future study should assess outcomes at hospitals with CPGs recommending selective CSF testing. Of note, infants 29 to 56 days of age were less likely to be hospitalized when cared for at a hospital with an established CPG prior to 2013 without increase in 3‐day revisits for SBI. This finding may indicate that longer duration of CPG implementation is associated with lower rates of hospitalization for low‐risk infants; this finding merits further study.

The presence of a CPG was not associated with lower costs for febrile infants in either age group. Although individual healthcare systems have achieved lower costs with CPG implementation,[12] the mere presence of a CPG is not associated with lower costs when assessed across institutions. Higher costs for admitted and discharged infants 29 to 56 days of age in the presence of a CPG likely reflects the higher rate of CSF testing at hospitals whose CPGs recommend testing for all febrile infants, as well as inpatient management strategies for hospitalized infants not captured in our study. Future investigation should include an assessment of the cost‐effectiveness of the various testing and treatment strategies employed for the febrile young infant.

Our study has several limitations. First, the validity of ICD‐9 diagnosis codes for identifying young infants with fever is not well established, and thus our study is subject to misclassification bias. To minimize missed patients, we included infants with either an ICD‐9 admission or discharge diagnosis of fever; however, utilization of diagnosis codes for patient identification may have resulted in undercapture of infants with a measured temperature of 38.0C. It is also possible that some patients who did not undergo testing were misclassified as having a fever or had temperatures below standard thresholds to prompt diagnostic testing. This is a potential reason that testing was not performed in 100% of infants, even at hospitals with CPGs that recommended testing for all patients. Additionally, some febrile infants diagnosed with SBI may not have an associated ICD‐9 diagnosis code for fever. Although the overall SBI rate observed in our study was similar to prior studies,[4, 31] the rate in neonates 28 days of age was lower than reported in recent investigations,[2, 3] which may indicate inclusion of a higher proportion of low‐risk febrile infants. With the exception of bronchiolitis, we also did not assess diagnostic testing in the presence of other identified sources of infection such as herpes simplex virus.

Second, we were unable to assess the presence or absence of a CPG at the 4 excluded EDs that did not respond to the survey or the institutions excluded for data‐quality issues. However, included and excluded hospitals did not differ in region or annual ED volume (data not shown).

Third, although we classified hospitals based upon the presence and content of CPGs, we were unable to fully evaluate adherence to the CPG at each site.

Last, though PHIS hospitals represent 85% of freestanding children's hospitals, many febrile infants are hospitalized at non‐PHIS institutions; our results may not be generalizable to care provided at nonchildren's hospitals.

CONCLUSIONS

Management of febrile neonates 28 days of age does not vary based on CPG presence. However, CPGs for the febrile infant aged 29 to 56 days vary in recommendations for CSF testing as well as ceftriaxone use for low‐risk patients, which significantly contributes to practice variation and healthcare costs across institutions.

Acknowledgements

The Febrile Young Infant Research Collaborative includes the following additional investigators who are acknowledged for their work on this study: Kao‐Ping Chua, MD, Harvard PhD Program in Health Policy, Harvard University, Cambridge, Massachusetts, and Division of Emergency Medicine, Department of Pediatrics, Boston Children's Hospital, Boston, Massachusetts; Elana A. Feldman, BA, University of Washington School of Medicine, Seattle, Washington; and Katie L. Hayes, BS, Division of Emergency Medicine, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania.

Disclosures

This project was funded in part by The Gerber Foundation Novice Researcher Award (Ref #18273835). Dr. Fran Balamuth received career development support from the National Institutes of Health (NHLBI K12‐HL109009). Funders were not involved in design or conduct of the study; collection, management, analysis, or interpretation of the data; preparation, review, or approval of the manuscript. The authors have no financial relationships relevant to this article to disclose. No payment was received for the production of this article. The authors have no conflicts of interest relevant to this article to disclose.

Febrile young infants are at high risk for serious bacterial infection (SBI) with reported rates of 8.5% to 12%, even higher in neonates 28 days of age.[1, 2, 3] As a result, febrile infants often undergo extensive diagnostic evaluation consisting of a combination of urine, blood, and cerebrospinal fluid (CSF) testing.[4, 5, 6] Several clinical prediction algorithms use this diagnostic testing to identify febrile infants at low risk for SBI, but they differ with respect to age range, recommended testing, antibiotic administration, and threshold for hospitalization.[4, 5, 6] Additionally, the optimal management strategy for this population has not been defined.[7] Consequently, laboratory testing, antibiotic use, and hospitalization for febrile young infants vary widely among hospitals.[8, 9, 10]

Clinical practice guidelines (CPGs) are designed to implement evidence‐based care and reduce practice variability, with the goal of improving quality of care and optimizing costs.[11] Implementation of a CPG for management of febrile young infants in the Intermountain Healthcare System was associated with greater adherence to evidence‐based care and lower costs.[12] However, when strong evidence is lacking, different interpretations of febrile infant risk classification incorporated into local CPGs may be a major driver of the across‐hospital practice variation observed in prior studies.[8, 9] Understanding sources of variability as well as determining the association of CPGs with clinicians' practice patterns can help identify quality improvement opportunities, either through national benchmarking or local efforts.

Our primary objectives were to compare (1) recommendations of pediatric emergency departmentbased institutional CPGs for febrile young infants and (2) rates of urine, blood, CSF testing, hospitalization, and ceftriaxone use at emergency department (ED) discharge based upon CPG presence and the specific CPG recommendations. Our secondary objectives were to describe the association of CPGs with healthcare costs and return visits for SBI.

METHODS

Study Design

We used the Pediatric Health Information System (PHIS) to identify febrile infants 56 days of age who presented to the ED between January 1, 2013 and December 31, 2013. We also surveyed ED providers at participating PHIS hospitals. Informed consent was obtained from survey respondents. The institutional review board at Boston Children's Hospital approved the study protocol.

Clinical Practice Guideline Survey

We sent an electronic survey to medical directors or division directors at 37 pediatric EDs to determine whether their ED utilized a CPG for the management of the febrile young infant in 2013. If no response was received after the second attempt, we queried ED fellowship directors or other ED attending physicians at nonresponding hospitals. Survey items included the presence of a febrile young infant CPG, and if present, the year of implementation, ages targeted, and CPG content. As applicable, respondents were asked to share their CPG and/or provide the specific CPG recommendations.

We collected and managed survey data using the Research Electronic Data Capture (REDCap) electronic data capture tools hosted at Boston Children's Hospital. REDCap is a secure, Web‐based application designed to support data capture for research studies.[13]

Data Source

The PHIS database contains administrative data from 44 US children's hospitals. These hospitals, affiliated with the Children's Hospital Association, represent 85% of freestanding US children's hospitals.[14] Encrypted patient identifiers permit tracking of patients across encounters.[15] Data quality and integrity are assured jointly by the Children's Hospital Association and participating hospitals.[16] For this study, 7 hospitals were excluded due to incomplete ED data or known data‐quality issues.[17]

Patients

We identified study infants using the following International Classification of Diseases, 9th Revision (ICD‐9) admission or discharge diagnosis codes for fever as defined previously[8, 9]: 780.6, 778.4, 780.60, or 780.61. We excluded infants with a complex chronic condition[18] and those transferred from another institution, as these infants may warrant a nonstandard evaluation and/or may have incomplete data. For infants with >1 ED visit for fever during the study period, repeat visits within 3 days of an index visit were considered a revisit for the same episode of illness; visits >3 days following an index visit were considered as a new index visit.

Study Definitions

From the PHIS database, we abstracted demographic characteristics (gender, race/ethnicity), insurance status, and region where the hospital was located (using US Census categories[19]). Billing codes were used to assess whether urine, blood, and CSF testing (as defined previously[9]) were performed during the ED evaluation. To account for ED visits that spanned the midnight hour, for hospitalized patients we considered any testing or treatment occurring on the initial or second hospital day to be performed in the ED; billing code data in PHIS are based upon calendar day and do not distinguish testing performed in the ED versus inpatient setting.[8, 9] Patients billed for observation care were classified as being hospitalized.[20, 21]

We identified the presence of an SBI using ICD‐9 diagnosis codes for the following infections as described previously[9]: urinary tract infection or pyelonephritis,[22] bacteremia or sepsis, bacterial meningitis,[16] pneumonia,[23] or bacterial enteritis. To assess return visits for SBI that required inpatient management, we defined an ED revisit for an SBI as a return visit within 3 days of ED discharge[24, 25] that resulted in hospitalization with an associated ICD‐9 discharge diagnosis code for an SBI.

Hospitals charges in PHIS database were adjusted for hospital location by using the Centers for Medicare and Medicaid Services price/wage index. Costs were estimated by applying hospital‐level cost‐to‐charge ratios to charge data.[26]

Measured Exposures

The primary exposure was the presence of an ED‐based CPG for management of the febrile young infant aged 28 days and 29 to 56 days; 56 days was used as the upper age limit as all of the CPGs included infants up to this age or beyond. Six institutions utilized CPGs with different thresholds to define the age categories (eg, dichotomized at 27 or 30 days); these CPGs were classified into the aforementioned age groups to permit comparisons across standardized age groups. We classified institutions based on the presence of a CPG. To assess differences in the application of low‐risk criteria, the CPGs were further classified a priori based upon specific recommendations around laboratory testing and hospitalization, as well as ceftriaxone use for infants aged 29 to 56 days discharged from the ED. CPGs were categorized based upon whether testing, hospitalization, and ceftriaxone use were: (1) recommended for all patients, (2) recommended only if patients were classified as high risk (absence of low‐risk criteria), (3) recommended against, or (4) recommended to consider at clinician discretion.

Outcome Measures

Measured outcomes were performance of urine, blood, CSF testing, and hospitalization rate, as well as rate of ceftriaxone use for discharged infants aged 29 to 56 days, 3‐day revisits for SBI, and costs per visit, which included hospitalization costs for admitted patients.

Data Analysis

We described continuous variables using median and interquartile range or range values and categorical variables using frequencies. We compared medians using Wilcoxon rank sum and categorical variables using a [2] test. We compared rates of testing, hospitalization, ceftriaxone use, and 3‐day revisits for SBI based on the presence of a CPG, and when present, the specific CPG recommendations. Costs per visit were compared between institutions with and without CPGs and assessed separately for admitted and discharged patients. To adjust for potential confounders and clustering of patients within hospitals, we used generalized estimating equations with logistic regression to generate adjusted odd ratios (aORs) and 95% confidence intervals (CIs). Models were adjusted for geographic region, payer, race, and gender. Statistical analyses were performed by using SAS version 9.3 (SAS Institute, Cary, NC). We determined statistical significance as a 2‐tailed P value <0.05.

Febrile infants with bronchiolitis or a history of prematurity may be managed differently from full‐term febrile young infants without bronchiolitis.[6, 27] Therefore, we performed a subgroup analysis after exclusion of infants with an ICD‐9 discharge diagnosis code for bronchiolitis (466.11 and 466.19)[28] or prematurity (765).

Because our study included ED encounters in 2013, we repeated our analyses after exclusion of hospitals with CPGs implemented during the 2013 calendar year.

RESULTS

CPG by Institution

Thirty‐three (89.2%) of the 37 EDs surveyed completed the questionnaire. Overall, 21 (63.6%) of the 33 EDs had a CPG; 15 (45.5%) had a CPG for all infants 56 days of age, 5 (15.2%) had a CPG for infants 28 days only, and 1 (3.0%) had a CPG for infants 29 to 56 days but not 28 days of age (Figure 1). Seventeen EDs had an established CPG prior to 2013, and 4 hospitals implemented a CPG during the 2013 calendar year, 2 with CPGs for neonates 28 days and 2 with CPGs for both 28 days and 29 to 56 days of age. Hospitals with CPGs were more likely to be located in the Northeast and West regions of the United States and provide care to a higher proportion of non‐Hispanic white patients, as well as those with commercial insurance (Table 1).

jhm2329-fig-0001-m.png
Specific clinical practice guideline (CPG) recommendations for diagnostic testing, hospitalization, and ceftriaxone use at ED discharge by institution among the 21 institutions with a CPG. Urine testing is defined as urine dipstick, urinalysis, or urine culture; blood testing as complete blood count or blood culture, and cerebrospinal fluid (CSF) testing as cell count, culture, or procedure code for lumbar puncture. Abbreviations: ED, emergency department.
Characteristics of Patients in Hospitals With and Without CPGs for the Febrile Young Infant 56 Days of Age
Characteristic28 Days2956 Days
No CPG, n=996, N (%)CPG, n=2,149, N (%)P ValueNo CPG, n=2,460, N (%)CPG, n=3,772, N (%)P Value
  • NOTE: Abbreviations: CPG, clinical practice guideline; IQR, interquartile range; UTI, urinary tract infection. *Includes UTI/pyelonephritis, bacteremia/sepsis, bacterial meningitis, pneumonia, and bacterial enteritis. Some infants had more than 1 site of infection.

Race      
Non‐Hispanic white325 (32.6)996 (46.3) 867 (35.2)1,728 (45.8) 
Non‐Hispanic black248 (24.9)381 (17.7) 593 (24.1)670 (17.8) 
Hispanic243 (24.4)531 (24.7) 655 (26.6)986 (26.1) 
Asian28 (2.8)78 (3.6) 40 (1.6)122 (3.2) 
Other Race152 (15.3)163 (7.6)<0.001305 (12.4)266 (7.1)<0.001
Gender      
Female435 (43.7)926 (43.1)0.761,067 (43.4)1,714 (45.4)0.22
Payer      
Commercial243 (24.4)738 (34.3) 554 (22.5)1,202 (31.9) 
Government664 (66.7)1,269 (59.1) 1,798 (73.1)2,342 (62.1) 
Other payer89 (8.9)142 (6.6)<0.001108 (4.4)228 (6.0)<0.001
Region      
Northeast39 (3.9)245 (11.4) 77 (3.1)572 (15.2) 
South648 (65.1)915 (42.6) 1,662 (67.6)1,462 (38.8) 
Midwest271 (27.2)462 (21.5) 506 (20.6)851 (22.6) 
West38 (3.8)527 (24.5)<0.001215 (8.7)887 (23.5)<0.001
Serious bacterial infection      
Overall*131 (13.2)242 (11.3)0.14191 (7.8)237 (6.3)0.03
UTI/pyelonephritis73 (7.3)153 (7.1) 103 (4.2)154 (4.1) 
Bacteremia/sepsis56 (5.6)91 (4.2) 78 (3.2)61 (1.6) 
Bacterial meningitis15 (1.5)15 (0.7) 4 (0.2)14 (0.4) 
Age, d, median (IQR)18 (11, 24)18 (11, 23)0.6746 (37, 53)45 (37, 53)0.11

All 20 CPGs for the febrile young infant 28 days of age recommended urine, blood, CSF testing, and hospitalization for all infants (Figure 1). Of the 16 hospitals with CPGs for febrile infants aged 29 to 56 days, all recommended urine and blood testing for all patients, except for 1 CPG, which recommended consideration of blood testing but not to obtain routinely. Hospitals varied in recommendations for CSF testing among infants aged 29 to 56 days: 8 (50%) recommended CSF testing in all patients and 8 (50%) recommended CSF testing only if the patient was high risk per defined criteria (based on history, physical examination, urine, and blood testing). In all 16 CPGs, hospitalization was recommended only for high‐risk infants. For low‐risk infants aged 2956 days being discharged from the ED, 3 hospitals recommended ceftriaxone for all, 9 recommended consideration of ceftriaxone, and 4 recommended against antibiotics (Figure 1).

Study Patients

During the study period, there were 10,415 infants 56 days old with a diagnosis of fever at the 33 participating hospitals. After exclusion of 635 (6.1%) infants with a complex chronic condition and 445 (4.3%) transferred from another institution (including 42 with a complex chronic condition), 9377 infants remained in our study cohort. Approximately one‐third of the cohort was 28 days of age and two‐thirds aged 29 to 56 days. The overall SBI rate was 8.5% but varied by age (11.9% in infants 28 days and 6.9% in infants 29 to 56 days of age) (Table 1).

CPGs and Use of Diagnostic Testing, Hospitalization Rates, Ceftriaxone Use, and Revisits for SBI

For infants 28 days of age, the presence of a CPG was not associated with urine, blood, CSF testing, or hospitalization after multivariable adjustment (Table 2). Among infants aged 29 to 56 days, urine testing did not differ based on the presence of a CPG, whereas blood testing was performed less often at the 1 hospital whose CPG recommended to consider, but not routinely obtain, testing (aOR: 0.4, 95% CI: 0.3‐0.7, P=0.001). Compared to hospitals without a CPG, CSF testing was performed less often at hospitals with CPG recommendations to only obtain CSF if high risk (aOR: 0.5, 95% CI: 0.3‐0.8, P=0.002). However, the odds of hospitalization did not differ at institutions with and without a febrile infant CPG (aOR: 0.7, 95% CI: 0.5‐1.1, P=0.10). For infants aged 29 to 56 days discharged from the ED, ceftriaxone was administered more often at hospitals with CPGs that recommended ceftriaxone for all discharged patients (aOR: 4.6, 95% CI: 2.39.3, P<0.001) and less often at hospitals whose CPGs recommended against antibiotics (aOR: 0.3, 95% CI: 0.1‐0.9, P=0.03) (Table 3). Our findings were similar in the subgroup of infants without bronchiolitis or prematurity (see Supporting Tables 1 and 2 in the online version of this article). After exclusion of hospitals with a CPG implemented during the 2013 calendar year (4 hospitals excluded in the 28 days age group and 2 hospitals excluded in the 29 to 56 days age group), infants aged 29 to 56 days cared for at a hospital with a CPG experienced a lower odds of hospitalization (aOR: 0.7, 95% CI: 0.4‐0.98, P=0.04). Otherwise, our findings in both age groups did not materially differ from the main analyses.

Variation in Testing and Hospitalization Based on CPG‐Specific Recommendations Among Infants 28 Days of Age With Diagnosis of Fever
Testing/HospitalizationNo. of HospitalsNo. of Patients% Received*aOR (95% CI)P Value
  • NOTE: Abbreviations: aOR, adjusted odds ratio; CI, confidence interval; CPG, clinical practice guideline; CSF, cerebrospinal fluid. *Percent of infants who received test or were hospitalized. Adjusted for hospital clustering, geographic region, payer, race, and gender. Urine testing defined as urine dipstick, urinalysis, or urine culture; Blood testing defined as complete blood count or blood culture. ‖CSF testing defined as cell count, culture, or procedure code for lumbar puncture

Laboratory testing     
Urine testing     
No CPG1399675.6Ref 
CPG: recommend for all202,14980.71.2 (0.9‐1.7)0.22
Blood testing     
No CPG1399676.9Ref 
CPG: recommend for all202,14981.81.2 (0.9‐1.7)0.25
CSF testing     
No CPG1399671.0Ref 
CPG: recommend for all202,14977.51.3 (1.01.7)0.08
Disposition     
Hospitalization     
No CPG1399675.4Ref 
CPG: recommend for all202,14981.61.2 (0.9‐1.8)0.26
Variation in Testing, Hospitalization, and Ceftriaxone Use Based on CPG‐Specific Recommendations Among Infants 29 to 56 Days of Age With Diagnosis of Fever
Testing/HospitalizationNo. of HospitalsNo. of Patients% Received*aOR (95% CI)P Value
  • NOTE: Abbreviations: aOR, adjusted odds ratio; CI, confidence interval; CPG, clinical practice guideline; CSF, cerebrospinal fluid. *Percent of infants who received test, were hospitalized, or received ceftriaxone. Adjusted for hospital clustering, geographic region, payer, race, and gender. Urine testing defined as urine dipstick, urinalysis, or urine culture. Blood testing defined as complete blood count or blood culture. CSF testing defined as cell count, culture, or procedure code for lumbar puncture. For low‐risk infants discharged from the emergency department.

Laboratory resting     
Urine testing     
No CPG172,46081.1Ref 
CPG: recommend for all163,77282.10.9 (0.7‐1.4)0.76
Blood testing     
No CPG172,46079.4Ref 
CPG: recommend for all153,62882.61.1 (0.7‐1.6)0.70
CPG: recommend consider114462.50.4 (0.3‐0.7)0.001
CSF testing     
No CPG172,46046.3Ref 
CPG: recommend for all81,51770.31.3 (0.9‐1.9)0.11
CPG: recommend if high‐risk82,25539.90.5 (0.3‐0.8)0.002
Disposition     
Hospitalization     
No CPG172,46047.0Ref 
CPG: recommend if high‐risk163,77242.00.7 (0.5‐1.1)0.10
Ceftriaxone if discharged     
No CPG171,30411.7Ref 
CPG: recommend against431310.90.3 (0.1‐0.9)0.03
CPG: recommend consider91,56714.41.5 (0.9‐2.4)0.09
CPG: recommend for all330664.14.6 (2.39.3)< 0.001

Three‐day revisits for SBI were similarly low at hospitals with and without CPGs among infants 28 days (1.5% vs 0.8%, P=0.44) and 29 to 56 days of age (1.4% vs 1.1%, P=0.44) and did not differ after exclusion of hospitals with a CPG implemented in 2013.

CPGs and Costs

Among infants 28 days of age, costs per visit did not differ for admitted and discharged patients based on CPG presence. The presence of an ED febrile infant CPG was associated with higher costs for both admitted and discharged infants 29 to 56 days of age (Table 4). The cost analysis did not significantly differ after exclusion of hospitals with CPGs implemented in 2013.

Costs per Visit for Febrile Young Infants 56 Days of Age at Institutions With and Without CPGs
 28 Days, Cost, Median (IQR)29 to 56 Days, Cost, Median (IQR)
No CPGCPGP ValueNo CPGCPGP Value
  • NOTE: Abbreviations: CPG, clinical practice guideline; IQR, interquartile range.

Admitted$4,979 ($3,408$6,607) [n=751]$4,715 ($3,472$6,526) [n=1,753]0.79$3,756 ($2,725$5,041) [n=1,156]$3,923 ($3,077$5,243) [n=1,586]<0.001
Discharged$298 ($166$510) [n=245]$231 ($160$464) [n=396]0.10$681($398$982) [n=1,304)]$764 ($412$1,100) [n=2,186]<0.001

DISCUSSION

We described the content and association of CPGs with management of the febrile infant 56 days of age across a large sample of children's hospitals. Nearly two‐thirds of included pediatric EDs have a CPG for the management of young febrile infants. Management of febrile infants 28 days was uniform, with a majority hospitalized after urine, blood, and CSF testing regardless of the presence of a CPG. In contrast, CPGs for infants 29 to 56 days of age varied in their recommendations for CSF testing as well as ceftriaxone use for infants discharged from the ED. Consequently, we observed considerable hospital variability in CSF testing and ceftriaxone use for discharged infants, which correlates with variation in the presence and content of CPGs. Institutional CPGs may be a source of the across‐hospital variation in care of febrile young infants observed in prior study.[9]

Febrile infants 28 days of age are at particularly high risk for SBI, with a prevalence of nearly 20% or higher.[2, 3, 29] The high prevalence of SBI, combined with the inherent difficulty in distinguishing neonates with and without SBI,[2, 30] has resulted in uniform CPG recommendations to perform the full‐sepsis workup in this young age group. Similar to prior studies,[8, 9] we observed that most febrile infants 28 days undergo the full sepsis evaluation, including CSF testing, and are hospitalized regardless of the presence of a CPG.

However, given the conflicting recommendations for febrile infants 29 to 56 days of age,[4, 5, 6] the optimal management strategy is less certain.[7] The Rochester, Philadelphia, and Boston criteria, 3 published models to identify infants at low risk for SBI, primarily differ in their recommendations for CSF testing and ceftriaxone use in this age group.[4, 5, 6] Half of the CPGs recommended CSF testing for all febrile infants, and half recommended CSF testing only if the infant was high risk. Institutional guidelines that recommended selective CSF testing for febrile infants aged 29 to 56 days were associated with lower rates of CSF testing. Furthermore, ceftriaxone use varied based on CPG recommendations for low‐risk infants discharged from the ED. Therefore, the influence of febrile infant CPGs mainly relates to the limiting of CSF testing and targeted ceftriaxone use in low‐risk infants. As the rate of return visits for SBI is low across hospitals, future study should assess outcomes at hospitals with CPGs recommending selective CSF testing. Of note, infants 29 to 56 days of age were less likely to be hospitalized when cared for at a hospital with an established CPG prior to 2013 without increase in 3‐day revisits for SBI. This finding may indicate that longer duration of CPG implementation is associated with lower rates of hospitalization for low‐risk infants; this finding merits further study.

The presence of a CPG was not associated with lower costs for febrile infants in either age group. Although individual healthcare systems have achieved lower costs with CPG implementation,[12] the mere presence of a CPG is not associated with lower costs when assessed across institutions. Higher costs for admitted and discharged infants 29 to 56 days of age in the presence of a CPG likely reflects the higher rate of CSF testing at hospitals whose CPGs recommend testing for all febrile infants, as well as inpatient management strategies for hospitalized infants not captured in our study. Future investigation should include an assessment of the cost‐effectiveness of the various testing and treatment strategies employed for the febrile young infant.

Our study has several limitations. First, the validity of ICD‐9 diagnosis codes for identifying young infants with fever is not well established, and thus our study is subject to misclassification bias. To minimize missed patients, we included infants with either an ICD‐9 admission or discharge diagnosis of fever; however, utilization of diagnosis codes for patient identification may have resulted in undercapture of infants with a measured temperature of 38.0C. It is also possible that some patients who did not undergo testing were misclassified as having a fever or had temperatures below standard thresholds to prompt diagnostic testing. This is a potential reason that testing was not performed in 100% of infants, even at hospitals with CPGs that recommended testing for all patients. Additionally, some febrile infants diagnosed with SBI may not have an associated ICD‐9 diagnosis code for fever. Although the overall SBI rate observed in our study was similar to prior studies,[4, 31] the rate in neonates 28 days of age was lower than reported in recent investigations,[2, 3] which may indicate inclusion of a higher proportion of low‐risk febrile infants. With the exception of bronchiolitis, we also did not assess diagnostic testing in the presence of other identified sources of infection such as herpes simplex virus.

Second, we were unable to assess the presence or absence of a CPG at the 4 excluded EDs that did not respond to the survey or the institutions excluded for data‐quality issues. However, included and excluded hospitals did not differ in region or annual ED volume (data not shown).

Third, although we classified hospitals based upon the presence and content of CPGs, we were unable to fully evaluate adherence to the CPG at each site.

Last, though PHIS hospitals represent 85% of freestanding children's hospitals, many febrile infants are hospitalized at non‐PHIS institutions; our results may not be generalizable to care provided at nonchildren's hospitals.

CONCLUSIONS

Management of febrile neonates 28 days of age does not vary based on CPG presence. However, CPGs for the febrile infant aged 29 to 56 days vary in recommendations for CSF testing as well as ceftriaxone use for low‐risk patients, which significantly contributes to practice variation and healthcare costs across institutions.

Acknowledgements

The Febrile Young Infant Research Collaborative includes the following additional investigators who are acknowledged for their work on this study: Kao‐Ping Chua, MD, Harvard PhD Program in Health Policy, Harvard University, Cambridge, Massachusetts, and Division of Emergency Medicine, Department of Pediatrics, Boston Children's Hospital, Boston, Massachusetts; Elana A. Feldman, BA, University of Washington School of Medicine, Seattle, Washington; and Katie L. Hayes, BS, Division of Emergency Medicine, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania.

Disclosures

This project was funded in part by The Gerber Foundation Novice Researcher Award (Ref #18273835). Dr. Fran Balamuth received career development support from the National Institutes of Health (NHLBI K12‐HL109009). Funders were not involved in design or conduct of the study; collection, management, analysis, or interpretation of the data; preparation, review, or approval of the manuscript. The authors have no financial relationships relevant to this article to disclose. No payment was received for the production of this article. The authors have no conflicts of interest relevant to this article to disclose.

References
  1. Huppler AR, Eickhoff JC, Wald ER. Performance of low‐risk criteria in the evaluation of young infants with fever: review of the literature. Pediatrics. 2010;125:228233.
  2. Schwartz S, Raveh D, Toker O, Segal G, Godovitch N, Schlesinger Y. A week‐by‐week analysis of the low‐risk criteria for serious bacterial infection in febrile neonates. Arch Dis Child. 2009;94:287292.
  3. Garcia S, Mintegi S, Gomez B, et al. Is 15 days an appropriate cut‐off age for considering serious bacterial infection in the management of febrile infants? Pediatr Infect Dis J. 2012;31:455458.
  4. Baker MD, Bell LM, Avner JR. Outpatient management without antibiotics of fever in selected infants. N Engl J Med. 1993;329:14371441.
  5. Baskin MN, Fleisher GR, O'Rourke EJ. Identifying febrile infants at risk for a serious bacterial infection. J Pediatr. 1993;123:489490.
  6. Jaskiewicz JA, McCarthy CA, Richardson AC, et al. Febrile infants at low risk for serious bacterial infection—an appraisal of the Rochester criteria and implications for management. Febrile Infant Collaborative Study Group. Pediatrics. 1994;94:390396.
  7. American College of Emergency Physicians Clinical Policies Committee; American College of Emergency Physicians Clinical Policies Subcommittee on Pediatric Fever. Clinical policy for children younger than three years presenting to the emergency department with fever. Ann Emerg Med. 2003;42:530545.
  8. Jain S, Cheng J, Alpern ER, et al. Management of febrile neonates in US pediatric emergency departments. Pediatrics. 2014;133:187195.
  9. Aronson PL, Thurm C, Alpern ER, et al. Variation in care of the febrile young infant <90 days in US pediatric emergency departments. Pediatrics. 2014;134:667677.
  10. Yarden‐Bilavsky H, Ashkenazi S, Amir J, Schlesinger Y, Bilavsky E. Fever survey highlights significant variations in how infants aged ≤60 days are evaluated and underline the need for guidelines. Acta Paediatr. 2014;103:379385.
  11. Bergman DA. Evidence‐based guidelines and critical pathways for quality improvement. Pediatrics. 1999;103:225232.
  12. Byington CL, Reynolds CC, Korgenski K, et al. Costs and infant outcomes after implementation of a care process model for febrile infants. Pediatrics. 2012;130:e16e24.
  13. Harris PA, Taylor R, Thielke R, Payne J, Gonzalez N, Conde JG. Research electronic data capture (REDCap)—a metadata‐driven methodology and workflow process for providing translational research informatics support. J Biomed Inform. 2009;42:377381.
  14. Wood JN, Feudtner C, Medina SP, Luan X, Localio R, Rubin DM. Variation in occult injury screening for children with suspected abuse in selected US children's hospitals. Pediatrics. 2012;130:853860.
  15. Fletcher DM. Achieving data quality. How data from a pediatric health information system earns the trust of its users. J AHIMA. 2004;75:2226.
  16. Mongelluzzo J, Mohamad Z, Have TR, Shah SS. Corticosteroids and mortality in children with bacterial meningitis. JAMA. 2008;299:20482055.
  17. Kharbanda AB, Hall M, Shah SS, et al. Variation in resource utilization across a national sample of pediatric emergency departments. J Pediatr. 2013;163:230236.
  18. Feudtner C, Hays RM, Haynes G, Geyer JR, Neff JM, Koepsell TD. Deaths attributed to pediatric complex chronic conditions: national trends and implications for supportive care services. Pediatrics. 2001;107:E99.
  19. US Census Bureau. Geographic terms and concepts—census divisions and census regions. Available at: https://www.census.gov/geo/reference/gtc/gtc_census_divreg.html. Accessed September 10, 2014.
  20. Macy ML, Hall M, Shah SS, et al. Pediatric observation status: are we overlooking a growing population in children's hospitals? J Hosp Med. 2012;7:530536.
  21. Macy ML, Hall M, Shah SS, et al. Differences in designations of observation care in US freestanding children's hospitals: are they virtual or real? J Hosp Med. 2012;7:287293.
  22. Tieder JS, Hall M, Auger KA, et al. Accuracy of administrative billing codes to detect urinary tract infection hospitalizations. Pediatrics. 2011;128:323330.
  23. Williams DJ, Shah SS, Myers A, et al. Identifying pediatric community‐acquired pneumonia hospitalizations: accuracy of administrative billing codes. JAMA Pediatr. 2013;167:851858.
  24. Gordon JA, An LC, Hayward RA, Williams BC. Initial emergency department diagnosis and return visits: risk versus perception. Ann Emerg Med. 1998;32:569573.
  25. Cho CS, Shapiro DJ, Cabana MD, Maselli JH, Hersh AL. A national depiction of children with return visits to the emergency department within 72 hours, 2001–2007. Pediatr Emerg Care. 2012;28:606610.
  26. Healthcare Cost and Utilization Project. Cost‐to‐charge ratio files. Available at: http://www.hcup‐us.ahrq.gov/db/state/costtocharge.jsp. Accessed September 11, 2014.
  27. Levine DA, Platt SL, Dayan PS, et al. Risk of serious bacterial infection in young febrile infants with respiratory syncytial virus infections. Pediatrics. 2004;113:17281734.
  28. Parikh K, Hall M, Mittal V, et al. Establishing benchmarks for the hospitalized care of children with asthma, bronchiolitis, and pneumonia. Pediatrics. 2014;134:555562.
  29. Mintegi S, Benito J, Astobiza E, Capape S, Gomez B, Eguireun A. Well appearing young infants with fever without known source in the emergency department: are lumbar punctures always necessary? Eur J Emerg Med. 2010;17:167169.
  30. Baker MD, Bell LM. Unpredictability of serious bacterial illness in febrile infants from birth to 1 month of age. Arch Pediatr Adolesc Med. 1999;153:508511.
  31. Pantell RH, Newman TB, Bernzweig J, et al. Management and outcomes of care of fever in early infancy. JAMA. 2004;291:12031212.
References
  1. Huppler AR, Eickhoff JC, Wald ER. Performance of low‐risk criteria in the evaluation of young infants with fever: review of the literature. Pediatrics. 2010;125:228233.
  2. Schwartz S, Raveh D, Toker O, Segal G, Godovitch N, Schlesinger Y. A week‐by‐week analysis of the low‐risk criteria for serious bacterial infection in febrile neonates. Arch Dis Child. 2009;94:287292.
  3. Garcia S, Mintegi S, Gomez B, et al. Is 15 days an appropriate cut‐off age for considering serious bacterial infection in the management of febrile infants? Pediatr Infect Dis J. 2012;31:455458.
  4. Baker MD, Bell LM, Avner JR. Outpatient management without antibiotics of fever in selected infants. N Engl J Med. 1993;329:14371441.
  5. Baskin MN, Fleisher GR, O'Rourke EJ. Identifying febrile infants at risk for a serious bacterial infection. J Pediatr. 1993;123:489490.
  6. Jaskiewicz JA, McCarthy CA, Richardson AC, et al. Febrile infants at low risk for serious bacterial infection—an appraisal of the Rochester criteria and implications for management. Febrile Infant Collaborative Study Group. Pediatrics. 1994;94:390396.
  7. American College of Emergency Physicians Clinical Policies Committee; American College of Emergency Physicians Clinical Policies Subcommittee on Pediatric Fever. Clinical policy for children younger than three years presenting to the emergency department with fever. Ann Emerg Med. 2003;42:530545.
  8. Jain S, Cheng J, Alpern ER, et al. Management of febrile neonates in US pediatric emergency departments. Pediatrics. 2014;133:187195.
  9. Aronson PL, Thurm C, Alpern ER, et al. Variation in care of the febrile young infant <90 days in US pediatric emergency departments. Pediatrics. 2014;134:667677.
  10. Yarden‐Bilavsky H, Ashkenazi S, Amir J, Schlesinger Y, Bilavsky E. Fever survey highlights significant variations in how infants aged ≤60 days are evaluated and underline the need for guidelines. Acta Paediatr. 2014;103:379385.
  11. Bergman DA. Evidence‐based guidelines and critical pathways for quality improvement. Pediatrics. 1999;103:225232.
  12. Byington CL, Reynolds CC, Korgenski K, et al. Costs and infant outcomes after implementation of a care process model for febrile infants. Pediatrics. 2012;130:e16e24.
  13. Harris PA, Taylor R, Thielke R, Payne J, Gonzalez N, Conde JG. Research electronic data capture (REDCap)—a metadata‐driven methodology and workflow process for providing translational research informatics support. J Biomed Inform. 2009;42:377381.
  14. Wood JN, Feudtner C, Medina SP, Luan X, Localio R, Rubin DM. Variation in occult injury screening for children with suspected abuse in selected US children's hospitals. Pediatrics. 2012;130:853860.
  15. Fletcher DM. Achieving data quality. How data from a pediatric health information system earns the trust of its users. J AHIMA. 2004;75:2226.
  16. Mongelluzzo J, Mohamad Z, Have TR, Shah SS. Corticosteroids and mortality in children with bacterial meningitis. JAMA. 2008;299:20482055.
  17. Kharbanda AB, Hall M, Shah SS, et al. Variation in resource utilization across a national sample of pediatric emergency departments. J Pediatr. 2013;163:230236.
  18. Feudtner C, Hays RM, Haynes G, Geyer JR, Neff JM, Koepsell TD. Deaths attributed to pediatric complex chronic conditions: national trends and implications for supportive care services. Pediatrics. 2001;107:E99.
  19. US Census Bureau. Geographic terms and concepts—census divisions and census regions. Available at: https://www.census.gov/geo/reference/gtc/gtc_census_divreg.html. Accessed September 10, 2014.
  20. Macy ML, Hall M, Shah SS, et al. Pediatric observation status: are we overlooking a growing population in children's hospitals? J Hosp Med. 2012;7:530536.
  21. Macy ML, Hall M, Shah SS, et al. Differences in designations of observation care in US freestanding children's hospitals: are they virtual or real? J Hosp Med. 2012;7:287293.
  22. Tieder JS, Hall M, Auger KA, et al. Accuracy of administrative billing codes to detect urinary tract infection hospitalizations. Pediatrics. 2011;128:323330.
  23. Williams DJ, Shah SS, Myers A, et al. Identifying pediatric community‐acquired pneumonia hospitalizations: accuracy of administrative billing codes. JAMA Pediatr. 2013;167:851858.
  24. Gordon JA, An LC, Hayward RA, Williams BC. Initial emergency department diagnosis and return visits: risk versus perception. Ann Emerg Med. 1998;32:569573.
  25. Cho CS, Shapiro DJ, Cabana MD, Maselli JH, Hersh AL. A national depiction of children with return visits to the emergency department within 72 hours, 2001–2007. Pediatr Emerg Care. 2012;28:606610.
  26. Healthcare Cost and Utilization Project. Cost‐to‐charge ratio files. Available at: http://www.hcup‐us.ahrq.gov/db/state/costtocharge.jsp. Accessed September 11, 2014.
  27. Levine DA, Platt SL, Dayan PS, et al. Risk of serious bacterial infection in young febrile infants with respiratory syncytial virus infections. Pediatrics. 2004;113:17281734.
  28. Parikh K, Hall M, Mittal V, et al. Establishing benchmarks for the hospitalized care of children with asthma, bronchiolitis, and pneumonia. Pediatrics. 2014;134:555562.
  29. Mintegi S, Benito J, Astobiza E, Capape S, Gomez B, Eguireun A. Well appearing young infants with fever without known source in the emergency department: are lumbar punctures always necessary? Eur J Emerg Med. 2010;17:167169.
  30. Baker MD, Bell LM. Unpredictability of serious bacterial illness in febrile infants from birth to 1 month of age. Arch Pediatr Adolesc Med. 1999;153:508511.
  31. Pantell RH, Newman TB, Bernzweig J, et al. Management and outcomes of care of fever in early infancy. JAMA. 2004;291:12031212.
Issue
Journal of Hospital Medicine - 10(6)
Issue
Journal of Hospital Medicine - 10(6)
Page Number
358-365
Page Number
358-365
Publications
Publications
Article Type
Display Headline
Association of clinical practice guidelines with emergency department management of febrile infants ≤56 days of age
Display Headline
Association of clinical practice guidelines with emergency department management of febrile infants ≤56 days of age
Sections
Article Source

© 2015 Society of Hospital Medicine

Disallow All Ads
Correspondence Location
Address for correspondence and reprint requests: Paul L. Aronson, MD, Section of Pediatric Emergency Medicine, Yale School of Medicine, 100 York Street, Suite 1F, New Haven, CT 06511; Telephone: 203–737‐7443; Fax: 203–737‐7447; E‐mail: paul.aronson@yale.edu
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Article PDF Media
Image
Disable zoom
Off
Media Files
Image
Disable zoom
Off
Image
Disable zoom
Off
Image
Disable zoom
Off

Return Visits to Pediatric EDs

Article Type
Changed
Sun, 05/21/2017 - 13:39
Display Headline
Prevalence and predictors of return visits to pediatric emergency departments

Returns to the hospital following recent encounters, such as an admission to the inpatient unit or evaluation in an emergency department (ED), may reflect the natural progression of a disease, the quality of care received during the initial admission or visit, or the quality of the underlying healthcare system.[1, 2, 3, 4, 5, 6, 7, 8, 9, 10] Although national attention has focused on hospital readmissions,[3, 4, 5, 6, 7, 11, 12] ED revisits are a source of concern to emergency physicians.[8, 9] Some ED revisits are medically necessary, but revisits that may be managed in the primary care setting contribute to ED crowding, can be stressful to patients and providers, and increase healthcare costs.[10, 11, 12] Approximately 27 million annual ED visits are made by children, accounting for over one‐quarter of all ED visits in the United States, with a reported ED revisit rate of 2.5% to 5.2%.[2, 13, 14, 15, 16, 17, 18, 19, 20] Improved understanding of the patient‐level or visit‐level factors associated with ED revisits may provide an opportunity to enhance disposition decision making at the index visit and optimize site of and communication around follow‐up care.

Previous studies on ED revisits have largely been conducted in single centers and have used variable visit intervals ranging between 48 hours and 30 days.[2, 13, 16, 18, 21, 22, 23, 24, 25] Two national studies used the National Hospital Ambulatory Medical Care Survey, which includes data from both general and pediatric EDs.[13, 14] Factors identified to be associated with increased odds of returning were: young age, higher acuity, chronic conditions, and public insurance. One national study identified some diagnoses associated with higher likelihood of returning,[13] whereas the other focused primarily on infectious diseaserelated diagnoses.[14]

The purpose of this study was to describe the prevalence of return visits specifically to pediatric EDs and to investigate patient‐level, visit‐level, and healthcare systemrelated factors that may be associated with return visits and hospitalization at return.

METHODS

Study Design and Data Source

This retrospective cohort study used data from the Pediatric Health Information System (PHIS), an administrative database with data from 44 tertiary care pediatric hospitals in 27 US states and the District of Columbia. This database contains patient demographics, diagnoses, and procedures as well as medications, diagnostic imaging, laboratory, and supply charges for each patient. Data are deidentified prior to inclusion; encrypted medical record numbers allow for the identification of individual patients across all ED visits and hospitalizations to the same hospital. The Children's Hospital Association (Overland Park, KS) and participating hospitals jointly assure the quality and integrity of the data. This study was approved by the institutional review board at Boston Children's Hospital with a waiver for informed consent granted.

Study Population and Protocol

To standardize comparisons across the hospitals, we included data from 23 of the 44 hospitals in PHIS; 7 were excluded for not including ED‐specific data. For institutions that collect information from multiple hospitals within their healthcare system, we included only records from the main campus or children's hospital when possible, leading to the exclusion of 9 hospitals where the data were not able to be segregated. As an additional level of data validation, we compared the hospital‐level ED volume and admission rates as reported in the PHIS to those reported to a separate database (the Pediatric Analysis and Comparison Tool). We further excluded 5 hospitals whose volume differed by >10% between these 2 data sources.

Patients <18 years of age who were discharged from these EDs following their index visit in 2012 formed the eligible cohort.

Key Outcome Measures

The primary outcomes were return visits within 72 hours of discharge from the ED, and return visits resulting in hospitalization, including observation status. We defined an ED revisit as a return within 72 hours of ED discharge regardless of whether the patient was subsequently discharged from the ED on the return visit or hospitalized. We assessed revisits within 72 hours of an index ED discharge, because return visits within this time frame are likely to be related to the index visit.[2, 13, 16, 21, 22, 24, 25, 26]

Factors Associated With ED Revisits

A priori, we chose to adjust for the following patient‐level factors: age (<30 days, 30 days<1 year, 14 years, 511 years, 1217 years), gender, and socioeconomic status (SES) measured as the zip codebased median household income, obtained from the 2010 US Census, with respect to the federal poverty level (FPL) (<1.5 FPL, 1.52 FPL, 23 FPL, and >3 FPL).[27] We also adjusted for insurance type (commercial, government, or other), proximity of patient's home zip code to hospital (modeled as the natural log of the geographical distance to patient's home address from the hospital), ED diagnosis‐based severity classification system score (1=low severity, 5=high severity),[28] presence of a complex chronic condition at the index or prior visits using a validated classification scheme,[15, 29, 30, 31] and primary care physician (PCP) density per 100,000 in the patient's residential area (modeled as quartiles: very low, <57.2; low, 57.267.9; medium, 68.078.7; high, >78.8). PCP density, defined by the Dartmouth Atlas of Health Care,[32, 33, 34] is the number of primary care physicians per 100,000 residents (PCP count) in federal health service areas (HSA). Patients were assigned to a corresponding HSA based on their home zip code.

Visit‐level factors included arrival time of index visit (8:01 am 4:00 pm, 4:01 pm12:00 am, 12:01 am8 am representing day, evening, and overnight arrival, respectively), day of the week, season, length of stay (LOS) in the ED during the index visit, and ED crowding (calculated as the average daily LOS/yearly average LOS for the individual ED).[35] We categorized the ED primary diagnosis for each visit using the major diagnosis groupings of a previously described pediatric ED‐specific classification scheme.[36] Using International Classification of Diseases, Ninth Revision (ICD‐9) codes, we identified the conditions with the highest ED revisit rates.

Statistical Analyses

Categorical variables describing the study cohort were summarized using frequencies and percentages. Continuous variables were summarized using mean, median, and interquartile range values, where appropriate. We used 2 different hierarchical logistic regression models to assess revisit rates by patient‐ and visit‐level characteristics. The initial model included all patients discharged from the ED following the index visit and assessed for the outcome of a revisit within 72 hours. The second model considered only patients who returned within 72 hours of an index visit and assessed for hospitalization on that return visit. We used generalized linear mixed effects models, with hospital as a random effect to account for the presence of correlated data (within hospitals), nonconstant variability (across hospitals), and binary responses. Adjusted odds ratios with 95% confidence intervals were used as summary measures of the effect of the individual adjusters. Adjusters were missing in fewer than 5% of patients across participating hospitals. Statistical analyses were performed using SAS version 9.3 (SAS Institute Inc., Cary, NC); 2‐sided P values <0.004 were considered statistically significant to account for multiple comparisons (Bonferroni‐adjusted level of significance=0.0038).

RESULTS

Patients

A total of 1,610,201 patients <18 years of age evaluated across the 23 PHIS EDs in 2012 were included in the study. Twenty‐one of the 23 EDs have academic affiliations; 10 are located in the South, 6 in the Midwest, 5 in the West, and 2 in the Northeast region of the United States. The annual ED volume for these EDs ranged from 25,090 to 136,160 (median, 65,075; interquartile range, 45,28085,206). Of the total patients, 1,415,721 (87.9%) were discharged following the index visit and comprised our study cohort. Of these patients, 47,294 (revisit rate: 3.3%) had an ED revisit within 72 hours. There were 4015 patients (0.3%) who returned more than once within 72 hours, and the largest proportion of these returned with infection‐related conditions. Of those returning, 37,999 (80.3%) were discharged again, whereas 9295 (19.7%) were admitted to the hospital (Figure 1). The demographic and clinical characteristics of study participants are displayed in Table 1.

jhm2273-fig-0001-m.png
Patient disposition from the emergency departments of study hospitals (n = 23) in 2012.
Characteristics of Patients Who Returned Within 72 Hours of ED Discharge to the Study EDs
 Index Visit, n=1,415,721, n (%)Return Visits Within 72 Hours of Discharge, n=47,294, 3.3%
Return to Discharge, n (%)Return to Admission, n (%)
  • NOTE: Abbreviations: CCC, complex chronic condition; ED, emergency department; FPL, federal poverty level; IQR, interquartile range; LOS, length of stay.

  • Socioeconomic status is relative to the federal poverty level for a family of 4.

Gender, female659,417 (46.6)17,665 (46.5)4,304 (46.3)
Payor   
Commercial379,403 (26.8)8,388 (22.1)3,214 (34.6)
Government925,147 (65.4)26,880 (70.7)5,786 (62.3)
Other111,171 (7.9)2,731 (7.2)295 (3.2)
Age   
<30 days19,217 (1.4)488 (1.3)253 (2.7)
30 days to <1 year216,967 (15.3)8,280 (21.8)2,372 (25.5)
1 year to 4 years547,083 (38.6)15,542 (40.9)3,187 (34.3)
5 years to 11 years409,463 (28.9)8,906 (23.4)1,964 (21.1)
12 years to 17 years222,991 (15.8)4,783 (12.6)1,519 (16.3)
Socioeconomic statusa   
<1.5 times FPL493,770 (34.9)13,851 (36.5)2,879 (31.0)
1.5 to 2 times FPL455,490 (32.2)12,364 (32.5)2,904 (31.2)
2 to 3 times FPL367,557 (26.0)9,560 (25.2)2,714 (29.2)
>3 times FPL98,904 (7.0)2,224 (5.9)798 (8.6)
Primary care physician density per 100,000 patients   
Very low351,798 (24.9)8,727 (23.0)2,628 (28.3)
Low357,099 (25.2)9,810 (25.8)2,067 (22.2)
Medium347,995 (24.6)10,186 (26.8)2,035 (21.9)
High358,829 (25.4)9,276 (24.4)2,565 (27.6)
CCC present, yes125,774 (8.9)4,446 (11.7)2,825 (30.4)
Severity score   
Low severity (0,1,2)721,061 (50.9)17,310 (45.6)2,955 (31.8)
High severity (3,4,5)694,660 (49.1)20,689 (54.5)6,340 (68.2)
Time of arrival   
Day533,328 (37.7)13,449 (35.4)3,396 (36.5)
Evening684,873 (48.4)18,417 (48.5)4,378 (47.1)
Overnight197,520 (14.0)6,133 (16.1)1,521 (16.4)
Season   
Winter384,957 (27.2)10,603 (27.9)2,844 (30.6)
Spring367,434 (26.0)9,923 (26.1)2,311 (24.9)
Summer303,872 (21.5)8,308 (21.9)1,875 (20.2)
Fall359,458 (25.4)9,165 (24.1)2,265 (24.4)
Weekday/weekend   
Monday217,774 (15.4)5,646 (14.9)1,394 (15)
Tuesday198,220 (14.0)5,054 (13.3)1,316 (14.2)
Wednesday194,295 (13.7)4,985 (13.1)1,333 (14.3)
Thursday191,950 (13.6)5,123 (13.5)1,234 (13.3)
Friday190,022 (13.4)5,449 (14.3)1,228 (13.2)
Saturday202,247 (14.3)5,766 (15.2)1,364 (14.7)
Sunday221,213 (15.6)5,976 (15.7)1,426 (15.3)
Distance from hospital in miles, median (IQR)8.3 (4.614.9)9.2 (4.917.4)8.3 (4.614.9)
ED crowding score at index visit, median (IQR)1.0 (0.91.1)1.0 (0.91.1)1.0 (0.91.1)
ED LOS in hours at index visit, median (IQR)2.0 (1.03.0)3.0 (2.05.0)2.0 (1.03.0)

ED Revisit Rates and Revisits Resulting in Admission

In multivariate analyses, compared to patients who did not return to the ED, patients who returned within 72 hours of discharge had higher odds of revisit if they had the following characteristics: a chronic condition, were <1 year old, a higher severity score, and public insurance. Visit‐level factors associated with higher odds of revisits included arrival for the index visit during the evening or overnight shift or on a Friday or Saturday, index visit during times of lower ED crowding, and living closer to the hospital. On return, patients were more likely to be hospitalized if they had a higher severity score, a chronic condition, private insurance, or were <30 days old. Visit‐level factors associated with higher odds of hospitalization at revisit included an index visit during the evening and overnight shift and living further from the hospital. Although the median SES and PCP density of a patient's area of residence were not associated with greater likelihood of returning, when they returned, patients residing in an area with a lower SES and higher PCP densities (>78.8 PCPs/100,000) had lower odds of being admitted to the hospital. Patients whose index visit was on a Sunday also had lower odds of being hospitalized upon return (Table 2).

Multivariate Analyses of Factors Associated With ED Revisits and Admission at Return
CharacteristicAdjusted OR of 72‐Hour Revisit (95% CI), n=1,380,723P ValueAdjusted OR of 72‐Hour Revisit Admissions (95% CI), n=46,364P Value
  • NOTE: Effects of continuous variables are assessed as 1‐unit offsets from the mean. Abbreviations: CCC, complex chronic condition; CI, confidence interval; ED, emergency department; FPL, federal poverty level; LOS, length of stay; OR, odds ratio, NA, not applicable.

  • Socioeconomic status is relative to the FPL for a family of 4.

  • ED crowding score and LOS are based on index visit. ED crowding score is calculated as the daily LOS (in hours)/overall LOS (in hours). Overall average across hospitals=1; a 1‐ unit increase translates into twice the duration for the daily LOS over the yearly average ED LOS.

  • Modeled as the natural log of the patient geographic distance from the hospital based on zip codes. Number in parentheses represents the exponential of the modeled variable.

Gender    
Male0.99 (0.971.01)0.28091.02 (0.971.07)0.5179
FemaleReference Reference 
Payor    
Government1.14 (1.111.17)<0.00010.68 (0.640.72)<0.0001
Other0.97 (0.921.01)0.11480.33 (0.280.39)<0.0001
PrivateReference Reference 
Age group    
30 days to <1 year1.32 (1.221.42)<0.00010.58 (0.490.69)<0.0001
1 year to 5 years0.89 (0.830.96)0.0030.41 (0.340.48)<0.0001
5 years to 11 years0.69 (0.640.74)<0.00010.40 (0.330.48)<0.0001
12 years to 17 years0.72 (0.660.77)<0.00010.50 (0.420.60)<0.0001
<30 daysReference Reference 
Socioeconomic statusa    
% <1.5 times FPL0.96 (0.921.01)0.09920.82 (0.740.92)0.0005
% 1.5 to 2 times FPL0.98 (0.941.02)0.29920.83 (0.750.92)0.0005
% 2 to 3 times FPL1.02 (0.981.07)0.2920.88 (0.790.97)0.01
% >3 times FPLReference Reference 
Severity score    
High severity, 4, 5, 61.43 (1.401.45)<0.00013.42 (3.233.62)<0.0001
Low severity, 1, 2, 3Reference Reference 
Presence of any CCC    
Yes1.90 (1.861.96)<0.00012.92 (2.753.10)<0.0001
NoReference Reference 
Time of arrival    
Evening1.05 (1.031.08)<0.00011.37 (1.291.44)<0.0001
Overnight1.19 (1.151.22)<0.00011.84 (1.711.97)<0.0001
DayReference Reference 
Season    
Winter1.09 (1.061.11)<0.00011.06 (0.991.14)0.0722
Spring1.07 (1.041.10)<0.00010.98 (0.911.046)0.4763
Summer1.05 (1.021.08)0.00110.93 (0.871.01)0.0729
FallReference Reference 
Weekday/weekend    
Thursday1.02 (0.9821.055)0.32970.983 (0.8971.078)0.7185
Friday1.08 (1.041.11)<0.00011.03 (0.941.13)0.5832
Saturday1.08 (1.041.12)<0.00010.89 (0.810.97)0.0112
Sunday1.02 (0.991.06)0.20540.81 (0.740.89)<0.0001
Monday1.00 (0.961.03)0.89280.98 (0.901.07)0.6647
Tuesday0.99 (0.951.03)0.53420.93 (0.851.02)0.1417
WednesdayReference Reference 
PCP ratio per 100,000 patients    
57.267.91.00 (0.961.04)0.88440.93 (0.841.03)0.1669
68.078.71.00 (0.951.04)0.81560.86 (0.770.96)0.0066
>78.81.00 (0.951.04)0.68830.82 (0.730.92)0.001
<57.2Reference Reference 
ED crowding score at index visitb    
20.92 (0.900.95)<0.00010.96 (0.881.05)0.3435
1Reference Reference 
Distance from hospitalc    
3.168, 23.6 miles0.95 (0.940.96)<0.00011.16 (1.121.19)<0.0001
2.168, 8.7 milesReference Reference 
ED LOS at index visitb    
3.7 hours1.003 (1.0011.005)0.0052NA 
2.7 hoursReference   

Diagnoses Associated With Return Visits

Patients with index visit diagnoses of sickle cell disease and leukemia had the highest proportion of return visits (10.7% and 7.3%, respectively). Other conditions with high revisit rates included infectious diseases such as cellulitis, bronchiolitis, and gastroenteritis. Patients with other chronic diseases such as diabetes and with devices, such as gastrostomy tubes, also had high rates of return visits. At return, the rate of hospitalization for these conditions ranged from a 1‐in‐6 chance of hospitalization for the diagnoses of a fever to a 1‐in‐2 chance of hospitalization for patients with sickle cell anemia (Table 3).

Major Diagnostic Subgroups With the Highest ED Revisit and Admission at Return Rates
Major Diagnostic SubgroupNo. of Index ED Visit Dischargesa72‐Hour Revisit, % (95% CI)Admitted on Return, % (95% CI)
  • NOTE: Abbreviations: CI, confidence interval; ED, emergency department; NOS, not otherwise specified.

  • Diagnoses with <500 index visits (ie, <2 visits per month across the 23 hospitals) or <30 revisits within entire study cohort excluded from analyses.

  • Most prevalent diagnoses as identified by International Classification of Diseases, Ninth Revision codes within specified major diagnostic subgroups: devices and complications of the circulatory system, complication of other vascular device, implant, and graft; other hematologic diseases, anemia NOS, neutropenia NOS, or thrombocytopenia NOS; other devices and complications, hemorrhage complicating a procedure; devices and complications of the gastrointestinal system, gastrostomy; other infectious diseases, perinatal infections.

Sickle cell anemia2,53110.7 (9.511.9)49.6 (43.755.6)
Neoplastic diseases, cancer5367.3 (5.19.5)36 (2151)
Infectious gastrointestinal diseases8027.2 (5.49.0)21 (1031)
Devices and complications of the circulatory systemb1,0336.9 (5.38.4)45 (3457)
Other hematologic diseasesb1,5386.1 (4.97.3)33 (2443)
Fever80,6265.9 (5.76.0)16.3 (15.217.3)
Dehydration7,3625.4 (5.25.5)34.6 (30.139)
Infectious respiratory diseases72,6525.4 (5.25.5)28.6 (27.230)
Seizures17,6375.3 (4.95.6)33.3 (30.336.4)
Other devices and complicationsb1,8965.3 (4.36.3)39.0 (29.448.6)
Infectious skin, dermatologic and soft tissue diseases40,2724.7 (4.55)20.0 (18.221.8)
Devices and complications of the gastrointestinal systemb4,6924.6 (4.05.2)24.7 (18.930.4)
Vomiting44,7304.4 (4.24.6)23.7 (21.825.6)
Infectious urinary tract diseases17,0204.4 (4.14.7)25.9 (22.729)
Headache19,0164.3 (4.14.6)28.2 (25.131.3)
Diabetes mellitus1,5314.5 (3.35.3)29 (1840)
Abdominal pain39,5944.2 (44.4)24.8 (22.726.8)
Other infectious diseasesb6474.2 (2.65.7)33 (1651)
Gastroenteritis55,6134.0 (3.84.1)20.6 (18.922.3)

DISCUSSION

In this nationally representative sample of free‐standing children's hospitals, 3.3% of patients discharged from the ED returned to the same ED within 72 hours. This rate is similar to rates previously published in studies of general EDs.[11, 15] Of the returning children, over 80% were discharged again, and 19.7% were hospitalized, which is two‐thirds more than the admission rate at index visit (12%). In accordance with previous studies,[14, 16, 25] we found higher disease severity, presence of a chronic condition, and younger age were strongly associated with both the odds of patients returning to the ED and of being hospitalized at return. Patients who were hospitalized lived further away from the hospital and were of a higher SES. In this study, we show that visit‐level and access‐related factors are also associated with increased risk of return, although to a lesser degree. Patients seen on a weekend (Friday or Saturday) were found to have higher odds of returning, whereas those seen initially on a Sunday had lower odds of hospitalization at return. In this study, we also found that patients seen on the evening or night shifts at the index presentation had a significant association with return visits and hospitalization at return. Additionally, we found that although PCP density was not associated with the odds of returning to the ED, patients from areas with a higher PCP density were less likely to be admitted at return. In addition, by evaluating the diagnoses of patients who returned, we found that many infectious conditions commonly seen in the ED also had high return rates.

As previously shown,[23] we found that patients with complex and chronic diseases were at risk for ED revisits, especially patients with sickle cell anemia and cancer (mainly acute leukemia). In addition, patients with a chronic condition were 3 times more likely to be hospitalized when they returned. These findings may indicate an opportunity for improved discharge planning and coordination of care with subspecialty care providers for particularly at‐risk populations, or stronger consideration of admission at the index visit. However, admission for these patients at revisit may be unavoidable.

Excluding patients with chronic and complex conditions, the majority of conditions with high revisit rates were acute infectious conditions. One national study showed that >70% of ED revisits by patients with infectious conditions had planned ED follow‐up.[13] Although this study was unable to assess the reasons for return or admission at return, children with infectious diseases often worsen over time (eg, those with bronchiolitis). The relatively low admission rates at return for these conditions, despite evidence that providers may have a lower threshold for admission when a patient returns to the ED shortly after discharge,[24] may reflect the potential for improving follow‐up at the PCP office. However, although some revisits may be prevented,[37, 38] we recognize that an ED visit could be appropriate and necessary for some of these children, especially those without primary care.

Access to primary care and insurance status influence ED utilization.[14, 39, 40, 41] A fragmented healthcare system with poor access to primary care is strongly associated with utilization of the ED for nonurgent care. A high ED revisit rate might be indicative of poor coordination between ED and outpatient services.[9, 39, 42, 43, 44, 45, 46] Our study's finding of increased risk of return visit if the index visit occurred on a Friday or Saturday, and a decreased likelihood of subsequent admission when a patient returns on a Sunday, may suggest limited or perceived limited access to the PCP over a weekend. Although insured patients tend to use the ED less often for nonemergent cases, even when patients have PCPs, they might still choose to return to the ED out of convenience.[47, 48] This may be reflected in our finding that, when adjusted for insurance status and PCP density, patients who lived closer to the hospital were more likely to return, but less likely to be admitted, thereby suggesting proximity as a factor in the decision to return. It is also possible that patients residing further away returned to another institution. Although PCP density did not seem to be associated with revisits, patients who lived in areas with higher PCP density were less likely to be admitted when they returned. In this study, there was a stepwise gradient in the effect of PCP density on the odds of being hospitalized on return with those patients in areas with fewer PCPs being admitted at higher rates on return. Guttmann et al.,[40] in a recent study conducted in Canada where there is universal health insurance, showed that children residing in areas with higher PCP densities had higher rates of PCP visits but lower rates of ED visits compared to children residing in areas with lower PCP densities. It is possible that emergency physicians have more confidence that patients will have dedicated follow‐up when a PCP can be identified. These findings suggest that the development of PCP networks with expanded access, such as alignment of office hours with parent need and patient/parent education about PCP availability, may reduce ED revisits. Alternatively, creation of centralized hospital‐based urgent care centers for evening, night, and weekend visits may benefit both the patient and the PCP and avoid ED revisits and associated costs.

Targeting and eliminating disparities in care might also play a role in reducing ED revisits. Prior studies have shown that publicly insured individuals, in particular, frequently use the ED as their usual source of care and are more likely to return to the ED within 72 hours of an initial visit.[23, 39, 44, 49, 50] Likewise, we found that patients with public insurance were more likely to return but less likely to be admitted on revisit. After controlling for disease severity and other demographic variables, patients with public insurance and of lower socioeconomic status still had lower odds of being hospitalized following a revisit. This might also signify an increase of avoidable hospitalizations among patients of higher SES or with private insurance. Further investigation is needed to explore the reasons for these differences and to identify effective interventions to eliminate disparities.

Our findings have implications for emergency care, ambulatory care, and the larger healthcare system. First, ED revisits are costly and contribute to already overburdened EDs.[10, 11] The average ED visit incurs charges that are 2 to 5 times more than an outpatient office visit.[49, 50] Careful coordination of ambulatory and ED services could not only ensure optimal care for patients, but could save the US healthcare system billions of dollars in potentially avoidable healthcare expenditures.[49, 50] Second, prior studies have demonstrated a consistent relationship between poor access to primary care and increased use of the ED for nonurgent conditions.[42] Publicly insured patients have been shown to have disproportionately increased difficulty acquiring and accessing primary care.[41, 42, 47, 51] Furthermore, conditions with high ED revisit rates are similar to conditions reported by Berry et al.4 as having the highest hospital readmission rates such as cancer, sickle cell anemia, seizure, pneumonia, asthma, and gastroenteritis. This might suggest a close relationship between 72‐hour ED revisits and 30‐day hospital readmissions. In light of the recent expansion of health insurance coverage to an additional 30 million individuals, the need for better coordination of services throughout the entire continuum of care, including primary care, ED, and inpatient services, has never been more important.[52] Future improvements could explore condition‐specific revisit or readmission rates to identify the most effective interventions to reduce the possibly preventable returns.

This study has several limitations. First, as an administrative database, PHIS has limited clinical data, and reasons for return visits could not be assessed. Variations between hospitals in diagnostic coding might also lead to misclassification bias. Second, we were unable to assess return visits to a different ED. Thus, we may have underestimated revisit frequency. However, because children are generally more likely to seek repeat care in the same hospital,[3] we believe our estimate of return visit rate approximates the actual return visit rate; our findings are also similar to previously reported rates. Third, for the PCP density factor, we were unable to account for types of insurance each physician accepted and influence on return rates. Fourth, return visits in our sample could have been for conditions unrelated to the diagnosis at index visit, though the short timeframe considered for revisits makes this less likely. In addition, the crowding index does not include the proportion of occupied beds at the precise moment of the index visit. Finally, this cohort includes only children seen in the EDs of pediatric hospitals, and our findings may not be generalizable to all EDs who provide care for ill and injured children.

We have shown that, in addition to previously identified patient level factors, there are visit‐level and access‐related factors associated with pediatric ED return visits. Eighty percent are discharged again, and almost one‐fifth of returning patients are admitted to the hospital. Admitted patients tend to be younger, sicker, chronically ill, and live farther from the hospital. By being aware of patients' comorbidities, PCP access, as well as certain diagnoses associated with high rates of return, physicians may better target interventions to optimize care. This may include having a lower threshold for hospitalization at the initial visit for children at high risk of return, and communication with the PCP at the time of discharge to ensure close follow‐up. Our study helps to provide benchmarks around ED revisit rates, and may serve as a starting point to better understand variation in care. Future efforts should aim to find creative solutions at individual institutions, with the goal of disseminating and replicating successes more broadly. For example, investigators in Boston have shown that the use of a comprehensive home‐based asthma management program has been successful in decreasing emergency department visits and hospitalization rates.[53] It is possible that this approach could be spread to other institutions to decrease revisits for patients with asthma. As a next step, the authors have undertaken an investigation to identify hospital‐level characteristics that may be associated with rates of return visits.

Acknowledgements

The authors thank the following members of the PHIS ED Return Visits Research Group for their contributions to the data analysis plan and interpretation of results of this study: Rustin Morse, MD, Children's Medical Center of Dallas; Catherine Perron, MD, Boston Children's Hospital; John Cheng, MD, Children's Healthcare of Atlanta; Shabnam Jain, MD, MPH, Children's Healthcare of Atlanta; and Amanda Montalbano, MD, MPH, Children's Mercy Hospitals and Clinics. These contributors did not receive compensation for their help with this work.

Disclosures

A.T.A. and A.M.S. conceived the study and developed the initial study design. All authors were involved in the development of the final study design and data analysis plan. C.W.T. collected and analyzed the data. A.T.A. and C.W.T. had full access to all of the data and take responsibility for the integrity of the data and the accuracy of the data analysis. All authors were involved in the interpretation of the data. A.T.A. drafted the article, and all authors made critical revisions to the initial draft and subsequent versions. A.T.A. and A.M.S. take full responsibility for the article as a whole. The authors report no conflicts of interest.

Files
References
  1. Joint policy statement—guidelines for care of children in the emergency department. Pediatrics. 2009;124:12331243.
  2. Alessandrini EA, Lavelle JM, Grenfell SM, Jacobstein CR, Shaw KN. Return visits to a pediatric emergency department. Pediatr Emerg Care. 2004;20:166171.
  3. Axon RN, Williams MV. Hospital readmission as an accountability measure. JAMA. 2011;305:504505.
  4. Berry JG, Hall DE, Kuo DZ, et al. Hospital utilization and characteristics of patients experiencing recurrent readmissions within children's hospitals. JAMA. 2011;305:682690.
  5. Berry JG, Toomey SL, Zaslavsky AM, et al. Pediatric readmission prevalence and variability across hospitals. JAMA. 2013;309:372380.
  6. Carrns A. Farewell, and don't come back. Health reform gives hospitals a big incentive to send patients home for good. US News World Rep. 2010;147:20, 2223.
  7. Coye MJ. CMS' stealth health reform. Plan to reduce readmissions and boost the continuum of care. Hosp Health Netw. 2008;82:24.
  8. Lerman B, Kobernick MS. Return visits to the emergency department. J Emerg Med. 1987;5:359362.
  9. Rising KL, White LF, Fernandez WG, Boutwell AE. Emergency department visits after hospital discharge: a missing part of the equation. Ann Emerg Med. 2013;62:145150.
  10. Stang AS, Straus SE, Crotts J, Johnson DW, Guttmann A. Quality indicators for high acuity pediatric conditions. Pediatrics. 2013;132:752762.
  11. Fontanarosa PB, McNutt RA. Revisiting hospital readmissions. JAMA. 2013;309:398400.
  12. Vaduganathan M, Bonow RO, Gheorghiade M. Thirty‐day readmissions: the clock is ticking. JAMA. 2013;309:345346.
  13. Adekoya N. Patients seen in emergency departments who had a prior visit within the previous 72 h‐National Hospital Ambulatory Medical Care Survey, 2002. Public Health. 2005;119:914918.
  14. Cho CS, Shapiro DJ, Cabana MD, Maselli JH, Hersh AL. A national depiction of children with return visits to the emergency department within 72 hours, 2001–2007. Pediatr Emerg Care. 2012;28:606610.
  15. Feudtner C, Levin JE, Srivastava R, et al. How well can hospital readmission be predicted in a cohort of hospitalized children? A retrospective, multicenter study. Pediatrics. 2009;123:286293.
  16. Goldman RD, Ong M, Macpherson A. Unscheduled return visits to the pediatric emergency department‐one‐year experience. Pediatr Emerg Care. 2006;22:545549.
  17. Klein‐Kremer A, Goldman RD. Return visits to the emergency department among febrile children 3 to 36 months of age. Pediatr Emerg Care. 2011;27:11261129.
  18. LeDuc K, Rosebrook H, Rannie M, Gao D. Pediatric emergency department recidivism: demographic characteristics and diagnostic predictors. J Emerg Nurs. 2006;32:131138.
  19. Healthcare Cost and Utilization Project. Pediatric emergency department visits in community hospitals from selected states, 2005. Statistical brief #52. Available at: http://www.ncbi.nlm.nih.gov/books/NBK56039. Accessed October 3, 2013.
  20. Sharma V, Simon SD, Bakewell JM, Ellerbeck EF, Fox MH, Wallace DD. Factors influencing infant visits to emergency departments. Pediatrics. 2000;106:10311039.
  21. Ali AB, Place R, Howell J, Malubay SM. Early pediatric emergency department return visits: a prospective patient‐centric assessment. Clin Pediatr (Phila). 2012;51:651658.
  22. Hu KW, Lu YH, Lin HJ, Guo HR, Foo NP. Unscheduled return visits with and without admission post emergency department discharge. J Emerg Med. 2012;43:11101118.
  23. Jacobstein CR, Alessandrini EA, Lavelle JM, Shaw KN. Unscheduled revisits to a pediatric emergency department: risk factors for children with fever or infection‐related complaints. Pediatr Emerg Care. 2005;21:816821.
  24. Sauvin G, Freund Y, Saidi K, Riou B, Hausfater P. Unscheduled return visits to the emergency department: consequences for triage. Acad Emerg Med. 2013;20:3339.
  25. Zimmerman DR, McCarten‐Gibbs KA, DeNoble DH, et al. Repeat pediatric visits to a general emergency department. Ann Emerg Med. 1996;28:467473.
  26. Keith KD, Bocka JJ, Kobernick MS, Krome RL, Ross MA. Emergency department revisits. Ann Emerg Med. 1989;18:964968.
  27. US Department of Health 19:7078.
  28. Feudtner C, Christakis DA, Connell FA. Pediatric deaths attributable to complex chronic conditions: a population‐based study of Washington State, 1980–1997. Pediatrics. 2000;106:205209.
  29. Feudtner C, Hays RM, Haynes G, Geyer JR, Neff JM, Koepsell TD. Deaths attributed to pediatric complex chronic conditions: national trends and implications for supportive care services. Pediatrics. 2001;107:E99.
  30. Feudtner C, Silveira MJ, Christakis DA. Where do children with complex chronic conditions die? Patterns in Washington State, 1980–1998. Pediatrics. 2002;109:656660.
  31. Dartmouth Atlas of Health Care. Hospital and physician capacity, 2006. Available at: http://www.dartmouthatlas.org/data/topic/topic.aspx?cat=24. Accessed October 7, 2013.
  32. Dartmouth Atlas of Health Care. Research methods. What is an HSA/HRR? Available at: http://www.dartmouthatlas.org/tools/faq/researchmethods.aspx. Accessed October 7, 2013,.
  33. Dartmouth Atlas of Health Care. Appendix on the geography of health care in the United States. Available at: http://www.dartmouthatlas.org/downloads/methods/geogappdx.pdf. Accessed October 7, 2013.
  34. Beniuk K, Boyle AA, Clarkson PJ. Emergency department crowding: prioritising quantified crowding measures using a Delphi study. Emerg Med J. 2012;29:868871.
  35. Alessandrini EA, Alpern ER, Chamberlain JM, Shea JA, Gorelick MH. A new diagnosis grouping system for child emergency department visits. Acad Emerg Med. 2010;17:204213.
  36. Guttmann A, Zagorski B, Austin PC, et al. Effectiveness of emergency department asthma management strategies on return visits in children: a population‐based study. Pediatrics. 2007;120:e1402e1410.
  37. Horwitz DA, Schwarz ES, Scott MG, Lewis LM. Emergency department patients with diabetes have better glycemic control when they have identifiable primary care providers. Acad Emerg Med. 2012;19:650655.
  38. Billings J, Zeitel L, Lukomnik J, Carey TS, Blank AE, Newman L. Impact of socioeconomic status on hospital use in New York City. Health Aff (Millwood). 1993;12:162173.
  39. Guttmann A, Shipman SA, Lam K, Goodman DC, Stukel TA. Primary care physician supply and children's health care use, access, and outcomes: findings from Canada. Pediatrics. 2010;125:11191126.
  40. Asplin BR, Rhodes KV, Levy H, et al. Insurance status and access to urgent ambulatory care follow‐up appointments. JAMA. 2005;294:12481254.
  41. Kellermann AL, Weinick RM. Emergency departments, Medicaid costs, and access to primary care—understanding the link. N Engl J Med. 2012;366:21412143.
  42. Committee on the Future of Emergency Care in the United States Health System. Emergency Care for Children: Growing Pains. Washington, DC: The National Academies Press; 2007.
  43. Committee on the Future of Emergency Care in the United States Health System. Hospital‐Based Emergency Care: At the Breaking Point. Washington, DC: The National Academies Press; 2007.
  44. Radley DC, Schoen C. Geographic variation in access to care—the relationship with quality. N Engl J Med. 2012;367:36.
  45. Tang N, Stein J, Hsia RY, Maselli JH, Gonzales R. Trends and characteristics of US emergency department visits, 1997–2007. JAMA. 2010;304:664670.
  46. Young GP, Wagner MB, Kellermann AL, Ellis J, Bouley D. Ambulatory visits to hospital emergency departments. Patterns and reasons for use. 24 Hours in the ED Study Group. JAMA. 1996;276:460465.
  47. Tranquada KE, Denninghoff KR, King ME, Davis SM, Rosen P. Emergency department workload increase: dependence on primary care? J Emerg Med. 2010;38:279285.
  48. Network for Excellence in Health Innovation. Leading healthcare research organizations to examine emergency department overuse. New England Research Institute, 2008. Available at: http://www.nehi.net/news/310‐leading‐health‐care‐research‐organizations‐to‐examine‐emergency‐department‐overuse/view. Accessed October 4, 2013.
  49. Robert Wood Johnson Foundation. Quality field notes: reducing inappropriate emergency department use. Available at: http://www.rwjf.org/en/research‐publications/find‐rwjf‐research/2013/09/quality‐field‐notes–reducing‐inappropriate‐emergency‐department.html.
  50. Access of Medicaid recipients to outpatient care. N Engl J Med. 1994;330:14261430.
  51. Medicaid policy statement. Pediatrics. 2013;131:e1697e1706.
  52. Woods ER, Bhaumik U, Sommer SJ, et al. Community asthma initiative: evaluation of a quality improvement program for comprehensive asthma care. Pediatrics. 2012;129:465472.
Article PDF
Issue
Journal of Hospital Medicine - 9(12)
Publications
Page Number
779-787
Sections
Files
Files
Article PDF
Article PDF

Returns to the hospital following recent encounters, such as an admission to the inpatient unit or evaluation in an emergency department (ED), may reflect the natural progression of a disease, the quality of care received during the initial admission or visit, or the quality of the underlying healthcare system.[1, 2, 3, 4, 5, 6, 7, 8, 9, 10] Although national attention has focused on hospital readmissions,[3, 4, 5, 6, 7, 11, 12] ED revisits are a source of concern to emergency physicians.[8, 9] Some ED revisits are medically necessary, but revisits that may be managed in the primary care setting contribute to ED crowding, can be stressful to patients and providers, and increase healthcare costs.[10, 11, 12] Approximately 27 million annual ED visits are made by children, accounting for over one‐quarter of all ED visits in the United States, with a reported ED revisit rate of 2.5% to 5.2%.[2, 13, 14, 15, 16, 17, 18, 19, 20] Improved understanding of the patient‐level or visit‐level factors associated with ED revisits may provide an opportunity to enhance disposition decision making at the index visit and optimize site of and communication around follow‐up care.

Previous studies on ED revisits have largely been conducted in single centers and have used variable visit intervals ranging between 48 hours and 30 days.[2, 13, 16, 18, 21, 22, 23, 24, 25] Two national studies used the National Hospital Ambulatory Medical Care Survey, which includes data from both general and pediatric EDs.[13, 14] Factors identified to be associated with increased odds of returning were: young age, higher acuity, chronic conditions, and public insurance. One national study identified some diagnoses associated with higher likelihood of returning,[13] whereas the other focused primarily on infectious diseaserelated diagnoses.[14]

The purpose of this study was to describe the prevalence of return visits specifically to pediatric EDs and to investigate patient‐level, visit‐level, and healthcare systemrelated factors that may be associated with return visits and hospitalization at return.

METHODS

Study Design and Data Source

This retrospective cohort study used data from the Pediatric Health Information System (PHIS), an administrative database with data from 44 tertiary care pediatric hospitals in 27 US states and the District of Columbia. This database contains patient demographics, diagnoses, and procedures as well as medications, diagnostic imaging, laboratory, and supply charges for each patient. Data are deidentified prior to inclusion; encrypted medical record numbers allow for the identification of individual patients across all ED visits and hospitalizations to the same hospital. The Children's Hospital Association (Overland Park, KS) and participating hospitals jointly assure the quality and integrity of the data. This study was approved by the institutional review board at Boston Children's Hospital with a waiver for informed consent granted.

Study Population and Protocol

To standardize comparisons across the hospitals, we included data from 23 of the 44 hospitals in PHIS; 7 were excluded for not including ED‐specific data. For institutions that collect information from multiple hospitals within their healthcare system, we included only records from the main campus or children's hospital when possible, leading to the exclusion of 9 hospitals where the data were not able to be segregated. As an additional level of data validation, we compared the hospital‐level ED volume and admission rates as reported in the PHIS to those reported to a separate database (the Pediatric Analysis and Comparison Tool). We further excluded 5 hospitals whose volume differed by >10% between these 2 data sources.

Patients <18 years of age who were discharged from these EDs following their index visit in 2012 formed the eligible cohort.

Key Outcome Measures

The primary outcomes were return visits within 72 hours of discharge from the ED, and return visits resulting in hospitalization, including observation status. We defined an ED revisit as a return within 72 hours of ED discharge regardless of whether the patient was subsequently discharged from the ED on the return visit or hospitalized. We assessed revisits within 72 hours of an index ED discharge, because return visits within this time frame are likely to be related to the index visit.[2, 13, 16, 21, 22, 24, 25, 26]

Factors Associated With ED Revisits

A priori, we chose to adjust for the following patient‐level factors: age (<30 days, 30 days<1 year, 14 years, 511 years, 1217 years), gender, and socioeconomic status (SES) measured as the zip codebased median household income, obtained from the 2010 US Census, with respect to the federal poverty level (FPL) (<1.5 FPL, 1.52 FPL, 23 FPL, and >3 FPL).[27] We also adjusted for insurance type (commercial, government, or other), proximity of patient's home zip code to hospital (modeled as the natural log of the geographical distance to patient's home address from the hospital), ED diagnosis‐based severity classification system score (1=low severity, 5=high severity),[28] presence of a complex chronic condition at the index or prior visits using a validated classification scheme,[15, 29, 30, 31] and primary care physician (PCP) density per 100,000 in the patient's residential area (modeled as quartiles: very low, <57.2; low, 57.267.9; medium, 68.078.7; high, >78.8). PCP density, defined by the Dartmouth Atlas of Health Care,[32, 33, 34] is the number of primary care physicians per 100,000 residents (PCP count) in federal health service areas (HSA). Patients were assigned to a corresponding HSA based on their home zip code.

Visit‐level factors included arrival time of index visit (8:01 am 4:00 pm, 4:01 pm12:00 am, 12:01 am8 am representing day, evening, and overnight arrival, respectively), day of the week, season, length of stay (LOS) in the ED during the index visit, and ED crowding (calculated as the average daily LOS/yearly average LOS for the individual ED).[35] We categorized the ED primary diagnosis for each visit using the major diagnosis groupings of a previously described pediatric ED‐specific classification scheme.[36] Using International Classification of Diseases, Ninth Revision (ICD‐9) codes, we identified the conditions with the highest ED revisit rates.

Statistical Analyses

Categorical variables describing the study cohort were summarized using frequencies and percentages. Continuous variables were summarized using mean, median, and interquartile range values, where appropriate. We used 2 different hierarchical logistic regression models to assess revisit rates by patient‐ and visit‐level characteristics. The initial model included all patients discharged from the ED following the index visit and assessed for the outcome of a revisit within 72 hours. The second model considered only patients who returned within 72 hours of an index visit and assessed for hospitalization on that return visit. We used generalized linear mixed effects models, with hospital as a random effect to account for the presence of correlated data (within hospitals), nonconstant variability (across hospitals), and binary responses. Adjusted odds ratios with 95% confidence intervals were used as summary measures of the effect of the individual adjusters. Adjusters were missing in fewer than 5% of patients across participating hospitals. Statistical analyses were performed using SAS version 9.3 (SAS Institute Inc., Cary, NC); 2‐sided P values <0.004 were considered statistically significant to account for multiple comparisons (Bonferroni‐adjusted level of significance=0.0038).

RESULTS

Patients

A total of 1,610,201 patients <18 years of age evaluated across the 23 PHIS EDs in 2012 were included in the study. Twenty‐one of the 23 EDs have academic affiliations; 10 are located in the South, 6 in the Midwest, 5 in the West, and 2 in the Northeast region of the United States. The annual ED volume for these EDs ranged from 25,090 to 136,160 (median, 65,075; interquartile range, 45,28085,206). Of the total patients, 1,415,721 (87.9%) were discharged following the index visit and comprised our study cohort. Of these patients, 47,294 (revisit rate: 3.3%) had an ED revisit within 72 hours. There were 4015 patients (0.3%) who returned more than once within 72 hours, and the largest proportion of these returned with infection‐related conditions. Of those returning, 37,999 (80.3%) were discharged again, whereas 9295 (19.7%) were admitted to the hospital (Figure 1). The demographic and clinical characteristics of study participants are displayed in Table 1.

jhm2273-fig-0001-m.png
Patient disposition from the emergency departments of study hospitals (n = 23) in 2012.
Characteristics of Patients Who Returned Within 72 Hours of ED Discharge to the Study EDs
 Index Visit, n=1,415,721, n (%)Return Visits Within 72 Hours of Discharge, n=47,294, 3.3%
Return to Discharge, n (%)Return to Admission, n (%)
  • NOTE: Abbreviations: CCC, complex chronic condition; ED, emergency department; FPL, federal poverty level; IQR, interquartile range; LOS, length of stay.

  • Socioeconomic status is relative to the federal poverty level for a family of 4.

Gender, female659,417 (46.6)17,665 (46.5)4,304 (46.3)
Payor   
Commercial379,403 (26.8)8,388 (22.1)3,214 (34.6)
Government925,147 (65.4)26,880 (70.7)5,786 (62.3)
Other111,171 (7.9)2,731 (7.2)295 (3.2)
Age   
<30 days19,217 (1.4)488 (1.3)253 (2.7)
30 days to <1 year216,967 (15.3)8,280 (21.8)2,372 (25.5)
1 year to 4 years547,083 (38.6)15,542 (40.9)3,187 (34.3)
5 years to 11 years409,463 (28.9)8,906 (23.4)1,964 (21.1)
12 years to 17 years222,991 (15.8)4,783 (12.6)1,519 (16.3)
Socioeconomic statusa   
<1.5 times FPL493,770 (34.9)13,851 (36.5)2,879 (31.0)
1.5 to 2 times FPL455,490 (32.2)12,364 (32.5)2,904 (31.2)
2 to 3 times FPL367,557 (26.0)9,560 (25.2)2,714 (29.2)
>3 times FPL98,904 (7.0)2,224 (5.9)798 (8.6)
Primary care physician density per 100,000 patients   
Very low351,798 (24.9)8,727 (23.0)2,628 (28.3)
Low357,099 (25.2)9,810 (25.8)2,067 (22.2)
Medium347,995 (24.6)10,186 (26.8)2,035 (21.9)
High358,829 (25.4)9,276 (24.4)2,565 (27.6)
CCC present, yes125,774 (8.9)4,446 (11.7)2,825 (30.4)
Severity score   
Low severity (0,1,2)721,061 (50.9)17,310 (45.6)2,955 (31.8)
High severity (3,4,5)694,660 (49.1)20,689 (54.5)6,340 (68.2)
Time of arrival   
Day533,328 (37.7)13,449 (35.4)3,396 (36.5)
Evening684,873 (48.4)18,417 (48.5)4,378 (47.1)
Overnight197,520 (14.0)6,133 (16.1)1,521 (16.4)
Season   
Winter384,957 (27.2)10,603 (27.9)2,844 (30.6)
Spring367,434 (26.0)9,923 (26.1)2,311 (24.9)
Summer303,872 (21.5)8,308 (21.9)1,875 (20.2)
Fall359,458 (25.4)9,165 (24.1)2,265 (24.4)
Weekday/weekend   
Monday217,774 (15.4)5,646 (14.9)1,394 (15)
Tuesday198,220 (14.0)5,054 (13.3)1,316 (14.2)
Wednesday194,295 (13.7)4,985 (13.1)1,333 (14.3)
Thursday191,950 (13.6)5,123 (13.5)1,234 (13.3)
Friday190,022 (13.4)5,449 (14.3)1,228 (13.2)
Saturday202,247 (14.3)5,766 (15.2)1,364 (14.7)
Sunday221,213 (15.6)5,976 (15.7)1,426 (15.3)
Distance from hospital in miles, median (IQR)8.3 (4.614.9)9.2 (4.917.4)8.3 (4.614.9)
ED crowding score at index visit, median (IQR)1.0 (0.91.1)1.0 (0.91.1)1.0 (0.91.1)
ED LOS in hours at index visit, median (IQR)2.0 (1.03.0)3.0 (2.05.0)2.0 (1.03.0)

ED Revisit Rates and Revisits Resulting in Admission

In multivariate analyses, compared to patients who did not return to the ED, patients who returned within 72 hours of discharge had higher odds of revisit if they had the following characteristics: a chronic condition, were <1 year old, a higher severity score, and public insurance. Visit‐level factors associated with higher odds of revisits included arrival for the index visit during the evening or overnight shift or on a Friday or Saturday, index visit during times of lower ED crowding, and living closer to the hospital. On return, patients were more likely to be hospitalized if they had a higher severity score, a chronic condition, private insurance, or were <30 days old. Visit‐level factors associated with higher odds of hospitalization at revisit included an index visit during the evening and overnight shift and living further from the hospital. Although the median SES and PCP density of a patient's area of residence were not associated with greater likelihood of returning, when they returned, patients residing in an area with a lower SES and higher PCP densities (>78.8 PCPs/100,000) had lower odds of being admitted to the hospital. Patients whose index visit was on a Sunday also had lower odds of being hospitalized upon return (Table 2).

Multivariate Analyses of Factors Associated With ED Revisits and Admission at Return
CharacteristicAdjusted OR of 72‐Hour Revisit (95% CI), n=1,380,723P ValueAdjusted OR of 72‐Hour Revisit Admissions (95% CI), n=46,364P Value
  • NOTE: Effects of continuous variables are assessed as 1‐unit offsets from the mean. Abbreviations: CCC, complex chronic condition; CI, confidence interval; ED, emergency department; FPL, federal poverty level; LOS, length of stay; OR, odds ratio, NA, not applicable.

  • Socioeconomic status is relative to the FPL for a family of 4.

  • ED crowding score and LOS are based on index visit. ED crowding score is calculated as the daily LOS (in hours)/overall LOS (in hours). Overall average across hospitals=1; a 1‐ unit increase translates into twice the duration for the daily LOS over the yearly average ED LOS.

  • Modeled as the natural log of the patient geographic distance from the hospital based on zip codes. Number in parentheses represents the exponential of the modeled variable.

Gender    
Male0.99 (0.971.01)0.28091.02 (0.971.07)0.5179
FemaleReference Reference 
Payor    
Government1.14 (1.111.17)<0.00010.68 (0.640.72)<0.0001
Other0.97 (0.921.01)0.11480.33 (0.280.39)<0.0001
PrivateReference Reference 
Age group    
30 days to <1 year1.32 (1.221.42)<0.00010.58 (0.490.69)<0.0001
1 year to 5 years0.89 (0.830.96)0.0030.41 (0.340.48)<0.0001
5 years to 11 years0.69 (0.640.74)<0.00010.40 (0.330.48)<0.0001
12 years to 17 years0.72 (0.660.77)<0.00010.50 (0.420.60)<0.0001
<30 daysReference Reference 
Socioeconomic statusa    
% <1.5 times FPL0.96 (0.921.01)0.09920.82 (0.740.92)0.0005
% 1.5 to 2 times FPL0.98 (0.941.02)0.29920.83 (0.750.92)0.0005
% 2 to 3 times FPL1.02 (0.981.07)0.2920.88 (0.790.97)0.01
% >3 times FPLReference Reference 
Severity score    
High severity, 4, 5, 61.43 (1.401.45)<0.00013.42 (3.233.62)<0.0001
Low severity, 1, 2, 3Reference Reference 
Presence of any CCC    
Yes1.90 (1.861.96)<0.00012.92 (2.753.10)<0.0001
NoReference Reference 
Time of arrival    
Evening1.05 (1.031.08)<0.00011.37 (1.291.44)<0.0001
Overnight1.19 (1.151.22)<0.00011.84 (1.711.97)<0.0001
DayReference Reference 
Season    
Winter1.09 (1.061.11)<0.00011.06 (0.991.14)0.0722
Spring1.07 (1.041.10)<0.00010.98 (0.911.046)0.4763
Summer1.05 (1.021.08)0.00110.93 (0.871.01)0.0729
FallReference Reference 
Weekday/weekend    
Thursday1.02 (0.9821.055)0.32970.983 (0.8971.078)0.7185
Friday1.08 (1.041.11)<0.00011.03 (0.941.13)0.5832
Saturday1.08 (1.041.12)<0.00010.89 (0.810.97)0.0112
Sunday1.02 (0.991.06)0.20540.81 (0.740.89)<0.0001
Monday1.00 (0.961.03)0.89280.98 (0.901.07)0.6647
Tuesday0.99 (0.951.03)0.53420.93 (0.851.02)0.1417
WednesdayReference Reference 
PCP ratio per 100,000 patients    
57.267.91.00 (0.961.04)0.88440.93 (0.841.03)0.1669
68.078.71.00 (0.951.04)0.81560.86 (0.770.96)0.0066
>78.81.00 (0.951.04)0.68830.82 (0.730.92)0.001
<57.2Reference Reference 
ED crowding score at index visitb    
20.92 (0.900.95)<0.00010.96 (0.881.05)0.3435
1Reference Reference 
Distance from hospitalc    
3.168, 23.6 miles0.95 (0.940.96)<0.00011.16 (1.121.19)<0.0001
2.168, 8.7 milesReference Reference 
ED LOS at index visitb    
3.7 hours1.003 (1.0011.005)0.0052NA 
2.7 hoursReference   

Diagnoses Associated With Return Visits

Patients with index visit diagnoses of sickle cell disease and leukemia had the highest proportion of return visits (10.7% and 7.3%, respectively). Other conditions with high revisit rates included infectious diseases such as cellulitis, bronchiolitis, and gastroenteritis. Patients with other chronic diseases such as diabetes and with devices, such as gastrostomy tubes, also had high rates of return visits. At return, the rate of hospitalization for these conditions ranged from a 1‐in‐6 chance of hospitalization for the diagnoses of a fever to a 1‐in‐2 chance of hospitalization for patients with sickle cell anemia (Table 3).

Major Diagnostic Subgroups With the Highest ED Revisit and Admission at Return Rates
Major Diagnostic SubgroupNo. of Index ED Visit Dischargesa72‐Hour Revisit, % (95% CI)Admitted on Return, % (95% CI)
  • NOTE: Abbreviations: CI, confidence interval; ED, emergency department; NOS, not otherwise specified.

  • Diagnoses with <500 index visits (ie, <2 visits per month across the 23 hospitals) or <30 revisits within entire study cohort excluded from analyses.

  • Most prevalent diagnoses as identified by International Classification of Diseases, Ninth Revision codes within specified major diagnostic subgroups: devices and complications of the circulatory system, complication of other vascular device, implant, and graft; other hematologic diseases, anemia NOS, neutropenia NOS, or thrombocytopenia NOS; other devices and complications, hemorrhage complicating a procedure; devices and complications of the gastrointestinal system, gastrostomy; other infectious diseases, perinatal infections.

Sickle cell anemia2,53110.7 (9.511.9)49.6 (43.755.6)
Neoplastic diseases, cancer5367.3 (5.19.5)36 (2151)
Infectious gastrointestinal diseases8027.2 (5.49.0)21 (1031)
Devices and complications of the circulatory systemb1,0336.9 (5.38.4)45 (3457)
Other hematologic diseasesb1,5386.1 (4.97.3)33 (2443)
Fever80,6265.9 (5.76.0)16.3 (15.217.3)
Dehydration7,3625.4 (5.25.5)34.6 (30.139)
Infectious respiratory diseases72,6525.4 (5.25.5)28.6 (27.230)
Seizures17,6375.3 (4.95.6)33.3 (30.336.4)
Other devices and complicationsb1,8965.3 (4.36.3)39.0 (29.448.6)
Infectious skin, dermatologic and soft tissue diseases40,2724.7 (4.55)20.0 (18.221.8)
Devices and complications of the gastrointestinal systemb4,6924.6 (4.05.2)24.7 (18.930.4)
Vomiting44,7304.4 (4.24.6)23.7 (21.825.6)
Infectious urinary tract diseases17,0204.4 (4.14.7)25.9 (22.729)
Headache19,0164.3 (4.14.6)28.2 (25.131.3)
Diabetes mellitus1,5314.5 (3.35.3)29 (1840)
Abdominal pain39,5944.2 (44.4)24.8 (22.726.8)
Other infectious diseasesb6474.2 (2.65.7)33 (1651)
Gastroenteritis55,6134.0 (3.84.1)20.6 (18.922.3)

DISCUSSION

In this nationally representative sample of free‐standing children's hospitals, 3.3% of patients discharged from the ED returned to the same ED within 72 hours. This rate is similar to rates previously published in studies of general EDs.[11, 15] Of the returning children, over 80% were discharged again, and 19.7% were hospitalized, which is two‐thirds more than the admission rate at index visit (12%). In accordance with previous studies,[14, 16, 25] we found higher disease severity, presence of a chronic condition, and younger age were strongly associated with both the odds of patients returning to the ED and of being hospitalized at return. Patients who were hospitalized lived further away from the hospital and were of a higher SES. In this study, we show that visit‐level and access‐related factors are also associated with increased risk of return, although to a lesser degree. Patients seen on a weekend (Friday or Saturday) were found to have higher odds of returning, whereas those seen initially on a Sunday had lower odds of hospitalization at return. In this study, we also found that patients seen on the evening or night shifts at the index presentation had a significant association with return visits and hospitalization at return. Additionally, we found that although PCP density was not associated with the odds of returning to the ED, patients from areas with a higher PCP density were less likely to be admitted at return. In addition, by evaluating the diagnoses of patients who returned, we found that many infectious conditions commonly seen in the ED also had high return rates.

As previously shown,[23] we found that patients with complex and chronic diseases were at risk for ED revisits, especially patients with sickle cell anemia and cancer (mainly acute leukemia). In addition, patients with a chronic condition were 3 times more likely to be hospitalized when they returned. These findings may indicate an opportunity for improved discharge planning and coordination of care with subspecialty care providers for particularly at‐risk populations, or stronger consideration of admission at the index visit. However, admission for these patients at revisit may be unavoidable.

Excluding patients with chronic and complex conditions, the majority of conditions with high revisit rates were acute infectious conditions. One national study showed that >70% of ED revisits by patients with infectious conditions had planned ED follow‐up.[13] Although this study was unable to assess the reasons for return or admission at return, children with infectious diseases often worsen over time (eg, those with bronchiolitis). The relatively low admission rates at return for these conditions, despite evidence that providers may have a lower threshold for admission when a patient returns to the ED shortly after discharge,[24] may reflect the potential for improving follow‐up at the PCP office. However, although some revisits may be prevented,[37, 38] we recognize that an ED visit could be appropriate and necessary for some of these children, especially those without primary care.

Access to primary care and insurance status influence ED utilization.[14, 39, 40, 41] A fragmented healthcare system with poor access to primary care is strongly associated with utilization of the ED for nonurgent care. A high ED revisit rate might be indicative of poor coordination between ED and outpatient services.[9, 39, 42, 43, 44, 45, 46] Our study's finding of increased risk of return visit if the index visit occurred on a Friday or Saturday, and a decreased likelihood of subsequent admission when a patient returns on a Sunday, may suggest limited or perceived limited access to the PCP over a weekend. Although insured patients tend to use the ED less often for nonemergent cases, even when patients have PCPs, they might still choose to return to the ED out of convenience.[47, 48] This may be reflected in our finding that, when adjusted for insurance status and PCP density, patients who lived closer to the hospital were more likely to return, but less likely to be admitted, thereby suggesting proximity as a factor in the decision to return. It is also possible that patients residing further away returned to another institution. Although PCP density did not seem to be associated with revisits, patients who lived in areas with higher PCP density were less likely to be admitted when they returned. In this study, there was a stepwise gradient in the effect of PCP density on the odds of being hospitalized on return with those patients in areas with fewer PCPs being admitted at higher rates on return. Guttmann et al.,[40] in a recent study conducted in Canada where there is universal health insurance, showed that children residing in areas with higher PCP densities had higher rates of PCP visits but lower rates of ED visits compared to children residing in areas with lower PCP densities. It is possible that emergency physicians have more confidence that patients will have dedicated follow‐up when a PCP can be identified. These findings suggest that the development of PCP networks with expanded access, such as alignment of office hours with parent need and patient/parent education about PCP availability, may reduce ED revisits. Alternatively, creation of centralized hospital‐based urgent care centers for evening, night, and weekend visits may benefit both the patient and the PCP and avoid ED revisits and associated costs.

Targeting and eliminating disparities in care might also play a role in reducing ED revisits. Prior studies have shown that publicly insured individuals, in particular, frequently use the ED as their usual source of care and are more likely to return to the ED within 72 hours of an initial visit.[23, 39, 44, 49, 50] Likewise, we found that patients with public insurance were more likely to return but less likely to be admitted on revisit. After controlling for disease severity and other demographic variables, patients with public insurance and of lower socioeconomic status still had lower odds of being hospitalized following a revisit. This might also signify an increase of avoidable hospitalizations among patients of higher SES or with private insurance. Further investigation is needed to explore the reasons for these differences and to identify effective interventions to eliminate disparities.

Our findings have implications for emergency care, ambulatory care, and the larger healthcare system. First, ED revisits are costly and contribute to already overburdened EDs.[10, 11] The average ED visit incurs charges that are 2 to 5 times more than an outpatient office visit.[49, 50] Careful coordination of ambulatory and ED services could not only ensure optimal care for patients, but could save the US healthcare system billions of dollars in potentially avoidable healthcare expenditures.[49, 50] Second, prior studies have demonstrated a consistent relationship between poor access to primary care and increased use of the ED for nonurgent conditions.[42] Publicly insured patients have been shown to have disproportionately increased difficulty acquiring and accessing primary care.[41, 42, 47, 51] Furthermore, conditions with high ED revisit rates are similar to conditions reported by Berry et al.4 as having the highest hospital readmission rates such as cancer, sickle cell anemia, seizure, pneumonia, asthma, and gastroenteritis. This might suggest a close relationship between 72‐hour ED revisits and 30‐day hospital readmissions. In light of the recent expansion of health insurance coverage to an additional 30 million individuals, the need for better coordination of services throughout the entire continuum of care, including primary care, ED, and inpatient services, has never been more important.[52] Future improvements could explore condition‐specific revisit or readmission rates to identify the most effective interventions to reduce the possibly preventable returns.

This study has several limitations. First, as an administrative database, PHIS has limited clinical data, and reasons for return visits could not be assessed. Variations between hospitals in diagnostic coding might also lead to misclassification bias. Second, we were unable to assess return visits to a different ED. Thus, we may have underestimated revisit frequency. However, because children are generally more likely to seek repeat care in the same hospital,[3] we believe our estimate of return visit rate approximates the actual return visit rate; our findings are also similar to previously reported rates. Third, for the PCP density factor, we were unable to account for types of insurance each physician accepted and influence on return rates. Fourth, return visits in our sample could have been for conditions unrelated to the diagnosis at index visit, though the short timeframe considered for revisits makes this less likely. In addition, the crowding index does not include the proportion of occupied beds at the precise moment of the index visit. Finally, this cohort includes only children seen in the EDs of pediatric hospitals, and our findings may not be generalizable to all EDs who provide care for ill and injured children.

We have shown that, in addition to previously identified patient level factors, there are visit‐level and access‐related factors associated with pediatric ED return visits. Eighty percent are discharged again, and almost one‐fifth of returning patients are admitted to the hospital. Admitted patients tend to be younger, sicker, chronically ill, and live farther from the hospital. By being aware of patients' comorbidities, PCP access, as well as certain diagnoses associated with high rates of return, physicians may better target interventions to optimize care. This may include having a lower threshold for hospitalization at the initial visit for children at high risk of return, and communication with the PCP at the time of discharge to ensure close follow‐up. Our study helps to provide benchmarks around ED revisit rates, and may serve as a starting point to better understand variation in care. Future efforts should aim to find creative solutions at individual institutions, with the goal of disseminating and replicating successes more broadly. For example, investigators in Boston have shown that the use of a comprehensive home‐based asthma management program has been successful in decreasing emergency department visits and hospitalization rates.[53] It is possible that this approach could be spread to other institutions to decrease revisits for patients with asthma. As a next step, the authors have undertaken an investigation to identify hospital‐level characteristics that may be associated with rates of return visits.

Acknowledgements

The authors thank the following members of the PHIS ED Return Visits Research Group for their contributions to the data analysis plan and interpretation of results of this study: Rustin Morse, MD, Children's Medical Center of Dallas; Catherine Perron, MD, Boston Children's Hospital; John Cheng, MD, Children's Healthcare of Atlanta; Shabnam Jain, MD, MPH, Children's Healthcare of Atlanta; and Amanda Montalbano, MD, MPH, Children's Mercy Hospitals and Clinics. These contributors did not receive compensation for their help with this work.

Disclosures

A.T.A. and A.M.S. conceived the study and developed the initial study design. All authors were involved in the development of the final study design and data analysis plan. C.W.T. collected and analyzed the data. A.T.A. and C.W.T. had full access to all of the data and take responsibility for the integrity of the data and the accuracy of the data analysis. All authors were involved in the interpretation of the data. A.T.A. drafted the article, and all authors made critical revisions to the initial draft and subsequent versions. A.T.A. and A.M.S. take full responsibility for the article as a whole. The authors report no conflicts of interest.

Returns to the hospital following recent encounters, such as an admission to the inpatient unit or evaluation in an emergency department (ED), may reflect the natural progression of a disease, the quality of care received during the initial admission or visit, or the quality of the underlying healthcare system.[1, 2, 3, 4, 5, 6, 7, 8, 9, 10] Although national attention has focused on hospital readmissions,[3, 4, 5, 6, 7, 11, 12] ED revisits are a source of concern to emergency physicians.[8, 9] Some ED revisits are medically necessary, but revisits that may be managed in the primary care setting contribute to ED crowding, can be stressful to patients and providers, and increase healthcare costs.[10, 11, 12] Approximately 27 million annual ED visits are made by children, accounting for over one‐quarter of all ED visits in the United States, with a reported ED revisit rate of 2.5% to 5.2%.[2, 13, 14, 15, 16, 17, 18, 19, 20] Improved understanding of the patient‐level or visit‐level factors associated with ED revisits may provide an opportunity to enhance disposition decision making at the index visit and optimize site of and communication around follow‐up care.

Previous studies on ED revisits have largely been conducted in single centers and have used variable visit intervals ranging between 48 hours and 30 days.[2, 13, 16, 18, 21, 22, 23, 24, 25] Two national studies used the National Hospital Ambulatory Medical Care Survey, which includes data from both general and pediatric EDs.[13, 14] Factors identified to be associated with increased odds of returning were: young age, higher acuity, chronic conditions, and public insurance. One national study identified some diagnoses associated with higher likelihood of returning,[13] whereas the other focused primarily on infectious diseaserelated diagnoses.[14]

The purpose of this study was to describe the prevalence of return visits specifically to pediatric EDs and to investigate patient‐level, visit‐level, and healthcare systemrelated factors that may be associated with return visits and hospitalization at return.

METHODS

Study Design and Data Source

This retrospective cohort study used data from the Pediatric Health Information System (PHIS), an administrative database with data from 44 tertiary care pediatric hospitals in 27 US states and the District of Columbia. This database contains patient demographics, diagnoses, and procedures as well as medications, diagnostic imaging, laboratory, and supply charges for each patient. Data are deidentified prior to inclusion; encrypted medical record numbers allow for the identification of individual patients across all ED visits and hospitalizations to the same hospital. The Children's Hospital Association (Overland Park, KS) and participating hospitals jointly assure the quality and integrity of the data. This study was approved by the institutional review board at Boston Children's Hospital with a waiver for informed consent granted.

Study Population and Protocol

To standardize comparisons across the hospitals, we included data from 23 of the 44 hospitals in PHIS; 7 were excluded for not including ED‐specific data. For institutions that collect information from multiple hospitals within their healthcare system, we included only records from the main campus or children's hospital when possible, leading to the exclusion of 9 hospitals where the data were not able to be segregated. As an additional level of data validation, we compared the hospital‐level ED volume and admission rates as reported in the PHIS to those reported to a separate database (the Pediatric Analysis and Comparison Tool). We further excluded 5 hospitals whose volume differed by >10% between these 2 data sources.

Patients <18 years of age who were discharged from these EDs following their index visit in 2012 formed the eligible cohort.

Key Outcome Measures

The primary outcomes were return visits within 72 hours of discharge from the ED, and return visits resulting in hospitalization, including observation status. We defined an ED revisit as a return within 72 hours of ED discharge regardless of whether the patient was subsequently discharged from the ED on the return visit or hospitalized. We assessed revisits within 72 hours of an index ED discharge, because return visits within this time frame are likely to be related to the index visit.[2, 13, 16, 21, 22, 24, 25, 26]

Factors Associated With ED Revisits

A priori, we chose to adjust for the following patient‐level factors: age (<30 days, 30 days<1 year, 14 years, 511 years, 1217 years), gender, and socioeconomic status (SES) measured as the zip codebased median household income, obtained from the 2010 US Census, with respect to the federal poverty level (FPL) (<1.5 FPL, 1.52 FPL, 23 FPL, and >3 FPL).[27] We also adjusted for insurance type (commercial, government, or other), proximity of patient's home zip code to hospital (modeled as the natural log of the geographical distance to patient's home address from the hospital), ED diagnosis‐based severity classification system score (1=low severity, 5=high severity),[28] presence of a complex chronic condition at the index or prior visits using a validated classification scheme,[15, 29, 30, 31] and primary care physician (PCP) density per 100,000 in the patient's residential area (modeled as quartiles: very low, <57.2; low, 57.267.9; medium, 68.078.7; high, >78.8). PCP density, defined by the Dartmouth Atlas of Health Care,[32, 33, 34] is the number of primary care physicians per 100,000 residents (PCP count) in federal health service areas (HSA). Patients were assigned to a corresponding HSA based on their home zip code.

Visit‐level factors included arrival time of index visit (8:01 am 4:00 pm, 4:01 pm12:00 am, 12:01 am8 am representing day, evening, and overnight arrival, respectively), day of the week, season, length of stay (LOS) in the ED during the index visit, and ED crowding (calculated as the average daily LOS/yearly average LOS for the individual ED).[35] We categorized the ED primary diagnosis for each visit using the major diagnosis groupings of a previously described pediatric ED‐specific classification scheme.[36] Using International Classification of Diseases, Ninth Revision (ICD‐9) codes, we identified the conditions with the highest ED revisit rates.

Statistical Analyses

Categorical variables describing the study cohort were summarized using frequencies and percentages. Continuous variables were summarized using mean, median, and interquartile range values, where appropriate. We used 2 different hierarchical logistic regression models to assess revisit rates by patient‐ and visit‐level characteristics. The initial model included all patients discharged from the ED following the index visit and assessed for the outcome of a revisit within 72 hours. The second model considered only patients who returned within 72 hours of an index visit and assessed for hospitalization on that return visit. We used generalized linear mixed effects models, with hospital as a random effect to account for the presence of correlated data (within hospitals), nonconstant variability (across hospitals), and binary responses. Adjusted odds ratios with 95% confidence intervals were used as summary measures of the effect of the individual adjusters. Adjusters were missing in fewer than 5% of patients across participating hospitals. Statistical analyses were performed using SAS version 9.3 (SAS Institute Inc., Cary, NC); 2‐sided P values <0.004 were considered statistically significant to account for multiple comparisons (Bonferroni‐adjusted level of significance=0.0038).

RESULTS

Patients

A total of 1,610,201 patients <18 years of age evaluated across the 23 PHIS EDs in 2012 were included in the study. Twenty‐one of the 23 EDs have academic affiliations; 10 are located in the South, 6 in the Midwest, 5 in the West, and 2 in the Northeast region of the United States. The annual ED volume for these EDs ranged from 25,090 to 136,160 (median, 65,075; interquartile range, 45,28085,206). Of the total patients, 1,415,721 (87.9%) were discharged following the index visit and comprised our study cohort. Of these patients, 47,294 (revisit rate: 3.3%) had an ED revisit within 72 hours. There were 4015 patients (0.3%) who returned more than once within 72 hours, and the largest proportion of these returned with infection‐related conditions. Of those returning, 37,999 (80.3%) were discharged again, whereas 9295 (19.7%) were admitted to the hospital (Figure 1). The demographic and clinical characteristics of study participants are displayed in Table 1.

jhm2273-fig-0001-m.png
Patient disposition from the emergency departments of study hospitals (n = 23) in 2012.
Characteristics of Patients Who Returned Within 72 Hours of ED Discharge to the Study EDs
 Index Visit, n=1,415,721, n (%)Return Visits Within 72 Hours of Discharge, n=47,294, 3.3%
Return to Discharge, n (%)Return to Admission, n (%)
  • NOTE: Abbreviations: CCC, complex chronic condition; ED, emergency department; FPL, federal poverty level; IQR, interquartile range; LOS, length of stay.

  • Socioeconomic status is relative to the federal poverty level for a family of 4.

Gender, female659,417 (46.6)17,665 (46.5)4,304 (46.3)
Payor   
Commercial379,403 (26.8)8,388 (22.1)3,214 (34.6)
Government925,147 (65.4)26,880 (70.7)5,786 (62.3)
Other111,171 (7.9)2,731 (7.2)295 (3.2)
Age   
<30 days19,217 (1.4)488 (1.3)253 (2.7)
30 days to <1 year216,967 (15.3)8,280 (21.8)2,372 (25.5)
1 year to 4 years547,083 (38.6)15,542 (40.9)3,187 (34.3)
5 years to 11 years409,463 (28.9)8,906 (23.4)1,964 (21.1)
12 years to 17 years222,991 (15.8)4,783 (12.6)1,519 (16.3)
Socioeconomic statusa   
<1.5 times FPL493,770 (34.9)13,851 (36.5)2,879 (31.0)
1.5 to 2 times FPL455,490 (32.2)12,364 (32.5)2,904 (31.2)
2 to 3 times FPL367,557 (26.0)9,560 (25.2)2,714 (29.2)
>3 times FPL98,904 (7.0)2,224 (5.9)798 (8.6)
Primary care physician density per 100,000 patients   
Very low351,798 (24.9)8,727 (23.0)2,628 (28.3)
Low357,099 (25.2)9,810 (25.8)2,067 (22.2)
Medium347,995 (24.6)10,186 (26.8)2,035 (21.9)
High358,829 (25.4)9,276 (24.4)2,565 (27.6)
CCC present, yes125,774 (8.9)4,446 (11.7)2,825 (30.4)
Severity score   
Low severity (0,1,2)721,061 (50.9)17,310 (45.6)2,955 (31.8)
High severity (3,4,5)694,660 (49.1)20,689 (54.5)6,340 (68.2)
Time of arrival   
Day533,328 (37.7)13,449 (35.4)3,396 (36.5)
Evening684,873 (48.4)18,417 (48.5)4,378 (47.1)
Overnight197,520 (14.0)6,133 (16.1)1,521 (16.4)
Season   
Winter384,957 (27.2)10,603 (27.9)2,844 (30.6)
Spring367,434 (26.0)9,923 (26.1)2,311 (24.9)
Summer303,872 (21.5)8,308 (21.9)1,875 (20.2)
Fall359,458 (25.4)9,165 (24.1)2,265 (24.4)
Weekday/weekend   
Monday217,774 (15.4)5,646 (14.9)1,394 (15)
Tuesday198,220 (14.0)5,054 (13.3)1,316 (14.2)
Wednesday194,295 (13.7)4,985 (13.1)1,333 (14.3)
Thursday191,950 (13.6)5,123 (13.5)1,234 (13.3)
Friday190,022 (13.4)5,449 (14.3)1,228 (13.2)
Saturday202,247 (14.3)5,766 (15.2)1,364 (14.7)
Sunday221,213 (15.6)5,976 (15.7)1,426 (15.3)
Distance from hospital in miles, median (IQR)8.3 (4.614.9)9.2 (4.917.4)8.3 (4.614.9)
ED crowding score at index visit, median (IQR)1.0 (0.91.1)1.0 (0.91.1)1.0 (0.91.1)
ED LOS in hours at index visit, median (IQR)2.0 (1.03.0)3.0 (2.05.0)2.0 (1.03.0)

ED Revisit Rates and Revisits Resulting in Admission

In multivariate analyses, compared to patients who did not return to the ED, patients who returned within 72 hours of discharge had higher odds of revisit if they had the following characteristics: a chronic condition, were <1 year old, a higher severity score, and public insurance. Visit‐level factors associated with higher odds of revisits included arrival for the index visit during the evening or overnight shift or on a Friday or Saturday, index visit during times of lower ED crowding, and living closer to the hospital. On return, patients were more likely to be hospitalized if they had a higher severity score, a chronic condition, private insurance, or were <30 days old. Visit‐level factors associated with higher odds of hospitalization at revisit included an index visit during the evening and overnight shift and living further from the hospital. Although the median SES and PCP density of a patient's area of residence were not associated with greater likelihood of returning, when they returned, patients residing in an area with a lower SES and higher PCP densities (>78.8 PCPs/100,000) had lower odds of being admitted to the hospital. Patients whose index visit was on a Sunday also had lower odds of being hospitalized upon return (Table 2).

Multivariate Analyses of Factors Associated With ED Revisits and Admission at Return
CharacteristicAdjusted OR of 72‐Hour Revisit (95% CI), n=1,380,723P ValueAdjusted OR of 72‐Hour Revisit Admissions (95% CI), n=46,364P Value
  • NOTE: Effects of continuous variables are assessed as 1‐unit offsets from the mean. Abbreviations: CCC, complex chronic condition; CI, confidence interval; ED, emergency department; FPL, federal poverty level; LOS, length of stay; OR, odds ratio, NA, not applicable.

  • Socioeconomic status is relative to the FPL for a family of 4.

  • ED crowding score and LOS are based on index visit. ED crowding score is calculated as the daily LOS (in hours)/overall LOS (in hours). Overall average across hospitals=1; a 1‐ unit increase translates into twice the duration for the daily LOS over the yearly average ED LOS.

  • Modeled as the natural log of the patient geographic distance from the hospital based on zip codes. Number in parentheses represents the exponential of the modeled variable.

Gender    
Male0.99 (0.971.01)0.28091.02 (0.971.07)0.5179
FemaleReference Reference 
Payor    
Government1.14 (1.111.17)<0.00010.68 (0.640.72)<0.0001
Other0.97 (0.921.01)0.11480.33 (0.280.39)<0.0001
PrivateReference Reference 
Age group    
30 days to <1 year1.32 (1.221.42)<0.00010.58 (0.490.69)<0.0001
1 year to 5 years0.89 (0.830.96)0.0030.41 (0.340.48)<0.0001
5 years to 11 years0.69 (0.640.74)<0.00010.40 (0.330.48)<0.0001
12 years to 17 years0.72 (0.660.77)<0.00010.50 (0.420.60)<0.0001
<30 daysReference Reference 
Socioeconomic statusa    
% <1.5 times FPL0.96 (0.921.01)0.09920.82 (0.740.92)0.0005
% 1.5 to 2 times FPL0.98 (0.941.02)0.29920.83 (0.750.92)0.0005
% 2 to 3 times FPL1.02 (0.981.07)0.2920.88 (0.790.97)0.01
% >3 times FPLReference Reference 
Severity score    
High severity, 4, 5, 61.43 (1.401.45)<0.00013.42 (3.233.62)<0.0001
Low severity, 1, 2, 3Reference Reference 
Presence of any CCC    
Yes1.90 (1.861.96)<0.00012.92 (2.753.10)<0.0001
NoReference Reference 
Time of arrival    
Evening1.05 (1.031.08)<0.00011.37 (1.291.44)<0.0001
Overnight1.19 (1.151.22)<0.00011.84 (1.711.97)<0.0001
DayReference Reference 
Season    
Winter1.09 (1.061.11)<0.00011.06 (0.991.14)0.0722
Spring1.07 (1.041.10)<0.00010.98 (0.911.046)0.4763
Summer1.05 (1.021.08)0.00110.93 (0.871.01)0.0729
FallReference Reference 
Weekday/weekend    
Thursday1.02 (0.9821.055)0.32970.983 (0.8971.078)0.7185
Friday1.08 (1.041.11)<0.00011.03 (0.941.13)0.5832
Saturday1.08 (1.041.12)<0.00010.89 (0.810.97)0.0112
Sunday1.02 (0.991.06)0.20540.81 (0.740.89)<0.0001
Monday1.00 (0.961.03)0.89280.98 (0.901.07)0.6647
Tuesday0.99 (0.951.03)0.53420.93 (0.851.02)0.1417
WednesdayReference Reference 
PCP ratio per 100,000 patients    
57.267.91.00 (0.961.04)0.88440.93 (0.841.03)0.1669
68.078.71.00 (0.951.04)0.81560.86 (0.770.96)0.0066
>78.81.00 (0.951.04)0.68830.82 (0.730.92)0.001
<57.2Reference Reference 
ED crowding score at index visitb    
20.92 (0.900.95)<0.00010.96 (0.881.05)0.3435
1Reference Reference 
Distance from hospitalc    
3.168, 23.6 miles0.95 (0.940.96)<0.00011.16 (1.121.19)<0.0001
2.168, 8.7 milesReference Reference 
ED LOS at index visitb    
3.7 hours1.003 (1.0011.005)0.0052NA 
2.7 hoursReference   

Diagnoses Associated With Return Visits

Patients with index visit diagnoses of sickle cell disease and leukemia had the highest proportion of return visits (10.7% and 7.3%, respectively). Other conditions with high revisit rates included infectious diseases such as cellulitis, bronchiolitis, and gastroenteritis. Patients with other chronic diseases such as diabetes and with devices, such as gastrostomy tubes, also had high rates of return visits. At return, the rate of hospitalization for these conditions ranged from a 1‐in‐6 chance of hospitalization for the diagnoses of a fever to a 1‐in‐2 chance of hospitalization for patients with sickle cell anemia (Table 3).

Major Diagnostic Subgroups With the Highest ED Revisit and Admission at Return Rates
Major Diagnostic SubgroupNo. of Index ED Visit Dischargesa72‐Hour Revisit, % (95% CI)Admitted on Return, % (95% CI)
  • NOTE: Abbreviations: CI, confidence interval; ED, emergency department; NOS, not otherwise specified.

  • Diagnoses with <500 index visits (ie, <2 visits per month across the 23 hospitals) or <30 revisits within entire study cohort excluded from analyses.

  • Most prevalent diagnoses as identified by International Classification of Diseases, Ninth Revision codes within specified major diagnostic subgroups: devices and complications of the circulatory system, complication of other vascular device, implant, and graft; other hematologic diseases, anemia NOS, neutropenia NOS, or thrombocytopenia NOS; other devices and complications, hemorrhage complicating a procedure; devices and complications of the gastrointestinal system, gastrostomy; other infectious diseases, perinatal infections.

Sickle cell anemia2,53110.7 (9.511.9)49.6 (43.755.6)
Neoplastic diseases, cancer5367.3 (5.19.5)36 (2151)
Infectious gastrointestinal diseases8027.2 (5.49.0)21 (1031)
Devices and complications of the circulatory systemb1,0336.9 (5.38.4)45 (3457)
Other hematologic diseasesb1,5386.1 (4.97.3)33 (2443)
Fever80,6265.9 (5.76.0)16.3 (15.217.3)
Dehydration7,3625.4 (5.25.5)34.6 (30.139)
Infectious respiratory diseases72,6525.4 (5.25.5)28.6 (27.230)
Seizures17,6375.3 (4.95.6)33.3 (30.336.4)
Other devices and complicationsb1,8965.3 (4.36.3)39.0 (29.448.6)
Infectious skin, dermatologic and soft tissue diseases40,2724.7 (4.55)20.0 (18.221.8)
Devices and complications of the gastrointestinal systemb4,6924.6 (4.05.2)24.7 (18.930.4)
Vomiting44,7304.4 (4.24.6)23.7 (21.825.6)
Infectious urinary tract diseases17,0204.4 (4.14.7)25.9 (22.729)
Headache19,0164.3 (4.14.6)28.2 (25.131.3)
Diabetes mellitus1,5314.5 (3.35.3)29 (1840)
Abdominal pain39,5944.2 (44.4)24.8 (22.726.8)
Other infectious diseasesb6474.2 (2.65.7)33 (1651)
Gastroenteritis55,6134.0 (3.84.1)20.6 (18.922.3)

DISCUSSION

In this nationally representative sample of free‐standing children's hospitals, 3.3% of patients discharged from the ED returned to the same ED within 72 hours. This rate is similar to rates previously published in studies of general EDs.[11, 15] Of the returning children, over 80% were discharged again, and 19.7% were hospitalized, which is two‐thirds more than the admission rate at index visit (12%). In accordance with previous studies,[14, 16, 25] we found higher disease severity, presence of a chronic condition, and younger age were strongly associated with both the odds of patients returning to the ED and of being hospitalized at return. Patients who were hospitalized lived further away from the hospital and were of a higher SES. In this study, we show that visit‐level and access‐related factors are also associated with increased risk of return, although to a lesser degree. Patients seen on a weekend (Friday or Saturday) were found to have higher odds of returning, whereas those seen initially on a Sunday had lower odds of hospitalization at return. In this study, we also found that patients seen on the evening or night shifts at the index presentation had a significant association with return visits and hospitalization at return. Additionally, we found that although PCP density was not associated with the odds of returning to the ED, patients from areas with a higher PCP density were less likely to be admitted at return. In addition, by evaluating the diagnoses of patients who returned, we found that many infectious conditions commonly seen in the ED also had high return rates.

As previously shown,[23] we found that patients with complex and chronic diseases were at risk for ED revisits, especially patients with sickle cell anemia and cancer (mainly acute leukemia). In addition, patients with a chronic condition were 3 times more likely to be hospitalized when they returned. These findings may indicate an opportunity for improved discharge planning and coordination of care with subspecialty care providers for particularly at‐risk populations, or stronger consideration of admission at the index visit. However, admission for these patients at revisit may be unavoidable.

Excluding patients with chronic and complex conditions, the majority of conditions with high revisit rates were acute infectious conditions. One national study showed that >70% of ED revisits by patients with infectious conditions had planned ED follow‐up.[13] Although this study was unable to assess the reasons for return or admission at return, children with infectious diseases often worsen over time (eg, those with bronchiolitis). The relatively low admission rates at return for these conditions, despite evidence that providers may have a lower threshold for admission when a patient returns to the ED shortly after discharge,[24] may reflect the potential for improving follow‐up at the PCP office. However, although some revisits may be prevented,[37, 38] we recognize that an ED visit could be appropriate and necessary for some of these children, especially those without primary care.

Access to primary care and insurance status influence ED utilization.[14, 39, 40, 41] A fragmented healthcare system with poor access to primary care is strongly associated with utilization of the ED for nonurgent care. A high ED revisit rate might be indicative of poor coordination between ED and outpatient services.[9, 39, 42, 43, 44, 45, 46] Our study's finding of increased risk of return visit if the index visit occurred on a Friday or Saturday, and a decreased likelihood of subsequent admission when a patient returns on a Sunday, may suggest limited or perceived limited access to the PCP over a weekend. Although insured patients tend to use the ED less often for nonemergent cases, even when patients have PCPs, they might still choose to return to the ED out of convenience.[47, 48] This may be reflected in our finding that, when adjusted for insurance status and PCP density, patients who lived closer to the hospital were more likely to return, but less likely to be admitted, thereby suggesting proximity as a factor in the decision to return. It is also possible that patients residing further away returned to another institution. Although PCP density did not seem to be associated with revisits, patients who lived in areas with higher PCP density were less likely to be admitted when they returned. In this study, there was a stepwise gradient in the effect of PCP density on the odds of being hospitalized on return with those patients in areas with fewer PCPs being admitted at higher rates on return. Guttmann et al.,[40] in a recent study conducted in Canada where there is universal health insurance, showed that children residing in areas with higher PCP densities had higher rates of PCP visits but lower rates of ED visits compared to children residing in areas with lower PCP densities. It is possible that emergency physicians have more confidence that patients will have dedicated follow‐up when a PCP can be identified. These findings suggest that the development of PCP networks with expanded access, such as alignment of office hours with parent need and patient/parent education about PCP availability, may reduce ED revisits. Alternatively, creation of centralized hospital‐based urgent care centers for evening, night, and weekend visits may benefit both the patient and the PCP and avoid ED revisits and associated costs.

Targeting and eliminating disparities in care might also play a role in reducing ED revisits. Prior studies have shown that publicly insured individuals, in particular, frequently use the ED as their usual source of care and are more likely to return to the ED within 72 hours of an initial visit.[23, 39, 44, 49, 50] Likewise, we found that patients with public insurance were more likely to return but less likely to be admitted on revisit. After controlling for disease severity and other demographic variables, patients with public insurance and of lower socioeconomic status still had lower odds of being hospitalized following a revisit. This might also signify an increase of avoidable hospitalizations among patients of higher SES or with private insurance. Further investigation is needed to explore the reasons for these differences and to identify effective interventions to eliminate disparities.

Our findings have implications for emergency care, ambulatory care, and the larger healthcare system. First, ED revisits are costly and contribute to already overburdened EDs.[10, 11] The average ED visit incurs charges that are 2 to 5 times more than an outpatient office visit.[49, 50] Careful coordination of ambulatory and ED services could not only ensure optimal care for patients, but could save the US healthcare system billions of dollars in potentially avoidable healthcare expenditures.[49, 50] Second, prior studies have demonstrated a consistent relationship between poor access to primary care and increased use of the ED for nonurgent conditions.[42] Publicly insured patients have been shown to have disproportionately increased difficulty acquiring and accessing primary care.[41, 42, 47, 51] Furthermore, conditions with high ED revisit rates are similar to conditions reported by Berry et al.4 as having the highest hospital readmission rates such as cancer, sickle cell anemia, seizure, pneumonia, asthma, and gastroenteritis. This might suggest a close relationship between 72‐hour ED revisits and 30‐day hospital readmissions. In light of the recent expansion of health insurance coverage to an additional 30 million individuals, the need for better coordination of services throughout the entire continuum of care, including primary care, ED, and inpatient services, has never been more important.[52] Future improvements could explore condition‐specific revisit or readmission rates to identify the most effective interventions to reduce the possibly preventable returns.

This study has several limitations. First, as an administrative database, PHIS has limited clinical data, and reasons for return visits could not be assessed. Variations between hospitals in diagnostic coding might also lead to misclassification bias. Second, we were unable to assess return visits to a different ED. Thus, we may have underestimated revisit frequency. However, because children are generally more likely to seek repeat care in the same hospital,[3] we believe our estimate of return visit rate approximates the actual return visit rate; our findings are also similar to previously reported rates. Third, for the PCP density factor, we were unable to account for types of insurance each physician accepted and influence on return rates. Fourth, return visits in our sample could have been for conditions unrelated to the diagnosis at index visit, though the short timeframe considered for revisits makes this less likely. In addition, the crowding index does not include the proportion of occupied beds at the precise moment of the index visit. Finally, this cohort includes only children seen in the EDs of pediatric hospitals, and our findings may not be generalizable to all EDs who provide care for ill and injured children.

We have shown that, in addition to previously identified patient level factors, there are visit‐level and access‐related factors associated with pediatric ED return visits. Eighty percent are discharged again, and almost one‐fifth of returning patients are admitted to the hospital. Admitted patients tend to be younger, sicker, chronically ill, and live farther from the hospital. By being aware of patients' comorbidities, PCP access, as well as certain diagnoses associated with high rates of return, physicians may better target interventions to optimize care. This may include having a lower threshold for hospitalization at the initial visit for children at high risk of return, and communication with the PCP at the time of discharge to ensure close follow‐up. Our study helps to provide benchmarks around ED revisit rates, and may serve as a starting point to better understand variation in care. Future efforts should aim to find creative solutions at individual institutions, with the goal of disseminating and replicating successes more broadly. For example, investigators in Boston have shown that the use of a comprehensive home‐based asthma management program has been successful in decreasing emergency department visits and hospitalization rates.[53] It is possible that this approach could be spread to other institutions to decrease revisits for patients with asthma. As a next step, the authors have undertaken an investigation to identify hospital‐level characteristics that may be associated with rates of return visits.

Acknowledgements

The authors thank the following members of the PHIS ED Return Visits Research Group for their contributions to the data analysis plan and interpretation of results of this study: Rustin Morse, MD, Children's Medical Center of Dallas; Catherine Perron, MD, Boston Children's Hospital; John Cheng, MD, Children's Healthcare of Atlanta; Shabnam Jain, MD, MPH, Children's Healthcare of Atlanta; and Amanda Montalbano, MD, MPH, Children's Mercy Hospitals and Clinics. These contributors did not receive compensation for their help with this work.

Disclosures

A.T.A. and A.M.S. conceived the study and developed the initial study design. All authors were involved in the development of the final study design and data analysis plan. C.W.T. collected and analyzed the data. A.T.A. and C.W.T. had full access to all of the data and take responsibility for the integrity of the data and the accuracy of the data analysis. All authors were involved in the interpretation of the data. A.T.A. drafted the article, and all authors made critical revisions to the initial draft and subsequent versions. A.T.A. and A.M.S. take full responsibility for the article as a whole. The authors report no conflicts of interest.

References
  1. Joint policy statement—guidelines for care of children in the emergency department. Pediatrics. 2009;124:12331243.
  2. Alessandrini EA, Lavelle JM, Grenfell SM, Jacobstein CR, Shaw KN. Return visits to a pediatric emergency department. Pediatr Emerg Care. 2004;20:166171.
  3. Axon RN, Williams MV. Hospital readmission as an accountability measure. JAMA. 2011;305:504505.
  4. Berry JG, Hall DE, Kuo DZ, et al. Hospital utilization and characteristics of patients experiencing recurrent readmissions within children's hospitals. JAMA. 2011;305:682690.
  5. Berry JG, Toomey SL, Zaslavsky AM, et al. Pediatric readmission prevalence and variability across hospitals. JAMA. 2013;309:372380.
  6. Carrns A. Farewell, and don't come back. Health reform gives hospitals a big incentive to send patients home for good. US News World Rep. 2010;147:20, 2223.
  7. Coye MJ. CMS' stealth health reform. Plan to reduce readmissions and boost the continuum of care. Hosp Health Netw. 2008;82:24.
  8. Lerman B, Kobernick MS. Return visits to the emergency department. J Emerg Med. 1987;5:359362.
  9. Rising KL, White LF, Fernandez WG, Boutwell AE. Emergency department visits after hospital discharge: a missing part of the equation. Ann Emerg Med. 2013;62:145150.
  10. Stang AS, Straus SE, Crotts J, Johnson DW, Guttmann A. Quality indicators for high acuity pediatric conditions. Pediatrics. 2013;132:752762.
  11. Fontanarosa PB, McNutt RA. Revisiting hospital readmissions. JAMA. 2013;309:398400.
  12. Vaduganathan M, Bonow RO, Gheorghiade M. Thirty‐day readmissions: the clock is ticking. JAMA. 2013;309:345346.
  13. Adekoya N. Patients seen in emergency departments who had a prior visit within the previous 72 h‐National Hospital Ambulatory Medical Care Survey, 2002. Public Health. 2005;119:914918.
  14. Cho CS, Shapiro DJ, Cabana MD, Maselli JH, Hersh AL. A national depiction of children with return visits to the emergency department within 72 hours, 2001–2007. Pediatr Emerg Care. 2012;28:606610.
  15. Feudtner C, Levin JE, Srivastava R, et al. How well can hospital readmission be predicted in a cohort of hospitalized children? A retrospective, multicenter study. Pediatrics. 2009;123:286293.
  16. Goldman RD, Ong M, Macpherson A. Unscheduled return visits to the pediatric emergency department‐one‐year experience. Pediatr Emerg Care. 2006;22:545549.
  17. Klein‐Kremer A, Goldman RD. Return visits to the emergency department among febrile children 3 to 36 months of age. Pediatr Emerg Care. 2011;27:11261129.
  18. LeDuc K, Rosebrook H, Rannie M, Gao D. Pediatric emergency department recidivism: demographic characteristics and diagnostic predictors. J Emerg Nurs. 2006;32:131138.
  19. Healthcare Cost and Utilization Project. Pediatric emergency department visits in community hospitals from selected states, 2005. Statistical brief #52. Available at: http://www.ncbi.nlm.nih.gov/books/NBK56039. Accessed October 3, 2013.
  20. Sharma V, Simon SD, Bakewell JM, Ellerbeck EF, Fox MH, Wallace DD. Factors influencing infant visits to emergency departments. Pediatrics. 2000;106:10311039.
  21. Ali AB, Place R, Howell J, Malubay SM. Early pediatric emergency department return visits: a prospective patient‐centric assessment. Clin Pediatr (Phila). 2012;51:651658.
  22. Hu KW, Lu YH, Lin HJ, Guo HR, Foo NP. Unscheduled return visits with and without admission post emergency department discharge. J Emerg Med. 2012;43:11101118.
  23. Jacobstein CR, Alessandrini EA, Lavelle JM, Shaw KN. Unscheduled revisits to a pediatric emergency department: risk factors for children with fever or infection‐related complaints. Pediatr Emerg Care. 2005;21:816821.
  24. Sauvin G, Freund Y, Saidi K, Riou B, Hausfater P. Unscheduled return visits to the emergency department: consequences for triage. Acad Emerg Med. 2013;20:3339.
  25. Zimmerman DR, McCarten‐Gibbs KA, DeNoble DH, et al. Repeat pediatric visits to a general emergency department. Ann Emerg Med. 1996;28:467473.
  26. Keith KD, Bocka JJ, Kobernick MS, Krome RL, Ross MA. Emergency department revisits. Ann Emerg Med. 1989;18:964968.
  27. US Department of Health 19:7078.
  28. Feudtner C, Christakis DA, Connell FA. Pediatric deaths attributable to complex chronic conditions: a population‐based study of Washington State, 1980–1997. Pediatrics. 2000;106:205209.
  29. Feudtner C, Hays RM, Haynes G, Geyer JR, Neff JM, Koepsell TD. Deaths attributed to pediatric complex chronic conditions: national trends and implications for supportive care services. Pediatrics. 2001;107:E99.
  30. Feudtner C, Silveira MJ, Christakis DA. Where do children with complex chronic conditions die? Patterns in Washington State, 1980–1998. Pediatrics. 2002;109:656660.
  31. Dartmouth Atlas of Health Care. Hospital and physician capacity, 2006. Available at: http://www.dartmouthatlas.org/data/topic/topic.aspx?cat=24. Accessed October 7, 2013.
  32. Dartmouth Atlas of Health Care. Research methods. What is an HSA/HRR? Available at: http://www.dartmouthatlas.org/tools/faq/researchmethods.aspx. Accessed October 7, 2013,.
  33. Dartmouth Atlas of Health Care. Appendix on the geography of health care in the United States. Available at: http://www.dartmouthatlas.org/downloads/methods/geogappdx.pdf. Accessed October 7, 2013.
  34. Beniuk K, Boyle AA, Clarkson PJ. Emergency department crowding: prioritising quantified crowding measures using a Delphi study. Emerg Med J. 2012;29:868871.
  35. Alessandrini EA, Alpern ER, Chamberlain JM, Shea JA, Gorelick MH. A new diagnosis grouping system for child emergency department visits. Acad Emerg Med. 2010;17:204213.
  36. Guttmann A, Zagorski B, Austin PC, et al. Effectiveness of emergency department asthma management strategies on return visits in children: a population‐based study. Pediatrics. 2007;120:e1402e1410.
  37. Horwitz DA, Schwarz ES, Scott MG, Lewis LM. Emergency department patients with diabetes have better glycemic control when they have identifiable primary care providers. Acad Emerg Med. 2012;19:650655.
  38. Billings J, Zeitel L, Lukomnik J, Carey TS, Blank AE, Newman L. Impact of socioeconomic status on hospital use in New York City. Health Aff (Millwood). 1993;12:162173.
  39. Guttmann A, Shipman SA, Lam K, Goodman DC, Stukel TA. Primary care physician supply and children's health care use, access, and outcomes: findings from Canada. Pediatrics. 2010;125:11191126.
  40. Asplin BR, Rhodes KV, Levy H, et al. Insurance status and access to urgent ambulatory care follow‐up appointments. JAMA. 2005;294:12481254.
  41. Kellermann AL, Weinick RM. Emergency departments, Medicaid costs, and access to primary care—understanding the link. N Engl J Med. 2012;366:21412143.
  42. Committee on the Future of Emergency Care in the United States Health System. Emergency Care for Children: Growing Pains. Washington, DC: The National Academies Press; 2007.
  43. Committee on the Future of Emergency Care in the United States Health System. Hospital‐Based Emergency Care: At the Breaking Point. Washington, DC: The National Academies Press; 2007.
  44. Radley DC, Schoen C. Geographic variation in access to care—the relationship with quality. N Engl J Med. 2012;367:36.
  45. Tang N, Stein J, Hsia RY, Maselli JH, Gonzales R. Trends and characteristics of US emergency department visits, 1997–2007. JAMA. 2010;304:664670.
  46. Young GP, Wagner MB, Kellermann AL, Ellis J, Bouley D. Ambulatory visits to hospital emergency departments. Patterns and reasons for use. 24 Hours in the ED Study Group. JAMA. 1996;276:460465.
  47. Tranquada KE, Denninghoff KR, King ME, Davis SM, Rosen P. Emergency department workload increase: dependence on primary care? J Emerg Med. 2010;38:279285.
  48. Network for Excellence in Health Innovation. Leading healthcare research organizations to examine emergency department overuse. New England Research Institute, 2008. Available at: http://www.nehi.net/news/310‐leading‐health‐care‐research‐organizations‐to‐examine‐emergency‐department‐overuse/view. Accessed October 4, 2013.
  49. Robert Wood Johnson Foundation. Quality field notes: reducing inappropriate emergency department use. Available at: http://www.rwjf.org/en/research‐publications/find‐rwjf‐research/2013/09/quality‐field‐notes–reducing‐inappropriate‐emergency‐department.html.
  50. Access of Medicaid recipients to outpatient care. N Engl J Med. 1994;330:14261430.
  51. Medicaid policy statement. Pediatrics. 2013;131:e1697e1706.
  52. Woods ER, Bhaumik U, Sommer SJ, et al. Community asthma initiative: evaluation of a quality improvement program for comprehensive asthma care. Pediatrics. 2012;129:465472.
References
  1. Joint policy statement—guidelines for care of children in the emergency department. Pediatrics. 2009;124:12331243.
  2. Alessandrini EA, Lavelle JM, Grenfell SM, Jacobstein CR, Shaw KN. Return visits to a pediatric emergency department. Pediatr Emerg Care. 2004;20:166171.
  3. Axon RN, Williams MV. Hospital readmission as an accountability measure. JAMA. 2011;305:504505.
  4. Berry JG, Hall DE, Kuo DZ, et al. Hospital utilization and characteristics of patients experiencing recurrent readmissions within children's hospitals. JAMA. 2011;305:682690.
  5. Berry JG, Toomey SL, Zaslavsky AM, et al. Pediatric readmission prevalence and variability across hospitals. JAMA. 2013;309:372380.
  6. Carrns A. Farewell, and don't come back. Health reform gives hospitals a big incentive to send patients home for good. US News World Rep. 2010;147:20, 2223.
  7. Coye MJ. CMS' stealth health reform. Plan to reduce readmissions and boost the continuum of care. Hosp Health Netw. 2008;82:24.
  8. Lerman B, Kobernick MS. Return visits to the emergency department. J Emerg Med. 1987;5:359362.
  9. Rising KL, White LF, Fernandez WG, Boutwell AE. Emergency department visits after hospital discharge: a missing part of the equation. Ann Emerg Med. 2013;62:145150.
  10. Stang AS, Straus SE, Crotts J, Johnson DW, Guttmann A. Quality indicators for high acuity pediatric conditions. Pediatrics. 2013;132:752762.
  11. Fontanarosa PB, McNutt RA. Revisiting hospital readmissions. JAMA. 2013;309:398400.
  12. Vaduganathan M, Bonow RO, Gheorghiade M. Thirty‐day readmissions: the clock is ticking. JAMA. 2013;309:345346.
  13. Adekoya N. Patients seen in emergency departments who had a prior visit within the previous 72 h‐National Hospital Ambulatory Medical Care Survey, 2002. Public Health. 2005;119:914918.
  14. Cho CS, Shapiro DJ, Cabana MD, Maselli JH, Hersh AL. A national depiction of children with return visits to the emergency department within 72 hours, 2001–2007. Pediatr Emerg Care. 2012;28:606610.
  15. Feudtner C, Levin JE, Srivastava R, et al. How well can hospital readmission be predicted in a cohort of hospitalized children? A retrospective, multicenter study. Pediatrics. 2009;123:286293.
  16. Goldman RD, Ong M, Macpherson A. Unscheduled return visits to the pediatric emergency department‐one‐year experience. Pediatr Emerg Care. 2006;22:545549.
  17. Klein‐Kremer A, Goldman RD. Return visits to the emergency department among febrile children 3 to 36 months of age. Pediatr Emerg Care. 2011;27:11261129.
  18. LeDuc K, Rosebrook H, Rannie M, Gao D. Pediatric emergency department recidivism: demographic characteristics and diagnostic predictors. J Emerg Nurs. 2006;32:131138.
  19. Healthcare Cost and Utilization Project. Pediatric emergency department visits in community hospitals from selected states, 2005. Statistical brief #52. Available at: http://www.ncbi.nlm.nih.gov/books/NBK56039. Accessed October 3, 2013.
  20. Sharma V, Simon SD, Bakewell JM, Ellerbeck EF, Fox MH, Wallace DD. Factors influencing infant visits to emergency departments. Pediatrics. 2000;106:10311039.
  21. Ali AB, Place R, Howell J, Malubay SM. Early pediatric emergency department return visits: a prospective patient‐centric assessment. Clin Pediatr (Phila). 2012;51:651658.
  22. Hu KW, Lu YH, Lin HJ, Guo HR, Foo NP. Unscheduled return visits with and without admission post emergency department discharge. J Emerg Med. 2012;43:11101118.
  23. Jacobstein CR, Alessandrini EA, Lavelle JM, Shaw KN. Unscheduled revisits to a pediatric emergency department: risk factors for children with fever or infection‐related complaints. Pediatr Emerg Care. 2005;21:816821.
  24. Sauvin G, Freund Y, Saidi K, Riou B, Hausfater P. Unscheduled return visits to the emergency department: consequences for triage. Acad Emerg Med. 2013;20:3339.
  25. Zimmerman DR, McCarten‐Gibbs KA, DeNoble DH, et al. Repeat pediatric visits to a general emergency department. Ann Emerg Med. 1996;28:467473.
  26. Keith KD, Bocka JJ, Kobernick MS, Krome RL, Ross MA. Emergency department revisits. Ann Emerg Med. 1989;18:964968.
  27. US Department of Health 19:7078.
  28. Feudtner C, Christakis DA, Connell FA. Pediatric deaths attributable to complex chronic conditions: a population‐based study of Washington State, 1980–1997. Pediatrics. 2000;106:205209.
  29. Feudtner C, Hays RM, Haynes G, Geyer JR, Neff JM, Koepsell TD. Deaths attributed to pediatric complex chronic conditions: national trends and implications for supportive care services. Pediatrics. 2001;107:E99.
  30. Feudtner C, Silveira MJ, Christakis DA. Where do children with complex chronic conditions die? Patterns in Washington State, 1980–1998. Pediatrics. 2002;109:656660.
  31. Dartmouth Atlas of Health Care. Hospital and physician capacity, 2006. Available at: http://www.dartmouthatlas.org/data/topic/topic.aspx?cat=24. Accessed October 7, 2013.
  32. Dartmouth Atlas of Health Care. Research methods. What is an HSA/HRR? Available at: http://www.dartmouthatlas.org/tools/faq/researchmethods.aspx. Accessed October 7, 2013,.
  33. Dartmouth Atlas of Health Care. Appendix on the geography of health care in the United States. Available at: http://www.dartmouthatlas.org/downloads/methods/geogappdx.pdf. Accessed October 7, 2013.
  34. Beniuk K, Boyle AA, Clarkson PJ. Emergency department crowding: prioritising quantified crowding measures using a Delphi study. Emerg Med J. 2012;29:868871.
  35. Alessandrini EA, Alpern ER, Chamberlain JM, Shea JA, Gorelick MH. A new diagnosis grouping system for child emergency department visits. Acad Emerg Med. 2010;17:204213.
  36. Guttmann A, Zagorski B, Austin PC, et al. Effectiveness of emergency department asthma management strategies on return visits in children: a population‐based study. Pediatrics. 2007;120:e1402e1410.
  37. Horwitz DA, Schwarz ES, Scott MG, Lewis LM. Emergency department patients with diabetes have better glycemic control when they have identifiable primary care providers. Acad Emerg Med. 2012;19:650655.
  38. Billings J, Zeitel L, Lukomnik J, Carey TS, Blank AE, Newman L. Impact of socioeconomic status on hospital use in New York City. Health Aff (Millwood). 1993;12:162173.
  39. Guttmann A, Shipman SA, Lam K, Goodman DC, Stukel TA. Primary care physician supply and children's health care use, access, and outcomes: findings from Canada. Pediatrics. 2010;125:11191126.
  40. Asplin BR, Rhodes KV, Levy H, et al. Insurance status and access to urgent ambulatory care follow‐up appointments. JAMA. 2005;294:12481254.
  41. Kellermann AL, Weinick RM. Emergency departments, Medicaid costs, and access to primary care—understanding the link. N Engl J Med. 2012;366:21412143.
  42. Committee on the Future of Emergency Care in the United States Health System. Emergency Care for Children: Growing Pains. Washington, DC: The National Academies Press; 2007.
  43. Committee on the Future of Emergency Care in the United States Health System. Hospital‐Based Emergency Care: At the Breaking Point. Washington, DC: The National Academies Press; 2007.
  44. Radley DC, Schoen C. Geographic variation in access to care—the relationship with quality. N Engl J Med. 2012;367:36.
  45. Tang N, Stein J, Hsia RY, Maselli JH, Gonzales R. Trends and characteristics of US emergency department visits, 1997–2007. JAMA. 2010;304:664670.
  46. Young GP, Wagner MB, Kellermann AL, Ellis J, Bouley D. Ambulatory visits to hospital emergency departments. Patterns and reasons for use. 24 Hours in the ED Study Group. JAMA. 1996;276:460465.
  47. Tranquada KE, Denninghoff KR, King ME, Davis SM, Rosen P. Emergency department workload increase: dependence on primary care? J Emerg Med. 2010;38:279285.
  48. Network for Excellence in Health Innovation. Leading healthcare research organizations to examine emergency department overuse. New England Research Institute, 2008. Available at: http://www.nehi.net/news/310‐leading‐health‐care‐research‐organizations‐to‐examine‐emergency‐department‐overuse/view. Accessed October 4, 2013.
  49. Robert Wood Johnson Foundation. Quality field notes: reducing inappropriate emergency department use. Available at: http://www.rwjf.org/en/research‐publications/find‐rwjf‐research/2013/09/quality‐field‐notes–reducing‐inappropriate‐emergency‐department.html.
  50. Access of Medicaid recipients to outpatient care. N Engl J Med. 1994;330:14261430.
  51. Medicaid policy statement. Pediatrics. 2013;131:e1697e1706.
  52. Woods ER, Bhaumik U, Sommer SJ, et al. Community asthma initiative: evaluation of a quality improvement program for comprehensive asthma care. Pediatrics. 2012;129:465472.
Issue
Journal of Hospital Medicine - 9(12)
Issue
Journal of Hospital Medicine - 9(12)
Page Number
779-787
Page Number
779-787
Publications
Publications
Article Type
Display Headline
Prevalence and predictors of return visits to pediatric emergency departments
Display Headline
Prevalence and predictors of return visits to pediatric emergency departments
Sections
Article Source

© 2014 Society of Hospital Medicine

Disallow All Ads
Correspondence Location
Address for correspondence and reprint requests: Anne Stack, MD, Division of Emergency Medicine, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115; Telephone: 617‐355‐6624; Fax: 617‐730‐4824; E‐mail: anne.stack@childrens.harvard.edu
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Article PDF Media
Image
Disable zoom
Off
Media Files
Image
Disable zoom
Off
Image
Disable zoom
Off
Image
Disable zoom
Off
Image
Disable zoom
Off
Image
Disable zoom
Off
Image
Disable zoom
Off
Image
Disable zoom
Off
Image
Disable zoom
Off
Image
Disable zoom
Off
Image
Disable zoom
Off
Image
Disable zoom
Off
Image
Disable zoom
Off
Image
Disable zoom
Off
Image
Disable zoom
Off
Image
Disable zoom
Off

Pneumonia Guideline Therapy Outcomes

Article Type
Changed
Sun, 05/21/2017 - 13:32
Display Headline
Hospital outcomes associated with guideline‐recommended antibiotic therapy for pediatric pneumonia

Community‐acquired pneumonia (CAP) is a common and serious infection in children. With more than 150,000 children requiring hospitalization annually, CAP is the fifth most prevalent and the second most costly diagnosis of all pediatric hospitalizations in the United States.[1, 2, 3]

In August 2011, the Pediatric Infectious Diseases Society (PIDS) and the Infectious Diseases Society of America (IDSA) published an evidence‐based guideline for the management of CAP in children. This guideline recommended that fully immunized children without underlying complications who require hospitalization receive an aminopenicillin as first‐line antibiotic therapy.[4] Additionally, the guideline recommends empirically adding a macrolide to an aminopenicillin when atypical pneumonia is a diagnostic consideration.

This recommendation was a substantial departure from practice for hospitals nationwide, as a multicenter study of children's hospitals (20052010) demonstrated that <10% of patients diagnosed with CAP received aminopenicillins as empiric therapy.[5] Since publication of the PIDS/IDSA guidelines, the use of aminopenicillins has increased significantly across institutions, but the majority of hospitalized patients still receive broad‐spectrum cephalosporin therapy for CAP.[6]

At baseline, 30% of patients hospitalized with CAP received guideline‐recommended antibiotic therapy at our institution. Through the use of quality‐improvement methods, the proportion of patients receiving guideline‐recommended therapy increased to 100%.[7] The objective of this study was to ensure that there were not unintended negative consequences to guideline implementation. Specifically, we sought to identify changes in length of stay (LOS), hospital costs, and treatment failures associated with use of guideline‐recommended antibiotic therapy for children hospitalized with uncomplicated CAP.

METHODS

Study Design and Study Population

This retrospective cohort study included children age 3 months to 18 years, hospitalized with CAP, between May 2, 2011 and July 30, 2012, at Cincinnati Children's Hospital Medical Center (CCHMC), a 512‐bed free‐standing children's hospital. The CCHMC Institutional Review Board approved this study with a waiver of informed consent.

Patients were eligible for inclusion if they were admitted to the hospital for inpatient or observation level care with a primary or secondary International Classification of Disease, 9th Revision discharge diagnosis code of pneumonia (480.02, 480.89, 481, 482.0, 482.30‐2, 482.41‐2, 482.83, 482.8990, 483.8, 484.3, 485, 486, 487.0) or effusion/empyema (510.0, 510.9, 511.01, 511.89, 513).[8] Patients with complex chronic conditions[9] were excluded. Medical records of eligible patients (n=260) were reviewed by 2 members of the study team to ensure that patients fell into the purview of the guideline. Patients who did not receive antibiotics (n=11) or for whom there was documented concern for aspiration (n=1) were excluded. Additionally, patients with immunodeficiency (n=1) or who had not received age‐appropriate vaccinations (n=2), and patients who required intensive care unit admission on presentation (n=17) or who had a complicated pneumonia, defined by presence of moderate or large pleural effusion at time of admission (n=8), were also excluded.[7] Finally, for patients with multiple pneumonia admissions, only the index visit was included; subsequent visits occurring within 30 days of discharge were considered readmissions.

Treatment Measure

The primary exposure of interest was empiric antibiotic therapy upon hospital admission. Antibiotic therapy was classified as guideline recommended or nonguideline recommended. Guideline‐recommended therapy was defined as follows:

  1. For children without drug allergies: ampicillin (200 mg/kg/day intravenously) or amoxicillin (90 mg/kg/day orally);
  2. For children with penicillin allergy: ceftriaxone (50100 mg/kg/day intravenously or intramuscularly) or cefdinir (14 mg/kg/day orally);
  3. For children with penicillin and cephalosporin allergy: clindamycin (40 mg/kg/day orally or intravenously); and
  4. Or azithromycin (10 mg/kg/day orally or intravenously on day 1) in combination with antibiotic category 1 or 2 or 3 above.

 

Outcome Measures

The primary outcomes examined were hospital LOS, total cost of hospitalization, and inpatient pharmacy costs. LOS was measured in hours and defined as the difference in time between departure from and arrival to the inpatient unit. Total cost of index hospitalization included both direct and indirect costs, obtained from the Centers for Medicare & Medicaid Services' Relative Value Units data for Current Procedural Terminology codes.[10]

Secondary outcomes included broadening of antibiotic therapy during the hospital course, pneumonia‐related emergency department (ED) revisits within 30 days, and pneumonia‐related inpatient readmissions within 30 days. Broadening of antibiotic therapy was defined as addition of a second antibiotic (eg, adding azithromycin on day 3 of hospitalization) or change in empiric antibiotic to a class with broader antimicrobial activity (eg, ampicillin to ceftriaxone) at any time during hospitalization. As our study population included only patients with uncomplicated pneumonia at the time of admission, this outcome was used to capture possible treatment failure. ED revisits and inpatient readmissions were reviewed by 3 investigators to identify pneumonia‐related visits. To encompass all possible treatment failures, all respiratory‐related complaints (eg, wheezing, respiratory distress) were considered as pneumonia‐related. Disagreements were resolved by group discussion.

Covariates

Severity of illness on presentation was evaluated using the Emergency Severity Index version 4,[11] abnormal vital signs on presentation (as defined by Pediatric Advanced Life Support age‐specific criteria[12]), and need for oxygen in the first 24 hours of hospitalization. Supplemental oxygen is administered for saturations <91% per protocol at our institution. The patient's highest Pediatric Early Warning Scale score[13] during hospitalization was used as a proxy for disease severity. Exam findings on presentation (eg, increased respiratory effort, rales, wheezing) were determined through chart review. Laboratory tests and radiologic imaging variables included complete blood cell count, blood culture, chest radiograph, chest ultrasound, and chest computed tomography. Abnormal white blood cell count was defined as <5000 or >15,000 cells/mL, the defined reference range for the CCHMC clinical laboratory.

Data Analysis

Continuous variables were described using median and interquartile range (IQR) and compared across groups using Wilcoxon rank sum test due to non‐normal distributions. Categorical variables were described by counts and frequencies and compared using the 2 test.

Multivariable linear regression analysis was performed to assess the independent effect of receipt of empiric guideline‐recommended antibiotic therapy on outcomes of LOS and costs while adjusting for covariates. As LOS and costs were non‐normally distributed, we logarithmically transformed these values to use as the dependent variables in our models. The resulting coefficients were back‐transformed to reflect the percent change in LOS and costs incurred between subjects who received empiric guideline therapy compared with those who did not.[14] Covariates were chosen a priori due to their clinical and biological relevance to the outcomes of LOS (eg, wheezing on presentation and need for supplemental oxygen), total cost of hospitalization (eg, LOS and need for repeat imaging), and inpatient pharmacy costs (eg, LOS and wheezing on presentation) (Table 1).

Cohort Characteristics
CharacteristicOverall Cohort, n=220Guideline Therapy, n=166Nonguideline Therapy, n=54P Value
  • NOTE: Abbreviations: CT, computed tomography; IQR, interquartile range; PEWS, Pediatric Early Warning Scale. *P<0.05.

Age, y, median (IQR)2.9 (1.36.3)2.5 (1.35.2)5.6 (2.38.8)<0.01*
Male, no. (%)122 (55.5%)89 (53.6%)33 (61.1%)0.34
Emergency Severity Index, no. (%)0.11
290 (40.9%)73 (44.0%)17 (31.5%) 
3116 (52.7%)85 (51.2%)31 (57.4%) 
414 (6.4%)8 (4.8%)6 (11.1%) 
Abnormal vital signs on presentation, no. (%)
Fever99 (45.0%)80 (48.2%)19 (35.2%)0.10
Tachycardia100 (45.5%)76 (45.8%)24 (44.4%)0.86
Tachypnea124 (56.4%)100 (60.2%)24 (44.4%)0.04*
Hypotension000 
Hypoxia27 (12.3%)24 (14.5%)3 (5.6%)0.08
Physical exam on presentation, no. (%)
Increased respiratory effort146 (66.4%)111 (66.9%)35 (64.8%)0.78
Distressed110 (50.0%)86 (51.8%)24 (44.4%)0.35
Retraction103 (46.8%)81 (48.8%)22 (40.7%)0.30
Grunting17 (7.7%)14 (8.4%)3 (5.6%)0.49
Nasal flaring19 (8.6%)17 (10.2%)2 (3.7%)0.14
Rales135 (61.4%)99 (59.6%)36 (66.7%)0.36
Wheeze91 (41.4%)66 (39.8%)25 (46.3%)0.40
Decreased breath sounds89 (40.5%)65 (39.2%)24 (44.4%)0.49
Dehydration21 (9.6%)13 (7.8%)8 (14.8%)0.13
PEWS 5 during admission, no. (%)43 (19.6%)34 (20.5%)9 (16.7%)0.54
Oxygen requirement in first 24 hours, no. (%)114 (51.8%)90 (53.6%)24 (46.2%)0.35
Complete blood count obtained, no. (%)99 (45.0%)72 (43.4%)27 (50.0%)0.40
Abnormal white blood cell count35 (35.7%)23 (32.4%)12 (44.4%)0.27
Blood culture obtained, no. (%)104 (47.3%)80 (48.2%)24 (44.4%)0.63
Positive2 (1.9%)1 (1.3%)1 (4.2%)0.36
Chest radiograph available, no. (%)214 (97.3%)161 (97.0%)53 (98.2%)0.65
Infiltrate178 (83.2%)139 (86.3%)39 (73.6%)0.03*
Bilateral29 (16.3%)20 (14.4%)9 (23.1%)0.19
Multilobar46 (25.8%)33 (23.7%)13 (33.3%)0.23
Effusion24 (11.2%)16 (9.9%)8 (15.1%)0.30
Additional imaging, no. (%)    
Repeat chest radiograph26 (11.8%)17 (10.2%)9 (16.7%)0.20
Chest ultrasound4 (1.8%)3 (1.8%)1 (1.9%)0.98
Chest CT2 (0.9%)1 (0.6%)1 (1.9%)0.40
Antibiotic, no. (%)   <0.01*
Aminopenicillin140 (63.6%)140 (84.3%)0 (0%) 
Third‐generation cephalosporin37 (16.8%)8 (4.8%)29 (53.7%) 
Macrolide monotherapy18 (8.2%)0 (0%)18 (33.3%) 
Clindamycin2 (0.9%)1 (0.6%)1 (1.9%) 
Levofloxacin1 (0.5%)0 (0%)1 (1.9%) 
Aminopenicillin+macrolide16 (7.3%)16 (9.6%)0 (0%) 
Cephalosporin+macrolide6 (2.7%)1 (0.6%)5 (9.3%) 

Secondary outcomes of broadened antibiotic therapy, ED revisits, and hospital readmissions were assessed using the Fisher exact test. Due to the small number of events, we were unable to evaluate these associations in models adjusted for potential confounders.

All analyses were performed with SAS version 9.3 (SAS Institute, Cary, NC), and P values <0.05 were considered significant.

RESULTS

Of the 220 unique patients included, 122 (55%) were male. The median age was 2.9 years (IQR: 1.36.3 years). Empiric guideline‐recommended therapy was prescribed to 168 (76%) patients (Table 1). Aminopenicillins were the most common guideline‐recommended therapy, accounting for 84% of guideline‐recommended antibiotics. An additional 10% of patients received the guideline‐recommended combination therapy with an aminopenicillin and a macrolide. Nonguideline‐recommended therapy included third‐generation cephalosporin antibiotics (54%) and macrolide monotherapy (33%).

Those who received empiric guideline‐recommended antibiotic therapy were similar to those who received nonguideline‐recommended therapy with respect to sex, Emergency Severity Index, physical exam findings on presentation, oxygen requirement in the first 24 hours, abnormal laboratory findings, presence of effusion on chest radiograph, and need for additional imaging (Table 1). However, patients in the guideline‐recommended therapy group were significantly younger (median 2.5 years vs 5.6 years, P0.01), more likely to have elevated respiratory rate on presentation (60.2% vs 44.4%, P=0.04), and more likely to have an infiltrate on chest radiograph (86.3% vs 73.6%, P=0.03) (Table 1). Patients who received nonguideline‐recommended macrolide monotherapy had a median age of 7.4 years (IQR: 5.89.8 years).

Median hospital LOS for the total cohort was 1.3 days (IQR: 0.91.9 days) (Table 2). There were no differences in LOS between patients who received and did not receive guideline‐recommended therapy in the unadjusted or the adjusted model (Table 3).

Unadjusted Outcomes
OutcomeGuideline Therapy, n=166Nonguideline Therapy, n=54P Value
  • NOTE: Abbreviations: IQR, interquartile range.

Length of stay, d, median (IQR)1.3 (0.91.9)1.3 (0.92.0)0.74
Total costs, median, (IQR)$4118 ($2,647$6,004)$4045 ($2,829$6,200)0.44
Pharmacy total costs, median, (IQR)$84 ($40$179)$106 ($58$217)0.12
Broadened therapy, no. (%)10 (6.0%)4 (7.4%)0.75
Emergency department revisit, no. (%)7 (4.2%)2 (3.7%)1.00
Readmission, no. (%)1 (0.6%)1 (1.9%)0.43
Univariate and Multivariate Analyses of Receipt of Empiric Guideline‐Recommended Therapy With Length of Stay, Total Costs, and Pharmacy Costs
OutcomeUnadjusted Coefficient (95% CI)Adjusted Coefficient (95% CI)Adjusted % Change in Outcome (95% CI)*
  • NOTE: Abbreviations: CI, confidence interval. *Negative adjusted percent change indicates decrease in outcome associated with guideline‐recommended therapy; positive adjusted percent change indicates increase in outcome associated with guideline‐recommended therapy. Model is adjusted for age, fever on presentation, tachypnea on presentation, wheezing on presentation, need for supplemental oxygen, Pediatric Early Warning Score 5, chest radiograph findings, and need for repeat imaging. Model is adjusted for age, wheezing on presentation, need for supplemental oxygen, Pediatric Early Warning Score 5, need for repeat imaging, and length of stay. Model is adjusted for age, wheezing on presentation, and length of stay.

Length of stay0.06 (0.27 to 0.15)0.06 (0.25 to 0.12)5.8 (22.1 to 12.8)
Total costs0.18 (0.40 to 0.04)0.11 (0.32 to 0.09)10.9 (27.4 to 9.4)
Pharmacy total costs0.44 (0.46 to 0.02)0.16 (0.57 to 0.24)14.8 (43.4 to 27.1)

Median total costs of the index hospitalization for the total cohort were $4097 (IQR: $2657$6054), with median inpatient pharmacy costs of $92 (IQR: $40$183) (Table 2). There were no differences in total or inpatient pharmacy costs for patients who received guideline‐recommended therapy compared with those who did not in unadjusted or adjusted analyses. Fourteen patients (6.4%) had antibiotic therapy broadened during hospitalization, 10 were initially prescribed guideline‐recommended therapy, and 4 were initially prescribed nonguideline‐recommended therapy (Table 4).

Clinical Details of Patients Who Had Antibiotic Therapy Broadened During Initial Hospitalization
Initial TherapyReasons for Antibiotic Change Identified From Chart Review
Guideline=10Ampicillin to ceftriaxone:
1 patient with clinical worsening
1 patient with coincident urinary tract infection due to resistant organism
4 patients without evidence of clinical worsening or documentation of rationale
Addition of a macrolide:
3 patients without evidence of clinical worsening or documentation of rationale
Addition of clindamycin:
1 patient with clinical worsening
Nonguideline=4Ceftriaxone to clindamycin:
1 patient with clinical worsening
Addition of a macrolide:
1 patient with clinical worsening
1 patients without evidence of clinical worsening or documentation of rationale
Addition of clindamycin:
1 patient with clinical worsening

Of the 9 pneumonia‐related ED revisits within 30 days of discharge, 7 occurred in patients prescribed empiric guideline‐recommended therapy (Table 5). No ED revisit resulted in hospital readmission or antibiotic change related to pneumonia. Two ED revisits resulted in new antibiotic prescriptions for diagnoses other than pneumonia.

Clinical Details of Patients With an Emergency Department Revisit or Inpatient Readmission Following Index Hospitalization
RevisitInitial TherapyDay PostdischargeClinical Symptoms at Return VisitClinical DiagnosisAntibiotic Prescription
  • NOTE: Abbreviations: ED, emergency department; IV, intravenous.

EDGuideline3Poor oral intake and feverPneumoniaContinued prior antibiotic
EDGuideline8Recurrent cough and feverResolving pneumoniaContinued prior antibiotic
EDGuideline13Follow‐upResolved pneumoniaNo further antibiotic
EDGuideline16Increased work of breathingReactive airway diseaseNo antibiotic
EDGuideline20Persistent coughViral illnessNo antibiotic
EDGuideline22Recurrent cough and congestionSinusitisAugmentin
EDGuideline26Increased work of breathingReactive airway diseaseNo antibiotic
EDNonguideline16Recurrent feverAcute otitis mediaAmoxicillin
EDNonguideline20Recurrent cough and feverViral illnessNo antibiotic
AdmissionGuideline3Increased work of breathingPneumoniaIV ampicillin
AdmissionNonguideline9Refusal to take oral clindamycinPneumoniaIV clindamycin

Two patients were readmitted for a pneumonia‐related illness within 30 days of discharge; 1 had received guideline‐recommended therapy (Table 5). Both patients were directly admitted to the inpatient ward without an associated ED visit. Antibiotic class was not changed for either patient upon readmission, despite the decision to convert to intravenous form.

DISCUSSION

In this retrospective cohort study, patients who received empiric guideline‐recommended antibiotic therapy on admission for CAP had no difference in LOS, total cost of hospitalization, or inpatient pharmacy costs compared with those who received therapy that varied from guideline recommendations. Our study suggests that prescribing narrow‐spectrum therapy and, in some circumstances, combination therapy, as recommended by the 2011 PIDS/IDSA pneumonia guideline, did not result in negative unintended consequences.

In our study, children receiving guideline‐recommended therapy were younger, more likely to have elevated respiratory rate on presentation, and more likely to have an infiltrate on chest radiograph. We hypothesize the age difference is a reflection of common use of nonguideline macrolide monotherapy in the older, school‐age child, as macrolides are commonly used for coverage of Mycoplasma pneumonia in older children with CAP.[15] Children receiving macrolide monotherapy were older than those receiving guideline‐recommended therapy (median age of 7.4 years and 2.5 years, respectively). We also hypothesize that some providers may prescribe macrolide monotherapy to children deemed less ill than expected for CAP (eg, normal percutaneous oxygen saturation). This hypothesis is supported by the finding that 60% of patients who had a normal respiratory rate and received nonguideline therapy were prescribed macrolide monotherapy. We did control for the characteristics that varied between the two treatment groups in our models to eliminate potential confounding.

One prior study evaluated the effects of guideline implementation in CAP. In evaluation of a clinical practice guideline that recommended ampicillin as first‐line therapy for CAP, no significant difference was found following guideline introduction in the number of treatment failures (defined as the need to broaden therapy or development of complicated pneumonia within 48 hours or 30‐day inpatient readmission).[16] Our study builds on these findings by directly comparing outcomes between recipients of guideline and nonguideline therapy, which was not done in the prestudy or poststudy design of prior work.[16] We believe that classifying patients based on empiric therapy received rather than timing of guideline introduction thoroughly examines the effect of following guideline recommendations. Additionally, outcomes other than treatment failures were examined in our study including LOS, costs, and ED revisits.

Our results are similar to other observational studies that compared narrow‐ and broad‐spectrum antibiotics for children hospitalized with CAP. Using administrative data, Williams et al. found no significant difference in LOS or cost between narrow‐ and broad‐spectrum intravenous antibiotic recipients ages 6 months to 18 years at 43 children's hospitals.[17] Queen et al. compared narrow‐ and broad‐spectrum antibiotic therapy for children ages 2 months to 18 years at 4 children's hospitals.[18] Differences in average daily cost were not significant, but children who received narrow‐spectrum antibiotics had a significantly shorter LOS. Finally, an observational study of 319 Israeli children <2 years of age found no significant difference in duration of oxygen requirement, LOS, or need for change in antibiotic therapy between the 66 children prescribed aminopenicillins and the 253 children prescribed cefuroxime.[19] These studies suggest that prescribing narrow‐spectrum antimicrobial therapy, as per the guideline recommendations, results in similar outcomes to broad‐spectrum antimicrobial therapy.

Our study adds to prior studies that compared narrow‐ and broad‐spectrum therapy by comparing outcomes associated with guideline‐recommended therapy to nonguideline therapy. We considered antibiotic therapy as guideline recommended if appropriately chosen per the guideline, not just by simple classification of antibiotic. For example, the use of a cephalosporin in a patient with aminopenicillin allergy was considered guideline‐recommended therapy. We chose to classify the exposure of empiric antibiotic therapy in this manner to reflect true clinical application of the guideline, as not all children are able to receive aminopenicillins. Additionally, as our study stemmed from our prior improvement work aimed at increasing guideline adherent therapy,[7] we have a higher frequency of narrow‐spectrum antibiotic use than prior studies. In our study, almost two‐thirds of patients received narrow‐spectrum therapy, whereas narrow‐spectrum use in prior studies ranged from 10% to 33%.[17, 18, 19] Finally, we were able to confirm the diagnosis of pneumonia via medical record review and to adjust for severity of illness using clinical variables including vital signs, physical exam findings, and laboratory and radiologic study results.

This study must be interpreted in the context of several limitations. First, our study population was defined through discharge diagnosis codes, and therefore dependent on the accuracy of coding. However, we minimized potential for misclassification through use of a previously validated approach to identify patients with CAP and through medical record review to confirm the diagnosis. Second, we may be unable to detect very small differences in outcomes given limited power, specifically for outcomes of LOS, ED revisits, hospital readmissions, and need to broaden antibiotic therapy. Third, residual confounding may be present. Although we controlled for many clinical variables in our analyses, antibiotic prescribing practices may be influenced by unmeasured factors. The potential of system level confounding is mitigated by standardized care for patients with CAP at our institution. Prior system‐level changes using quality‐improvement science have resulted in a high level of adherence with guideline‐recommended antimicrobials as well as standardized medical discharge criteria.[7, 20] Additionally, nonmedical factors may influence LOS, limiting its use as an outcome measure. This limitation was minimized in our study by standardizing medical discharge criteria. Prior work at our institution demonstrated that the majority of patients, including those with CAP, were discharged within 2 hours of meeting medical discharge criteria.[20] Fourth, discharged patients who experienced adverse outcomes may have received care at a nearby adult emergency department or at their pediatrician's office. Although these events would not have been captured in our electronic health record, serious complications due to treatment failure (eg, empyema) would require hospitalization. As our hospital is the only children's hospital in the Greater Cincinnati metropolitan area, these patents would receive care at our institution. Therefore, any misclassification of revisits or readmissions is likely to be minimal. Finally, study patients were admitted to a large tertiary academic children's hospital, warranting further investigation to determine if these findings can be translated to smaller community settings.

In conclusion, receipt of guideline‐recommended antibiotic therapy for patients hospitalized with CAP was not associated with increases in LOS, total costs of hospitalization, or inpatient pharmacy costs. Our findings highlight the importance of changing antibiotic prescribing practices to reflect guideline recommendations, as there was no evidence of negative unintended consequences with our local practice change.

Acknowledgments

Disclosures: Dr. Ambroggio and Dr. Thomson were supported by funds from the NRSA T32HP10027‐14. Dr. Shah was supported by funds from NIAID K01A173729. The authors report no conflicts of interest.

Files
References
  1. Kronman MP, Hersh AL, Feng R, Huang YS, Lee GE, Shah SS. Ambulatory visit rates and antibiotic prescribing for children with pneumonia, 1994–2007. Pediatrics. 2011;127(3):411418.
  2. Lee GE, Lorch SA, Sheffler‐Collins S, Kronman MP, Shah SS. National hospitalization trends for pediatric pneumonia and associated complications. Pediatrics. 2010;126(2):204213.
  3. Keren R, Luan X, Localio R, et al. Prioritization of comparative effectiveness research topics in hospital pediatrics. Arch Pediatr Adolesc Med. 2012;166(12):11551164.
  4. Bradley JS, Byington CL, Shah SS, et al. The management of community‐acquired pneumonia in infants and children older than 3 months of age: clinical practice guidelines by the Pediatric Infectious Diseases Society and the Infectious Diseases Society of America. Clin Infect Dis. 2011;53(7):e25e76.
  5. Brogan TV, Hall M, Williams DJ, et al. Variability in processes of care and outcomes among children hospitalized with community‐acquired pneumonia. Pediatr Infect Dis J. 2012;31(10):10361041.
  6. Ross RK, Hersh AL, Kronman MP, et al. Impact of IDSA/PIDS guidelines on treatment of community‐acquired pneumonia in hospitalized children. Clin Infect Dis. 2014;58(6):834838.
  7. Ambroggio L, Thomson J, Murtagh Kurowski E, et al. Quality improvement methods increase appropriate antibiotic prescribing for childhood pneumonia. Pediatrics. 2013;131(5):e1623e1631.
  8. Williams DJ, Shah SS, Myers A, et al. Identifying pediatric community‐acquired pneumonia hospitalizations: accuracy of administrative billing codes. JAMA Pediatr. 2013;167(9):851858.
  9. Feudtner C, Hays RM, Haynes G, Geyer JR, Neff JM, Koepsell TD. Deaths attributed to pediatric complex chronic conditions: national trends and implications for supportive care services. Pediatrics. 2001;107(6):E99.
  10. Centers for Medicare 2011.
  11. Kleinman ME, Chameides L, Schexnayder SM, et al. Pediatric advanced life support: 2010 American Heart Association guidelines for cardiopulmonary resuscitation and emergency cardiovascular care. Pediatrics. 2010;126(5):e1361e1399.
  12. Tucker KM, Brewer TL, Baker RB, Demeritt B, Vossmeyer MT. Prospective evaluation of a pediatric inpatient early warning scoring system. J Spec Pediatr Nurs. 2009;14(2):7985.
  13. Vittinghoff E, Glidden DV, Shiboski SC, McCulloch CE. Regression Methods in Biostatistics: Linear, Logistic, Survival, and Repeated Measures Models. New York, NY: Springer; 2005.
  14. Biondi E, McCulloh R, Alverson B, Klein A, Dixon A. Treatment of mycoplasma pneumonia: a systematic review. Pediatrics. 2014;133(6):10811090.
  15. Newman RE, Hedican EB, Herigon JC, Williams DD, Williams AR, Newland JG. Impact of a guideline on management of children hospitalized with community‐acquired pneumonia. Pediatrics. 2012;129(3):e597e604.
  16. Williams DJ, Hall M, Shah SS, et al. Narrow vs broad‐spectrum antimicrobial therapy for children hospitalized with pneumonia. Pediatrics. 2013;132(5):e1141e1148.
  17. Queen MA, Myers AL, Hall M, et al. Comparative effectiveness of empiric antibiotics for community‐acquired pneumonia. Pediatrics. 2014;133(1):e23e29.
  18. Dinur‐Schejter Y, Cohen‐Cymberknoh M, Tenenbaum A, et al. Antibiotic treatment of children with community‐acquired pneumonia: comparison of penicillin or ampicillin versus cefuroxime. Pediatr Pulmonol. 2013;48(1):5258.
  19. White CM, Statile AM, White DL, et al. Using quality improvement to optimise paediatric discharge efficiency. BMJ Qual Saf. 2014;23(5):428436.
Article PDF
Issue
Journal of Hospital Medicine - 10(1)
Publications
Page Number
13-18
Sections
Files
Files
Article PDF
Article PDF

Community‐acquired pneumonia (CAP) is a common and serious infection in children. With more than 150,000 children requiring hospitalization annually, CAP is the fifth most prevalent and the second most costly diagnosis of all pediatric hospitalizations in the United States.[1, 2, 3]

In August 2011, the Pediatric Infectious Diseases Society (PIDS) and the Infectious Diseases Society of America (IDSA) published an evidence‐based guideline for the management of CAP in children. This guideline recommended that fully immunized children without underlying complications who require hospitalization receive an aminopenicillin as first‐line antibiotic therapy.[4] Additionally, the guideline recommends empirically adding a macrolide to an aminopenicillin when atypical pneumonia is a diagnostic consideration.

This recommendation was a substantial departure from practice for hospitals nationwide, as a multicenter study of children's hospitals (20052010) demonstrated that <10% of patients diagnosed with CAP received aminopenicillins as empiric therapy.[5] Since publication of the PIDS/IDSA guidelines, the use of aminopenicillins has increased significantly across institutions, but the majority of hospitalized patients still receive broad‐spectrum cephalosporin therapy for CAP.[6]

At baseline, 30% of patients hospitalized with CAP received guideline‐recommended antibiotic therapy at our institution. Through the use of quality‐improvement methods, the proportion of patients receiving guideline‐recommended therapy increased to 100%.[7] The objective of this study was to ensure that there were not unintended negative consequences to guideline implementation. Specifically, we sought to identify changes in length of stay (LOS), hospital costs, and treatment failures associated with use of guideline‐recommended antibiotic therapy for children hospitalized with uncomplicated CAP.

METHODS

Study Design and Study Population

This retrospective cohort study included children age 3 months to 18 years, hospitalized with CAP, between May 2, 2011 and July 30, 2012, at Cincinnati Children's Hospital Medical Center (CCHMC), a 512‐bed free‐standing children's hospital. The CCHMC Institutional Review Board approved this study with a waiver of informed consent.

Patients were eligible for inclusion if they were admitted to the hospital for inpatient or observation level care with a primary or secondary International Classification of Disease, 9th Revision discharge diagnosis code of pneumonia (480.02, 480.89, 481, 482.0, 482.30‐2, 482.41‐2, 482.83, 482.8990, 483.8, 484.3, 485, 486, 487.0) or effusion/empyema (510.0, 510.9, 511.01, 511.89, 513).[8] Patients with complex chronic conditions[9] were excluded. Medical records of eligible patients (n=260) were reviewed by 2 members of the study team to ensure that patients fell into the purview of the guideline. Patients who did not receive antibiotics (n=11) or for whom there was documented concern for aspiration (n=1) were excluded. Additionally, patients with immunodeficiency (n=1) or who had not received age‐appropriate vaccinations (n=2), and patients who required intensive care unit admission on presentation (n=17) or who had a complicated pneumonia, defined by presence of moderate or large pleural effusion at time of admission (n=8), were also excluded.[7] Finally, for patients with multiple pneumonia admissions, only the index visit was included; subsequent visits occurring within 30 days of discharge were considered readmissions.

Treatment Measure

The primary exposure of interest was empiric antibiotic therapy upon hospital admission. Antibiotic therapy was classified as guideline recommended or nonguideline recommended. Guideline‐recommended therapy was defined as follows:

  1. For children without drug allergies: ampicillin (200 mg/kg/day intravenously) or amoxicillin (90 mg/kg/day orally);
  2. For children with penicillin allergy: ceftriaxone (50100 mg/kg/day intravenously or intramuscularly) or cefdinir (14 mg/kg/day orally);
  3. For children with penicillin and cephalosporin allergy: clindamycin (40 mg/kg/day orally or intravenously); and
  4. Or azithromycin (10 mg/kg/day orally or intravenously on day 1) in combination with antibiotic category 1 or 2 or 3 above.

 

Outcome Measures

The primary outcomes examined were hospital LOS, total cost of hospitalization, and inpatient pharmacy costs. LOS was measured in hours and defined as the difference in time between departure from and arrival to the inpatient unit. Total cost of index hospitalization included both direct and indirect costs, obtained from the Centers for Medicare & Medicaid Services' Relative Value Units data for Current Procedural Terminology codes.[10]

Secondary outcomes included broadening of antibiotic therapy during the hospital course, pneumonia‐related emergency department (ED) revisits within 30 days, and pneumonia‐related inpatient readmissions within 30 days. Broadening of antibiotic therapy was defined as addition of a second antibiotic (eg, adding azithromycin on day 3 of hospitalization) or change in empiric antibiotic to a class with broader antimicrobial activity (eg, ampicillin to ceftriaxone) at any time during hospitalization. As our study population included only patients with uncomplicated pneumonia at the time of admission, this outcome was used to capture possible treatment failure. ED revisits and inpatient readmissions were reviewed by 3 investigators to identify pneumonia‐related visits. To encompass all possible treatment failures, all respiratory‐related complaints (eg, wheezing, respiratory distress) were considered as pneumonia‐related. Disagreements were resolved by group discussion.

Covariates

Severity of illness on presentation was evaluated using the Emergency Severity Index version 4,[11] abnormal vital signs on presentation (as defined by Pediatric Advanced Life Support age‐specific criteria[12]), and need for oxygen in the first 24 hours of hospitalization. Supplemental oxygen is administered for saturations <91% per protocol at our institution. The patient's highest Pediatric Early Warning Scale score[13] during hospitalization was used as a proxy for disease severity. Exam findings on presentation (eg, increased respiratory effort, rales, wheezing) were determined through chart review. Laboratory tests and radiologic imaging variables included complete blood cell count, blood culture, chest radiograph, chest ultrasound, and chest computed tomography. Abnormal white blood cell count was defined as <5000 or >15,000 cells/mL, the defined reference range for the CCHMC clinical laboratory.

Data Analysis

Continuous variables were described using median and interquartile range (IQR) and compared across groups using Wilcoxon rank sum test due to non‐normal distributions. Categorical variables were described by counts and frequencies and compared using the 2 test.

Multivariable linear regression analysis was performed to assess the independent effect of receipt of empiric guideline‐recommended antibiotic therapy on outcomes of LOS and costs while adjusting for covariates. As LOS and costs were non‐normally distributed, we logarithmically transformed these values to use as the dependent variables in our models. The resulting coefficients were back‐transformed to reflect the percent change in LOS and costs incurred between subjects who received empiric guideline therapy compared with those who did not.[14] Covariates were chosen a priori due to their clinical and biological relevance to the outcomes of LOS (eg, wheezing on presentation and need for supplemental oxygen), total cost of hospitalization (eg, LOS and need for repeat imaging), and inpatient pharmacy costs (eg, LOS and wheezing on presentation) (Table 1).

Cohort Characteristics
CharacteristicOverall Cohort, n=220Guideline Therapy, n=166Nonguideline Therapy, n=54P Value
  • NOTE: Abbreviations: CT, computed tomography; IQR, interquartile range; PEWS, Pediatric Early Warning Scale. *P<0.05.

Age, y, median (IQR)2.9 (1.36.3)2.5 (1.35.2)5.6 (2.38.8)<0.01*
Male, no. (%)122 (55.5%)89 (53.6%)33 (61.1%)0.34
Emergency Severity Index, no. (%)0.11
290 (40.9%)73 (44.0%)17 (31.5%) 
3116 (52.7%)85 (51.2%)31 (57.4%) 
414 (6.4%)8 (4.8%)6 (11.1%) 
Abnormal vital signs on presentation, no. (%)
Fever99 (45.0%)80 (48.2%)19 (35.2%)0.10
Tachycardia100 (45.5%)76 (45.8%)24 (44.4%)0.86
Tachypnea124 (56.4%)100 (60.2%)24 (44.4%)0.04*
Hypotension000 
Hypoxia27 (12.3%)24 (14.5%)3 (5.6%)0.08
Physical exam on presentation, no. (%)
Increased respiratory effort146 (66.4%)111 (66.9%)35 (64.8%)0.78
Distressed110 (50.0%)86 (51.8%)24 (44.4%)0.35
Retraction103 (46.8%)81 (48.8%)22 (40.7%)0.30
Grunting17 (7.7%)14 (8.4%)3 (5.6%)0.49
Nasal flaring19 (8.6%)17 (10.2%)2 (3.7%)0.14
Rales135 (61.4%)99 (59.6%)36 (66.7%)0.36
Wheeze91 (41.4%)66 (39.8%)25 (46.3%)0.40
Decreased breath sounds89 (40.5%)65 (39.2%)24 (44.4%)0.49
Dehydration21 (9.6%)13 (7.8%)8 (14.8%)0.13
PEWS 5 during admission, no. (%)43 (19.6%)34 (20.5%)9 (16.7%)0.54
Oxygen requirement in first 24 hours, no. (%)114 (51.8%)90 (53.6%)24 (46.2%)0.35
Complete blood count obtained, no. (%)99 (45.0%)72 (43.4%)27 (50.0%)0.40
Abnormal white blood cell count35 (35.7%)23 (32.4%)12 (44.4%)0.27
Blood culture obtained, no. (%)104 (47.3%)80 (48.2%)24 (44.4%)0.63
Positive2 (1.9%)1 (1.3%)1 (4.2%)0.36
Chest radiograph available, no. (%)214 (97.3%)161 (97.0%)53 (98.2%)0.65
Infiltrate178 (83.2%)139 (86.3%)39 (73.6%)0.03*
Bilateral29 (16.3%)20 (14.4%)9 (23.1%)0.19
Multilobar46 (25.8%)33 (23.7%)13 (33.3%)0.23
Effusion24 (11.2%)16 (9.9%)8 (15.1%)0.30
Additional imaging, no. (%)    
Repeat chest radiograph26 (11.8%)17 (10.2%)9 (16.7%)0.20
Chest ultrasound4 (1.8%)3 (1.8%)1 (1.9%)0.98
Chest CT2 (0.9%)1 (0.6%)1 (1.9%)0.40
Antibiotic, no. (%)   <0.01*
Aminopenicillin140 (63.6%)140 (84.3%)0 (0%) 
Third‐generation cephalosporin37 (16.8%)8 (4.8%)29 (53.7%) 
Macrolide monotherapy18 (8.2%)0 (0%)18 (33.3%) 
Clindamycin2 (0.9%)1 (0.6%)1 (1.9%) 
Levofloxacin1 (0.5%)0 (0%)1 (1.9%) 
Aminopenicillin+macrolide16 (7.3%)16 (9.6%)0 (0%) 
Cephalosporin+macrolide6 (2.7%)1 (0.6%)5 (9.3%) 

Secondary outcomes of broadened antibiotic therapy, ED revisits, and hospital readmissions were assessed using the Fisher exact test. Due to the small number of events, we were unable to evaluate these associations in models adjusted for potential confounders.

All analyses were performed with SAS version 9.3 (SAS Institute, Cary, NC), and P values <0.05 were considered significant.

RESULTS

Of the 220 unique patients included, 122 (55%) were male. The median age was 2.9 years (IQR: 1.36.3 years). Empiric guideline‐recommended therapy was prescribed to 168 (76%) patients (Table 1). Aminopenicillins were the most common guideline‐recommended therapy, accounting for 84% of guideline‐recommended antibiotics. An additional 10% of patients received the guideline‐recommended combination therapy with an aminopenicillin and a macrolide. Nonguideline‐recommended therapy included third‐generation cephalosporin antibiotics (54%) and macrolide monotherapy (33%).

Those who received empiric guideline‐recommended antibiotic therapy were similar to those who received nonguideline‐recommended therapy with respect to sex, Emergency Severity Index, physical exam findings on presentation, oxygen requirement in the first 24 hours, abnormal laboratory findings, presence of effusion on chest radiograph, and need for additional imaging (Table 1). However, patients in the guideline‐recommended therapy group were significantly younger (median 2.5 years vs 5.6 years, P0.01), more likely to have elevated respiratory rate on presentation (60.2% vs 44.4%, P=0.04), and more likely to have an infiltrate on chest radiograph (86.3% vs 73.6%, P=0.03) (Table 1). Patients who received nonguideline‐recommended macrolide monotherapy had a median age of 7.4 years (IQR: 5.89.8 years).

Median hospital LOS for the total cohort was 1.3 days (IQR: 0.91.9 days) (Table 2). There were no differences in LOS between patients who received and did not receive guideline‐recommended therapy in the unadjusted or the adjusted model (Table 3).

Unadjusted Outcomes
OutcomeGuideline Therapy, n=166Nonguideline Therapy, n=54P Value
  • NOTE: Abbreviations: IQR, interquartile range.

Length of stay, d, median (IQR)1.3 (0.91.9)1.3 (0.92.0)0.74
Total costs, median, (IQR)$4118 ($2,647$6,004)$4045 ($2,829$6,200)0.44
Pharmacy total costs, median, (IQR)$84 ($40$179)$106 ($58$217)0.12
Broadened therapy, no. (%)10 (6.0%)4 (7.4%)0.75
Emergency department revisit, no. (%)7 (4.2%)2 (3.7%)1.00
Readmission, no. (%)1 (0.6%)1 (1.9%)0.43
Univariate and Multivariate Analyses of Receipt of Empiric Guideline‐Recommended Therapy With Length of Stay, Total Costs, and Pharmacy Costs
OutcomeUnadjusted Coefficient (95% CI)Adjusted Coefficient (95% CI)Adjusted % Change in Outcome (95% CI)*
  • NOTE: Abbreviations: CI, confidence interval. *Negative adjusted percent change indicates decrease in outcome associated with guideline‐recommended therapy; positive adjusted percent change indicates increase in outcome associated with guideline‐recommended therapy. Model is adjusted for age, fever on presentation, tachypnea on presentation, wheezing on presentation, need for supplemental oxygen, Pediatric Early Warning Score 5, chest radiograph findings, and need for repeat imaging. Model is adjusted for age, wheezing on presentation, need for supplemental oxygen, Pediatric Early Warning Score 5, need for repeat imaging, and length of stay. Model is adjusted for age, wheezing on presentation, and length of stay.

Length of stay0.06 (0.27 to 0.15)0.06 (0.25 to 0.12)5.8 (22.1 to 12.8)
Total costs0.18 (0.40 to 0.04)0.11 (0.32 to 0.09)10.9 (27.4 to 9.4)
Pharmacy total costs0.44 (0.46 to 0.02)0.16 (0.57 to 0.24)14.8 (43.4 to 27.1)

Median total costs of the index hospitalization for the total cohort were $4097 (IQR: $2657$6054), with median inpatient pharmacy costs of $92 (IQR: $40$183) (Table 2). There were no differences in total or inpatient pharmacy costs for patients who received guideline‐recommended therapy compared with those who did not in unadjusted or adjusted analyses. Fourteen patients (6.4%) had antibiotic therapy broadened during hospitalization, 10 were initially prescribed guideline‐recommended therapy, and 4 were initially prescribed nonguideline‐recommended therapy (Table 4).

Clinical Details of Patients Who Had Antibiotic Therapy Broadened During Initial Hospitalization
Initial TherapyReasons for Antibiotic Change Identified From Chart Review
Guideline=10Ampicillin to ceftriaxone:
1 patient with clinical worsening
1 patient with coincident urinary tract infection due to resistant organism
4 patients without evidence of clinical worsening or documentation of rationale
Addition of a macrolide:
3 patients without evidence of clinical worsening or documentation of rationale
Addition of clindamycin:
1 patient with clinical worsening
Nonguideline=4Ceftriaxone to clindamycin:
1 patient with clinical worsening
Addition of a macrolide:
1 patient with clinical worsening
1 patients without evidence of clinical worsening or documentation of rationale
Addition of clindamycin:
1 patient with clinical worsening

Of the 9 pneumonia‐related ED revisits within 30 days of discharge, 7 occurred in patients prescribed empiric guideline‐recommended therapy (Table 5). No ED revisit resulted in hospital readmission or antibiotic change related to pneumonia. Two ED revisits resulted in new antibiotic prescriptions for diagnoses other than pneumonia.

Clinical Details of Patients With an Emergency Department Revisit or Inpatient Readmission Following Index Hospitalization
RevisitInitial TherapyDay PostdischargeClinical Symptoms at Return VisitClinical DiagnosisAntibiotic Prescription
  • NOTE: Abbreviations: ED, emergency department; IV, intravenous.

EDGuideline3Poor oral intake and feverPneumoniaContinued prior antibiotic
EDGuideline8Recurrent cough and feverResolving pneumoniaContinued prior antibiotic
EDGuideline13Follow‐upResolved pneumoniaNo further antibiotic
EDGuideline16Increased work of breathingReactive airway diseaseNo antibiotic
EDGuideline20Persistent coughViral illnessNo antibiotic
EDGuideline22Recurrent cough and congestionSinusitisAugmentin
EDGuideline26Increased work of breathingReactive airway diseaseNo antibiotic
EDNonguideline16Recurrent feverAcute otitis mediaAmoxicillin
EDNonguideline20Recurrent cough and feverViral illnessNo antibiotic
AdmissionGuideline3Increased work of breathingPneumoniaIV ampicillin
AdmissionNonguideline9Refusal to take oral clindamycinPneumoniaIV clindamycin

Two patients were readmitted for a pneumonia‐related illness within 30 days of discharge; 1 had received guideline‐recommended therapy (Table 5). Both patients were directly admitted to the inpatient ward without an associated ED visit. Antibiotic class was not changed for either patient upon readmission, despite the decision to convert to intravenous form.

DISCUSSION

In this retrospective cohort study, patients who received empiric guideline‐recommended antibiotic therapy on admission for CAP had no difference in LOS, total cost of hospitalization, or inpatient pharmacy costs compared with those who received therapy that varied from guideline recommendations. Our study suggests that prescribing narrow‐spectrum therapy and, in some circumstances, combination therapy, as recommended by the 2011 PIDS/IDSA pneumonia guideline, did not result in negative unintended consequences.

In our study, children receiving guideline‐recommended therapy were younger, more likely to have elevated respiratory rate on presentation, and more likely to have an infiltrate on chest radiograph. We hypothesize the age difference is a reflection of common use of nonguideline macrolide monotherapy in the older, school‐age child, as macrolides are commonly used for coverage of Mycoplasma pneumonia in older children with CAP.[15] Children receiving macrolide monotherapy were older than those receiving guideline‐recommended therapy (median age of 7.4 years and 2.5 years, respectively). We also hypothesize that some providers may prescribe macrolide monotherapy to children deemed less ill than expected for CAP (eg, normal percutaneous oxygen saturation). This hypothesis is supported by the finding that 60% of patients who had a normal respiratory rate and received nonguideline therapy were prescribed macrolide monotherapy. We did control for the characteristics that varied between the two treatment groups in our models to eliminate potential confounding.

One prior study evaluated the effects of guideline implementation in CAP. In evaluation of a clinical practice guideline that recommended ampicillin as first‐line therapy for CAP, no significant difference was found following guideline introduction in the number of treatment failures (defined as the need to broaden therapy or development of complicated pneumonia within 48 hours or 30‐day inpatient readmission).[16] Our study builds on these findings by directly comparing outcomes between recipients of guideline and nonguideline therapy, which was not done in the prestudy or poststudy design of prior work.[16] We believe that classifying patients based on empiric therapy received rather than timing of guideline introduction thoroughly examines the effect of following guideline recommendations. Additionally, outcomes other than treatment failures were examined in our study including LOS, costs, and ED revisits.

Our results are similar to other observational studies that compared narrow‐ and broad‐spectrum antibiotics for children hospitalized with CAP. Using administrative data, Williams et al. found no significant difference in LOS or cost between narrow‐ and broad‐spectrum intravenous antibiotic recipients ages 6 months to 18 years at 43 children's hospitals.[17] Queen et al. compared narrow‐ and broad‐spectrum antibiotic therapy for children ages 2 months to 18 years at 4 children's hospitals.[18] Differences in average daily cost were not significant, but children who received narrow‐spectrum antibiotics had a significantly shorter LOS. Finally, an observational study of 319 Israeli children <2 years of age found no significant difference in duration of oxygen requirement, LOS, or need for change in antibiotic therapy between the 66 children prescribed aminopenicillins and the 253 children prescribed cefuroxime.[19] These studies suggest that prescribing narrow‐spectrum antimicrobial therapy, as per the guideline recommendations, results in similar outcomes to broad‐spectrum antimicrobial therapy.

Our study adds to prior studies that compared narrow‐ and broad‐spectrum therapy by comparing outcomes associated with guideline‐recommended therapy to nonguideline therapy. We considered antibiotic therapy as guideline recommended if appropriately chosen per the guideline, not just by simple classification of antibiotic. For example, the use of a cephalosporin in a patient with aminopenicillin allergy was considered guideline‐recommended therapy. We chose to classify the exposure of empiric antibiotic therapy in this manner to reflect true clinical application of the guideline, as not all children are able to receive aminopenicillins. Additionally, as our study stemmed from our prior improvement work aimed at increasing guideline adherent therapy,[7] we have a higher frequency of narrow‐spectrum antibiotic use than prior studies. In our study, almost two‐thirds of patients received narrow‐spectrum therapy, whereas narrow‐spectrum use in prior studies ranged from 10% to 33%.[17, 18, 19] Finally, we were able to confirm the diagnosis of pneumonia via medical record review and to adjust for severity of illness using clinical variables including vital signs, physical exam findings, and laboratory and radiologic study results.

This study must be interpreted in the context of several limitations. First, our study population was defined through discharge diagnosis codes, and therefore dependent on the accuracy of coding. However, we minimized potential for misclassification through use of a previously validated approach to identify patients with CAP and through medical record review to confirm the diagnosis. Second, we may be unable to detect very small differences in outcomes given limited power, specifically for outcomes of LOS, ED revisits, hospital readmissions, and need to broaden antibiotic therapy. Third, residual confounding may be present. Although we controlled for many clinical variables in our analyses, antibiotic prescribing practices may be influenced by unmeasured factors. The potential of system level confounding is mitigated by standardized care for patients with CAP at our institution. Prior system‐level changes using quality‐improvement science have resulted in a high level of adherence with guideline‐recommended antimicrobials as well as standardized medical discharge criteria.[7, 20] Additionally, nonmedical factors may influence LOS, limiting its use as an outcome measure. This limitation was minimized in our study by standardizing medical discharge criteria. Prior work at our institution demonstrated that the majority of patients, including those with CAP, were discharged within 2 hours of meeting medical discharge criteria.[20] Fourth, discharged patients who experienced adverse outcomes may have received care at a nearby adult emergency department or at their pediatrician's office. Although these events would not have been captured in our electronic health record, serious complications due to treatment failure (eg, empyema) would require hospitalization. As our hospital is the only children's hospital in the Greater Cincinnati metropolitan area, these patents would receive care at our institution. Therefore, any misclassification of revisits or readmissions is likely to be minimal. Finally, study patients were admitted to a large tertiary academic children's hospital, warranting further investigation to determine if these findings can be translated to smaller community settings.

In conclusion, receipt of guideline‐recommended antibiotic therapy for patients hospitalized with CAP was not associated with increases in LOS, total costs of hospitalization, or inpatient pharmacy costs. Our findings highlight the importance of changing antibiotic prescribing practices to reflect guideline recommendations, as there was no evidence of negative unintended consequences with our local practice change.

Acknowledgments

Disclosures: Dr. Ambroggio and Dr. Thomson were supported by funds from the NRSA T32HP10027‐14. Dr. Shah was supported by funds from NIAID K01A173729. The authors report no conflicts of interest.

Community‐acquired pneumonia (CAP) is a common and serious infection in children. With more than 150,000 children requiring hospitalization annually, CAP is the fifth most prevalent and the second most costly diagnosis of all pediatric hospitalizations in the United States.[1, 2, 3]

In August 2011, the Pediatric Infectious Diseases Society (PIDS) and the Infectious Diseases Society of America (IDSA) published an evidence‐based guideline for the management of CAP in children. This guideline recommended that fully immunized children without underlying complications who require hospitalization receive an aminopenicillin as first‐line antibiotic therapy.[4] Additionally, the guideline recommends empirically adding a macrolide to an aminopenicillin when atypical pneumonia is a diagnostic consideration.

This recommendation was a substantial departure from practice for hospitals nationwide, as a multicenter study of children's hospitals (20052010) demonstrated that <10% of patients diagnosed with CAP received aminopenicillins as empiric therapy.[5] Since publication of the PIDS/IDSA guidelines, the use of aminopenicillins has increased significantly across institutions, but the majority of hospitalized patients still receive broad‐spectrum cephalosporin therapy for CAP.[6]

At baseline, 30% of patients hospitalized with CAP received guideline‐recommended antibiotic therapy at our institution. Through the use of quality‐improvement methods, the proportion of patients receiving guideline‐recommended therapy increased to 100%.[7] The objective of this study was to ensure that there were not unintended negative consequences to guideline implementation. Specifically, we sought to identify changes in length of stay (LOS), hospital costs, and treatment failures associated with use of guideline‐recommended antibiotic therapy for children hospitalized with uncomplicated CAP.

METHODS

Study Design and Study Population

This retrospective cohort study included children age 3 months to 18 years, hospitalized with CAP, between May 2, 2011 and July 30, 2012, at Cincinnati Children's Hospital Medical Center (CCHMC), a 512‐bed free‐standing children's hospital. The CCHMC Institutional Review Board approved this study with a waiver of informed consent.

Patients were eligible for inclusion if they were admitted to the hospital for inpatient or observation level care with a primary or secondary International Classification of Disease, 9th Revision discharge diagnosis code of pneumonia (480.02, 480.89, 481, 482.0, 482.30‐2, 482.41‐2, 482.83, 482.8990, 483.8, 484.3, 485, 486, 487.0) or effusion/empyema (510.0, 510.9, 511.01, 511.89, 513).[8] Patients with complex chronic conditions[9] were excluded. Medical records of eligible patients (n=260) were reviewed by 2 members of the study team to ensure that patients fell into the purview of the guideline. Patients who did not receive antibiotics (n=11) or for whom there was documented concern for aspiration (n=1) were excluded. Additionally, patients with immunodeficiency (n=1) or who had not received age‐appropriate vaccinations (n=2), and patients who required intensive care unit admission on presentation (n=17) or who had a complicated pneumonia, defined by presence of moderate or large pleural effusion at time of admission (n=8), were also excluded.[7] Finally, for patients with multiple pneumonia admissions, only the index visit was included; subsequent visits occurring within 30 days of discharge were considered readmissions.

Treatment Measure

The primary exposure of interest was empiric antibiotic therapy upon hospital admission. Antibiotic therapy was classified as guideline recommended or nonguideline recommended. Guideline‐recommended therapy was defined as follows:

  1. For children without drug allergies: ampicillin (200 mg/kg/day intravenously) or amoxicillin (90 mg/kg/day orally);
  2. For children with penicillin allergy: ceftriaxone (50100 mg/kg/day intravenously or intramuscularly) or cefdinir (14 mg/kg/day orally);
  3. For children with penicillin and cephalosporin allergy: clindamycin (40 mg/kg/day orally or intravenously); and
  4. Or azithromycin (10 mg/kg/day orally or intravenously on day 1) in combination with antibiotic category 1 or 2 or 3 above.

 

Outcome Measures

The primary outcomes examined were hospital LOS, total cost of hospitalization, and inpatient pharmacy costs. LOS was measured in hours and defined as the difference in time between departure from and arrival to the inpatient unit. Total cost of index hospitalization included both direct and indirect costs, obtained from the Centers for Medicare & Medicaid Services' Relative Value Units data for Current Procedural Terminology codes.[10]

Secondary outcomes included broadening of antibiotic therapy during the hospital course, pneumonia‐related emergency department (ED) revisits within 30 days, and pneumonia‐related inpatient readmissions within 30 days. Broadening of antibiotic therapy was defined as addition of a second antibiotic (eg, adding azithromycin on day 3 of hospitalization) or change in empiric antibiotic to a class with broader antimicrobial activity (eg, ampicillin to ceftriaxone) at any time during hospitalization. As our study population included only patients with uncomplicated pneumonia at the time of admission, this outcome was used to capture possible treatment failure. ED revisits and inpatient readmissions were reviewed by 3 investigators to identify pneumonia‐related visits. To encompass all possible treatment failures, all respiratory‐related complaints (eg, wheezing, respiratory distress) were considered as pneumonia‐related. Disagreements were resolved by group discussion.

Covariates

Severity of illness on presentation was evaluated using the Emergency Severity Index version 4,[11] abnormal vital signs on presentation (as defined by Pediatric Advanced Life Support age‐specific criteria[12]), and need for oxygen in the first 24 hours of hospitalization. Supplemental oxygen is administered for saturations <91% per protocol at our institution. The patient's highest Pediatric Early Warning Scale score[13] during hospitalization was used as a proxy for disease severity. Exam findings on presentation (eg, increased respiratory effort, rales, wheezing) were determined through chart review. Laboratory tests and radiologic imaging variables included complete blood cell count, blood culture, chest radiograph, chest ultrasound, and chest computed tomography. Abnormal white blood cell count was defined as <5000 or >15,000 cells/mL, the defined reference range for the CCHMC clinical laboratory.

Data Analysis

Continuous variables were described using median and interquartile range (IQR) and compared across groups using Wilcoxon rank sum test due to non‐normal distributions. Categorical variables were described by counts and frequencies and compared using the 2 test.

Multivariable linear regression analysis was performed to assess the independent effect of receipt of empiric guideline‐recommended antibiotic therapy on outcomes of LOS and costs while adjusting for covariates. As LOS and costs were non‐normally distributed, we logarithmically transformed these values to use as the dependent variables in our models. The resulting coefficients were back‐transformed to reflect the percent change in LOS and costs incurred between subjects who received empiric guideline therapy compared with those who did not.[14] Covariates were chosen a priori due to their clinical and biological relevance to the outcomes of LOS (eg, wheezing on presentation and need for supplemental oxygen), total cost of hospitalization (eg, LOS and need for repeat imaging), and inpatient pharmacy costs (eg, LOS and wheezing on presentation) (Table 1).

Cohort Characteristics
CharacteristicOverall Cohort, n=220Guideline Therapy, n=166Nonguideline Therapy, n=54P Value
  • NOTE: Abbreviations: CT, computed tomography; IQR, interquartile range; PEWS, Pediatric Early Warning Scale. *P<0.05.

Age, y, median (IQR)2.9 (1.36.3)2.5 (1.35.2)5.6 (2.38.8)<0.01*
Male, no. (%)122 (55.5%)89 (53.6%)33 (61.1%)0.34
Emergency Severity Index, no. (%)0.11
290 (40.9%)73 (44.0%)17 (31.5%) 
3116 (52.7%)85 (51.2%)31 (57.4%) 
414 (6.4%)8 (4.8%)6 (11.1%) 
Abnormal vital signs on presentation, no. (%)
Fever99 (45.0%)80 (48.2%)19 (35.2%)0.10
Tachycardia100 (45.5%)76 (45.8%)24 (44.4%)0.86
Tachypnea124 (56.4%)100 (60.2%)24 (44.4%)0.04*
Hypotension000 
Hypoxia27 (12.3%)24 (14.5%)3 (5.6%)0.08
Physical exam on presentation, no. (%)
Increased respiratory effort146 (66.4%)111 (66.9%)35 (64.8%)0.78
Distressed110 (50.0%)86 (51.8%)24 (44.4%)0.35
Retraction103 (46.8%)81 (48.8%)22 (40.7%)0.30
Grunting17 (7.7%)14 (8.4%)3 (5.6%)0.49
Nasal flaring19 (8.6%)17 (10.2%)2 (3.7%)0.14
Rales135 (61.4%)99 (59.6%)36 (66.7%)0.36
Wheeze91 (41.4%)66 (39.8%)25 (46.3%)0.40
Decreased breath sounds89 (40.5%)65 (39.2%)24 (44.4%)0.49
Dehydration21 (9.6%)13 (7.8%)8 (14.8%)0.13
PEWS 5 during admission, no. (%)43 (19.6%)34 (20.5%)9 (16.7%)0.54
Oxygen requirement in first 24 hours, no. (%)114 (51.8%)90 (53.6%)24 (46.2%)0.35
Complete blood count obtained, no. (%)99 (45.0%)72 (43.4%)27 (50.0%)0.40
Abnormal white blood cell count35 (35.7%)23 (32.4%)12 (44.4%)0.27
Blood culture obtained, no. (%)104 (47.3%)80 (48.2%)24 (44.4%)0.63
Positive2 (1.9%)1 (1.3%)1 (4.2%)0.36
Chest radiograph available, no. (%)214 (97.3%)161 (97.0%)53 (98.2%)0.65
Infiltrate178 (83.2%)139 (86.3%)39 (73.6%)0.03*
Bilateral29 (16.3%)20 (14.4%)9 (23.1%)0.19
Multilobar46 (25.8%)33 (23.7%)13 (33.3%)0.23
Effusion24 (11.2%)16 (9.9%)8 (15.1%)0.30
Additional imaging, no. (%)    
Repeat chest radiograph26 (11.8%)17 (10.2%)9 (16.7%)0.20
Chest ultrasound4 (1.8%)3 (1.8%)1 (1.9%)0.98
Chest CT2 (0.9%)1 (0.6%)1 (1.9%)0.40
Antibiotic, no. (%)   <0.01*
Aminopenicillin140 (63.6%)140 (84.3%)0 (0%) 
Third‐generation cephalosporin37 (16.8%)8 (4.8%)29 (53.7%) 
Macrolide monotherapy18 (8.2%)0 (0%)18 (33.3%) 
Clindamycin2 (0.9%)1 (0.6%)1 (1.9%) 
Levofloxacin1 (0.5%)0 (0%)1 (1.9%) 
Aminopenicillin+macrolide16 (7.3%)16 (9.6%)0 (0%) 
Cephalosporin+macrolide6 (2.7%)1 (0.6%)5 (9.3%) 

Secondary outcomes of broadened antibiotic therapy, ED revisits, and hospital readmissions were assessed using the Fisher exact test. Due to the small number of events, we were unable to evaluate these associations in models adjusted for potential confounders.

All analyses were performed with SAS version 9.3 (SAS Institute, Cary, NC), and P values <0.05 were considered significant.

RESULTS

Of the 220 unique patients included, 122 (55%) were male. The median age was 2.9 years (IQR: 1.36.3 years). Empiric guideline‐recommended therapy was prescribed to 168 (76%) patients (Table 1). Aminopenicillins were the most common guideline‐recommended therapy, accounting for 84% of guideline‐recommended antibiotics. An additional 10% of patients received the guideline‐recommended combination therapy with an aminopenicillin and a macrolide. Nonguideline‐recommended therapy included third‐generation cephalosporin antibiotics (54%) and macrolide monotherapy (33%).

Those who received empiric guideline‐recommended antibiotic therapy were similar to those who received nonguideline‐recommended therapy with respect to sex, Emergency Severity Index, physical exam findings on presentation, oxygen requirement in the first 24 hours, abnormal laboratory findings, presence of effusion on chest radiograph, and need for additional imaging (Table 1). However, patients in the guideline‐recommended therapy group were significantly younger (median 2.5 years vs 5.6 years, P0.01), more likely to have elevated respiratory rate on presentation (60.2% vs 44.4%, P=0.04), and more likely to have an infiltrate on chest radiograph (86.3% vs 73.6%, P=0.03) (Table 1). Patients who received nonguideline‐recommended macrolide monotherapy had a median age of 7.4 years (IQR: 5.89.8 years).

Median hospital LOS for the total cohort was 1.3 days (IQR: 0.91.9 days) (Table 2). There were no differences in LOS between patients who received and did not receive guideline‐recommended therapy in the unadjusted or the adjusted model (Table 3).

Unadjusted Outcomes
OutcomeGuideline Therapy, n=166Nonguideline Therapy, n=54P Value
  • NOTE: Abbreviations: IQR, interquartile range.

Length of stay, d, median (IQR)1.3 (0.91.9)1.3 (0.92.0)0.74
Total costs, median, (IQR)$4118 ($2,647$6,004)$4045 ($2,829$6,200)0.44
Pharmacy total costs, median, (IQR)$84 ($40$179)$106 ($58$217)0.12
Broadened therapy, no. (%)10 (6.0%)4 (7.4%)0.75
Emergency department revisit, no. (%)7 (4.2%)2 (3.7%)1.00
Readmission, no. (%)1 (0.6%)1 (1.9%)0.43
Univariate and Multivariate Analyses of Receipt of Empiric Guideline‐Recommended Therapy With Length of Stay, Total Costs, and Pharmacy Costs
OutcomeUnadjusted Coefficient (95% CI)Adjusted Coefficient (95% CI)Adjusted % Change in Outcome (95% CI)*
  • NOTE: Abbreviations: CI, confidence interval. *Negative adjusted percent change indicates decrease in outcome associated with guideline‐recommended therapy; positive adjusted percent change indicates increase in outcome associated with guideline‐recommended therapy. Model is adjusted for age, fever on presentation, tachypnea on presentation, wheezing on presentation, need for supplemental oxygen, Pediatric Early Warning Score 5, chest radiograph findings, and need for repeat imaging. Model is adjusted for age, wheezing on presentation, need for supplemental oxygen, Pediatric Early Warning Score 5, need for repeat imaging, and length of stay. Model is adjusted for age, wheezing on presentation, and length of stay.

Length of stay0.06 (0.27 to 0.15)0.06 (0.25 to 0.12)5.8 (22.1 to 12.8)
Total costs0.18 (0.40 to 0.04)0.11 (0.32 to 0.09)10.9 (27.4 to 9.4)
Pharmacy total costs0.44 (0.46 to 0.02)0.16 (0.57 to 0.24)14.8 (43.4 to 27.1)

Median total costs of the index hospitalization for the total cohort were $4097 (IQR: $2657$6054), with median inpatient pharmacy costs of $92 (IQR: $40$183) (Table 2). There were no differences in total or inpatient pharmacy costs for patients who received guideline‐recommended therapy compared with those who did not in unadjusted or adjusted analyses. Fourteen patients (6.4%) had antibiotic therapy broadened during hospitalization, 10 were initially prescribed guideline‐recommended therapy, and 4 were initially prescribed nonguideline‐recommended therapy (Table 4).

Clinical Details of Patients Who Had Antibiotic Therapy Broadened During Initial Hospitalization
Initial TherapyReasons for Antibiotic Change Identified From Chart Review
Guideline=10Ampicillin to ceftriaxone:
1 patient with clinical worsening
1 patient with coincident urinary tract infection due to resistant organism
4 patients without evidence of clinical worsening or documentation of rationale
Addition of a macrolide:
3 patients without evidence of clinical worsening or documentation of rationale
Addition of clindamycin:
1 patient with clinical worsening
Nonguideline=4Ceftriaxone to clindamycin:
1 patient with clinical worsening
Addition of a macrolide:
1 patient with clinical worsening
1 patients without evidence of clinical worsening or documentation of rationale
Addition of clindamycin:
1 patient with clinical worsening

Of the 9 pneumonia‐related ED revisits within 30 days of discharge, 7 occurred in patients prescribed empiric guideline‐recommended therapy (Table 5). No ED revisit resulted in hospital readmission or antibiotic change related to pneumonia. Two ED revisits resulted in new antibiotic prescriptions for diagnoses other than pneumonia.

Clinical Details of Patients With an Emergency Department Revisit or Inpatient Readmission Following Index Hospitalization
RevisitInitial TherapyDay PostdischargeClinical Symptoms at Return VisitClinical DiagnosisAntibiotic Prescription
  • NOTE: Abbreviations: ED, emergency department; IV, intravenous.

EDGuideline3Poor oral intake and feverPneumoniaContinued prior antibiotic
EDGuideline8Recurrent cough and feverResolving pneumoniaContinued prior antibiotic
EDGuideline13Follow‐upResolved pneumoniaNo further antibiotic
EDGuideline16Increased work of breathingReactive airway diseaseNo antibiotic
EDGuideline20Persistent coughViral illnessNo antibiotic
EDGuideline22Recurrent cough and congestionSinusitisAugmentin
EDGuideline26Increased work of breathingReactive airway diseaseNo antibiotic
EDNonguideline16Recurrent feverAcute otitis mediaAmoxicillin
EDNonguideline20Recurrent cough and feverViral illnessNo antibiotic
AdmissionGuideline3Increased work of breathingPneumoniaIV ampicillin
AdmissionNonguideline9Refusal to take oral clindamycinPneumoniaIV clindamycin

Two patients were readmitted for a pneumonia‐related illness within 30 days of discharge; 1 had received guideline‐recommended therapy (Table 5). Both patients were directly admitted to the inpatient ward without an associated ED visit. Antibiotic class was not changed for either patient upon readmission, despite the decision to convert to intravenous form.

DISCUSSION

In this retrospective cohort study, patients who received empiric guideline‐recommended antibiotic therapy on admission for CAP had no difference in LOS, total cost of hospitalization, or inpatient pharmacy costs compared with those who received therapy that varied from guideline recommendations. Our study suggests that prescribing narrow‐spectrum therapy and, in some circumstances, combination therapy, as recommended by the 2011 PIDS/IDSA pneumonia guideline, did not result in negative unintended consequences.

In our study, children receiving guideline‐recommended therapy were younger, more likely to have elevated respiratory rate on presentation, and more likely to have an infiltrate on chest radiograph. We hypothesize the age difference is a reflection of common use of nonguideline macrolide monotherapy in the older, school‐age child, as macrolides are commonly used for coverage of Mycoplasma pneumonia in older children with CAP.[15] Children receiving macrolide monotherapy were older than those receiving guideline‐recommended therapy (median age of 7.4 years and 2.5 years, respectively). We also hypothesize that some providers may prescribe macrolide monotherapy to children deemed less ill than expected for CAP (eg, normal percutaneous oxygen saturation). This hypothesis is supported by the finding that 60% of patients who had a normal respiratory rate and received nonguideline therapy were prescribed macrolide monotherapy. We did control for the characteristics that varied between the two treatment groups in our models to eliminate potential confounding.

One prior study evaluated the effects of guideline implementation in CAP. In evaluation of a clinical practice guideline that recommended ampicillin as first‐line therapy for CAP, no significant difference was found following guideline introduction in the number of treatment failures (defined as the need to broaden therapy or development of complicated pneumonia within 48 hours or 30‐day inpatient readmission).[16] Our study builds on these findings by directly comparing outcomes between recipients of guideline and nonguideline therapy, which was not done in the prestudy or poststudy design of prior work.[16] We believe that classifying patients based on empiric therapy received rather than timing of guideline introduction thoroughly examines the effect of following guideline recommendations. Additionally, outcomes other than treatment failures were examined in our study including LOS, costs, and ED revisits.

Our results are similar to other observational studies that compared narrow‐ and broad‐spectrum antibiotics for children hospitalized with CAP. Using administrative data, Williams et al. found no significant difference in LOS or cost between narrow‐ and broad‐spectrum intravenous antibiotic recipients ages 6 months to 18 years at 43 children's hospitals.[17] Queen et al. compared narrow‐ and broad‐spectrum antibiotic therapy for children ages 2 months to 18 years at 4 children's hospitals.[18] Differences in average daily cost were not significant, but children who received narrow‐spectrum antibiotics had a significantly shorter LOS. Finally, an observational study of 319 Israeli children <2 years of age found no significant difference in duration of oxygen requirement, LOS, or need for change in antibiotic therapy between the 66 children prescribed aminopenicillins and the 253 children prescribed cefuroxime.[19] These studies suggest that prescribing narrow‐spectrum antimicrobial therapy, as per the guideline recommendations, results in similar outcomes to broad‐spectrum antimicrobial therapy.

Our study adds to prior studies that compared narrow‐ and broad‐spectrum therapy by comparing outcomes associated with guideline‐recommended therapy to nonguideline therapy. We considered antibiotic therapy as guideline recommended if appropriately chosen per the guideline, not just by simple classification of antibiotic. For example, the use of a cephalosporin in a patient with aminopenicillin allergy was considered guideline‐recommended therapy. We chose to classify the exposure of empiric antibiotic therapy in this manner to reflect true clinical application of the guideline, as not all children are able to receive aminopenicillins. Additionally, as our study stemmed from our prior improvement work aimed at increasing guideline adherent therapy,[7] we have a higher frequency of narrow‐spectrum antibiotic use than prior studies. In our study, almost two‐thirds of patients received narrow‐spectrum therapy, whereas narrow‐spectrum use in prior studies ranged from 10% to 33%.[17, 18, 19] Finally, we were able to confirm the diagnosis of pneumonia via medical record review and to adjust for severity of illness using clinical variables including vital signs, physical exam findings, and laboratory and radiologic study results.

This study must be interpreted in the context of several limitations. First, our study population was defined through discharge diagnosis codes, and therefore dependent on the accuracy of coding. However, we minimized potential for misclassification through use of a previously validated approach to identify patients with CAP and through medical record review to confirm the diagnosis. Second, we may be unable to detect very small differences in outcomes given limited power, specifically for outcomes of LOS, ED revisits, hospital readmissions, and need to broaden antibiotic therapy. Third, residual confounding may be present. Although we controlled for many clinical variables in our analyses, antibiotic prescribing practices may be influenced by unmeasured factors. The potential of system level confounding is mitigated by standardized care for patients with CAP at our institution. Prior system‐level changes using quality‐improvement science have resulted in a high level of adherence with guideline‐recommended antimicrobials as well as standardized medical discharge criteria.[7, 20] Additionally, nonmedical factors may influence LOS, limiting its use as an outcome measure. This limitation was minimized in our study by standardizing medical discharge criteria. Prior work at our institution demonstrated that the majority of patients, including those with CAP, were discharged within 2 hours of meeting medical discharge criteria.[20] Fourth, discharged patients who experienced adverse outcomes may have received care at a nearby adult emergency department or at their pediatrician's office. Although these events would not have been captured in our electronic health record, serious complications due to treatment failure (eg, empyema) would require hospitalization. As our hospital is the only children's hospital in the Greater Cincinnati metropolitan area, these patents would receive care at our institution. Therefore, any misclassification of revisits or readmissions is likely to be minimal. Finally, study patients were admitted to a large tertiary academic children's hospital, warranting further investigation to determine if these findings can be translated to smaller community settings.

In conclusion, receipt of guideline‐recommended antibiotic therapy for patients hospitalized with CAP was not associated with increases in LOS, total costs of hospitalization, or inpatient pharmacy costs. Our findings highlight the importance of changing antibiotic prescribing practices to reflect guideline recommendations, as there was no evidence of negative unintended consequences with our local practice change.

Acknowledgments

Disclosures: Dr. Ambroggio and Dr. Thomson were supported by funds from the NRSA T32HP10027‐14. Dr. Shah was supported by funds from NIAID K01A173729. The authors report no conflicts of interest.

References
  1. Kronman MP, Hersh AL, Feng R, Huang YS, Lee GE, Shah SS. Ambulatory visit rates and antibiotic prescribing for children with pneumonia, 1994–2007. Pediatrics. 2011;127(3):411418.
  2. Lee GE, Lorch SA, Sheffler‐Collins S, Kronman MP, Shah SS. National hospitalization trends for pediatric pneumonia and associated complications. Pediatrics. 2010;126(2):204213.
  3. Keren R, Luan X, Localio R, et al. Prioritization of comparative effectiveness research topics in hospital pediatrics. Arch Pediatr Adolesc Med. 2012;166(12):11551164.
  4. Bradley JS, Byington CL, Shah SS, et al. The management of community‐acquired pneumonia in infants and children older than 3 months of age: clinical practice guidelines by the Pediatric Infectious Diseases Society and the Infectious Diseases Society of America. Clin Infect Dis. 2011;53(7):e25e76.
  5. Brogan TV, Hall M, Williams DJ, et al. Variability in processes of care and outcomes among children hospitalized with community‐acquired pneumonia. Pediatr Infect Dis J. 2012;31(10):10361041.
  6. Ross RK, Hersh AL, Kronman MP, et al. Impact of IDSA/PIDS guidelines on treatment of community‐acquired pneumonia in hospitalized children. Clin Infect Dis. 2014;58(6):834838.
  7. Ambroggio L, Thomson J, Murtagh Kurowski E, et al. Quality improvement methods increase appropriate antibiotic prescribing for childhood pneumonia. Pediatrics. 2013;131(5):e1623e1631.
  8. Williams DJ, Shah SS, Myers A, et al. Identifying pediatric community‐acquired pneumonia hospitalizations: accuracy of administrative billing codes. JAMA Pediatr. 2013;167(9):851858.
  9. Feudtner C, Hays RM, Haynes G, Geyer JR, Neff JM, Koepsell TD. Deaths attributed to pediatric complex chronic conditions: national trends and implications for supportive care services. Pediatrics. 2001;107(6):E99.
  10. Centers for Medicare 2011.
  11. Kleinman ME, Chameides L, Schexnayder SM, et al. Pediatric advanced life support: 2010 American Heart Association guidelines for cardiopulmonary resuscitation and emergency cardiovascular care. Pediatrics. 2010;126(5):e1361e1399.
  12. Tucker KM, Brewer TL, Baker RB, Demeritt B, Vossmeyer MT. Prospective evaluation of a pediatric inpatient early warning scoring system. J Spec Pediatr Nurs. 2009;14(2):7985.
  13. Vittinghoff E, Glidden DV, Shiboski SC, McCulloch CE. Regression Methods in Biostatistics: Linear, Logistic, Survival, and Repeated Measures Models. New York, NY: Springer; 2005.
  14. Biondi E, McCulloh R, Alverson B, Klein A, Dixon A. Treatment of mycoplasma pneumonia: a systematic review. Pediatrics. 2014;133(6):10811090.
  15. Newman RE, Hedican EB, Herigon JC, Williams DD, Williams AR, Newland JG. Impact of a guideline on management of children hospitalized with community‐acquired pneumonia. Pediatrics. 2012;129(3):e597e604.
  16. Williams DJ, Hall M, Shah SS, et al. Narrow vs broad‐spectrum antimicrobial therapy for children hospitalized with pneumonia. Pediatrics. 2013;132(5):e1141e1148.
  17. Queen MA, Myers AL, Hall M, et al. Comparative effectiveness of empiric antibiotics for community‐acquired pneumonia. Pediatrics. 2014;133(1):e23e29.
  18. Dinur‐Schejter Y, Cohen‐Cymberknoh M, Tenenbaum A, et al. Antibiotic treatment of children with community‐acquired pneumonia: comparison of penicillin or ampicillin versus cefuroxime. Pediatr Pulmonol. 2013;48(1):5258.
  19. White CM, Statile AM, White DL, et al. Using quality improvement to optimise paediatric discharge efficiency. BMJ Qual Saf. 2014;23(5):428436.
References
  1. Kronman MP, Hersh AL, Feng R, Huang YS, Lee GE, Shah SS. Ambulatory visit rates and antibiotic prescribing for children with pneumonia, 1994–2007. Pediatrics. 2011;127(3):411418.
  2. Lee GE, Lorch SA, Sheffler‐Collins S, Kronman MP, Shah SS. National hospitalization trends for pediatric pneumonia and associated complications. Pediatrics. 2010;126(2):204213.
  3. Keren R, Luan X, Localio R, et al. Prioritization of comparative effectiveness research topics in hospital pediatrics. Arch Pediatr Adolesc Med. 2012;166(12):11551164.
  4. Bradley JS, Byington CL, Shah SS, et al. The management of community‐acquired pneumonia in infants and children older than 3 months of age: clinical practice guidelines by the Pediatric Infectious Diseases Society and the Infectious Diseases Society of America. Clin Infect Dis. 2011;53(7):e25e76.
  5. Brogan TV, Hall M, Williams DJ, et al. Variability in processes of care and outcomes among children hospitalized with community‐acquired pneumonia. Pediatr Infect Dis J. 2012;31(10):10361041.
  6. Ross RK, Hersh AL, Kronman MP, et al. Impact of IDSA/PIDS guidelines on treatment of community‐acquired pneumonia in hospitalized children. Clin Infect Dis. 2014;58(6):834838.
  7. Ambroggio L, Thomson J, Murtagh Kurowski E, et al. Quality improvement methods increase appropriate antibiotic prescribing for childhood pneumonia. Pediatrics. 2013;131(5):e1623e1631.
  8. Williams DJ, Shah SS, Myers A, et al. Identifying pediatric community‐acquired pneumonia hospitalizations: accuracy of administrative billing codes. JAMA Pediatr. 2013;167(9):851858.
  9. Feudtner C, Hays RM, Haynes G, Geyer JR, Neff JM, Koepsell TD. Deaths attributed to pediatric complex chronic conditions: national trends and implications for supportive care services. Pediatrics. 2001;107(6):E99.
  10. Centers for Medicare 2011.
  11. Kleinman ME, Chameides L, Schexnayder SM, et al. Pediatric advanced life support: 2010 American Heart Association guidelines for cardiopulmonary resuscitation and emergency cardiovascular care. Pediatrics. 2010;126(5):e1361e1399.
  12. Tucker KM, Brewer TL, Baker RB, Demeritt B, Vossmeyer MT. Prospective evaluation of a pediatric inpatient early warning scoring system. J Spec Pediatr Nurs. 2009;14(2):7985.
  13. Vittinghoff E, Glidden DV, Shiboski SC, McCulloch CE. Regression Methods in Biostatistics: Linear, Logistic, Survival, and Repeated Measures Models. New York, NY: Springer; 2005.
  14. Biondi E, McCulloh R, Alverson B, Klein A, Dixon A. Treatment of mycoplasma pneumonia: a systematic review. Pediatrics. 2014;133(6):10811090.
  15. Newman RE, Hedican EB, Herigon JC, Williams DD, Williams AR, Newland JG. Impact of a guideline on management of children hospitalized with community‐acquired pneumonia. Pediatrics. 2012;129(3):e597e604.
  16. Williams DJ, Hall M, Shah SS, et al. Narrow vs broad‐spectrum antimicrobial therapy for children hospitalized with pneumonia. Pediatrics. 2013;132(5):e1141e1148.
  17. Queen MA, Myers AL, Hall M, et al. Comparative effectiveness of empiric antibiotics for community‐acquired pneumonia. Pediatrics. 2014;133(1):e23e29.
  18. Dinur‐Schejter Y, Cohen‐Cymberknoh M, Tenenbaum A, et al. Antibiotic treatment of children with community‐acquired pneumonia: comparison of penicillin or ampicillin versus cefuroxime. Pediatr Pulmonol. 2013;48(1):5258.
  19. White CM, Statile AM, White DL, et al. Using quality improvement to optimise paediatric discharge efficiency. BMJ Qual Saf. 2014;23(5):428436.
Issue
Journal of Hospital Medicine - 10(1)
Issue
Journal of Hospital Medicine - 10(1)
Page Number
13-18
Page Number
13-18
Publications
Publications
Article Type
Display Headline
Hospital outcomes associated with guideline‐recommended antibiotic therapy for pediatric pneumonia
Display Headline
Hospital outcomes associated with guideline‐recommended antibiotic therapy for pediatric pneumonia
Sections
Article Source

© 2014 Society of Hospital Medicine

Disallow All Ads
Correspondence Location
Address for correspondence and reprint requests: Joanna Thomson, MD, Division of Hospital Medicine, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, ML 9016, Cincinnati, OH, 45220; Telephone: 513‐803‐8092; Fax: 13‐803‐9244; E‐mail: joanna.thomson@cchmc.org
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Article PDF Media
Image
Disable zoom
Off
Media Files
Image
Disable zoom
Off

Radiographs Predict Pneumonia Severity

Article Type
Changed
Sun, 05/21/2017 - 14:01
Display Headline
Admission chest radiographs predict illness severity for children hospitalized with pneumonia

The 2011 Pediatric Infectious Diseases Society and Infectious Diseases Society of America (PIDS/IDSA) guidelines for management of pediatric community‐acquired pneumonia (CAP) recommend that admission chest radiographs be obtained in all children hospitalized with CAP to document the presence and extent of infiltrates and to identify complications.[1] Findings from chest radiographs may also provide clues to etiology and assist with predicting disease outcomes. In adults with CAP, clinical prediction tools use radiographic findings to inform triage decisions, guide management strategies, and predict outcomes.[2, 3, 4, 5, 6, 7] Whether or not radiographic findings could have similar utility among children with CAP is unknown.

Several retrospective studies have examined the ability of chest radiographs to predict pediatric pneumonia disease severity.[8, 9, 10, 11, 12] However, these studies used several different measures of severe pneumonia and/or were limited to young children <5 years of age, leading to inconsistent findings. These studies also rarely considered very severe disease (eg, need for invasive mechanical ventilation) or longitudinal outcome measures such as hospital length of stay. Finally, all of these prior studies were conducted outside of the United States, and most were single‐center investigations, potentially limiting generalizability. We sought to examine associations between admission chest radiographic findings and subsequent hospital care processes and clinical outcomes, including length of stay and resource utilization measures, among children hospitalized with CAP at 4 children's hospitals in the United States.

METHODS

Design and Setting

This study was nested within a multicenter retrospective cohort designed to validate International Classification of Diseases, 9th Revision, Clinical Modification (ICD9‐CM) diagnostic codes for pediatric CAP hospitalizations.[13] The Pediatric Health Information System database (Children's Hospital Association, Overland Park, KS) was used to identify children from 4 freestanding pediatric hospitals (Monroe Carell, Jr. Children's Hospital at Vanderbilt, Nashville, Tennessee; Children's Mercy Hospitals & Clinics, Kansas City, Missouri; Seattle Children's Hospital, Seattle, Washington; and Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio). The institutional review boards at each participating institution approved the study. The validation study included a 25% random sampling of children 60 days to 18 years of age (n=998) who were hospitalized between January 1, 2010 and December 31, 2010 with at least 1 ICD9‐CM discharge code indicating pneumonia. The diagnosis of CAP was confirmed by medical record review.

Study Population

This study was limited to children from the validation study who met criteria for clinical and radiographic CAP, defined as: (1) abnormal temperature or white blood cell count, (2) signs and symptoms of acute respiratory illness (eg, cough, tachypnea), and (3) chest radiograph indicating pneumonia within 48 hours of admission. Children with atelectasis as the only abnormal radiographic finding and those with complex chronic conditions (eg, cystic fibrosis, malignancy) were excluded using a previously described algorithm.[14]

Outcomes

Several measures of disease severity were assessed. Dichotomous outcomes included supplemental oxygen use, need for intensive care unit (ICU) admission, and need for invasive mechanical ventilation. Continuous outcomes included hospital length of stay, and for those requiring supplemental oxygen, duration of oxygen supplementation, measured in hours.

Exposure

To categorize infiltrate patterns and the presence and size of pleural effusions, we reviewed the final report from admission chest radiographs to obtain the final clinical interpretation performed by the attending pediatric radiologist. Infiltrate patterns were classified as single lobar (reference), unilateral multilobar, bilateral multilobar, or interstitial. Children with both lobar and interstitial infiltrates, and those with mention of atelectasis, were classified according to the type of lobar infiltrate. Those with atelectasis only were excluded. Pleural effusions were classified as absent, small, or moderate/large.

Analysis

Descriptive statistics were summarized using frequencies and percentages for categorical variables and median and interquartile range (IQR) values for continuous variables. Our primary exposures were infiltrate pattern and presence and size of pleural effusion on admission chest radiograph. Associations between radiographic findings and disease outcomes were analyzed using logistic and linear regression for dichotomous and continuous variables, respectively. Continuous outcomes were log‐transformed and normality assumptions verified prior to model development.

Due to the large number of covariates relative to outcome events, we used propensity score methods to adjust for potential confounding. The propensity score estimates the likelihood of a given exposure (ie, infiltrate pattern) conditional on a set of covariates. In this way, the propensity score summarizes potential confounding effects from a large number of covariates into a single variable. Including the propensity score as a covariate in multivariable regression improves model efficiency and helps protect against overfitting.[15] Covariates included in the estimation of the propensity score included age, sex, race/ethnicity, payer, hospital, asthma history, hospital transfer, recent hospitalization (within 30 days), recent emergency department or clinic visit (within 2 weeks), recent antibiotics for acute illness (within 5 days), illness duration prior to admission, tachypnea and/or increased work of breathing (retractions, nasal flaring, or grunting) at presentation, receipt of albuterol and/or corticosteroids during the first 2 calendar days of hospitalization, and concurrent diagnosis of bronchiolitis. All analyses included the estimated propensity score, infiltrate pattern, and pleural effusion (absent, small, or moderate/large).

RESULTS

Study Population

The median age of the 406 children with clinical and radiographic CAP was 3 years (IQR, 16 years) (Table 1). Single lobar infiltrate was the most common radiographic pattern (61%). Children with interstitial infiltrates (10%) were younger than those with lobar infiltrates of any type (median age 1 vs 3 years, P=0.02). A concomitant diagnosis of bronchiolitis was assigned to 34% of children with interstitial infiltrates but only 17% of those with lobar infiltrate patterns (range, 11%20%, P=0.03). Pleural effusion was present in 21% of children and was more common among those with lobar infiltrates, particularly multilobar disease. Only 1 child with interstitial infiltrate had a pleural effusion. Overall, 63% of children required supplemental oxygen, 8% required ICU admission, and 3% required invasive mechanical ventilation. Median length of stay was 51.5 hours (IQR, 3991) and median oxygen duration was 31.5 hours [IQR, 1365]. There were no deaths.

Characteristics of Children Hospitalized With Community‐Acquired Pneumonia According to Admission Radiographic Findings
CharacteristicInfiltrate PatternaP Valueb
Single LobarMultilobar, UnilateralMultilobar, BilateralInterstitial
  • NOTE: Data are presented as number (%) or median [IQR]. Abbreviations: ICU, intensive care unit; IQR, interquartile range; O2, oxygen.

  • Children with both lobar and interstitial infiltrates were classified according to the type of lobar infiltrate

  • P values are from 2 statistics for categorical variables and Kruskal‐Wallis tests for continuous variables.

No.247 (60.8)54 (13.3)64 (15.8)41 (10.1) 
Median age, y3 [16]3 [17]3 [15]1 [03]0.02
Male sex124 (50.2)32 (59.3)41 (64.1)30 (73.2)0.02
Race     
Non‐Hispanic white133 (53.8)36 (66.7)37 (57.8)17 (41.5)0.69
Non‐Hispanic black40 (16.2)6 (11.1)9 (14.1)8 (19.5) 
Hispanic25 (10.1)4 (7.4)5 (7.8)7 (17.1) 
Other49 (19.9)8 (14.8)13 (20.4)9 (22) 
Insurance     
Public130 (52.6)26 (48.1)33 (51.6)25 (61)0.90
Private116 (47)28 (51.9)31 (48.4)16 (39) 
Concurrent diagnosis     
Asthma80 (32.4)16 (29.6)17 (26.6)12 (29.3)0.82
Bronchiolitis43 (17.4)6 (11.1)13 (20.3)14 (34.1)0.03
Effusion     
None201 (81.4)31 (57.4)48 (75)40 (97.6)<.01
Small34 (13.8)20 (37)11 (17.2)0 
Moderate/large12 (4.9)3 (5.6)5 (7.8)1 (2.4) 

Outcomes According to Radiographic Infiltrate Pattern

Compared to children with single lobar infiltrates, the odds of ICU admission was significantly increased for those with either unilateral or bilateral multilobar infiltrates (unilateral, adjusted odds ratio [aOR]: 8.0, 95% confidence interval [CI]: 2.922.2; bilateral, aOR: 6.6, 95% CI: 2.14.5) (Figure 1, Table 2). Patients with bilateral multilobar infiltrates also had higher odds for supplemental oxygen use (aOR: 2.7, 95% CI: 1.25.8) and need for invasive mechanical ventilation (aOR: 3.0, 95% CI: 1.27.9). There were no differences in duration of oxygen supplementation or hospital length of stay for children with single versus multilobar infiltrates.

jhm2227-fig-0001-m.png
Propensity‐adjusted odds ratios for severe outcomes for children hospitalized with community‐acquired pneumonia according to admission radiographic findings. Single lobar infiltrate is the reference. Children with both lobar and interstitial infiltrates were classified according to the type of lobar infiltrate. Covariates included in the propensity score included: age, sex, race/ethnicity, payer, hospital, asthma history, hospital transfer, recent hospitalization (within 30 days), recent emergency department or clinic visit (within 2 weeks), recent antibiotics for acute illness (within 5 days), illness duration prior to admission, tachypnea and/or increased work of breathing (retractions, nasal flaring, or grunting) at presentation, receipt of albuterol and/or corticosteroids during the first 2 calendar days, and concurrent diagnosis of bronchiolitis. Pleural effusion (absent, small, or moderate/large) was included as a separate covariate. **Indicates that confidence interval (CIs) extends beyond the graph. The upper 95% CI for the odds ratio (OR) for infiltrates that were multilobar and unilateral was 22.2 for intensive care unit (ICU) admission and 37.8 for mechanical ventilation. Abbreviations: O2, oxygen.
Severe Outcomes for Children Hospitalized With Community‐Acquired Pneumonia According to Admission Radiographic Findings
OutcomeInfiltrate PatternaP Valueb
Single Lobar, n=247Multilobar, Unilateral, n=54Multilobar, Bilateral, n=64Interstitial, n=41
  • NOTE: Data are presented as number (%) or median [IQR]. Abbreviations: ICU, intensive care unit; IQR, interquartile range, O2, oxygen.

  • Children with both lobar and interstitial infiltrates were classified according to the type of lobar infiltrate.

  • P values are from 2 statistics for categorical variables and Kruskal‐Wallis tests for continuous variables.

Supplemental O2 requirement143 (57.9)34 (63)46 (71.9)31 (75.6)0.05
ICU admission10 (4)9 (16.7)9 (14.1)4 (9.8)<0.01
Mechanical ventilation5 (2)4 (7.4)4 (6.3)1 (2.4)0.13
Hospital length of stay, h47 [3779]63 [45114]56.5 [39.5101]62 [3993]<0.01
O2 duration, h27 [1059]38 [1777]38 [2381]34.5 [1765]0.18

Compared to those with single lobar infiltrates, children with interstitial infiltrates had higher odds of need for supplemental oxygen (aOR: 3.1, 95% CI: 1.37.6) and ICU admission (aOR: 4.4, 95% CI: 1.314.3) but not invasive mechanical ventilation. There were also no differences in duration of oxygen supplementation or hospital length of stay.

Outcomes According to Presence and Size of Pleural Effusion

Compared to those without pleural effusion, children with moderate to large effusion had a higher odds of ICU admission (aOR: 3.2, 95% CI: 1.18.9) and invasive mechanical ventilation (aOR: 14.8, 95% CI: 9.822.4), and also had a longer duration of oxygen supplementation (aOR: 3.0, 95% CI: 1.46.5) and hospital length of stay (aOR: 2.6, 95% CI: 1.9‐3.6) (Table 3, Figure 2). The presence of a small pleural effusion was not associated with increased need for supplemental oxygen, ICU admission, or mechanical ventilation compared to those without effusion. However, small effusion was associated with a longer duration of oxygen supplementation (aOR: 1.7, 95% CI: 12.7) and hospital length of stay (aOR: 1.6, 95% CI: 1.3‐1.9).

Severe Outcomes for Children Hospitalized With Community‐Acquired Pneumonia According to Presence and Size of Pleural Effusion
OutcomePleural EffusionP Valuea
None, n=320Small, n=65Moderate/Large, n=21
  • NOTE: Data are presented as number (%) or median [IQR]. Abbreviations: ICU, intensive care unit; IQR, interquartile range; O2, oxygen.

  • P values are from 2 statistics for categorical variables and Kruskal‐Wallis tests for continuous variables.

Supplemental O2 requirement200 (62.5)40 (61.5)14 (66.7)0.91
ICU admission22 (6.9)6 (9.2)4 (19)0.12
Mechanical ventilation5 (1.6)5 (7.7)4 (19)<0.01
Hospital length of stay, h48 [37.576]72 [45142]160 [82191]<0.01
Oxygen duration, h31 [1157]38.5 [1887]111 [27154]<0.01
jhm2227-fig-0002-m.png
Propensity‐adjusted odds ratios for severe outcomes for children hospitalized with community‐acquired pneumonia according to presence and size of effusion. No effusion is the reference. Covariates included in the propensity score included: age, sex, race/ethnicity, payer, hospital, asthma history, hospital transfer, recent hospitalization (within 30 days), recent emergency department or clinic visit (within 2 weeks), recent antibiotics for acute illness (within 5 days), illness duration prior to admission, tachypnea and/or increased work of breathing (retractions, nasal flaring, or grunting) at presentation, receipt of albuterol and/or corticosteroids during the first 2 calendar days, and concurrent diagnosis of bronchiolitis. Infiltrate pattern was included as a separate covariate. **Indicates confidence interval (CI) extends beyond the graph. The upper 95% CI for the odds ratio (OR) for mechanical ventilation was 34.2 for small effusion and 22.4 for moderate/large effusion. Abbreviations: ICU, intensive care unit; O2, oxygen.

DISCUSSION

We evaluated the association between admission chest radiographic findings and subsequent clinical outcomes and hospital care processes for children hospitalized with CAP at 4 children's hospitals in the United States. We conclude that radiographic findings are associated with important inpatient outcomes. Similar to data from adults, findings of moderate to large pleural effusions and bilateral multilobar infiltrates had the strongest associations with severe disease. Such information, in combination with other prognostic factors, may help clinicians identify high‐risk patients and support management decisions, while also helping to inform families about the expected hospital course.

Previous pediatric studies examining the association between radiographic findings and outcomes have produced inconsistent results.[8, 9, 10, 11, 12] All but 1 of these studies documented 1 radiographic characteristics associated with pneumonia disease severity.[11] Further, although most contrasted lobar/alveolar and interstitial infiltrates, only Patria et al. distinguished among lobar infiltrate patterns (eg, single lobar vs multilobar).[12] Similar to our findings, that study demonstrated increased disease severity among children with bilateral multifocal lobar infiltrates. Of the studies that considered the presence of pleural effusion, only 1 demonstrated this finding to be associated with more severe disease.[9] However, none of these prior studies examined size of the pleural effusion.

In our study, the strongest association with severe pneumonia outcomes was among children with moderate to large pleural effusion. Significant pleural effusions are much more commonly due to infection with bacterial pathogens, particularly Streptococcus pneumoniae, Staphylococcus aureus, and Streptococcus pyogenes, and may also indicate infection with more virulent and/or difficult to treat strains.[16, 17, 18, 19] Surgical intervention is also often required. As such, children with significant pleural effusions are often more ill on presentation and may have a prolonged period of recovery.[20, 21, 22]

Similarly, multilobar infiltrates, particularly bilateral, were associated with increased disease severity in terms of need for supplemental oxygen, ICU admission, and need for invasive mechanical ventilation. Although this finding may be expected, it is interesting to note that the duration of supplemental oxygen and hospital length of stay were similar to those with single lobar disease. One potential explanation is that, although children with multilobar disease are more severe at presentation, rates of recovery are similar to those with less extensive radiographic findings, owing to rapidly effective antimicrobials for uncomplicated bacterial pneumonia. This hypothesis also agrees with the 2011 PIDS/IDSA guidelines, which state that children receiving adequate therapy typically show signs of improvement within 48 to 72 hours regardless of initial severity.[1]

Interstitial infiltrate was also associated with increased severity at presentation but similar length of stay and duration of oxygen requirement compared with single lobar disease. We note that these children were substantially younger than those presenting with any pattern of lobar disease (median age, 1 vs 3 years), were more likely to have a concurrent diagnosis of bronchiolitis (34% vs 17%), and only 1 child with interstitial infiltrates had a documented pleural effusion (vs 23% of children with lobar infiltrates). Primary viral pneumonia is considered more likely to produce interstitial infiltrates on chest radiograph compared to bacterial disease, and although detailed etiologic data are unavailable for this study, our findings above strongly support this assertion.[23, 24]

The 2011 PIDS/IDSA guidelines recommend admission chest radiographs for all children hospitalized with pneumonia to assess extent of disease and identify complications that may requiring additional evaluation or surgical intervention.[1] Our findings highlight additional potential benefits of admission radiographs in terms of disease prognosis and management decisions. In the initial evaluation of a sick child with pneumonia, clinicians are often presented with a number of potential prognostic factors that may influence disease outcomes. However, it is sometimes difficult for providers to consider all available information and/or the relative importance of a single factor, resulting in inaccurate risk perceptions and management decisions that may contribute to poor outcomes.[25] Similar to adults, the development of clinical prediction rules, which incorporate a variety of important predictors including admission radiographic findings, likely would improve risk assessments and potentially outcomes for children with pneumonia. Such prognostic information is also helpful for clinicians who may use these data to inform and prepare families regarding the expected course of hospitalization.

Our study has several limitations. This study was retrospective and only included a sample of pneumonia hospitalizations during the study period, which may raise confounding concerns and potential for selection bias. However, detailed medical record reviews using standardized case definitions for radiographic CAP were used, and a large sample of children was randomly selected from each institution. In addition, a large number of potential confounders were selected a priori and included in multivariable analyses; propensity score adjustment was used to reduce model complexity and avoid overfitting. Radiographic findings were based on clinical interpretation by pediatric radiologists independent of a study protocol. Prior studies have demonstrated good agreement for identification of alveolar/lobar infiltrates and pleural effusion by trained radiologists, although agreement for interstitial infiltrate is poor.[26, 27] This limitation could result in either over‐ or underestimation of the prevalence of interstitial infiltrates likely resulting in a nondifferential bias toward the null. Microbiologic information, which may inform radiographic findings and disease severity, was also not available. However, because pneumonia etiology is frequently unknown in the clinical setting, our study reflects typical practice. We also did not include children from community or nonteaching hospitals. Thus, although findings may have relevance to community or nonteaching hospitals, our results cannot be generalized.

CONCLUSION

Our study demonstrates that among children hospitalized with CAP, admission chest radiographic findings are associated with important clinical outcomes and hospital care processes, highlighting additional benefits of the 2011 PIDS/IDSA guidelines' recommendation for admission chest radiographs for all children hospitalized with pneumonia. These data, in conjunction with other important prognostic information, may help clinicians more rapidly identify children at increased risk for severe illness, and could also offer guidance regarding disease management strategies and facilitate shared decision making with families. Thus, routine admission chest radiography in this population represents a valuable tool that contributes to improved quality of care.

Disclosures

Dr. Williams is supported by funds from the National Institutes of HealthNational Institute of Allergy and Infectious Diseases (K23AI104779). The authors report no conflicts of interest.

Files
References
  1. Bradley JS, Byington CL, Shah SS, et al. The management of community‐acquired pneumonia in infants and children older than 3 months of age: clinical practice guidelines by the Pediatric Infectious Diseases Society and the Infectious Diseases Society of America. Clin Infect Dis. 2011;53(7):e25e76.
  2. Fine MJ, Auble TE, Yealy DM, et al. A prediction rule to identify low‐risk patients with community‐acquired pneumonia. N Engl J Med. 1997;336(4):243250.
  3. Charles PG, Wolfe R, Whitby M, et al. SMART‐COP: a tool for predicting the need for intensive respiratory or vasopressor support in community‐acquired pneumonia. Clin Infect Dis. 2008;47(3):375384.
  4. Espana PP, Capelastegui A, Gorordo I, et al. Development and validation of a clinical prediction rule for severe community‐acquired pneumonia. Am J Respir Crit Care Med. 2006;174(11):12491256.
  5. Renaud B, Labarere J, Coma E, et al. Risk stratification of early admission to the intensive care unit of patients with no major criteria of severe community‐acquired pneumonia: development of an international prediction rule. Crit Care. 2009;13(2):R54.
  6. Hasley PB, Albaum MN, Li YH, et al. Do pulmonary radiographic findings at presentation predict mortality in patients with community‐acquired pneumonia? Arch Intern Med. 1996;156(19):22062212.
  7. Chalmers JD, Singanayagam A, Akram AR, Choudhury G, Mandal P, Hill AT. Safety and efficacy of CURB65‐guided antibiotic therapy in community‐acquired pneumonia. J Antimicrob Chemother. 2011;66(2):416423.
  8. Kin Key N, Araujo‐Neto CA, Nascimento‐Carvalho CM. Severity of childhood community‐acquired pneumonia and chest radiographic findings. Pediatr Pulmonol. 2009;44(3):249252.
  9. Grafakou O, Moustaki M, Tsolia M, et al. Can chest x‐ray predict pneumonia severity? Pediatr Pulmonol. 2004;38(6):465469.
  10. Clark JE, Hammal D, Spencer D, Hampton F. Children with pneumonia: how do they present and how are they managed? Arch Dis Child. 2007;92(5):394398.
  11. Bharti B, Kaur L, Bharti S. Role of chest X‐ray in predicting outcome of acute severe pneumonia. Indian Pediatr. 2008;45(11):893898.
  12. Patria MF, Longhi B, Lelii M, Galeone C, Pavesi MA, Esposito S. Association between radiological findings and severity of community‐acquired pneumonia in children. Ital J Pediatr. 2013;39:56.
  13. Williams DJ, Shah SS, Myers AM, et al. Identifying pediatric community‐acquired pneumonia hospitalizations: accuracy of administrative billing codes. JAMA Pediatrics. 2013;167(9):851858.
  14. Feudtner C, Hays RM, Haynes G, Geyer JR, Neff JM, Koepsell TD. Deaths attributed to pediatric complex chronic conditions: national trends and implications for supportive care services. Pediatrics. 2001;107(6):E99.
  15. Joffe MM, Rosenbaum PR. Invited commentary: propensity scores. Am J Epidemiol. 1999;150(4):327333.
  16. Grijalva CG, Nuorti JP, Zhu Y, Griffin MR. Increasing incidence of empyema complicating childhood community‐acquired pneumonia in the United States. Clin Infect Dis. 2010;50(6):805813.
  17. Michelow IC, Olsen K, Lozano J, et al. Epidemiology and clinical characteristics of community‐acquired pneumonia in hospitalized children. Pediatrics. 2004;113(4):701707.
  18. Blaschke AJ, Heyrend C, Byington CL, et al. Molecular analysis improves pathogen identification and epidemiologic study of pediatric parapneumonic empyema. Pediatr Infect Dis J. 2011;30(4):289294.
  19. Chonmaitree T, Powell KR. Parapneumonic pleural effusion and empyema in children. Review of a 19‐year experience, 1962–1980. Clin Pediatr (Phila). 1983;22(6):414419.
  20. Huang CY, Chang L, Liu CC, et al. Risk factors of progressive community‐acquired pneumonia in hospitalized children: a prospective study [published online ahead of print August 28, 2013]. J Microbiol Immunol Infect. doi: 10.1016/j.jmii.2013.06.009.
  21. Rowan‐Legg A, Barrowman N, Shenouda N, Koujok K, Saux N. Community‐acquired lobar pneumonia in children in the era of universal 7‐valent pneumococcal vaccination: a review of clinical presentations and antimicrobial treatment from a Canadian pediatric hospital. BMC Pediatr. 2012;12:133.
  22. Wexler ID, Knoll S, Picard E, et al. Clinical characteristics and outcome of complicated pneumococcal pneumonia in a pediatric population. Pediatr Pulmonol. 2006;41(8):726734.
  23. Virkki R, Juven T, Rikalainen H, Svedstrom E, Mertsola J, Ruuskanen O. Differentiation of bacterial and viral pneumonia in children. Thorax. 2002;57(5):438441.
  24. Harris M, Clark J, Coote N, et al. British Thoracic Society guidelines for the management of community acquired pneumonia in children: update 2011. Thorax. 2011;66(suppl 2):ii1ii23.
  25. Neill AM, Martin IR, Weir R, et al. Community acquired pneumonia: aetiology and usefulness of severity criteria on admission. Thorax. 1996;51(10):10101016.
  26. Neuman MI, Lee EY, Bixby S, et al. Variability in the interpretation of chest radiographs for the diagnosis of pneumonia in children. J Hosp Med. 2012;7(4):294298.
  27. Albaum MN, Hill LC, Murphy M, et al. Interobserver reliability of the chest radiograph in community‐acquired pneumonia. PORT Investigators. Chest. 1996;110(2):343350.
Article PDF
Issue
Journal of Hospital Medicine - 9(9)
Publications
Page Number
559-564
Sections
Files
Files
Article PDF
Article PDF

The 2011 Pediatric Infectious Diseases Society and Infectious Diseases Society of America (PIDS/IDSA) guidelines for management of pediatric community‐acquired pneumonia (CAP) recommend that admission chest radiographs be obtained in all children hospitalized with CAP to document the presence and extent of infiltrates and to identify complications.[1] Findings from chest radiographs may also provide clues to etiology and assist with predicting disease outcomes. In adults with CAP, clinical prediction tools use radiographic findings to inform triage decisions, guide management strategies, and predict outcomes.[2, 3, 4, 5, 6, 7] Whether or not radiographic findings could have similar utility among children with CAP is unknown.

Several retrospective studies have examined the ability of chest radiographs to predict pediatric pneumonia disease severity.[8, 9, 10, 11, 12] However, these studies used several different measures of severe pneumonia and/or were limited to young children <5 years of age, leading to inconsistent findings. These studies also rarely considered very severe disease (eg, need for invasive mechanical ventilation) or longitudinal outcome measures such as hospital length of stay. Finally, all of these prior studies were conducted outside of the United States, and most were single‐center investigations, potentially limiting generalizability. We sought to examine associations between admission chest radiographic findings and subsequent hospital care processes and clinical outcomes, including length of stay and resource utilization measures, among children hospitalized with CAP at 4 children's hospitals in the United States.

METHODS

Design and Setting

This study was nested within a multicenter retrospective cohort designed to validate International Classification of Diseases, 9th Revision, Clinical Modification (ICD9‐CM) diagnostic codes for pediatric CAP hospitalizations.[13] The Pediatric Health Information System database (Children's Hospital Association, Overland Park, KS) was used to identify children from 4 freestanding pediatric hospitals (Monroe Carell, Jr. Children's Hospital at Vanderbilt, Nashville, Tennessee; Children's Mercy Hospitals & Clinics, Kansas City, Missouri; Seattle Children's Hospital, Seattle, Washington; and Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio). The institutional review boards at each participating institution approved the study. The validation study included a 25% random sampling of children 60 days to 18 years of age (n=998) who were hospitalized between January 1, 2010 and December 31, 2010 with at least 1 ICD9‐CM discharge code indicating pneumonia. The diagnosis of CAP was confirmed by medical record review.

Study Population

This study was limited to children from the validation study who met criteria for clinical and radiographic CAP, defined as: (1) abnormal temperature or white blood cell count, (2) signs and symptoms of acute respiratory illness (eg, cough, tachypnea), and (3) chest radiograph indicating pneumonia within 48 hours of admission. Children with atelectasis as the only abnormal radiographic finding and those with complex chronic conditions (eg, cystic fibrosis, malignancy) were excluded using a previously described algorithm.[14]

Outcomes

Several measures of disease severity were assessed. Dichotomous outcomes included supplemental oxygen use, need for intensive care unit (ICU) admission, and need for invasive mechanical ventilation. Continuous outcomes included hospital length of stay, and for those requiring supplemental oxygen, duration of oxygen supplementation, measured in hours.

Exposure

To categorize infiltrate patterns and the presence and size of pleural effusions, we reviewed the final report from admission chest radiographs to obtain the final clinical interpretation performed by the attending pediatric radiologist. Infiltrate patterns were classified as single lobar (reference), unilateral multilobar, bilateral multilobar, or interstitial. Children with both lobar and interstitial infiltrates, and those with mention of atelectasis, were classified according to the type of lobar infiltrate. Those with atelectasis only were excluded. Pleural effusions were classified as absent, small, or moderate/large.

Analysis

Descriptive statistics were summarized using frequencies and percentages for categorical variables and median and interquartile range (IQR) values for continuous variables. Our primary exposures were infiltrate pattern and presence and size of pleural effusion on admission chest radiograph. Associations between radiographic findings and disease outcomes were analyzed using logistic and linear regression for dichotomous and continuous variables, respectively. Continuous outcomes were log‐transformed and normality assumptions verified prior to model development.

Due to the large number of covariates relative to outcome events, we used propensity score methods to adjust for potential confounding. The propensity score estimates the likelihood of a given exposure (ie, infiltrate pattern) conditional on a set of covariates. In this way, the propensity score summarizes potential confounding effects from a large number of covariates into a single variable. Including the propensity score as a covariate in multivariable regression improves model efficiency and helps protect against overfitting.[15] Covariates included in the estimation of the propensity score included age, sex, race/ethnicity, payer, hospital, asthma history, hospital transfer, recent hospitalization (within 30 days), recent emergency department or clinic visit (within 2 weeks), recent antibiotics for acute illness (within 5 days), illness duration prior to admission, tachypnea and/or increased work of breathing (retractions, nasal flaring, or grunting) at presentation, receipt of albuterol and/or corticosteroids during the first 2 calendar days of hospitalization, and concurrent diagnosis of bronchiolitis. All analyses included the estimated propensity score, infiltrate pattern, and pleural effusion (absent, small, or moderate/large).

RESULTS

Study Population

The median age of the 406 children with clinical and radiographic CAP was 3 years (IQR, 16 years) (Table 1). Single lobar infiltrate was the most common radiographic pattern (61%). Children with interstitial infiltrates (10%) were younger than those with lobar infiltrates of any type (median age 1 vs 3 years, P=0.02). A concomitant diagnosis of bronchiolitis was assigned to 34% of children with interstitial infiltrates but only 17% of those with lobar infiltrate patterns (range, 11%20%, P=0.03). Pleural effusion was present in 21% of children and was more common among those with lobar infiltrates, particularly multilobar disease. Only 1 child with interstitial infiltrate had a pleural effusion. Overall, 63% of children required supplemental oxygen, 8% required ICU admission, and 3% required invasive mechanical ventilation. Median length of stay was 51.5 hours (IQR, 3991) and median oxygen duration was 31.5 hours [IQR, 1365]. There were no deaths.

Characteristics of Children Hospitalized With Community‐Acquired Pneumonia According to Admission Radiographic Findings
CharacteristicInfiltrate PatternaP Valueb
Single LobarMultilobar, UnilateralMultilobar, BilateralInterstitial
  • NOTE: Data are presented as number (%) or median [IQR]. Abbreviations: ICU, intensive care unit; IQR, interquartile range; O2, oxygen.

  • Children with both lobar and interstitial infiltrates were classified according to the type of lobar infiltrate

  • P values are from 2 statistics for categorical variables and Kruskal‐Wallis tests for continuous variables.

No.247 (60.8)54 (13.3)64 (15.8)41 (10.1) 
Median age, y3 [16]3 [17]3 [15]1 [03]0.02
Male sex124 (50.2)32 (59.3)41 (64.1)30 (73.2)0.02
Race     
Non‐Hispanic white133 (53.8)36 (66.7)37 (57.8)17 (41.5)0.69
Non‐Hispanic black40 (16.2)6 (11.1)9 (14.1)8 (19.5) 
Hispanic25 (10.1)4 (7.4)5 (7.8)7 (17.1) 
Other49 (19.9)8 (14.8)13 (20.4)9 (22) 
Insurance     
Public130 (52.6)26 (48.1)33 (51.6)25 (61)0.90
Private116 (47)28 (51.9)31 (48.4)16 (39) 
Concurrent diagnosis     
Asthma80 (32.4)16 (29.6)17 (26.6)12 (29.3)0.82
Bronchiolitis43 (17.4)6 (11.1)13 (20.3)14 (34.1)0.03
Effusion     
None201 (81.4)31 (57.4)48 (75)40 (97.6)<.01
Small34 (13.8)20 (37)11 (17.2)0 
Moderate/large12 (4.9)3 (5.6)5 (7.8)1 (2.4) 

Outcomes According to Radiographic Infiltrate Pattern

Compared to children with single lobar infiltrates, the odds of ICU admission was significantly increased for those with either unilateral or bilateral multilobar infiltrates (unilateral, adjusted odds ratio [aOR]: 8.0, 95% confidence interval [CI]: 2.922.2; bilateral, aOR: 6.6, 95% CI: 2.14.5) (Figure 1, Table 2). Patients with bilateral multilobar infiltrates also had higher odds for supplemental oxygen use (aOR: 2.7, 95% CI: 1.25.8) and need for invasive mechanical ventilation (aOR: 3.0, 95% CI: 1.27.9). There were no differences in duration of oxygen supplementation or hospital length of stay for children with single versus multilobar infiltrates.

jhm2227-fig-0001-m.png
Propensity‐adjusted odds ratios for severe outcomes for children hospitalized with community‐acquired pneumonia according to admission radiographic findings. Single lobar infiltrate is the reference. Children with both lobar and interstitial infiltrates were classified according to the type of lobar infiltrate. Covariates included in the propensity score included: age, sex, race/ethnicity, payer, hospital, asthma history, hospital transfer, recent hospitalization (within 30 days), recent emergency department or clinic visit (within 2 weeks), recent antibiotics for acute illness (within 5 days), illness duration prior to admission, tachypnea and/or increased work of breathing (retractions, nasal flaring, or grunting) at presentation, receipt of albuterol and/or corticosteroids during the first 2 calendar days, and concurrent diagnosis of bronchiolitis. Pleural effusion (absent, small, or moderate/large) was included as a separate covariate. **Indicates that confidence interval (CIs) extends beyond the graph. The upper 95% CI for the odds ratio (OR) for infiltrates that were multilobar and unilateral was 22.2 for intensive care unit (ICU) admission and 37.8 for mechanical ventilation. Abbreviations: O2, oxygen.
Severe Outcomes for Children Hospitalized With Community‐Acquired Pneumonia According to Admission Radiographic Findings
OutcomeInfiltrate PatternaP Valueb
Single Lobar, n=247Multilobar, Unilateral, n=54Multilobar, Bilateral, n=64Interstitial, n=41
  • NOTE: Data are presented as number (%) or median [IQR]. Abbreviations: ICU, intensive care unit; IQR, interquartile range, O2, oxygen.

  • Children with both lobar and interstitial infiltrates were classified according to the type of lobar infiltrate.

  • P values are from 2 statistics for categorical variables and Kruskal‐Wallis tests for continuous variables.

Supplemental O2 requirement143 (57.9)34 (63)46 (71.9)31 (75.6)0.05
ICU admission10 (4)9 (16.7)9 (14.1)4 (9.8)<0.01
Mechanical ventilation5 (2)4 (7.4)4 (6.3)1 (2.4)0.13
Hospital length of stay, h47 [3779]63 [45114]56.5 [39.5101]62 [3993]<0.01
O2 duration, h27 [1059]38 [1777]38 [2381]34.5 [1765]0.18

Compared to those with single lobar infiltrates, children with interstitial infiltrates had higher odds of need for supplemental oxygen (aOR: 3.1, 95% CI: 1.37.6) and ICU admission (aOR: 4.4, 95% CI: 1.314.3) but not invasive mechanical ventilation. There were also no differences in duration of oxygen supplementation or hospital length of stay.

Outcomes According to Presence and Size of Pleural Effusion

Compared to those without pleural effusion, children with moderate to large effusion had a higher odds of ICU admission (aOR: 3.2, 95% CI: 1.18.9) and invasive mechanical ventilation (aOR: 14.8, 95% CI: 9.822.4), and also had a longer duration of oxygen supplementation (aOR: 3.0, 95% CI: 1.46.5) and hospital length of stay (aOR: 2.6, 95% CI: 1.9‐3.6) (Table 3, Figure 2). The presence of a small pleural effusion was not associated with increased need for supplemental oxygen, ICU admission, or mechanical ventilation compared to those without effusion. However, small effusion was associated with a longer duration of oxygen supplementation (aOR: 1.7, 95% CI: 12.7) and hospital length of stay (aOR: 1.6, 95% CI: 1.3‐1.9).

Severe Outcomes for Children Hospitalized With Community‐Acquired Pneumonia According to Presence and Size of Pleural Effusion
OutcomePleural EffusionP Valuea
None, n=320Small, n=65Moderate/Large, n=21
  • NOTE: Data are presented as number (%) or median [IQR]. Abbreviations: ICU, intensive care unit; IQR, interquartile range; O2, oxygen.

  • P values are from 2 statistics for categorical variables and Kruskal‐Wallis tests for continuous variables.

Supplemental O2 requirement200 (62.5)40 (61.5)14 (66.7)0.91
ICU admission22 (6.9)6 (9.2)4 (19)0.12
Mechanical ventilation5 (1.6)5 (7.7)4 (19)<0.01
Hospital length of stay, h48 [37.576]72 [45142]160 [82191]<0.01
Oxygen duration, h31 [1157]38.5 [1887]111 [27154]<0.01
jhm2227-fig-0002-m.png
Propensity‐adjusted odds ratios for severe outcomes for children hospitalized with community‐acquired pneumonia according to presence and size of effusion. No effusion is the reference. Covariates included in the propensity score included: age, sex, race/ethnicity, payer, hospital, asthma history, hospital transfer, recent hospitalization (within 30 days), recent emergency department or clinic visit (within 2 weeks), recent antibiotics for acute illness (within 5 days), illness duration prior to admission, tachypnea and/or increased work of breathing (retractions, nasal flaring, or grunting) at presentation, receipt of albuterol and/or corticosteroids during the first 2 calendar days, and concurrent diagnosis of bronchiolitis. Infiltrate pattern was included as a separate covariate. **Indicates confidence interval (CI) extends beyond the graph. The upper 95% CI for the odds ratio (OR) for mechanical ventilation was 34.2 for small effusion and 22.4 for moderate/large effusion. Abbreviations: ICU, intensive care unit; O2, oxygen.

DISCUSSION

We evaluated the association between admission chest radiographic findings and subsequent clinical outcomes and hospital care processes for children hospitalized with CAP at 4 children's hospitals in the United States. We conclude that radiographic findings are associated with important inpatient outcomes. Similar to data from adults, findings of moderate to large pleural effusions and bilateral multilobar infiltrates had the strongest associations with severe disease. Such information, in combination with other prognostic factors, may help clinicians identify high‐risk patients and support management decisions, while also helping to inform families about the expected hospital course.

Previous pediatric studies examining the association between radiographic findings and outcomes have produced inconsistent results.[8, 9, 10, 11, 12] All but 1 of these studies documented 1 radiographic characteristics associated with pneumonia disease severity.[11] Further, although most contrasted lobar/alveolar and interstitial infiltrates, only Patria et al. distinguished among lobar infiltrate patterns (eg, single lobar vs multilobar).[12] Similar to our findings, that study demonstrated increased disease severity among children with bilateral multifocal lobar infiltrates. Of the studies that considered the presence of pleural effusion, only 1 demonstrated this finding to be associated with more severe disease.[9] However, none of these prior studies examined size of the pleural effusion.

In our study, the strongest association with severe pneumonia outcomes was among children with moderate to large pleural effusion. Significant pleural effusions are much more commonly due to infection with bacterial pathogens, particularly Streptococcus pneumoniae, Staphylococcus aureus, and Streptococcus pyogenes, and may also indicate infection with more virulent and/or difficult to treat strains.[16, 17, 18, 19] Surgical intervention is also often required. As such, children with significant pleural effusions are often more ill on presentation and may have a prolonged period of recovery.[20, 21, 22]

Similarly, multilobar infiltrates, particularly bilateral, were associated with increased disease severity in terms of need for supplemental oxygen, ICU admission, and need for invasive mechanical ventilation. Although this finding may be expected, it is interesting to note that the duration of supplemental oxygen and hospital length of stay were similar to those with single lobar disease. One potential explanation is that, although children with multilobar disease are more severe at presentation, rates of recovery are similar to those with less extensive radiographic findings, owing to rapidly effective antimicrobials for uncomplicated bacterial pneumonia. This hypothesis also agrees with the 2011 PIDS/IDSA guidelines, which state that children receiving adequate therapy typically show signs of improvement within 48 to 72 hours regardless of initial severity.[1]

Interstitial infiltrate was also associated with increased severity at presentation but similar length of stay and duration of oxygen requirement compared with single lobar disease. We note that these children were substantially younger than those presenting with any pattern of lobar disease (median age, 1 vs 3 years), were more likely to have a concurrent diagnosis of bronchiolitis (34% vs 17%), and only 1 child with interstitial infiltrates had a documented pleural effusion (vs 23% of children with lobar infiltrates). Primary viral pneumonia is considered more likely to produce interstitial infiltrates on chest radiograph compared to bacterial disease, and although detailed etiologic data are unavailable for this study, our findings above strongly support this assertion.[23, 24]

The 2011 PIDS/IDSA guidelines recommend admission chest radiographs for all children hospitalized with pneumonia to assess extent of disease and identify complications that may requiring additional evaluation or surgical intervention.[1] Our findings highlight additional potential benefits of admission radiographs in terms of disease prognosis and management decisions. In the initial evaluation of a sick child with pneumonia, clinicians are often presented with a number of potential prognostic factors that may influence disease outcomes. However, it is sometimes difficult for providers to consider all available information and/or the relative importance of a single factor, resulting in inaccurate risk perceptions and management decisions that may contribute to poor outcomes.[25] Similar to adults, the development of clinical prediction rules, which incorporate a variety of important predictors including admission radiographic findings, likely would improve risk assessments and potentially outcomes for children with pneumonia. Such prognostic information is also helpful for clinicians who may use these data to inform and prepare families regarding the expected course of hospitalization.

Our study has several limitations. This study was retrospective and only included a sample of pneumonia hospitalizations during the study period, which may raise confounding concerns and potential for selection bias. However, detailed medical record reviews using standardized case definitions for radiographic CAP were used, and a large sample of children was randomly selected from each institution. In addition, a large number of potential confounders were selected a priori and included in multivariable analyses; propensity score adjustment was used to reduce model complexity and avoid overfitting. Radiographic findings were based on clinical interpretation by pediatric radiologists independent of a study protocol. Prior studies have demonstrated good agreement for identification of alveolar/lobar infiltrates and pleural effusion by trained radiologists, although agreement for interstitial infiltrate is poor.[26, 27] This limitation could result in either over‐ or underestimation of the prevalence of interstitial infiltrates likely resulting in a nondifferential bias toward the null. Microbiologic information, which may inform radiographic findings and disease severity, was also not available. However, because pneumonia etiology is frequently unknown in the clinical setting, our study reflects typical practice. We also did not include children from community or nonteaching hospitals. Thus, although findings may have relevance to community or nonteaching hospitals, our results cannot be generalized.

CONCLUSION

Our study demonstrates that among children hospitalized with CAP, admission chest radiographic findings are associated with important clinical outcomes and hospital care processes, highlighting additional benefits of the 2011 PIDS/IDSA guidelines' recommendation for admission chest radiographs for all children hospitalized with pneumonia. These data, in conjunction with other important prognostic information, may help clinicians more rapidly identify children at increased risk for severe illness, and could also offer guidance regarding disease management strategies and facilitate shared decision making with families. Thus, routine admission chest radiography in this population represents a valuable tool that contributes to improved quality of care.

Disclosures

Dr. Williams is supported by funds from the National Institutes of HealthNational Institute of Allergy and Infectious Diseases (K23AI104779). The authors report no conflicts of interest.

The 2011 Pediatric Infectious Diseases Society and Infectious Diseases Society of America (PIDS/IDSA) guidelines for management of pediatric community‐acquired pneumonia (CAP) recommend that admission chest radiographs be obtained in all children hospitalized with CAP to document the presence and extent of infiltrates and to identify complications.[1] Findings from chest radiographs may also provide clues to etiology and assist with predicting disease outcomes. In adults with CAP, clinical prediction tools use radiographic findings to inform triage decisions, guide management strategies, and predict outcomes.[2, 3, 4, 5, 6, 7] Whether or not radiographic findings could have similar utility among children with CAP is unknown.

Several retrospective studies have examined the ability of chest radiographs to predict pediatric pneumonia disease severity.[8, 9, 10, 11, 12] However, these studies used several different measures of severe pneumonia and/or were limited to young children <5 years of age, leading to inconsistent findings. These studies also rarely considered very severe disease (eg, need for invasive mechanical ventilation) or longitudinal outcome measures such as hospital length of stay. Finally, all of these prior studies were conducted outside of the United States, and most were single‐center investigations, potentially limiting generalizability. We sought to examine associations between admission chest radiographic findings and subsequent hospital care processes and clinical outcomes, including length of stay and resource utilization measures, among children hospitalized with CAP at 4 children's hospitals in the United States.

METHODS

Design and Setting

This study was nested within a multicenter retrospective cohort designed to validate International Classification of Diseases, 9th Revision, Clinical Modification (ICD9‐CM) diagnostic codes for pediatric CAP hospitalizations.[13] The Pediatric Health Information System database (Children's Hospital Association, Overland Park, KS) was used to identify children from 4 freestanding pediatric hospitals (Monroe Carell, Jr. Children's Hospital at Vanderbilt, Nashville, Tennessee; Children's Mercy Hospitals & Clinics, Kansas City, Missouri; Seattle Children's Hospital, Seattle, Washington; and Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio). The institutional review boards at each participating institution approved the study. The validation study included a 25% random sampling of children 60 days to 18 years of age (n=998) who were hospitalized between January 1, 2010 and December 31, 2010 with at least 1 ICD9‐CM discharge code indicating pneumonia. The diagnosis of CAP was confirmed by medical record review.

Study Population

This study was limited to children from the validation study who met criteria for clinical and radiographic CAP, defined as: (1) abnormal temperature or white blood cell count, (2) signs and symptoms of acute respiratory illness (eg, cough, tachypnea), and (3) chest radiograph indicating pneumonia within 48 hours of admission. Children with atelectasis as the only abnormal radiographic finding and those with complex chronic conditions (eg, cystic fibrosis, malignancy) were excluded using a previously described algorithm.[14]

Outcomes

Several measures of disease severity were assessed. Dichotomous outcomes included supplemental oxygen use, need for intensive care unit (ICU) admission, and need for invasive mechanical ventilation. Continuous outcomes included hospital length of stay, and for those requiring supplemental oxygen, duration of oxygen supplementation, measured in hours.

Exposure

To categorize infiltrate patterns and the presence and size of pleural effusions, we reviewed the final report from admission chest radiographs to obtain the final clinical interpretation performed by the attending pediatric radiologist. Infiltrate patterns were classified as single lobar (reference), unilateral multilobar, bilateral multilobar, or interstitial. Children with both lobar and interstitial infiltrates, and those with mention of atelectasis, were classified according to the type of lobar infiltrate. Those with atelectasis only were excluded. Pleural effusions were classified as absent, small, or moderate/large.

Analysis

Descriptive statistics were summarized using frequencies and percentages for categorical variables and median and interquartile range (IQR) values for continuous variables. Our primary exposures were infiltrate pattern and presence and size of pleural effusion on admission chest radiograph. Associations between radiographic findings and disease outcomes were analyzed using logistic and linear regression for dichotomous and continuous variables, respectively. Continuous outcomes were log‐transformed and normality assumptions verified prior to model development.

Due to the large number of covariates relative to outcome events, we used propensity score methods to adjust for potential confounding. The propensity score estimates the likelihood of a given exposure (ie, infiltrate pattern) conditional on a set of covariates. In this way, the propensity score summarizes potential confounding effects from a large number of covariates into a single variable. Including the propensity score as a covariate in multivariable regression improves model efficiency and helps protect against overfitting.[15] Covariates included in the estimation of the propensity score included age, sex, race/ethnicity, payer, hospital, asthma history, hospital transfer, recent hospitalization (within 30 days), recent emergency department or clinic visit (within 2 weeks), recent antibiotics for acute illness (within 5 days), illness duration prior to admission, tachypnea and/or increased work of breathing (retractions, nasal flaring, or grunting) at presentation, receipt of albuterol and/or corticosteroids during the first 2 calendar days of hospitalization, and concurrent diagnosis of bronchiolitis. All analyses included the estimated propensity score, infiltrate pattern, and pleural effusion (absent, small, or moderate/large).

RESULTS

Study Population

The median age of the 406 children with clinical and radiographic CAP was 3 years (IQR, 16 years) (Table 1). Single lobar infiltrate was the most common radiographic pattern (61%). Children with interstitial infiltrates (10%) were younger than those with lobar infiltrates of any type (median age 1 vs 3 years, P=0.02). A concomitant diagnosis of bronchiolitis was assigned to 34% of children with interstitial infiltrates but only 17% of those with lobar infiltrate patterns (range, 11%20%, P=0.03). Pleural effusion was present in 21% of children and was more common among those with lobar infiltrates, particularly multilobar disease. Only 1 child with interstitial infiltrate had a pleural effusion. Overall, 63% of children required supplemental oxygen, 8% required ICU admission, and 3% required invasive mechanical ventilation. Median length of stay was 51.5 hours (IQR, 3991) and median oxygen duration was 31.5 hours [IQR, 1365]. There were no deaths.

Characteristics of Children Hospitalized With Community‐Acquired Pneumonia According to Admission Radiographic Findings
CharacteristicInfiltrate PatternaP Valueb
Single LobarMultilobar, UnilateralMultilobar, BilateralInterstitial
  • NOTE: Data are presented as number (%) or median [IQR]. Abbreviations: ICU, intensive care unit; IQR, interquartile range; O2, oxygen.

  • Children with both lobar and interstitial infiltrates were classified according to the type of lobar infiltrate

  • P values are from 2 statistics for categorical variables and Kruskal‐Wallis tests for continuous variables.

No.247 (60.8)54 (13.3)64 (15.8)41 (10.1) 
Median age, y3 [16]3 [17]3 [15]1 [03]0.02
Male sex124 (50.2)32 (59.3)41 (64.1)30 (73.2)0.02
Race     
Non‐Hispanic white133 (53.8)36 (66.7)37 (57.8)17 (41.5)0.69
Non‐Hispanic black40 (16.2)6 (11.1)9 (14.1)8 (19.5) 
Hispanic25 (10.1)4 (7.4)5 (7.8)7 (17.1) 
Other49 (19.9)8 (14.8)13 (20.4)9 (22) 
Insurance     
Public130 (52.6)26 (48.1)33 (51.6)25 (61)0.90
Private116 (47)28 (51.9)31 (48.4)16 (39) 
Concurrent diagnosis     
Asthma80 (32.4)16 (29.6)17 (26.6)12 (29.3)0.82
Bronchiolitis43 (17.4)6 (11.1)13 (20.3)14 (34.1)0.03
Effusion     
None201 (81.4)31 (57.4)48 (75)40 (97.6)<.01
Small34 (13.8)20 (37)11 (17.2)0 
Moderate/large12 (4.9)3 (5.6)5 (7.8)1 (2.4) 

Outcomes According to Radiographic Infiltrate Pattern

Compared to children with single lobar infiltrates, the odds of ICU admission was significantly increased for those with either unilateral or bilateral multilobar infiltrates (unilateral, adjusted odds ratio [aOR]: 8.0, 95% confidence interval [CI]: 2.922.2; bilateral, aOR: 6.6, 95% CI: 2.14.5) (Figure 1, Table 2). Patients with bilateral multilobar infiltrates also had higher odds for supplemental oxygen use (aOR: 2.7, 95% CI: 1.25.8) and need for invasive mechanical ventilation (aOR: 3.0, 95% CI: 1.27.9). There were no differences in duration of oxygen supplementation or hospital length of stay for children with single versus multilobar infiltrates.

jhm2227-fig-0001-m.png
Propensity‐adjusted odds ratios for severe outcomes for children hospitalized with community‐acquired pneumonia according to admission radiographic findings. Single lobar infiltrate is the reference. Children with both lobar and interstitial infiltrates were classified according to the type of lobar infiltrate. Covariates included in the propensity score included: age, sex, race/ethnicity, payer, hospital, asthma history, hospital transfer, recent hospitalization (within 30 days), recent emergency department or clinic visit (within 2 weeks), recent antibiotics for acute illness (within 5 days), illness duration prior to admission, tachypnea and/or increased work of breathing (retractions, nasal flaring, or grunting) at presentation, receipt of albuterol and/or corticosteroids during the first 2 calendar days, and concurrent diagnosis of bronchiolitis. Pleural effusion (absent, small, or moderate/large) was included as a separate covariate. **Indicates that confidence interval (CIs) extends beyond the graph. The upper 95% CI for the odds ratio (OR) for infiltrates that were multilobar and unilateral was 22.2 for intensive care unit (ICU) admission and 37.8 for mechanical ventilation. Abbreviations: O2, oxygen.
Severe Outcomes for Children Hospitalized With Community‐Acquired Pneumonia According to Admission Radiographic Findings
OutcomeInfiltrate PatternaP Valueb
Single Lobar, n=247Multilobar, Unilateral, n=54Multilobar, Bilateral, n=64Interstitial, n=41
  • NOTE: Data are presented as number (%) or median [IQR]. Abbreviations: ICU, intensive care unit; IQR, interquartile range, O2, oxygen.

  • Children with both lobar and interstitial infiltrates were classified according to the type of lobar infiltrate.

  • P values are from 2 statistics for categorical variables and Kruskal‐Wallis tests for continuous variables.

Supplemental O2 requirement143 (57.9)34 (63)46 (71.9)31 (75.6)0.05
ICU admission10 (4)9 (16.7)9 (14.1)4 (9.8)<0.01
Mechanical ventilation5 (2)4 (7.4)4 (6.3)1 (2.4)0.13
Hospital length of stay, h47 [3779]63 [45114]56.5 [39.5101]62 [3993]<0.01
O2 duration, h27 [1059]38 [1777]38 [2381]34.5 [1765]0.18

Compared to those with single lobar infiltrates, children with interstitial infiltrates had higher odds of need for supplemental oxygen (aOR: 3.1, 95% CI: 1.37.6) and ICU admission (aOR: 4.4, 95% CI: 1.314.3) but not invasive mechanical ventilation. There were also no differences in duration of oxygen supplementation or hospital length of stay.

Outcomes According to Presence and Size of Pleural Effusion

Compared to those without pleural effusion, children with moderate to large effusion had a higher odds of ICU admission (aOR: 3.2, 95% CI: 1.18.9) and invasive mechanical ventilation (aOR: 14.8, 95% CI: 9.822.4), and also had a longer duration of oxygen supplementation (aOR: 3.0, 95% CI: 1.46.5) and hospital length of stay (aOR: 2.6, 95% CI: 1.9‐3.6) (Table 3, Figure 2). The presence of a small pleural effusion was not associated with increased need for supplemental oxygen, ICU admission, or mechanical ventilation compared to those without effusion. However, small effusion was associated with a longer duration of oxygen supplementation (aOR: 1.7, 95% CI: 12.7) and hospital length of stay (aOR: 1.6, 95% CI: 1.3‐1.9).

Severe Outcomes for Children Hospitalized With Community‐Acquired Pneumonia According to Presence and Size of Pleural Effusion
OutcomePleural EffusionP Valuea
None, n=320Small, n=65Moderate/Large, n=21
  • NOTE: Data are presented as number (%) or median [IQR]. Abbreviations: ICU, intensive care unit; IQR, interquartile range; O2, oxygen.

  • P values are from 2 statistics for categorical variables and Kruskal‐Wallis tests for continuous variables.

Supplemental O2 requirement200 (62.5)40 (61.5)14 (66.7)0.91
ICU admission22 (6.9)6 (9.2)4 (19)0.12
Mechanical ventilation5 (1.6)5 (7.7)4 (19)<0.01
Hospital length of stay, h48 [37.576]72 [45142]160 [82191]<0.01
Oxygen duration, h31 [1157]38.5 [1887]111 [27154]<0.01
jhm2227-fig-0002-m.png
Propensity‐adjusted odds ratios for severe outcomes for children hospitalized with community‐acquired pneumonia according to presence and size of effusion. No effusion is the reference. Covariates included in the propensity score included: age, sex, race/ethnicity, payer, hospital, asthma history, hospital transfer, recent hospitalization (within 30 days), recent emergency department or clinic visit (within 2 weeks), recent antibiotics for acute illness (within 5 days), illness duration prior to admission, tachypnea and/or increased work of breathing (retractions, nasal flaring, or grunting) at presentation, receipt of albuterol and/or corticosteroids during the first 2 calendar days, and concurrent diagnosis of bronchiolitis. Infiltrate pattern was included as a separate covariate. **Indicates confidence interval (CI) extends beyond the graph. The upper 95% CI for the odds ratio (OR) for mechanical ventilation was 34.2 for small effusion and 22.4 for moderate/large effusion. Abbreviations: ICU, intensive care unit; O2, oxygen.

DISCUSSION

We evaluated the association between admission chest radiographic findings and subsequent clinical outcomes and hospital care processes for children hospitalized with CAP at 4 children's hospitals in the United States. We conclude that radiographic findings are associated with important inpatient outcomes. Similar to data from adults, findings of moderate to large pleural effusions and bilateral multilobar infiltrates had the strongest associations with severe disease. Such information, in combination with other prognostic factors, may help clinicians identify high‐risk patients and support management decisions, while also helping to inform families about the expected hospital course.

Previous pediatric studies examining the association between radiographic findings and outcomes have produced inconsistent results.[8, 9, 10, 11, 12] All but 1 of these studies documented 1 radiographic characteristics associated with pneumonia disease severity.[11] Further, although most contrasted lobar/alveolar and interstitial infiltrates, only Patria et al. distinguished among lobar infiltrate patterns (eg, single lobar vs multilobar).[12] Similar to our findings, that study demonstrated increased disease severity among children with bilateral multifocal lobar infiltrates. Of the studies that considered the presence of pleural effusion, only 1 demonstrated this finding to be associated with more severe disease.[9] However, none of these prior studies examined size of the pleural effusion.

In our study, the strongest association with severe pneumonia outcomes was among children with moderate to large pleural effusion. Significant pleural effusions are much more commonly due to infection with bacterial pathogens, particularly Streptococcus pneumoniae, Staphylococcus aureus, and Streptococcus pyogenes, and may also indicate infection with more virulent and/or difficult to treat strains.[16, 17, 18, 19] Surgical intervention is also often required. As such, children with significant pleural effusions are often more ill on presentation and may have a prolonged period of recovery.[20, 21, 22]

Similarly, multilobar infiltrates, particularly bilateral, were associated with increased disease severity in terms of need for supplemental oxygen, ICU admission, and need for invasive mechanical ventilation. Although this finding may be expected, it is interesting to note that the duration of supplemental oxygen and hospital length of stay were similar to those with single lobar disease. One potential explanation is that, although children with multilobar disease are more severe at presentation, rates of recovery are similar to those with less extensive radiographic findings, owing to rapidly effective antimicrobials for uncomplicated bacterial pneumonia. This hypothesis also agrees with the 2011 PIDS/IDSA guidelines, which state that children receiving adequate therapy typically show signs of improvement within 48 to 72 hours regardless of initial severity.[1]

Interstitial infiltrate was also associated with increased severity at presentation but similar length of stay and duration of oxygen requirement compared with single lobar disease. We note that these children were substantially younger than those presenting with any pattern of lobar disease (median age, 1 vs 3 years), were more likely to have a concurrent diagnosis of bronchiolitis (34% vs 17%), and only 1 child with interstitial infiltrates had a documented pleural effusion (vs 23% of children with lobar infiltrates). Primary viral pneumonia is considered more likely to produce interstitial infiltrates on chest radiograph compared to bacterial disease, and although detailed etiologic data are unavailable for this study, our findings above strongly support this assertion.[23, 24]

The 2011 PIDS/IDSA guidelines recommend admission chest radiographs for all children hospitalized with pneumonia to assess extent of disease and identify complications that may requiring additional evaluation or surgical intervention.[1] Our findings highlight additional potential benefits of admission radiographs in terms of disease prognosis and management decisions. In the initial evaluation of a sick child with pneumonia, clinicians are often presented with a number of potential prognostic factors that may influence disease outcomes. However, it is sometimes difficult for providers to consider all available information and/or the relative importance of a single factor, resulting in inaccurate risk perceptions and management decisions that may contribute to poor outcomes.[25] Similar to adults, the development of clinical prediction rules, which incorporate a variety of important predictors including admission radiographic findings, likely would improve risk assessments and potentially outcomes for children with pneumonia. Such prognostic information is also helpful for clinicians who may use these data to inform and prepare families regarding the expected course of hospitalization.

Our study has several limitations. This study was retrospective and only included a sample of pneumonia hospitalizations during the study period, which may raise confounding concerns and potential for selection bias. However, detailed medical record reviews using standardized case definitions for radiographic CAP were used, and a large sample of children was randomly selected from each institution. In addition, a large number of potential confounders were selected a priori and included in multivariable analyses; propensity score adjustment was used to reduce model complexity and avoid overfitting. Radiographic findings were based on clinical interpretation by pediatric radiologists independent of a study protocol. Prior studies have demonstrated good agreement for identification of alveolar/lobar infiltrates and pleural effusion by trained radiologists, although agreement for interstitial infiltrate is poor.[26, 27] This limitation could result in either over‐ or underestimation of the prevalence of interstitial infiltrates likely resulting in a nondifferential bias toward the null. Microbiologic information, which may inform radiographic findings and disease severity, was also not available. However, because pneumonia etiology is frequently unknown in the clinical setting, our study reflects typical practice. We also did not include children from community or nonteaching hospitals. Thus, although findings may have relevance to community or nonteaching hospitals, our results cannot be generalized.

CONCLUSION

Our study demonstrates that among children hospitalized with CAP, admission chest radiographic findings are associated with important clinical outcomes and hospital care processes, highlighting additional benefits of the 2011 PIDS/IDSA guidelines' recommendation for admission chest radiographs for all children hospitalized with pneumonia. These data, in conjunction with other important prognostic information, may help clinicians more rapidly identify children at increased risk for severe illness, and could also offer guidance regarding disease management strategies and facilitate shared decision making with families. Thus, routine admission chest radiography in this population represents a valuable tool that contributes to improved quality of care.

Disclosures

Dr. Williams is supported by funds from the National Institutes of HealthNational Institute of Allergy and Infectious Diseases (K23AI104779). The authors report no conflicts of interest.

References
  1. Bradley JS, Byington CL, Shah SS, et al. The management of community‐acquired pneumonia in infants and children older than 3 months of age: clinical practice guidelines by the Pediatric Infectious Diseases Society and the Infectious Diseases Society of America. Clin Infect Dis. 2011;53(7):e25e76.
  2. Fine MJ, Auble TE, Yealy DM, et al. A prediction rule to identify low‐risk patients with community‐acquired pneumonia. N Engl J Med. 1997;336(4):243250.
  3. Charles PG, Wolfe R, Whitby M, et al. SMART‐COP: a tool for predicting the need for intensive respiratory or vasopressor support in community‐acquired pneumonia. Clin Infect Dis. 2008;47(3):375384.
  4. Espana PP, Capelastegui A, Gorordo I, et al. Development and validation of a clinical prediction rule for severe community‐acquired pneumonia. Am J Respir Crit Care Med. 2006;174(11):12491256.
  5. Renaud B, Labarere J, Coma E, et al. Risk stratification of early admission to the intensive care unit of patients with no major criteria of severe community‐acquired pneumonia: development of an international prediction rule. Crit Care. 2009;13(2):R54.
  6. Hasley PB, Albaum MN, Li YH, et al. Do pulmonary radiographic findings at presentation predict mortality in patients with community‐acquired pneumonia? Arch Intern Med. 1996;156(19):22062212.
  7. Chalmers JD, Singanayagam A, Akram AR, Choudhury G, Mandal P, Hill AT. Safety and efficacy of CURB65‐guided antibiotic therapy in community‐acquired pneumonia. J Antimicrob Chemother. 2011;66(2):416423.
  8. Kin Key N, Araujo‐Neto CA, Nascimento‐Carvalho CM. Severity of childhood community‐acquired pneumonia and chest radiographic findings. Pediatr Pulmonol. 2009;44(3):249252.
  9. Grafakou O, Moustaki M, Tsolia M, et al. Can chest x‐ray predict pneumonia severity? Pediatr Pulmonol. 2004;38(6):465469.
  10. Clark JE, Hammal D, Spencer D, Hampton F. Children with pneumonia: how do they present and how are they managed? Arch Dis Child. 2007;92(5):394398.
  11. Bharti B, Kaur L, Bharti S. Role of chest X‐ray in predicting outcome of acute severe pneumonia. Indian Pediatr. 2008;45(11):893898.
  12. Patria MF, Longhi B, Lelii M, Galeone C, Pavesi MA, Esposito S. Association between radiological findings and severity of community‐acquired pneumonia in children. Ital J Pediatr. 2013;39:56.
  13. Williams DJ, Shah SS, Myers AM, et al. Identifying pediatric community‐acquired pneumonia hospitalizations: accuracy of administrative billing codes. JAMA Pediatrics. 2013;167(9):851858.
  14. Feudtner C, Hays RM, Haynes G, Geyer JR, Neff JM, Koepsell TD. Deaths attributed to pediatric complex chronic conditions: national trends and implications for supportive care services. Pediatrics. 2001;107(6):E99.
  15. Joffe MM, Rosenbaum PR. Invited commentary: propensity scores. Am J Epidemiol. 1999;150(4):327333.
  16. Grijalva CG, Nuorti JP, Zhu Y, Griffin MR. Increasing incidence of empyema complicating childhood community‐acquired pneumonia in the United States. Clin Infect Dis. 2010;50(6):805813.
  17. Michelow IC, Olsen K, Lozano J, et al. Epidemiology and clinical characteristics of community‐acquired pneumonia in hospitalized children. Pediatrics. 2004;113(4):701707.
  18. Blaschke AJ, Heyrend C, Byington CL, et al. Molecular analysis improves pathogen identification and epidemiologic study of pediatric parapneumonic empyema. Pediatr Infect Dis J. 2011;30(4):289294.
  19. Chonmaitree T, Powell KR. Parapneumonic pleural effusion and empyema in children. Review of a 19‐year experience, 1962–1980. Clin Pediatr (Phila). 1983;22(6):414419.
  20. Huang CY, Chang L, Liu CC, et al. Risk factors of progressive community‐acquired pneumonia in hospitalized children: a prospective study [published online ahead of print August 28, 2013]. J Microbiol Immunol Infect. doi: 10.1016/j.jmii.2013.06.009.
  21. Rowan‐Legg A, Barrowman N, Shenouda N, Koujok K, Saux N. Community‐acquired lobar pneumonia in children in the era of universal 7‐valent pneumococcal vaccination: a review of clinical presentations and antimicrobial treatment from a Canadian pediatric hospital. BMC Pediatr. 2012;12:133.
  22. Wexler ID, Knoll S, Picard E, et al. Clinical characteristics and outcome of complicated pneumococcal pneumonia in a pediatric population. Pediatr Pulmonol. 2006;41(8):726734.
  23. Virkki R, Juven T, Rikalainen H, Svedstrom E, Mertsola J, Ruuskanen O. Differentiation of bacterial and viral pneumonia in children. Thorax. 2002;57(5):438441.
  24. Harris M, Clark J, Coote N, et al. British Thoracic Society guidelines for the management of community acquired pneumonia in children: update 2011. Thorax. 2011;66(suppl 2):ii1ii23.
  25. Neill AM, Martin IR, Weir R, et al. Community acquired pneumonia: aetiology and usefulness of severity criteria on admission. Thorax. 1996;51(10):10101016.
  26. Neuman MI, Lee EY, Bixby S, et al. Variability in the interpretation of chest radiographs for the diagnosis of pneumonia in children. J Hosp Med. 2012;7(4):294298.
  27. Albaum MN, Hill LC, Murphy M, et al. Interobserver reliability of the chest radiograph in community‐acquired pneumonia. PORT Investigators. Chest. 1996;110(2):343350.
References
  1. Bradley JS, Byington CL, Shah SS, et al. The management of community‐acquired pneumonia in infants and children older than 3 months of age: clinical practice guidelines by the Pediatric Infectious Diseases Society and the Infectious Diseases Society of America. Clin Infect Dis. 2011;53(7):e25e76.
  2. Fine MJ, Auble TE, Yealy DM, et al. A prediction rule to identify low‐risk patients with community‐acquired pneumonia. N Engl J Med. 1997;336(4):243250.
  3. Charles PG, Wolfe R, Whitby M, et al. SMART‐COP: a tool for predicting the need for intensive respiratory or vasopressor support in community‐acquired pneumonia. Clin Infect Dis. 2008;47(3):375384.
  4. Espana PP, Capelastegui A, Gorordo I, et al. Development and validation of a clinical prediction rule for severe community‐acquired pneumonia. Am J Respir Crit Care Med. 2006;174(11):12491256.
  5. Renaud B, Labarere J, Coma E, et al. Risk stratification of early admission to the intensive care unit of patients with no major criteria of severe community‐acquired pneumonia: development of an international prediction rule. Crit Care. 2009;13(2):R54.
  6. Hasley PB, Albaum MN, Li YH, et al. Do pulmonary radiographic findings at presentation predict mortality in patients with community‐acquired pneumonia? Arch Intern Med. 1996;156(19):22062212.
  7. Chalmers JD, Singanayagam A, Akram AR, Choudhury G, Mandal P, Hill AT. Safety and efficacy of CURB65‐guided antibiotic therapy in community‐acquired pneumonia. J Antimicrob Chemother. 2011;66(2):416423.
  8. Kin Key N, Araujo‐Neto CA, Nascimento‐Carvalho CM. Severity of childhood community‐acquired pneumonia and chest radiographic findings. Pediatr Pulmonol. 2009;44(3):249252.
  9. Grafakou O, Moustaki M, Tsolia M, et al. Can chest x‐ray predict pneumonia severity? Pediatr Pulmonol. 2004;38(6):465469.
  10. Clark JE, Hammal D, Spencer D, Hampton F. Children with pneumonia: how do they present and how are they managed? Arch Dis Child. 2007;92(5):394398.
  11. Bharti B, Kaur L, Bharti S. Role of chest X‐ray in predicting outcome of acute severe pneumonia. Indian Pediatr. 2008;45(11):893898.
  12. Patria MF, Longhi B, Lelii M, Galeone C, Pavesi MA, Esposito S. Association between radiological findings and severity of community‐acquired pneumonia in children. Ital J Pediatr. 2013;39:56.
  13. Williams DJ, Shah SS, Myers AM, et al. Identifying pediatric community‐acquired pneumonia hospitalizations: accuracy of administrative billing codes. JAMA Pediatrics. 2013;167(9):851858.
  14. Feudtner C, Hays RM, Haynes G, Geyer JR, Neff JM, Koepsell TD. Deaths attributed to pediatric complex chronic conditions: national trends and implications for supportive care services. Pediatrics. 2001;107(6):E99.
  15. Joffe MM, Rosenbaum PR. Invited commentary: propensity scores. Am J Epidemiol. 1999;150(4):327333.
  16. Grijalva CG, Nuorti JP, Zhu Y, Griffin MR. Increasing incidence of empyema complicating childhood community‐acquired pneumonia in the United States. Clin Infect Dis. 2010;50(6):805813.
  17. Michelow IC, Olsen K, Lozano J, et al. Epidemiology and clinical characteristics of community‐acquired pneumonia in hospitalized children. Pediatrics. 2004;113(4):701707.
  18. Blaschke AJ, Heyrend C, Byington CL, et al. Molecular analysis improves pathogen identification and epidemiologic study of pediatric parapneumonic empyema. Pediatr Infect Dis J. 2011;30(4):289294.
  19. Chonmaitree T, Powell KR. Parapneumonic pleural effusion and empyema in children. Review of a 19‐year experience, 1962–1980. Clin Pediatr (Phila). 1983;22(6):414419.
  20. Huang CY, Chang L, Liu CC, et al. Risk factors of progressive community‐acquired pneumonia in hospitalized children: a prospective study [published online ahead of print August 28, 2013]. J Microbiol Immunol Infect. doi: 10.1016/j.jmii.2013.06.009.
  21. Rowan‐Legg A, Barrowman N, Shenouda N, Koujok K, Saux N. Community‐acquired lobar pneumonia in children in the era of universal 7‐valent pneumococcal vaccination: a review of clinical presentations and antimicrobial treatment from a Canadian pediatric hospital. BMC Pediatr. 2012;12:133.
  22. Wexler ID, Knoll S, Picard E, et al. Clinical characteristics and outcome of complicated pneumococcal pneumonia in a pediatric population. Pediatr Pulmonol. 2006;41(8):726734.
  23. Virkki R, Juven T, Rikalainen H, Svedstrom E, Mertsola J, Ruuskanen O. Differentiation of bacterial and viral pneumonia in children. Thorax. 2002;57(5):438441.
  24. Harris M, Clark J, Coote N, et al. British Thoracic Society guidelines for the management of community acquired pneumonia in children: update 2011. Thorax. 2011;66(suppl 2):ii1ii23.
  25. Neill AM, Martin IR, Weir R, et al. Community acquired pneumonia: aetiology and usefulness of severity criteria on admission. Thorax. 1996;51(10):10101016.
  26. Neuman MI, Lee EY, Bixby S, et al. Variability in the interpretation of chest radiographs for the diagnosis of pneumonia in children. J Hosp Med. 2012;7(4):294298.
  27. Albaum MN, Hill LC, Murphy M, et al. Interobserver reliability of the chest radiograph in community‐acquired pneumonia. PORT Investigators. Chest. 1996;110(2):343350.
Issue
Journal of Hospital Medicine - 9(9)
Issue
Journal of Hospital Medicine - 9(9)
Page Number
559-564
Page Number
559-564
Publications
Publications
Article Type
Display Headline
Admission chest radiographs predict illness severity for children hospitalized with pneumonia
Display Headline
Admission chest radiographs predict illness severity for children hospitalized with pneumonia
Sections
Article Source

© 2014 Society of Hospital Medicine

Disallow All Ads
Correspondence Location
Address for correspondence and reprint requests: Derek J. Williams, MD, 1161 21st Ave S. S2323 MCN, Nashville, TN 37232; Telephone: 615‐322‐2744; Fax: 615-322-4399; E‐mail: derek.williams@vanderbilt.edu
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Article PDF Media
Image
Disable zoom
Off
Media Files
Image
Disable zoom
Off

Hospital to Home Transitions

Article Type
Changed
Mon, 01/02/2017 - 19:34
Display Headline
The successes and challenges of hospital to home transitions

Hospital readmissions, which account for a substantial proportion of healthcare expenditures, have increasingly become a focus for hospitals and health systems. Hospitals now assume greater responsibility for population health, and face financial penalties by federal and state agencies that consider readmissions a key measure of the quality of care provided during hospitalization. Consequently, there is broad interest in identifying approaches to reduce hospital reutilization, including emergency department (ED) revisits and hospital readmissions. In this issue of the Journal of Hospital Medicine, Auger et al.[1] report the results of a systematic review, which evaluates the effect of discharge interventions on hospital reutilization among children.

As Auger et al. note, the transition from hospital to home is a vulnerable time for children and their families, with 1 in 5 parents reporting major challenges with such transitions.[2] Auger and colleagues identified 14 studies spanning 3 pediatric disease processes that addressed this issue. The authors concluded that several interventions were potentially effective, but individual studies frequently used multifactorial interventions, precluding determination of discrete elements essential to success. The larger body of care transitions literature in adult populations provides insights for interventions that may benefit pediatric patients, as well as informs future research and quality improvement priorities.

The authors identified some distinct interventions that may successfully decrease hospital reutilization, which share common themes from the adult literature. The first is the use of a dedicated transition coordinator (eg, nurse) or coordinating center to assist with the patient's transition home after discharge. In adult studies, this bridging strategy[3, 4] (ie, use of a dedicated transition coordinator or provider) is initiated during the hospitalization and continues postdischarge in the form of phone calls or home visits. The second theme illustrated in both this pediatric review[1] and adult reviews[3, 4, 5] focuses on enhanced or individualized patient education. Most studies have used a combination of these strategies. For example, the Care Transitions Intervention (one of the best validated adult discharge approaches) uses a transition coach to aid the patient in medication self‐management, creation of a patient‐centered record, scheduling follow‐up appointments, and understanding signs and symptoms of a worsening condition.[6] In a randomized study, this intervention demonstrated a reduction in readmissions within 90 days to 16.7% in the intervention group, compared with 22.5% in the control group.[6] One of the pediatric studies highlighted in the review by Auger et al. achieved a decrease in 14‐day ED revisits from 8% prior to implementation of the program to 2.7% following implementation of the program.[7] This program was for patients discharged from the neonatal intensive care unit and involved a nurse coordinator (similar to a transition coach) who worked closely with families and ensured adequate resources prior to discharge as well as a home visitation program.[7]

Although Auger et al. identify some effective approaches to reducing hospital reutilization after discharge in children, their review and the complementary adult literature bring to light 4 main unresolved questions for hospitalists seeking to improve care transitions: (1) how to dissect diverse and heterogeneous interventions to determine the key driver of success, (2) how to interpret and generally apply interventions from single centers where they may have been tailored to a specific healthcare environment, (3) how to generalize the findings of many disease‐specific interventions to other populations, and (4) how to evaluate the cost and assess the costbenefit of implementing many of the more resource intensive interventions. An example of a heterogeneous intervention addressed in this pediatric systematic review was described by Ng et al.,[8] in which the intervention group received a combination of an enhanced discharge education session, disease‐specific nurse evaluation, an animated education booklet, and postdischarge telephone follow‐up, whereas the control group received a shorter discharge education session, a disease‐specific nurse evaluation only if referred by a physician, a written education booklet, and no telephone follow‐up. Investigators found that intervention patients were less likely to be readmitted or revisit the ED as compared with controls. A similarly multifaceted intervention introduced by Taggart et al.[9] was unable to detect a difference in readmissions or ED revisits. It is unclear whether or not the differences in outcomes were related to differences in the intervention bundle itself or institutional or local contextual factors, thus limiting application to other hospitals. Generalizability of interventions is similarly complicated in adults.

The studies presented in this pediatric review article are specific to 3 disease processes: cancer, asthma, and neonatal intensive care (ie, premature) populations. Beyond these populations, there were no other pediatric conditions that met inclusion criteria, thus limiting the generalizability of the findings. As described by Rennke et al.,[3] adult systematic reviews that have focused only on disease‐specific interventions to reduce hospital reutilization are also difficult to generalize to broader populations. Two of the 3 recent adult transition intervention systematic reviews excluded disease‐specific interventions in an attempt to find more broadly applicable interventions but struggled with the same heterogeneity discussed in this review by Auger et al.[3, 4] Although disease‐specific interventions were included in the third adult systematic review and the evaluation was restricted to randomized controlled trials, the authors still grappled with finding 1 or 2 common, successful intervention components.[5] The fourth unresolved question involves understanding the financial burden of implementing more resource‐intensive interventions such as postdischarge home nurse visits. For example, it may be difficult to justify the business case for hiring a transition coach or initiating home nurse visits when the cost and financial implications are unclear. Neither the pediatric nor adult literature describes this well.

Some of the challenges in identifying effective interventions differ between adult and pediatric populations. Adults tend to have multiple comorbid conditions, making them more medically complex and at greater risk for adverse outcomes, medication errors, and hospital utilization.[10] Although a small subset of the pediatric population with complex chronic medical conditions accounts for a majority of hospital reutilization and cost,[11] most hospitalized pediatric patients are otherwise healthy with acute illnesses.[12] Additionally, pediatric patients have lower overall hospital reutilization rates when compared with adults. Adult 30‐day readmission rates are approximately 20%[13] compared with pediatric patients whose mean 30‐day readmission rate is 6.5%.[14] With readmission being an outcome upon which studies are basing intervention success or failure, the relatively low readmission rates in the pediatric population make shifting that outcome more challenging.

There is also controversy about whether policymakers should be focusing on decreasing 30‐day readmission rates as a measure of success. We believe that efforts should focus on identifying more meaningful outcomes, especially outcomes important to patients and their families. No single metric is likely to be an adequate measure of the quality of care transitions, but a combination of outcome measures could potentially be more informative both for patients and clinicians. Patient satisfaction with the discharge process is measured as part of standard patient experience surveys, and the 3‐question Care Transitions Measure[15] has been validated and endorsed as a measure of patient perception of discharge safety in adult populations. There is a growing consensus that 30‐day readmission rates are lacking as a measure of discharge quality, and therefore, measuring shorter‐term7‐ or 14‐dayreadmission rates along with short‐term ED utilization after discharge would likely be more helpful for identifying care transitions problems. Attention should also be paid to measuring rates of specific adverse events in the postdischarge period, such as adverse drug events or failure to follow up on pending test results, as these failures are often implicated in reutilization.

In reflecting upon the published data on adult and pediatric transitions of care interventions and the lingering unanswered questions, we propose a few considerations for future direction of the field. First, engagement of the primary care provider may be beneficial. In many interventions describing a care transition coordinator, nursing fulfilled this role; however, there are opportunities for the primary care provider to play a greater role in this arena. Second, the use of factorial design in future studies may help elucidate which specific parts of each intervention may be the most crucial.[16] Finally, readmission rates are a controversial quality measure in adults. Pediatric readmissions are relatively uncommon, making it difficult to track measurements and show improvement. Clinicians, patients, and policymakers should prioritize outcome measures that are most meaningful to patients and their families that occur at a much higher rate than that of readmissions.

References
  1. Auger KA, Kenyon CC, Feudtner C, Davis MM. Pediatric hospital discharge interventions to reduce subsequent utilization: a systematic review. J Hosp Med. 2014;9(0):000000.
  2. Co JP, Ferris TG, Marino BL, Homer CJ, Perrin JM. Are hospital characteristics associated with parental views of pediatric inpatient care quality? Pediatrics. 2003;111(2):308314.
  3. Rennke S, Nguyen OK, Shoeb MH, Magan Y, Wachter RM, Ranji SR. Hospital‐initiated transitional care interventions as a patient safety strategy: a systematic review. Ann Intern Med. 2013;158(5 pt 2):433440.
  4. Hansen LO, Young RS, Hinami K, Leung A, Williams MV. Interventions to reduce 30‐day rehospitalization: a systematic review. Ann Intern Med. 2011;155(8):520528.
  5. Hesselink G, Schoonhoven L, Barach P, et al. Improving patient handovers from hospital to primary care: a systematic review. Ann Intern Med. 2012;157(6):417428.
  6. Coleman EA, Parry C, Chalmers S, Min SJ. The care transitions intervention: results of a randomized controlled trial. Arch Intern Med. 2006;166(17):18221828.
  7. Kotagal UR, Perlstein PH, Gamblian V, Donovan EF, Atherton HD. Description and evaluation of a program for the early discharge of infants from a neonatal intensive care unit. J Pediatr. 1995;127(2):285290.
  8. Ng DK, Chow PY, Lai WP, Chan KC, And BL, So HY. Effect of a structured asthma education program on hospitalized asthmatic children: a randomized controlled study. Pediatr Int. 2006;48(2):158162.
  9. Taggart VS, Zuckerman AE, Sly RM, et al. You Can Control Asthma: evaluation of an asthma education‐program for hospitalized inner‐city children. Patient Educ Couns. 1991;17(1):3547.
  10. Forster AJ, Clark HD, Menard A, et al. Adverse events among medical patients after discharge from hospital. CMAJ. 2004;170(3):345349.
  11. Berry JG, Hall DE, Kuo DZ, et al. Hospital utilization and characteristics of patients experiencing recurrent readmissions within children's hospitals. JAMA. 2011;305(7):682690.
  12. Keren R, Luan X, Localio R, et al. Prioritization of comparative effectiveness research topics in hospital pediatrics. Arch Pediatr Adolesc Med. 2012;166(12):11551164.
  13. Joynt KE, Orav EJ, Jha AK. Thirty‐day readmission rates for Medicare beneficiaries by race and site of care. JAMA. 2011;305(7):675681.
  14. Berry JG, Toomey SL, Zaslavsky AM, et al. Pediatric readmission prevalence and variability across hospitals. JAMA. 2013;309(4):372380.
  15. Parry C, Mahoney E, Chalmers SA, Coleman EA. Assessing the quality of transitional care: further applications of the care transitions measure. Med Care. 2008;46(3):317322.
  16. Moen RD, Nolan TW, Provost LP. Quality Improvement Through Planned Experimentation. 2nd ed. New York, NY: McGraw‐Hill; 1999.
Article PDF
Issue
Journal of Hospital Medicine - 9(4)
Publications
Page Number
271-273
Sections
Article PDF
Article PDF

Hospital readmissions, which account for a substantial proportion of healthcare expenditures, have increasingly become a focus for hospitals and health systems. Hospitals now assume greater responsibility for population health, and face financial penalties by federal and state agencies that consider readmissions a key measure of the quality of care provided during hospitalization. Consequently, there is broad interest in identifying approaches to reduce hospital reutilization, including emergency department (ED) revisits and hospital readmissions. In this issue of the Journal of Hospital Medicine, Auger et al.[1] report the results of a systematic review, which evaluates the effect of discharge interventions on hospital reutilization among children.

As Auger et al. note, the transition from hospital to home is a vulnerable time for children and their families, with 1 in 5 parents reporting major challenges with such transitions.[2] Auger and colleagues identified 14 studies spanning 3 pediatric disease processes that addressed this issue. The authors concluded that several interventions were potentially effective, but individual studies frequently used multifactorial interventions, precluding determination of discrete elements essential to success. The larger body of care transitions literature in adult populations provides insights for interventions that may benefit pediatric patients, as well as informs future research and quality improvement priorities.

The authors identified some distinct interventions that may successfully decrease hospital reutilization, which share common themes from the adult literature. The first is the use of a dedicated transition coordinator (eg, nurse) or coordinating center to assist with the patient's transition home after discharge. In adult studies, this bridging strategy[3, 4] (ie, use of a dedicated transition coordinator or provider) is initiated during the hospitalization and continues postdischarge in the form of phone calls or home visits. The second theme illustrated in both this pediatric review[1] and adult reviews[3, 4, 5] focuses on enhanced or individualized patient education. Most studies have used a combination of these strategies. For example, the Care Transitions Intervention (one of the best validated adult discharge approaches) uses a transition coach to aid the patient in medication self‐management, creation of a patient‐centered record, scheduling follow‐up appointments, and understanding signs and symptoms of a worsening condition.[6] In a randomized study, this intervention demonstrated a reduction in readmissions within 90 days to 16.7% in the intervention group, compared with 22.5% in the control group.[6] One of the pediatric studies highlighted in the review by Auger et al. achieved a decrease in 14‐day ED revisits from 8% prior to implementation of the program to 2.7% following implementation of the program.[7] This program was for patients discharged from the neonatal intensive care unit and involved a nurse coordinator (similar to a transition coach) who worked closely with families and ensured adequate resources prior to discharge as well as a home visitation program.[7]

Although Auger et al. identify some effective approaches to reducing hospital reutilization after discharge in children, their review and the complementary adult literature bring to light 4 main unresolved questions for hospitalists seeking to improve care transitions: (1) how to dissect diverse and heterogeneous interventions to determine the key driver of success, (2) how to interpret and generally apply interventions from single centers where they may have been tailored to a specific healthcare environment, (3) how to generalize the findings of many disease‐specific interventions to other populations, and (4) how to evaluate the cost and assess the costbenefit of implementing many of the more resource intensive interventions. An example of a heterogeneous intervention addressed in this pediatric systematic review was described by Ng et al.,[8] in which the intervention group received a combination of an enhanced discharge education session, disease‐specific nurse evaluation, an animated education booklet, and postdischarge telephone follow‐up, whereas the control group received a shorter discharge education session, a disease‐specific nurse evaluation only if referred by a physician, a written education booklet, and no telephone follow‐up. Investigators found that intervention patients were less likely to be readmitted or revisit the ED as compared with controls. A similarly multifaceted intervention introduced by Taggart et al.[9] was unable to detect a difference in readmissions or ED revisits. It is unclear whether or not the differences in outcomes were related to differences in the intervention bundle itself or institutional or local contextual factors, thus limiting application to other hospitals. Generalizability of interventions is similarly complicated in adults.

The studies presented in this pediatric review article are specific to 3 disease processes: cancer, asthma, and neonatal intensive care (ie, premature) populations. Beyond these populations, there were no other pediatric conditions that met inclusion criteria, thus limiting the generalizability of the findings. As described by Rennke et al.,[3] adult systematic reviews that have focused only on disease‐specific interventions to reduce hospital reutilization are also difficult to generalize to broader populations. Two of the 3 recent adult transition intervention systematic reviews excluded disease‐specific interventions in an attempt to find more broadly applicable interventions but struggled with the same heterogeneity discussed in this review by Auger et al.[3, 4] Although disease‐specific interventions were included in the third adult systematic review and the evaluation was restricted to randomized controlled trials, the authors still grappled with finding 1 or 2 common, successful intervention components.[5] The fourth unresolved question involves understanding the financial burden of implementing more resource‐intensive interventions such as postdischarge home nurse visits. For example, it may be difficult to justify the business case for hiring a transition coach or initiating home nurse visits when the cost and financial implications are unclear. Neither the pediatric nor adult literature describes this well.

Some of the challenges in identifying effective interventions differ between adult and pediatric populations. Adults tend to have multiple comorbid conditions, making them more medically complex and at greater risk for adverse outcomes, medication errors, and hospital utilization.[10] Although a small subset of the pediatric population with complex chronic medical conditions accounts for a majority of hospital reutilization and cost,[11] most hospitalized pediatric patients are otherwise healthy with acute illnesses.[12] Additionally, pediatric patients have lower overall hospital reutilization rates when compared with adults. Adult 30‐day readmission rates are approximately 20%[13] compared with pediatric patients whose mean 30‐day readmission rate is 6.5%.[14] With readmission being an outcome upon which studies are basing intervention success or failure, the relatively low readmission rates in the pediatric population make shifting that outcome more challenging.

There is also controversy about whether policymakers should be focusing on decreasing 30‐day readmission rates as a measure of success. We believe that efforts should focus on identifying more meaningful outcomes, especially outcomes important to patients and their families. No single metric is likely to be an adequate measure of the quality of care transitions, but a combination of outcome measures could potentially be more informative both for patients and clinicians. Patient satisfaction with the discharge process is measured as part of standard patient experience surveys, and the 3‐question Care Transitions Measure[15] has been validated and endorsed as a measure of patient perception of discharge safety in adult populations. There is a growing consensus that 30‐day readmission rates are lacking as a measure of discharge quality, and therefore, measuring shorter‐term7‐ or 14‐dayreadmission rates along with short‐term ED utilization after discharge would likely be more helpful for identifying care transitions problems. Attention should also be paid to measuring rates of specific adverse events in the postdischarge period, such as adverse drug events or failure to follow up on pending test results, as these failures are often implicated in reutilization.

In reflecting upon the published data on adult and pediatric transitions of care interventions and the lingering unanswered questions, we propose a few considerations for future direction of the field. First, engagement of the primary care provider may be beneficial. In many interventions describing a care transition coordinator, nursing fulfilled this role; however, there are opportunities for the primary care provider to play a greater role in this arena. Second, the use of factorial design in future studies may help elucidate which specific parts of each intervention may be the most crucial.[16] Finally, readmission rates are a controversial quality measure in adults. Pediatric readmissions are relatively uncommon, making it difficult to track measurements and show improvement. Clinicians, patients, and policymakers should prioritize outcome measures that are most meaningful to patients and their families that occur at a much higher rate than that of readmissions.

Hospital readmissions, which account for a substantial proportion of healthcare expenditures, have increasingly become a focus for hospitals and health systems. Hospitals now assume greater responsibility for population health, and face financial penalties by federal and state agencies that consider readmissions a key measure of the quality of care provided during hospitalization. Consequently, there is broad interest in identifying approaches to reduce hospital reutilization, including emergency department (ED) revisits and hospital readmissions. In this issue of the Journal of Hospital Medicine, Auger et al.[1] report the results of a systematic review, which evaluates the effect of discharge interventions on hospital reutilization among children.

As Auger et al. note, the transition from hospital to home is a vulnerable time for children and their families, with 1 in 5 parents reporting major challenges with such transitions.[2] Auger and colleagues identified 14 studies spanning 3 pediatric disease processes that addressed this issue. The authors concluded that several interventions were potentially effective, but individual studies frequently used multifactorial interventions, precluding determination of discrete elements essential to success. The larger body of care transitions literature in adult populations provides insights for interventions that may benefit pediatric patients, as well as informs future research and quality improvement priorities.

The authors identified some distinct interventions that may successfully decrease hospital reutilization, which share common themes from the adult literature. The first is the use of a dedicated transition coordinator (eg, nurse) or coordinating center to assist with the patient's transition home after discharge. In adult studies, this bridging strategy[3, 4] (ie, use of a dedicated transition coordinator or provider) is initiated during the hospitalization and continues postdischarge in the form of phone calls or home visits. The second theme illustrated in both this pediatric review[1] and adult reviews[3, 4, 5] focuses on enhanced or individualized patient education. Most studies have used a combination of these strategies. For example, the Care Transitions Intervention (one of the best validated adult discharge approaches) uses a transition coach to aid the patient in medication self‐management, creation of a patient‐centered record, scheduling follow‐up appointments, and understanding signs and symptoms of a worsening condition.[6] In a randomized study, this intervention demonstrated a reduction in readmissions within 90 days to 16.7% in the intervention group, compared with 22.5% in the control group.[6] One of the pediatric studies highlighted in the review by Auger et al. achieved a decrease in 14‐day ED revisits from 8% prior to implementation of the program to 2.7% following implementation of the program.[7] This program was for patients discharged from the neonatal intensive care unit and involved a nurse coordinator (similar to a transition coach) who worked closely with families and ensured adequate resources prior to discharge as well as a home visitation program.[7]

Although Auger et al. identify some effective approaches to reducing hospital reutilization after discharge in children, their review and the complementary adult literature bring to light 4 main unresolved questions for hospitalists seeking to improve care transitions: (1) how to dissect diverse and heterogeneous interventions to determine the key driver of success, (2) how to interpret and generally apply interventions from single centers where they may have been tailored to a specific healthcare environment, (3) how to generalize the findings of many disease‐specific interventions to other populations, and (4) how to evaluate the cost and assess the costbenefit of implementing many of the more resource intensive interventions. An example of a heterogeneous intervention addressed in this pediatric systematic review was described by Ng et al.,[8] in which the intervention group received a combination of an enhanced discharge education session, disease‐specific nurse evaluation, an animated education booklet, and postdischarge telephone follow‐up, whereas the control group received a shorter discharge education session, a disease‐specific nurse evaluation only if referred by a physician, a written education booklet, and no telephone follow‐up. Investigators found that intervention patients were less likely to be readmitted or revisit the ED as compared with controls. A similarly multifaceted intervention introduced by Taggart et al.[9] was unable to detect a difference in readmissions or ED revisits. It is unclear whether or not the differences in outcomes were related to differences in the intervention bundle itself or institutional or local contextual factors, thus limiting application to other hospitals. Generalizability of interventions is similarly complicated in adults.

The studies presented in this pediatric review article are specific to 3 disease processes: cancer, asthma, and neonatal intensive care (ie, premature) populations. Beyond these populations, there were no other pediatric conditions that met inclusion criteria, thus limiting the generalizability of the findings. As described by Rennke et al.,[3] adult systematic reviews that have focused only on disease‐specific interventions to reduce hospital reutilization are also difficult to generalize to broader populations. Two of the 3 recent adult transition intervention systematic reviews excluded disease‐specific interventions in an attempt to find more broadly applicable interventions but struggled with the same heterogeneity discussed in this review by Auger et al.[3, 4] Although disease‐specific interventions were included in the third adult systematic review and the evaluation was restricted to randomized controlled trials, the authors still grappled with finding 1 or 2 common, successful intervention components.[5] The fourth unresolved question involves understanding the financial burden of implementing more resource‐intensive interventions such as postdischarge home nurse visits. For example, it may be difficult to justify the business case for hiring a transition coach or initiating home nurse visits when the cost and financial implications are unclear. Neither the pediatric nor adult literature describes this well.

Some of the challenges in identifying effective interventions differ between adult and pediatric populations. Adults tend to have multiple comorbid conditions, making them more medically complex and at greater risk for adverse outcomes, medication errors, and hospital utilization.[10] Although a small subset of the pediatric population with complex chronic medical conditions accounts for a majority of hospital reutilization and cost,[11] most hospitalized pediatric patients are otherwise healthy with acute illnesses.[12] Additionally, pediatric patients have lower overall hospital reutilization rates when compared with adults. Adult 30‐day readmission rates are approximately 20%[13] compared with pediatric patients whose mean 30‐day readmission rate is 6.5%.[14] With readmission being an outcome upon which studies are basing intervention success or failure, the relatively low readmission rates in the pediatric population make shifting that outcome more challenging.

There is also controversy about whether policymakers should be focusing on decreasing 30‐day readmission rates as a measure of success. We believe that efforts should focus on identifying more meaningful outcomes, especially outcomes important to patients and their families. No single metric is likely to be an adequate measure of the quality of care transitions, but a combination of outcome measures could potentially be more informative both for patients and clinicians. Patient satisfaction with the discharge process is measured as part of standard patient experience surveys, and the 3‐question Care Transitions Measure[15] has been validated and endorsed as a measure of patient perception of discharge safety in adult populations. There is a growing consensus that 30‐day readmission rates are lacking as a measure of discharge quality, and therefore, measuring shorter‐term7‐ or 14‐dayreadmission rates along with short‐term ED utilization after discharge would likely be more helpful for identifying care transitions problems. Attention should also be paid to measuring rates of specific adverse events in the postdischarge period, such as adverse drug events or failure to follow up on pending test results, as these failures are often implicated in reutilization.

In reflecting upon the published data on adult and pediatric transitions of care interventions and the lingering unanswered questions, we propose a few considerations for future direction of the field. First, engagement of the primary care provider may be beneficial. In many interventions describing a care transition coordinator, nursing fulfilled this role; however, there are opportunities for the primary care provider to play a greater role in this arena. Second, the use of factorial design in future studies may help elucidate which specific parts of each intervention may be the most crucial.[16] Finally, readmission rates are a controversial quality measure in adults. Pediatric readmissions are relatively uncommon, making it difficult to track measurements and show improvement. Clinicians, patients, and policymakers should prioritize outcome measures that are most meaningful to patients and their families that occur at a much higher rate than that of readmissions.

References
  1. Auger KA, Kenyon CC, Feudtner C, Davis MM. Pediatric hospital discharge interventions to reduce subsequent utilization: a systematic review. J Hosp Med. 2014;9(0):000000.
  2. Co JP, Ferris TG, Marino BL, Homer CJ, Perrin JM. Are hospital characteristics associated with parental views of pediatric inpatient care quality? Pediatrics. 2003;111(2):308314.
  3. Rennke S, Nguyen OK, Shoeb MH, Magan Y, Wachter RM, Ranji SR. Hospital‐initiated transitional care interventions as a patient safety strategy: a systematic review. Ann Intern Med. 2013;158(5 pt 2):433440.
  4. Hansen LO, Young RS, Hinami K, Leung A, Williams MV. Interventions to reduce 30‐day rehospitalization: a systematic review. Ann Intern Med. 2011;155(8):520528.
  5. Hesselink G, Schoonhoven L, Barach P, et al. Improving patient handovers from hospital to primary care: a systematic review. Ann Intern Med. 2012;157(6):417428.
  6. Coleman EA, Parry C, Chalmers S, Min SJ. The care transitions intervention: results of a randomized controlled trial. Arch Intern Med. 2006;166(17):18221828.
  7. Kotagal UR, Perlstein PH, Gamblian V, Donovan EF, Atherton HD. Description and evaluation of a program for the early discharge of infants from a neonatal intensive care unit. J Pediatr. 1995;127(2):285290.
  8. Ng DK, Chow PY, Lai WP, Chan KC, And BL, So HY. Effect of a structured asthma education program on hospitalized asthmatic children: a randomized controlled study. Pediatr Int. 2006;48(2):158162.
  9. Taggart VS, Zuckerman AE, Sly RM, et al. You Can Control Asthma: evaluation of an asthma education‐program for hospitalized inner‐city children. Patient Educ Couns. 1991;17(1):3547.
  10. Forster AJ, Clark HD, Menard A, et al. Adverse events among medical patients after discharge from hospital. CMAJ. 2004;170(3):345349.
  11. Berry JG, Hall DE, Kuo DZ, et al. Hospital utilization and characteristics of patients experiencing recurrent readmissions within children's hospitals. JAMA. 2011;305(7):682690.
  12. Keren R, Luan X, Localio R, et al. Prioritization of comparative effectiveness research topics in hospital pediatrics. Arch Pediatr Adolesc Med. 2012;166(12):11551164.
  13. Joynt KE, Orav EJ, Jha AK. Thirty‐day readmission rates for Medicare beneficiaries by race and site of care. JAMA. 2011;305(7):675681.
  14. Berry JG, Toomey SL, Zaslavsky AM, et al. Pediatric readmission prevalence and variability across hospitals. JAMA. 2013;309(4):372380.
  15. Parry C, Mahoney E, Chalmers SA, Coleman EA. Assessing the quality of transitional care: further applications of the care transitions measure. Med Care. 2008;46(3):317322.
  16. Moen RD, Nolan TW, Provost LP. Quality Improvement Through Planned Experimentation. 2nd ed. New York, NY: McGraw‐Hill; 1999.
References
  1. Auger KA, Kenyon CC, Feudtner C, Davis MM. Pediatric hospital discharge interventions to reduce subsequent utilization: a systematic review. J Hosp Med. 2014;9(0):000000.
  2. Co JP, Ferris TG, Marino BL, Homer CJ, Perrin JM. Are hospital characteristics associated with parental views of pediatric inpatient care quality? Pediatrics. 2003;111(2):308314.
  3. Rennke S, Nguyen OK, Shoeb MH, Magan Y, Wachter RM, Ranji SR. Hospital‐initiated transitional care interventions as a patient safety strategy: a systematic review. Ann Intern Med. 2013;158(5 pt 2):433440.
  4. Hansen LO, Young RS, Hinami K, Leung A, Williams MV. Interventions to reduce 30‐day rehospitalization: a systematic review. Ann Intern Med. 2011;155(8):520528.
  5. Hesselink G, Schoonhoven L, Barach P, et al. Improving patient handovers from hospital to primary care: a systematic review. Ann Intern Med. 2012;157(6):417428.
  6. Coleman EA, Parry C, Chalmers S, Min SJ. The care transitions intervention: results of a randomized controlled trial. Arch Intern Med. 2006;166(17):18221828.
  7. Kotagal UR, Perlstein PH, Gamblian V, Donovan EF, Atherton HD. Description and evaluation of a program for the early discharge of infants from a neonatal intensive care unit. J Pediatr. 1995;127(2):285290.
  8. Ng DK, Chow PY, Lai WP, Chan KC, And BL, So HY. Effect of a structured asthma education program on hospitalized asthmatic children: a randomized controlled study. Pediatr Int. 2006;48(2):158162.
  9. Taggart VS, Zuckerman AE, Sly RM, et al. You Can Control Asthma: evaluation of an asthma education‐program for hospitalized inner‐city children. Patient Educ Couns. 1991;17(1):3547.
  10. Forster AJ, Clark HD, Menard A, et al. Adverse events among medical patients after discharge from hospital. CMAJ. 2004;170(3):345349.
  11. Berry JG, Hall DE, Kuo DZ, et al. Hospital utilization and characteristics of patients experiencing recurrent readmissions within children's hospitals. JAMA. 2011;305(7):682690.
  12. Keren R, Luan X, Localio R, et al. Prioritization of comparative effectiveness research topics in hospital pediatrics. Arch Pediatr Adolesc Med. 2012;166(12):11551164.
  13. Joynt KE, Orav EJ, Jha AK. Thirty‐day readmission rates for Medicare beneficiaries by race and site of care. JAMA. 2011;305(7):675681.
  14. Berry JG, Toomey SL, Zaslavsky AM, et al. Pediatric readmission prevalence and variability across hospitals. JAMA. 2013;309(4):372380.
  15. Parry C, Mahoney E, Chalmers SA, Coleman EA. Assessing the quality of transitional care: further applications of the care transitions measure. Med Care. 2008;46(3):317322.
  16. Moen RD, Nolan TW, Provost LP. Quality Improvement Through Planned Experimentation. 2nd ed. New York, NY: McGraw‐Hill; 1999.
Issue
Journal of Hospital Medicine - 9(4)
Issue
Journal of Hospital Medicine - 9(4)
Page Number
271-273
Page Number
271-273
Publications
Publications
Article Type
Display Headline
The successes and challenges of hospital to home transitions
Display Headline
The successes and challenges of hospital to home transitions
Sections
Article Source
© 2014 Society of Hospital Medicine
Disallow All Ads
Correspondence Location
Address for correspondence and reprint requests: Samir S. Shah, MD, Division of Hospital Medicine, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave., MLC 9016, Cincinnati, OH 45229‐3039; Telephone: 513–636‐0409; E‐mail: samir.shah@cchmc.org
Content Gating
Gated (full article locked unless allowed per User)
Gating Strategy
First Peek Free
Article PDF Media
Image
Disable zoom
Off

Chest Radiograph Interpretation

Article Type
Changed
Sun, 05/21/2017 - 18:10
Display Headline
Impact of clinical history on chest radiograph interpretation

The inclusion of clinical information in diagnostic testing may influence the interpretation of the clinical findings. Historical and clinical findings may focus the reader's attention to the relevant details, thereby improving the accuracy of the interpretation. However, such information may cause the reader to have preconceived notions about the results, biasing the overall interpretation.

The impact of clinical information on the interpretation of radiographic studies remains an issue of debate. Previous studies have found that clinical information improves the accuracy of radiographic interpretation for a broad range of diagnoses,[1, 2, 3, 4] whereas others do not show improvement.[5, 6, 7] Additionally, clinical information may serve as a distraction that leads to more false‐positive interpretations.[8] For this reason, many radiologists prefer to review radiographs without knowledge of the clinical scenario prompting the study to avoid focusing on the expected findings and potentially missing other important abnormalities.[9]

The chest radiograph (CXR) is the most commonly used diagnostic imaging modality. Nevertheless, poor agreement exists among radiologists in the interpretation of chest radiographs for the diagnosis of pneumonia in both adults and children.[10, 11, 12, 13, 14, 15] Recent studies have found a high degree of agreement among pediatric radiologists with implementation of the World Health Organization (WHO) criteria for standardized CXR interpretation for diagnosis of bacterial pneumonia in children.[16, 17, 18] In these studies, participants were blinded to the clinical presentation. Data investigating the impact of clinical history on CXR interpretation in the pediatric population are limited.[19]

We conducted this prospective case‐based study to evaluate the impact of clinical information on the reliability of radiographic diagnosis of pneumonia among children presenting to a pediatric emergency department (ED) with clinical suspicion of pneumonia.

METHODS

Study Subjects

Six board‐certified radiologists at 2 academic children's hospitals (Children's Hospital of Philadelphia [n = 3] and Boston Children's Hospital [n = 3]) interpreted the same 110 chest radiographs (100 original and 10 duplicates) on 2 separate occasions. Clinical information was withheld during the first interpretation. The inter‐ and inter‐rater reliability for the interpretation of these 110 radiographs without clinical information have been previously reported.[18] After a period of 6 months, the radiologists reviewed the radiographs with access to clinical information provided by the physician ordering the CXR. This clinical information included age, sex, clinical indication for obtaining the radiograph, relevant history, and physical examination findings. The radiologists did not have access to the patients' medical records. The radiologists varied with respect to the number of years practicing pediatric radiology (median, 8 years; range, 336 years).

Radiographs were selected from children who presented to the ED at Boston Children's Hospital with concern of pneumonia. We selected radiographs with a spectrum of respiratory disease processes encountered in a pediatric population. The final radiographs included 50 radiographs with a final reading in the medical record without suspicion for pneumonia and 50 radiographs with suspicion of pneumonia. In the latter group, 25 radiographs had a final reading suggestive of an alveolar infiltrate, and 25 radiographs had a final reading suggestive of an interstitial infiltrate. Ten duplicate radiographs were included.

Radiograph Interpretation

The radiologists interpreted both anterior‐posterior and lateral views for each subject. Digital Imaging and Communications in Medicine images were downloaded from a registry at Boston Children's Hospital, and were copied to DVDs that were provided to each radiologist. Standardized radiographic imaging software (eFilm Lite; Merge Healthcare, Chicago, Illinois) was used by each radiologist.

Each radiologist completed a study questionnaire for each radiograph (see Supporting Information, Appendix 1, in the online version of this article). The questionnaire utilized radiographic descriptors of primary endpoint pneumonia described by the WHO to standardize the radiographic diagnosis of pneumonia.[20, 21] No additional training was provided to the radiologists. The main outcome of interest was the presence or absence of an infiltrate. Among radiographs in which an infiltrate was identified, radiologists selected whether there was an alveolar infiltrate, interstitial infiltrate, or both. Alveolar infiltrate and interstitial infiltrate are defined on the study questionnaire (Appendix 1). A radiograph classified as having either an alveolar infiltrate or interstitial infiltrate (not atelectasis) was considered to have any infiltrate. Additional findings including air bronchograms, hilar adenopathy, pleural effusion, and location of abnormalities were also recorded.

Statistical Analysis

Inter‐rater reliability was assessed using the kappa statistic to determine the overall agreement among the 6 radiologists for each outcome (eg, presence or absence of alveolar infiltrate). The kappa statistic for more than 2 raters utilizes an analysis of variance approach.[22] To calculate 95% confidence intervals (CI) for kappa statistics with more than 2 raters, we employed a bootstrapping method with 1000 replications of samples equal in size to the study sample. Intra‐rater reliability was evaluated by examining the agreement within each radiologist upon review of 10 duplicate radiographs. We used the following benchmarks to classify the strength of agreement: poor (<0.0), slight (00.20), fair (0.210.40), moderate (0.410.60), substantial (0.610.80), almost perfect (0.811.0).[23] Negative kappa values represent agreement less than would be predicted by chance alone.[24, 25] To calculate the kappa, a value must be recorded in 3 of 4 of the following categories: negative to positive, positive to negative, concordant negative, and concordant positive reporting of pneumonia. If raters did not fulfill 3 categories, the kappa could not be calculated.

The inter‐rater concordance for identification of an alveolar infiltrate was calculated for each radiologist by comparing their reporting of alveolar infiltrate with and without clinical history for each of the 100 radiographs. Radiographs that were identified by an individual rater as no alveolar infiltrate when read without clinical history, but those subsequently identified as alveolar infiltrate with clinical history were categorized as negative to positive reporting of pneumonia with clinical history. Those that were identified as alveolar infiltrate but subsequently identified as no alveolar infiltrate were categorized as positive to negative reporting of pneumonia with clinical history. Those radiographs in which there was no change in identification of alveolar infiltrate with clinical information were categorized as concordant reporting of pneumonia.

The study was approved by the institutional review boards at both children's hospitals.

RESULTS

Patient Sample

The radiographs were from patients ranging in age from 1 week to 19 years (median, 3.5 years; interquartile range, 1.66.0 years). Fifty (50%) patients were male.

Inter‐rater Reliability

The kappa coefficients of inter‐rater reliability between the radiologists across the 6 clinical measures of interest with and without access to clinical history are plotted in Figure 1. Reliability improved from fair (k = 0.32, 95% CI: 0.24 to 0.42) to moderate (k = 0.53, 95% CI: 0.43 to 0.64) for identification of air bronchograms with the addition of clinical history. Although there was an increase in kappa values for identification of any infiltrate, alveolar infiltrate, interstitial infiltrate, and pleural effusion, and a decrease in the kappa value for identification of hilar adenopathy with the addition of clinical information, there was substantial overlap of the 95% CIs, suggesting that inclusion of clinical history did not result in a statistically significant change in the reliability of these findings.

jhm1991-fig-0001-m.png
Inter‐rater reliability of radiologists (n = 6) evaluating chest radiographs with and without access to clinical history data in children presenting to the emergency department with suspected pneumonia (n = 100).

Intra‐rater Reliability

The estimates of inter‐rater reliability for the interpretation of the 10 duplicate images with and without clinical history are shown in Table 1. The inter‐rater reliability in the identification of alveolar infiltrate remained substantial to almost perfect for each rater with and without access to clinical history. Rater 1 had a decrease in inter‐rater reliability from almost perfect (k = 1.0, 95% CI: 1.0 to 1.0) to fair (k = 0.21, 95% CI: 0.43 to 0.85) in the identification of interstitial infiltrate with the addition of clinical history. This rater also had a decrease in agreement from almost perfect (k = 1.0, 95% CI: 1.0 to 1.0) to fair (k = 0.4, 95% CI: 0.16 to 0.96) in the identification of any infiltrate.

Intra‐rater Reliability of Radiologists With and Without Access to Clinical History While Evaluating Chest Radiographs (n = 10) for Pneumonia in Children
 Phase 1No Clinical HistoryPhase 2Access to Clinical History
Kappa95% Confidence IntervalKappa95% Confidence Interval
  • NOTE: Abbreviations: N/A, not applicable.
  • Too few categories of agreement to calculate kappa. Both responses are negative for all 10 paired radiographs; kappa cannot be calculated.
Any infiltrate    
Rater 11.001.00 to 1.000.400.16 to 0.96
Rater 20.600.10 to 1.000.580.07 to 1.00
Rater 30.800.44 to 1.000.800.44 to 1.00
Rater 41.001.00 to 1.000.780.39 to 1.00
Rater 5N/Aa 0.110.36 to 0.14
Rater 61.001.00 to 1.001.001.00 to 1.00
Alveolar infiltrate    
Rater 11.001.00 to 1.001.001.00 to 1.00
Rater 21.001.00 to 1.001.001.00 to 1.00
Rater 31.001.00 to 1.001.001.00 to 1.00
Rater 41.001.00 to 1.000.780.39 to 1.00
Rater 50.780.39 to 1.001.001.00 to 1.00
Rater 60.740.27 to 1.000.780.39 to 1.00
Interstitial infiltrate    
Rater 11.001.00 to 1.000.210.43 to 0.85
Rater 20.210.43 to 0.850.110.36 to 0.14
Rater 30.740.27 to 1.000.780.39 to 1.00
Rater 4N/A N/A 
Rater 50.580.07 to 1.000.520.05 to 1.00
Rater 60.620.5 to 1.00N/Aa 

Intra‐rater Concordance

The inter‐rater concordance of the radiologists for the identification of alveolar infiltrate during the interpretation of the 100 chest radiographs with and without access to clinical history is shown in Figure 2. The availability of clinical information impacted physicians differently in the evaluation of alveolar infiltrates. Raters 1, 4, and 6 appeared more likely to identify an alveolar infiltrate with access to the clinical information, whereas raters 3 and 5 appeared less likely to identify an alveolar infiltrate. Of the 100 films that were interpreted with and without clinical information, the mean number of discordant interpretations per rater was 10, with values ranging from 6 to 19 for the individual raters. Radiographs in which more than 3 raters changed their interpretation regarding the presence of an alveolar infiltrate are shown in Figure 3. For Figure 3D, 4 radiologists changed their interpretation from no alveolar infiltrate to alveolar infiltrate, and 1 radiologist changed from alveolar infiltrate to no alveolar infiltrate with the addition of clinical history.

jhm1991-fig-0002-m.png
Intra‐rater concordance of radiologists before and after access to clinical history while evaluating chest radiographs (n = 100) for alveolar infiltrate in children.
jhm1991-fig-0003-m.png
Chest radiographs of children in which 3 or more radiologists changed their interpretation in regard to the presence or absence of an alveolar infiltrate with the addition of clinical information. (A, B, and C) Three of 6 radiologists changed their interpretation. (D) Five of 6 radiologists changed their interpretation. (A) Female, 2 years old. (B) Male, 9 months old. (C) Male, 3 years old. (D) Male, 3 years old. The clinical history provided for (D) read as follows: “3‐year‐old male with cough and difficulty breathing. Rales at left base.”

Comment

We investigated the impact of the availability of clinical information on the reliability of chest radiographic interpretation in the diagnosis of pneumonia. There was improved inter‐rater reliability in the identification of air bronchograms with the addition of clinical information; however, clinical history did not have a substantial impact on the inter‐rater reliability of other findings. The addition of clinical information did not alter the inter‐rater reliability in the identification of alveolar infiltrate. Clinical history affected individual raters differently in their interpretation of alveolar infiltrate, with 3 raters more likely to identify an alveolar infiltrate and 2 raters less likely to identify an alveolar infiltrate.

Most studies addressing the impact of clinical history on radiographic interpretation evaluated accuracy. In many of these studies, accuracy was defined as the raters' agreement with the final interpretation of each film as documented in the medical record or their agreement with the interpretation of the radiologists selecting the cases.[1, 2, 3, 5, 6, 7] Given the known inter‐rater variability in radiographic interpretation,[10, 11, 12, 13, 14, 15] accuracy of a radiologist's interpretation cannot be appropriately assessed through agreement with their peers. Because a true measure of accuracy in the radiographic diagnosis of pneumonia can only be determined through invasive testing, such as lung biopsy, reliability serves as a more appropriate measure of performance. Inclusion of clinical information in chest radiograph interpretation has been shown to improve reliability in the radiographic diagnosis of a broad range of conditions.[15]

The primary outcome in this study was the identification of an infiltrate. Previous studies have noted consistent identification of the radiographic features that are most suggestive of bacterial pneumonia, such as alveolar infiltrate, and less consistent identification of other radiographic findings, including interstitial infiltrate.[18, 26, 27] Among the radiologists in this study, the addition of clinical information did not have a meaningful impact on the reliability of either of these findings, as there was substantial inter‐rater agreement for the identification of alveolar infiltrate and only slight agreement for the identification of interstitial infiltrate, both with and without clinical history. Additionally, inter‐rater reliability for the identification of alveolar infiltrate remained substantial to almost perfect for all 6 raters with the addition of clinical information.

Clinical information impacted the raters differently in their pattern of alveolar infiltrate identification, suggesting that radiologists may differ in their approach to incorporating clinical history in the interpretation of chest radiographs. The inclusion of clinical information may impact a radiologist's perception, leading to improved identification of abnormalities; however, it may also guide their decision making about the relevance of previously identified abnormalities.[28] Some radiologists may use clinical information to support or suggest possible radiographic findings, whereas others may use the information to challenge potential findings. This study did not address the manner in which the individual raters utilized the clinical history. There were also several radiographs in which the clinical information resulted in a change in the identification of an alveolar infiltrate by 3 or more raters, with as many as 5 of 6 raters changing their interpretation for 1 particular radiograph. These changes in identification of an infiltrate suggest that unidentified aspects of a history may be likely to influence a rater's interpretation of a radiograph. Nevertheless, these changes did not result in improved reliability and it is not possible to determine if these changes resulted in improved accuracy in interpretation.

This study had several limitations. First, radiographs were purposefully selected to encompass a broad spectrum of radiographic findings. Thus, the prevalence of pneumonia and other abnormal findings was artificially higher than typically observed among a cohort of children for whom pneumonia is considered. Second, the radiologists recruited for this study all practice in an academic children's hospital setting. These factors may limit the generalizability of our findings. However, we would expect these results to be generalizable to pediatric radiologists from other academic institutions. Third, this study does not meet the criteria of a balanced study design as defined by Loy and Irwig.[19] A study was characterized as balanced if half of the radiographs were read with and half without clinical information in each of the 2 reading sessions. The proposed benefit of such a design is to control for possible changes in ability or reporting practices of the raters that may have occurred between study periods. The use of a standardized reporting tool likely minimized changes in reporting practices. Also, it is unlikely that the ability or reporting practices of an experienced radiologist would change over the study period. Fourth, the radiologists interpreted the films outside of their standard workflow and utilized a standardized reporting tool that focused on the presence or absence of pneumonia indicators. These factors may have increased the radiologists' suspicion for pneumonia even in the absence of clinical information. This may have biased the results toward finding no difference in the identification of pneumonia with the addition of detailed clinical history. Thus, the inclusion of clinical information in radiograph interpretation in clinical practice may have greater impact on the identification of these pneumonia indicators than was found in this study.[29] Finally, reliability does not imply accuracy, and it is unknown if changes in the identification of pneumonia indicators led to more accurate interpretation with respect to the clinical or pathologic diagnosis of pneumonia.

In conclusion, we observed high intra‐ and inter‐rater reliability among radiologists in the identification of an alveolar infiltrate, the radiographic finding most suggestive of bacterial pneumonia.[16, 17, 18, 30] The addition of clinical information did not have a substantial impact on the reliability of its identification.

Files
References
  1. Berbaum KS, Franken EA, Dorfman DD, et al. Tentative diagnoses facilitate the detection of diverse lesions in chest radiographs. Invest Radiol. 1986;21(7):532539.
  2. Berbaum KS, Franken EA, Dorfman DD, Barloon TJ. Influence of clinical history upon detection of nodules and other lesions. Invest Radiol. 1988;23(1):4855.
  3. Berbaum KS, Franken EA, Dorfman DD, Lueben KR. Influence of clinical history on perception of abnormalities in pediatric radiographs. Acad Radiol. 1994;1(3):217223.
  4. Song KS, Song HH, Park SH, et al. Impact of clinical history on film interpretation. Yonsei Med J. 1992;33(2):168172.
  5. Cooperstein LA, Good BC, Eelkema EA, et al. The effect of clinical history on chest radiograph interpretations in a PACS environment. Invest Radiol. 1990;25(6):670674.
  6. Good BC, Cooperstein LA, DeMarino GB, et al. Does knowledge of the clinical history affect the accuracy of chest radiograph interpretation?AJR Am J Roentgenol. 1990;154(4):709712.
  7. Quekel LG, Goei R, Kessels AG, Engelshoven JM. Detection of lung cancer on the chest radiograph: impact of previous films, clinical information, double reading, and dual reading. J Clin Epidemiol. 2001;54(11):11461150.
  8. Eldevik OP, Dugstad G, Orrison WW, Haughton VM. The effect of clinical bias on the interpretation of myelography and spinal computed tomography. Radiology. 1982;145(1):8589.
  9. Griscom NT. A suggestion: look at the images first, before you read the history. Radiology. 2002;223(1):910.
  10. Albaum MN, Hill LC, Murphy M, et al. Interobserver reliability of the chest radiograph in community‐acquired pneumonia. PORT Investigators. Chest. 1996;110(2):343350.
  11. Bloomfield FH, Teele RL, Voss M, Knight DB, Harding JE. Inter‐ and intra‐observer variability in the assessment of atelectasis and consolidation in neonatal chest radiographs. Pediatr Radiol. 1999;29(6):459462.
  12. Gatt ME, Spectre G, Paltiel O, Hiller N, Stalnikowicz R. Chest radiographs in the emergency department: is the radiologist really necessary?Postgrad Med J. 2003;79(930):214217.
  13. Hopstaken RM, Witbraad T, Engelshoven JM, Dinant GJ. Inter‐observer variation in the interpretation of chest radiographs for pneumonia in community‐acquired lower respiratory tract infections. Clin Radiol. 2004;59(8):743752.
  14. Novack V, Avnon LS, Smolyakov A, Barnea R, Jotkowitz A, Schlaeffer F. Disagreement in the interpretation of chest radiographs among specialists and clinical outcomes of patients hospitalized with suspected pneumonia. Eur J Intern Med. 2006;17(1):4347.
  15. Tudor GR, Finlay D, Taub N. An assessment of inter‐observer agreement and accuracy when reporting plain radiographs. Clin Radiol. 1997;52(3):235238.
  16. Shimol BS, Dagan R, Givon‐Lavi N, et al. Evaluation of the World Health Organization criteria for chest radiographs for pneumonia diagnosis in children. Eur J Pediatr. 2011;171(2):369374.
  17. Cherian T, Mulholland EK, Carlin JB, et al. Standardized interpretation of paediatric chest radiographs for the diagnosis of pneumonia in epidemiological studies. Bull World Health Organ. 2005;83(5):353359.
  18. Neuman MI, Lee EY, Bixby S, et al. Variability in the interpretation of chest radiographs for the diagnosis of pneumonia in children. J Hosp Med. 2012;7(4):294298.
  19. Loy CT, Irwig L. Accuracy of diagnostic tests read with and without clinical information: a systematic review. JAMA. 2004;292(13):16021609.
  20. Standardization of interpretation of chest radiographs for the diagnosis of pneumonia in children. In:World Health Organization: Pneumonia Vaccine Trial Investigators' Group.Geneva: Department of Vaccine and Biologics;2001.
  21. Hansen J, Black S, Shinefield H, et al. Effectiveness of heptavalent pneumococcal conjugate vaccine in children younger than 5 years of age for prevention of pneumonia: updated analysis using World Health Organization standardized interpretation of chest radiographs. Pediatr Infect Dis J. 2006;25(9):779781.
  22. Landis JR, Koch GG. A one‐way components of variance model for categorical data. Biometrics. 1977;33:671679.
  23. Landis JR, Koch GG. The measurement of observer agreement for categorical data. Biometrics. 1977;33(1):159174.
  24. Juurlink DN, Detsky AS. Kappa statistic. CMAJ. 2005;173(1):16.
  25. Kramer MS, Feinstein AR. Clinical biostatistics. LIV. The biostatistics of concordance. Clin Pharmacol Ther. 1981;29(1):111123.
  26. Bartlett JG, Dowell SF, Mandell LA, File TM, Musher DM, Fine MJ. Practice guidelines for the management of community‐acquired pneumonia in adults. Infectious Diseases Society of America. Clin Infect Dis. 2000;31(2):347382.
  27. Niederman MS, Mandell LA, Anzueto A, et al. Guidelines for the management of adults with community‐acquired pneumonia. Diagnosis, assessment of severity, antimicrobial therapy, and prevention. Am J Respir Crit Care Med. 2001;163(7):17301754.
  28. Berbaum KS, Franken EA. Commentary does clinical history affect perception?Acad Radiol. 2006;13(3):402403.
  29. Berbaum KS, Franken EA. The effect of clinical history on chest radiograph interpretations in a PACS environment. Invest Radiol. 1991;26(5):512514.
  30. Korppi M, Kiekara O, Heiskanen‐Kosma T, Soimakallio S. Comparison of radiological findings and microbial aetiology of childhood pneumonia. Acta Paediatr. 1993;82(4):360363.
Article PDF
Issue
Journal of Hospital Medicine - 8(7)
Publications
Page Number
359-364
Sections
Files
Files
Article PDF
Article PDF

The inclusion of clinical information in diagnostic testing may influence the interpretation of the clinical findings. Historical and clinical findings may focus the reader's attention to the relevant details, thereby improving the accuracy of the interpretation. However, such information may cause the reader to have preconceived notions about the results, biasing the overall interpretation.

The impact of clinical information on the interpretation of radiographic studies remains an issue of debate. Previous studies have found that clinical information improves the accuracy of radiographic interpretation for a broad range of diagnoses,[1, 2, 3, 4] whereas others do not show improvement.[5, 6, 7] Additionally, clinical information may serve as a distraction that leads to more false‐positive interpretations.[8] For this reason, many radiologists prefer to review radiographs without knowledge of the clinical scenario prompting the study to avoid focusing on the expected findings and potentially missing other important abnormalities.[9]

The chest radiograph (CXR) is the most commonly used diagnostic imaging modality. Nevertheless, poor agreement exists among radiologists in the interpretation of chest radiographs for the diagnosis of pneumonia in both adults and children.[10, 11, 12, 13, 14, 15] Recent studies have found a high degree of agreement among pediatric radiologists with implementation of the World Health Organization (WHO) criteria for standardized CXR interpretation for diagnosis of bacterial pneumonia in children.[16, 17, 18] In these studies, participants were blinded to the clinical presentation. Data investigating the impact of clinical history on CXR interpretation in the pediatric population are limited.[19]

We conducted this prospective case‐based study to evaluate the impact of clinical information on the reliability of radiographic diagnosis of pneumonia among children presenting to a pediatric emergency department (ED) with clinical suspicion of pneumonia.

METHODS

Study Subjects

Six board‐certified radiologists at 2 academic children's hospitals (Children's Hospital of Philadelphia [n = 3] and Boston Children's Hospital [n = 3]) interpreted the same 110 chest radiographs (100 original and 10 duplicates) on 2 separate occasions. Clinical information was withheld during the first interpretation. The inter‐ and inter‐rater reliability for the interpretation of these 110 radiographs without clinical information have been previously reported.[18] After a period of 6 months, the radiologists reviewed the radiographs with access to clinical information provided by the physician ordering the CXR. This clinical information included age, sex, clinical indication for obtaining the radiograph, relevant history, and physical examination findings. The radiologists did not have access to the patients' medical records. The radiologists varied with respect to the number of years practicing pediatric radiology (median, 8 years; range, 336 years).

Radiographs were selected from children who presented to the ED at Boston Children's Hospital with concern of pneumonia. We selected radiographs with a spectrum of respiratory disease processes encountered in a pediatric population. The final radiographs included 50 radiographs with a final reading in the medical record without suspicion for pneumonia and 50 radiographs with suspicion of pneumonia. In the latter group, 25 radiographs had a final reading suggestive of an alveolar infiltrate, and 25 radiographs had a final reading suggestive of an interstitial infiltrate. Ten duplicate radiographs were included.

Radiograph Interpretation

The radiologists interpreted both anterior‐posterior and lateral views for each subject. Digital Imaging and Communications in Medicine images were downloaded from a registry at Boston Children's Hospital, and were copied to DVDs that were provided to each radiologist. Standardized radiographic imaging software (eFilm Lite; Merge Healthcare, Chicago, Illinois) was used by each radiologist.

Each radiologist completed a study questionnaire for each radiograph (see Supporting Information, Appendix 1, in the online version of this article). The questionnaire utilized radiographic descriptors of primary endpoint pneumonia described by the WHO to standardize the radiographic diagnosis of pneumonia.[20, 21] No additional training was provided to the radiologists. The main outcome of interest was the presence or absence of an infiltrate. Among radiographs in which an infiltrate was identified, radiologists selected whether there was an alveolar infiltrate, interstitial infiltrate, or both. Alveolar infiltrate and interstitial infiltrate are defined on the study questionnaire (Appendix 1). A radiograph classified as having either an alveolar infiltrate or interstitial infiltrate (not atelectasis) was considered to have any infiltrate. Additional findings including air bronchograms, hilar adenopathy, pleural effusion, and location of abnormalities were also recorded.

Statistical Analysis

Inter‐rater reliability was assessed using the kappa statistic to determine the overall agreement among the 6 radiologists for each outcome (eg, presence or absence of alveolar infiltrate). The kappa statistic for more than 2 raters utilizes an analysis of variance approach.[22] To calculate 95% confidence intervals (CI) for kappa statistics with more than 2 raters, we employed a bootstrapping method with 1000 replications of samples equal in size to the study sample. Intra‐rater reliability was evaluated by examining the agreement within each radiologist upon review of 10 duplicate radiographs. We used the following benchmarks to classify the strength of agreement: poor (<0.0), slight (00.20), fair (0.210.40), moderate (0.410.60), substantial (0.610.80), almost perfect (0.811.0).[23] Negative kappa values represent agreement less than would be predicted by chance alone.[24, 25] To calculate the kappa, a value must be recorded in 3 of 4 of the following categories: negative to positive, positive to negative, concordant negative, and concordant positive reporting of pneumonia. If raters did not fulfill 3 categories, the kappa could not be calculated.

The inter‐rater concordance for identification of an alveolar infiltrate was calculated for each radiologist by comparing their reporting of alveolar infiltrate with and without clinical history for each of the 100 radiographs. Radiographs that were identified by an individual rater as no alveolar infiltrate when read without clinical history, but those subsequently identified as alveolar infiltrate with clinical history were categorized as negative to positive reporting of pneumonia with clinical history. Those that were identified as alveolar infiltrate but subsequently identified as no alveolar infiltrate were categorized as positive to negative reporting of pneumonia with clinical history. Those radiographs in which there was no change in identification of alveolar infiltrate with clinical information were categorized as concordant reporting of pneumonia.

The study was approved by the institutional review boards at both children's hospitals.

RESULTS

Patient Sample

The radiographs were from patients ranging in age from 1 week to 19 years (median, 3.5 years; interquartile range, 1.66.0 years). Fifty (50%) patients were male.

Inter‐rater Reliability

The kappa coefficients of inter‐rater reliability between the radiologists across the 6 clinical measures of interest with and without access to clinical history are plotted in Figure 1. Reliability improved from fair (k = 0.32, 95% CI: 0.24 to 0.42) to moderate (k = 0.53, 95% CI: 0.43 to 0.64) for identification of air bronchograms with the addition of clinical history. Although there was an increase in kappa values for identification of any infiltrate, alveolar infiltrate, interstitial infiltrate, and pleural effusion, and a decrease in the kappa value for identification of hilar adenopathy with the addition of clinical information, there was substantial overlap of the 95% CIs, suggesting that inclusion of clinical history did not result in a statistically significant change in the reliability of these findings.

jhm1991-fig-0001-m.png
Inter‐rater reliability of radiologists (n = 6) evaluating chest radiographs with and without access to clinical history data in children presenting to the emergency department with suspected pneumonia (n = 100).

Intra‐rater Reliability

The estimates of inter‐rater reliability for the interpretation of the 10 duplicate images with and without clinical history are shown in Table 1. The inter‐rater reliability in the identification of alveolar infiltrate remained substantial to almost perfect for each rater with and without access to clinical history. Rater 1 had a decrease in inter‐rater reliability from almost perfect (k = 1.0, 95% CI: 1.0 to 1.0) to fair (k = 0.21, 95% CI: 0.43 to 0.85) in the identification of interstitial infiltrate with the addition of clinical history. This rater also had a decrease in agreement from almost perfect (k = 1.0, 95% CI: 1.0 to 1.0) to fair (k = 0.4, 95% CI: 0.16 to 0.96) in the identification of any infiltrate.

Intra‐rater Reliability of Radiologists With and Without Access to Clinical History While Evaluating Chest Radiographs (n = 10) for Pneumonia in Children
 Phase 1No Clinical HistoryPhase 2Access to Clinical History
Kappa95% Confidence IntervalKappa95% Confidence Interval
  • NOTE: Abbreviations: N/A, not applicable.
  • Too few categories of agreement to calculate kappa. Both responses are negative for all 10 paired radiographs; kappa cannot be calculated.
Any infiltrate    
Rater 11.001.00 to 1.000.400.16 to 0.96
Rater 20.600.10 to 1.000.580.07 to 1.00
Rater 30.800.44 to 1.000.800.44 to 1.00
Rater 41.001.00 to 1.000.780.39 to 1.00
Rater 5N/Aa 0.110.36 to 0.14
Rater 61.001.00 to 1.001.001.00 to 1.00
Alveolar infiltrate    
Rater 11.001.00 to 1.001.001.00 to 1.00
Rater 21.001.00 to 1.001.001.00 to 1.00
Rater 31.001.00 to 1.001.001.00 to 1.00
Rater 41.001.00 to 1.000.780.39 to 1.00
Rater 50.780.39 to 1.001.001.00 to 1.00
Rater 60.740.27 to 1.000.780.39 to 1.00
Interstitial infiltrate    
Rater 11.001.00 to 1.000.210.43 to 0.85
Rater 20.210.43 to 0.850.110.36 to 0.14
Rater 30.740.27 to 1.000.780.39 to 1.00
Rater 4N/A N/A 
Rater 50.580.07 to 1.000.520.05 to 1.00
Rater 60.620.5 to 1.00N/Aa 

Intra‐rater Concordance

The inter‐rater concordance of the radiologists for the identification of alveolar infiltrate during the interpretation of the 100 chest radiographs with and without access to clinical history is shown in Figure 2. The availability of clinical information impacted physicians differently in the evaluation of alveolar infiltrates. Raters 1, 4, and 6 appeared more likely to identify an alveolar infiltrate with access to the clinical information, whereas raters 3 and 5 appeared less likely to identify an alveolar infiltrate. Of the 100 films that were interpreted with and without clinical information, the mean number of discordant interpretations per rater was 10, with values ranging from 6 to 19 for the individual raters. Radiographs in which more than 3 raters changed their interpretation regarding the presence of an alveolar infiltrate are shown in Figure 3. For Figure 3D, 4 radiologists changed their interpretation from no alveolar infiltrate to alveolar infiltrate, and 1 radiologist changed from alveolar infiltrate to no alveolar infiltrate with the addition of clinical history.

jhm1991-fig-0002-m.png
Intra‐rater concordance of radiologists before and after access to clinical history while evaluating chest radiographs (n = 100) for alveolar infiltrate in children.
jhm1991-fig-0003-m.png
Chest radiographs of children in which 3 or more radiologists changed their interpretation in regard to the presence or absence of an alveolar infiltrate with the addition of clinical information. (A, B, and C) Three of 6 radiologists changed their interpretation. (D) Five of 6 radiologists changed their interpretation. (A) Female, 2 years old. (B) Male, 9 months old. (C) Male, 3 years old. (D) Male, 3 years old. The clinical history provided for (D) read as follows: “3‐year‐old male with cough and difficulty breathing. Rales at left base.”

Comment

We investigated the impact of the availability of clinical information on the reliability of chest radiographic interpretation in the diagnosis of pneumonia. There was improved inter‐rater reliability in the identification of air bronchograms with the addition of clinical information; however, clinical history did not have a substantial impact on the inter‐rater reliability of other findings. The addition of clinical information did not alter the inter‐rater reliability in the identification of alveolar infiltrate. Clinical history affected individual raters differently in their interpretation of alveolar infiltrate, with 3 raters more likely to identify an alveolar infiltrate and 2 raters less likely to identify an alveolar infiltrate.

Most studies addressing the impact of clinical history on radiographic interpretation evaluated accuracy. In many of these studies, accuracy was defined as the raters' agreement with the final interpretation of each film as documented in the medical record or their agreement with the interpretation of the radiologists selecting the cases.[1, 2, 3, 5, 6, 7] Given the known inter‐rater variability in radiographic interpretation,[10, 11, 12, 13, 14, 15] accuracy of a radiologist's interpretation cannot be appropriately assessed through agreement with their peers. Because a true measure of accuracy in the radiographic diagnosis of pneumonia can only be determined through invasive testing, such as lung biopsy, reliability serves as a more appropriate measure of performance. Inclusion of clinical information in chest radiograph interpretation has been shown to improve reliability in the radiographic diagnosis of a broad range of conditions.[15]

The primary outcome in this study was the identification of an infiltrate. Previous studies have noted consistent identification of the radiographic features that are most suggestive of bacterial pneumonia, such as alveolar infiltrate, and less consistent identification of other radiographic findings, including interstitial infiltrate.[18, 26, 27] Among the radiologists in this study, the addition of clinical information did not have a meaningful impact on the reliability of either of these findings, as there was substantial inter‐rater agreement for the identification of alveolar infiltrate and only slight agreement for the identification of interstitial infiltrate, both with and without clinical history. Additionally, inter‐rater reliability for the identification of alveolar infiltrate remained substantial to almost perfect for all 6 raters with the addition of clinical information.

Clinical information impacted the raters differently in their pattern of alveolar infiltrate identification, suggesting that radiologists may differ in their approach to incorporating clinical history in the interpretation of chest radiographs. The inclusion of clinical information may impact a radiologist's perception, leading to improved identification of abnormalities; however, it may also guide their decision making about the relevance of previously identified abnormalities.[28] Some radiologists may use clinical information to support or suggest possible radiographic findings, whereas others may use the information to challenge potential findings. This study did not address the manner in which the individual raters utilized the clinical history. There were also several radiographs in which the clinical information resulted in a change in the identification of an alveolar infiltrate by 3 or more raters, with as many as 5 of 6 raters changing their interpretation for 1 particular radiograph. These changes in identification of an infiltrate suggest that unidentified aspects of a history may be likely to influence a rater's interpretation of a radiograph. Nevertheless, these changes did not result in improved reliability and it is not possible to determine if these changes resulted in improved accuracy in interpretation.

This study had several limitations. First, radiographs were purposefully selected to encompass a broad spectrum of radiographic findings. Thus, the prevalence of pneumonia and other abnormal findings was artificially higher than typically observed among a cohort of children for whom pneumonia is considered. Second, the radiologists recruited for this study all practice in an academic children's hospital setting. These factors may limit the generalizability of our findings. However, we would expect these results to be generalizable to pediatric radiologists from other academic institutions. Third, this study does not meet the criteria of a balanced study design as defined by Loy and Irwig.[19] A study was characterized as balanced if half of the radiographs were read with and half without clinical information in each of the 2 reading sessions. The proposed benefit of such a design is to control for possible changes in ability or reporting practices of the raters that may have occurred between study periods. The use of a standardized reporting tool likely minimized changes in reporting practices. Also, it is unlikely that the ability or reporting practices of an experienced radiologist would change over the study period. Fourth, the radiologists interpreted the films outside of their standard workflow and utilized a standardized reporting tool that focused on the presence or absence of pneumonia indicators. These factors may have increased the radiologists' suspicion for pneumonia even in the absence of clinical information. This may have biased the results toward finding no difference in the identification of pneumonia with the addition of detailed clinical history. Thus, the inclusion of clinical information in radiograph interpretation in clinical practice may have greater impact on the identification of these pneumonia indicators than was found in this study.[29] Finally, reliability does not imply accuracy, and it is unknown if changes in the identification of pneumonia indicators led to more accurate interpretation with respect to the clinical or pathologic diagnosis of pneumonia.

In conclusion, we observed high intra‐ and inter‐rater reliability among radiologists in the identification of an alveolar infiltrate, the radiographic finding most suggestive of bacterial pneumonia.[16, 17, 18, 30] The addition of clinical information did not have a substantial impact on the reliability of its identification.

The inclusion of clinical information in diagnostic testing may influence the interpretation of the clinical findings. Historical and clinical findings may focus the reader's attention to the relevant details, thereby improving the accuracy of the interpretation. However, such information may cause the reader to have preconceived notions about the results, biasing the overall interpretation.

The impact of clinical information on the interpretation of radiographic studies remains an issue of debate. Previous studies have found that clinical information improves the accuracy of radiographic interpretation for a broad range of diagnoses,[1, 2, 3, 4] whereas others do not show improvement.[5, 6, 7] Additionally, clinical information may serve as a distraction that leads to more false‐positive interpretations.[8] For this reason, many radiologists prefer to review radiographs without knowledge of the clinical scenario prompting the study to avoid focusing on the expected findings and potentially missing other important abnormalities.[9]

The chest radiograph (CXR) is the most commonly used diagnostic imaging modality. Nevertheless, poor agreement exists among radiologists in the interpretation of chest radiographs for the diagnosis of pneumonia in both adults and children.[10, 11, 12, 13, 14, 15] Recent studies have found a high degree of agreement among pediatric radiologists with implementation of the World Health Organization (WHO) criteria for standardized CXR interpretation for diagnosis of bacterial pneumonia in children.[16, 17, 18] In these studies, participants were blinded to the clinical presentation. Data investigating the impact of clinical history on CXR interpretation in the pediatric population are limited.[19]

We conducted this prospective case‐based study to evaluate the impact of clinical information on the reliability of radiographic diagnosis of pneumonia among children presenting to a pediatric emergency department (ED) with clinical suspicion of pneumonia.

METHODS

Study Subjects

Six board‐certified radiologists at 2 academic children's hospitals (Children's Hospital of Philadelphia [n = 3] and Boston Children's Hospital [n = 3]) interpreted the same 110 chest radiographs (100 original and 10 duplicates) on 2 separate occasions. Clinical information was withheld during the first interpretation. The inter‐ and inter‐rater reliability for the interpretation of these 110 radiographs without clinical information have been previously reported.[18] After a period of 6 months, the radiologists reviewed the radiographs with access to clinical information provided by the physician ordering the CXR. This clinical information included age, sex, clinical indication for obtaining the radiograph, relevant history, and physical examination findings. The radiologists did not have access to the patients' medical records. The radiologists varied with respect to the number of years practicing pediatric radiology (median, 8 years; range, 336 years).

Radiographs were selected from children who presented to the ED at Boston Children's Hospital with concern of pneumonia. We selected radiographs with a spectrum of respiratory disease processes encountered in a pediatric population. The final radiographs included 50 radiographs with a final reading in the medical record without suspicion for pneumonia and 50 radiographs with suspicion of pneumonia. In the latter group, 25 radiographs had a final reading suggestive of an alveolar infiltrate, and 25 radiographs had a final reading suggestive of an interstitial infiltrate. Ten duplicate radiographs were included.

Radiograph Interpretation

The radiologists interpreted both anterior‐posterior and lateral views for each subject. Digital Imaging and Communications in Medicine images were downloaded from a registry at Boston Children's Hospital, and were copied to DVDs that were provided to each radiologist. Standardized radiographic imaging software (eFilm Lite; Merge Healthcare, Chicago, Illinois) was used by each radiologist.

Each radiologist completed a study questionnaire for each radiograph (see Supporting Information, Appendix 1, in the online version of this article). The questionnaire utilized radiographic descriptors of primary endpoint pneumonia described by the WHO to standardize the radiographic diagnosis of pneumonia.[20, 21] No additional training was provided to the radiologists. The main outcome of interest was the presence or absence of an infiltrate. Among radiographs in which an infiltrate was identified, radiologists selected whether there was an alveolar infiltrate, interstitial infiltrate, or both. Alveolar infiltrate and interstitial infiltrate are defined on the study questionnaire (Appendix 1). A radiograph classified as having either an alveolar infiltrate or interstitial infiltrate (not atelectasis) was considered to have any infiltrate. Additional findings including air bronchograms, hilar adenopathy, pleural effusion, and location of abnormalities were also recorded.

Statistical Analysis

Inter‐rater reliability was assessed using the kappa statistic to determine the overall agreement among the 6 radiologists for each outcome (eg, presence or absence of alveolar infiltrate). The kappa statistic for more than 2 raters utilizes an analysis of variance approach.[22] To calculate 95% confidence intervals (CI) for kappa statistics with more than 2 raters, we employed a bootstrapping method with 1000 replications of samples equal in size to the study sample. Intra‐rater reliability was evaluated by examining the agreement within each radiologist upon review of 10 duplicate radiographs. We used the following benchmarks to classify the strength of agreement: poor (<0.0), slight (00.20), fair (0.210.40), moderate (0.410.60), substantial (0.610.80), almost perfect (0.811.0).[23] Negative kappa values represent agreement less than would be predicted by chance alone.[24, 25] To calculate the kappa, a value must be recorded in 3 of 4 of the following categories: negative to positive, positive to negative, concordant negative, and concordant positive reporting of pneumonia. If raters did not fulfill 3 categories, the kappa could not be calculated.

The inter‐rater concordance for identification of an alveolar infiltrate was calculated for each radiologist by comparing their reporting of alveolar infiltrate with and without clinical history for each of the 100 radiographs. Radiographs that were identified by an individual rater as no alveolar infiltrate when read without clinical history, but those subsequently identified as alveolar infiltrate with clinical history were categorized as negative to positive reporting of pneumonia with clinical history. Those that were identified as alveolar infiltrate but subsequently identified as no alveolar infiltrate were categorized as positive to negative reporting of pneumonia with clinical history. Those radiographs in which there was no change in identification of alveolar infiltrate with clinical information were categorized as concordant reporting of pneumonia.

The study was approved by the institutional review boards at both children's hospitals.

RESULTS

Patient Sample

The radiographs were from patients ranging in age from 1 week to 19 years (median, 3.5 years; interquartile range, 1.66.0 years). Fifty (50%) patients were male.

Inter‐rater Reliability

The kappa coefficients of inter‐rater reliability between the radiologists across the 6 clinical measures of interest with and without access to clinical history are plotted in Figure 1. Reliability improved from fair (k = 0.32, 95% CI: 0.24 to 0.42) to moderate (k = 0.53, 95% CI: 0.43 to 0.64) for identification of air bronchograms with the addition of clinical history. Although there was an increase in kappa values for identification of any infiltrate, alveolar infiltrate, interstitial infiltrate, and pleural effusion, and a decrease in the kappa value for identification of hilar adenopathy with the addition of clinical information, there was substantial overlap of the 95% CIs, suggesting that inclusion of clinical history did not result in a statistically significant change in the reliability of these findings.

jhm1991-fig-0001-m.png
Inter‐rater reliability of radiologists (n = 6) evaluating chest radiographs with and without access to clinical history data in children presenting to the emergency department with suspected pneumonia (n = 100).

Intra‐rater Reliability

The estimates of inter‐rater reliability for the interpretation of the 10 duplicate images with and without clinical history are shown in Table 1. The inter‐rater reliability in the identification of alveolar infiltrate remained substantial to almost perfect for each rater with and without access to clinical history. Rater 1 had a decrease in inter‐rater reliability from almost perfect (k = 1.0, 95% CI: 1.0 to 1.0) to fair (k = 0.21, 95% CI: 0.43 to 0.85) in the identification of interstitial infiltrate with the addition of clinical history. This rater also had a decrease in agreement from almost perfect (k = 1.0, 95% CI: 1.0 to 1.0) to fair (k = 0.4, 95% CI: 0.16 to 0.96) in the identification of any infiltrate.

Intra‐rater Reliability of Radiologists With and Without Access to Clinical History While Evaluating Chest Radiographs (n = 10) for Pneumonia in Children
 Phase 1No Clinical HistoryPhase 2Access to Clinical History
Kappa95% Confidence IntervalKappa95% Confidence Interval
  • NOTE: Abbreviations: N/A, not applicable.
  • Too few categories of agreement to calculate kappa. Both responses are negative for all 10 paired radiographs; kappa cannot be calculated.
Any infiltrate    
Rater 11.001.00 to 1.000.400.16 to 0.96
Rater 20.600.10 to 1.000.580.07 to 1.00
Rater 30.800.44 to 1.000.800.44 to 1.00
Rater 41.001.00 to 1.000.780.39 to 1.00
Rater 5N/Aa 0.110.36 to 0.14
Rater 61.001.00 to 1.001.001.00 to 1.00
Alveolar infiltrate    
Rater 11.001.00 to 1.001.001.00 to 1.00
Rater 21.001.00 to 1.001.001.00 to 1.00
Rater 31.001.00 to 1.001.001.00 to 1.00
Rater 41.001.00 to 1.000.780.39 to 1.00
Rater 50.780.39 to 1.001.001.00 to 1.00
Rater 60.740.27 to 1.000.780.39 to 1.00
Interstitial infiltrate    
Rater 11.001.00 to 1.000.210.43 to 0.85
Rater 20.210.43 to 0.850.110.36 to 0.14
Rater 30.740.27 to 1.000.780.39 to 1.00
Rater 4N/A N/A 
Rater 50.580.07 to 1.000.520.05 to 1.00
Rater 60.620.5 to 1.00N/Aa 

Intra‐rater Concordance

The inter‐rater concordance of the radiologists for the identification of alveolar infiltrate during the interpretation of the 100 chest radiographs with and without access to clinical history is shown in Figure 2. The availability of clinical information impacted physicians differently in the evaluation of alveolar infiltrates. Raters 1, 4, and 6 appeared more likely to identify an alveolar infiltrate with access to the clinical information, whereas raters 3 and 5 appeared less likely to identify an alveolar infiltrate. Of the 100 films that were interpreted with and without clinical information, the mean number of discordant interpretations per rater was 10, with values ranging from 6 to 19 for the individual raters. Radiographs in which more than 3 raters changed their interpretation regarding the presence of an alveolar infiltrate are shown in Figure 3. For Figure 3D, 4 radiologists changed their interpretation from no alveolar infiltrate to alveolar infiltrate, and 1 radiologist changed from alveolar infiltrate to no alveolar infiltrate with the addition of clinical history.

jhm1991-fig-0002-m.png
Intra‐rater concordance of radiologists before and after access to clinical history while evaluating chest radiographs (n = 100) for alveolar infiltrate in children.
jhm1991-fig-0003-m.png
Chest radiographs of children in which 3 or more radiologists changed their interpretation in regard to the presence or absence of an alveolar infiltrate with the addition of clinical information. (A, B, and C) Three of 6 radiologists changed their interpretation. (D) Five of 6 radiologists changed their interpretation. (A) Female, 2 years old. (B) Male, 9 months old. (C) Male, 3 years old. (D) Male, 3 years old. The clinical history provided for (D) read as follows: “3‐year‐old male with cough and difficulty breathing. Rales at left base.”

Comment

We investigated the impact of the availability of clinical information on the reliability of chest radiographic interpretation in the diagnosis of pneumonia. There was improved inter‐rater reliability in the identification of air bronchograms with the addition of clinical information; however, clinical history did not have a substantial impact on the inter‐rater reliability of other findings. The addition of clinical information did not alter the inter‐rater reliability in the identification of alveolar infiltrate. Clinical history affected individual raters differently in their interpretation of alveolar infiltrate, with 3 raters more likely to identify an alveolar infiltrate and 2 raters less likely to identify an alveolar infiltrate.

Most studies addressing the impact of clinical history on radiographic interpretation evaluated accuracy. In many of these studies, accuracy was defined as the raters' agreement with the final interpretation of each film as documented in the medical record or their agreement with the interpretation of the radiologists selecting the cases.[1, 2, 3, 5, 6, 7] Given the known inter‐rater variability in radiographic interpretation,[10, 11, 12, 13, 14, 15] accuracy of a radiologist's interpretation cannot be appropriately assessed through agreement with their peers. Because a true measure of accuracy in the radiographic diagnosis of pneumonia can only be determined through invasive testing, such as lung biopsy, reliability serves as a more appropriate measure of performance. Inclusion of clinical information in chest radiograph interpretation has been shown to improve reliability in the radiographic diagnosis of a broad range of conditions.[15]

The primary outcome in this study was the identification of an infiltrate. Previous studies have noted consistent identification of the radiographic features that are most suggestive of bacterial pneumonia, such as alveolar infiltrate, and less consistent identification of other radiographic findings, including interstitial infiltrate.[18, 26, 27] Among the radiologists in this study, the addition of clinical information did not have a meaningful impact on the reliability of either of these findings, as there was substantial inter‐rater agreement for the identification of alveolar infiltrate and only slight agreement for the identification of interstitial infiltrate, both with and without clinical history. Additionally, inter‐rater reliability for the identification of alveolar infiltrate remained substantial to almost perfect for all 6 raters with the addition of clinical information.

Clinical information impacted the raters differently in their pattern of alveolar infiltrate identification, suggesting that radiologists may differ in their approach to incorporating clinical history in the interpretation of chest radiographs. The inclusion of clinical information may impact a radiologist's perception, leading to improved identification of abnormalities; however, it may also guide their decision making about the relevance of previously identified abnormalities.[28] Some radiologists may use clinical information to support or suggest possible radiographic findings, whereas others may use the information to challenge potential findings. This study did not address the manner in which the individual raters utilized the clinical history. There were also several radiographs in which the clinical information resulted in a change in the identification of an alveolar infiltrate by 3 or more raters, with as many as 5 of 6 raters changing their interpretation for 1 particular radiograph. These changes in identification of an infiltrate suggest that unidentified aspects of a history may be likely to influence a rater's interpretation of a radiograph. Nevertheless, these changes did not result in improved reliability and it is not possible to determine if these changes resulted in improved accuracy in interpretation.

This study had several limitations. First, radiographs were purposefully selected to encompass a broad spectrum of radiographic findings. Thus, the prevalence of pneumonia and other abnormal findings was artificially higher than typically observed among a cohort of children for whom pneumonia is considered. Second, the radiologists recruited for this study all practice in an academic children's hospital setting. These factors may limit the generalizability of our findings. However, we would expect these results to be generalizable to pediatric radiologists from other academic institutions. Third, this study does not meet the criteria of a balanced study design as defined by Loy and Irwig.[19] A study was characterized as balanced if half of the radiographs were read with and half without clinical information in each of the 2 reading sessions. The proposed benefit of such a design is to control for possible changes in ability or reporting practices of the raters that may have occurred between study periods. The use of a standardized reporting tool likely minimized changes in reporting practices. Also, it is unlikely that the ability or reporting practices of an experienced radiologist would change over the study period. Fourth, the radiologists interpreted the films outside of their standard workflow and utilized a standardized reporting tool that focused on the presence or absence of pneumonia indicators. These factors may have increased the radiologists' suspicion for pneumonia even in the absence of clinical information. This may have biased the results toward finding no difference in the identification of pneumonia with the addition of detailed clinical history. Thus, the inclusion of clinical information in radiograph interpretation in clinical practice may have greater impact on the identification of these pneumonia indicators than was found in this study.[29] Finally, reliability does not imply accuracy, and it is unknown if changes in the identification of pneumonia indicators led to more accurate interpretation with respect to the clinical or pathologic diagnosis of pneumonia.

In conclusion, we observed high intra‐ and inter‐rater reliability among radiologists in the identification of an alveolar infiltrate, the radiographic finding most suggestive of bacterial pneumonia.[16, 17, 18, 30] The addition of clinical information did not have a substantial impact on the reliability of its identification.

References
  1. Berbaum KS, Franken EA, Dorfman DD, et al. Tentative diagnoses facilitate the detection of diverse lesions in chest radiographs. Invest Radiol. 1986;21(7):532539.
  2. Berbaum KS, Franken EA, Dorfman DD, Barloon TJ. Influence of clinical history upon detection of nodules and other lesions. Invest Radiol. 1988;23(1):4855.
  3. Berbaum KS, Franken EA, Dorfman DD, Lueben KR. Influence of clinical history on perception of abnormalities in pediatric radiographs. Acad Radiol. 1994;1(3):217223.
  4. Song KS, Song HH, Park SH, et al. Impact of clinical history on film interpretation. Yonsei Med J. 1992;33(2):168172.
  5. Cooperstein LA, Good BC, Eelkema EA, et al. The effect of clinical history on chest radiograph interpretations in a PACS environment. Invest Radiol. 1990;25(6):670674.
  6. Good BC, Cooperstein LA, DeMarino GB, et al. Does knowledge of the clinical history affect the accuracy of chest radiograph interpretation?AJR Am J Roentgenol. 1990;154(4):709712.
  7. Quekel LG, Goei R, Kessels AG, Engelshoven JM. Detection of lung cancer on the chest radiograph: impact of previous films, clinical information, double reading, and dual reading. J Clin Epidemiol. 2001;54(11):11461150.
  8. Eldevik OP, Dugstad G, Orrison WW, Haughton VM. The effect of clinical bias on the interpretation of myelography and spinal computed tomography. Radiology. 1982;145(1):8589.
  9. Griscom NT. A suggestion: look at the images first, before you read the history. Radiology. 2002;223(1):910.
  10. Albaum MN, Hill LC, Murphy M, et al. Interobserver reliability of the chest radiograph in community‐acquired pneumonia. PORT Investigators. Chest. 1996;110(2):343350.
  11. Bloomfield FH, Teele RL, Voss M, Knight DB, Harding JE. Inter‐ and intra‐observer variability in the assessment of atelectasis and consolidation in neonatal chest radiographs. Pediatr Radiol. 1999;29(6):459462.
  12. Gatt ME, Spectre G, Paltiel O, Hiller N, Stalnikowicz R. Chest radiographs in the emergency department: is the radiologist really necessary?Postgrad Med J. 2003;79(930):214217.
  13. Hopstaken RM, Witbraad T, Engelshoven JM, Dinant GJ. Inter‐observer variation in the interpretation of chest radiographs for pneumonia in community‐acquired lower respiratory tract infections. Clin Radiol. 2004;59(8):743752.
  14. Novack V, Avnon LS, Smolyakov A, Barnea R, Jotkowitz A, Schlaeffer F. Disagreement in the interpretation of chest radiographs among specialists and clinical outcomes of patients hospitalized with suspected pneumonia. Eur J Intern Med. 2006;17(1):4347.
  15. Tudor GR, Finlay D, Taub N. An assessment of inter‐observer agreement and accuracy when reporting plain radiographs. Clin Radiol. 1997;52(3):235238.
  16. Shimol BS, Dagan R, Givon‐Lavi N, et al. Evaluation of the World Health Organization criteria for chest radiographs for pneumonia diagnosis in children. Eur J Pediatr. 2011;171(2):369374.
  17. Cherian T, Mulholland EK, Carlin JB, et al. Standardized interpretation of paediatric chest radiographs for the diagnosis of pneumonia in epidemiological studies. Bull World Health Organ. 2005;83(5):353359.
  18. Neuman MI, Lee EY, Bixby S, et al. Variability in the interpretation of chest radiographs for the diagnosis of pneumonia in children. J Hosp Med. 2012;7(4):294298.
  19. Loy CT, Irwig L. Accuracy of diagnostic tests read with and without clinical information: a systematic review. JAMA. 2004;292(13):16021609.
  20. Standardization of interpretation of chest radiographs for the diagnosis of pneumonia in children. In:World Health Organization: Pneumonia Vaccine Trial Investigators' Group.Geneva: Department of Vaccine and Biologics;2001.
  21. Hansen J, Black S, Shinefield H, et al. Effectiveness of heptavalent pneumococcal conjugate vaccine in children younger than 5 years of age for prevention of pneumonia: updated analysis using World Health Organization standardized interpretation of chest radiographs. Pediatr Infect Dis J. 2006;25(9):779781.
  22. Landis JR, Koch GG. A one‐way components of variance model for categorical data. Biometrics. 1977;33:671679.
  23. Landis JR, Koch GG. The measurement of observer agreement for categorical data. Biometrics. 1977;33(1):159174.
  24. Juurlink DN, Detsky AS. Kappa statistic. CMAJ. 2005;173(1):16.
  25. Kramer MS, Feinstein AR. Clinical biostatistics. LIV. The biostatistics of concordance. Clin Pharmacol Ther. 1981;29(1):111123.
  26. Bartlett JG, Dowell SF, Mandell LA, File TM, Musher DM, Fine MJ. Practice guidelines for the management of community‐acquired pneumonia in adults. Infectious Diseases Society of America. Clin Infect Dis. 2000;31(2):347382.
  27. Niederman MS, Mandell LA, Anzueto A, et al. Guidelines for the management of adults with community‐acquired pneumonia. Diagnosis, assessment of severity, antimicrobial therapy, and prevention. Am J Respir Crit Care Med. 2001;163(7):17301754.
  28. Berbaum KS, Franken EA. Commentary does clinical history affect perception?Acad Radiol. 2006;13(3):402403.
  29. Berbaum KS, Franken EA. The effect of clinical history on chest radiograph interpretations in a PACS environment. Invest Radiol. 1991;26(5):512514.
  30. Korppi M, Kiekara O, Heiskanen‐Kosma T, Soimakallio S. Comparison of radiological findings and microbial aetiology of childhood pneumonia. Acta Paediatr. 1993;82(4):360363.
References
  1. Berbaum KS, Franken EA, Dorfman DD, et al. Tentative diagnoses facilitate the detection of diverse lesions in chest radiographs. Invest Radiol. 1986;21(7):532539.
  2. Berbaum KS, Franken EA, Dorfman DD, Barloon TJ. Influence of clinical history upon detection of nodules and other lesions. Invest Radiol. 1988;23(1):4855.
  3. Berbaum KS, Franken EA, Dorfman DD, Lueben KR. Influence of clinical history on perception of abnormalities in pediatric radiographs. Acad Radiol. 1994;1(3):217223.
  4. Song KS, Song HH, Park SH, et al. Impact of clinical history on film interpretation. Yonsei Med J. 1992;33(2):168172.
  5. Cooperstein LA, Good BC, Eelkema EA, et al. The effect of clinical history on chest radiograph interpretations in a PACS environment. Invest Radiol. 1990;25(6):670674.
  6. Good BC, Cooperstein LA, DeMarino GB, et al. Does knowledge of the clinical history affect the accuracy of chest radiograph interpretation?AJR Am J Roentgenol. 1990;154(4):709712.
  7. Quekel LG, Goei R, Kessels AG, Engelshoven JM. Detection of lung cancer on the chest radiograph: impact of previous films, clinical information, double reading, and dual reading. J Clin Epidemiol. 2001;54(11):11461150.
  8. Eldevik OP, Dugstad G, Orrison WW, Haughton VM. The effect of clinical bias on the interpretation of myelography and spinal computed tomography. Radiology. 1982;145(1):8589.
  9. Griscom NT. A suggestion: look at the images first, before you read the history. Radiology. 2002;223(1):910.
  10. Albaum MN, Hill LC, Murphy M, et al. Interobserver reliability of the chest radiograph in community‐acquired pneumonia. PORT Investigators. Chest. 1996;110(2):343350.
  11. Bloomfield FH, Teele RL, Voss M, Knight DB, Harding JE. Inter‐ and intra‐observer variability in the assessment of atelectasis and consolidation in neonatal chest radiographs. Pediatr Radiol. 1999;29(6):459462.
  12. Gatt ME, Spectre G, Paltiel O, Hiller N, Stalnikowicz R. Chest radiographs in the emergency department: is the radiologist really necessary?Postgrad Med J. 2003;79(930):214217.
  13. Hopstaken RM, Witbraad T, Engelshoven JM, Dinant GJ. Inter‐observer variation in the interpretation of chest radiographs for pneumonia in community‐acquired lower respiratory tract infections. Clin Radiol. 2004;59(8):743752.
  14. Novack V, Avnon LS, Smolyakov A, Barnea R, Jotkowitz A, Schlaeffer F. Disagreement in the interpretation of chest radiographs among specialists and clinical outcomes of patients hospitalized with suspected pneumonia. Eur J Intern Med. 2006;17(1):4347.
  15. Tudor GR, Finlay D, Taub N. An assessment of inter‐observer agreement and accuracy when reporting plain radiographs. Clin Radiol. 1997;52(3):235238.
  16. Shimol BS, Dagan R, Givon‐Lavi N, et al. Evaluation of the World Health Organization criteria for chest radiographs for pneumonia diagnosis in children. Eur J Pediatr. 2011;171(2):369374.
  17. Cherian T, Mulholland EK, Carlin JB, et al. Standardized interpretation of paediatric chest radiographs for the diagnosis of pneumonia in epidemiological studies. Bull World Health Organ. 2005;83(5):353359.
  18. Neuman MI, Lee EY, Bixby S, et al. Variability in the interpretation of chest radiographs for the diagnosis of pneumonia in children. J Hosp Med. 2012;7(4):294298.
  19. Loy CT, Irwig L. Accuracy of diagnostic tests read with and without clinical information: a systematic review. JAMA. 2004;292(13):16021609.
  20. Standardization of interpretation of chest radiographs for the diagnosis of pneumonia in children. In:World Health Organization: Pneumonia Vaccine Trial Investigators' Group.Geneva: Department of Vaccine and Biologics;2001.
  21. Hansen J, Black S, Shinefield H, et al. Effectiveness of heptavalent pneumococcal conjugate vaccine in children younger than 5 years of age for prevention of pneumonia: updated analysis using World Health Organization standardized interpretation of chest radiographs. Pediatr Infect Dis J. 2006;25(9):779781.
  22. Landis JR, Koch GG. A one‐way components of variance model for categorical data. Biometrics. 1977;33:671679.
  23. Landis JR, Koch GG. The measurement of observer agreement for categorical data. Biometrics. 1977;33(1):159174.
  24. Juurlink DN, Detsky AS. Kappa statistic. CMAJ. 2005;173(1):16.
  25. Kramer MS, Feinstein AR. Clinical biostatistics. LIV. The biostatistics of concordance. Clin Pharmacol Ther. 1981;29(1):111123.
  26. Bartlett JG, Dowell SF, Mandell LA, File TM, Musher DM, Fine MJ. Practice guidelines for the management of community‐acquired pneumonia in adults. Infectious Diseases Society of America. Clin Infect Dis. 2000;31(2):347382.
  27. Niederman MS, Mandell LA, Anzueto A, et al. Guidelines for the management of adults with community‐acquired pneumonia. Diagnosis, assessment of severity, antimicrobial therapy, and prevention. Am J Respir Crit Care Med. 2001;163(7):17301754.
  28. Berbaum KS, Franken EA. Commentary does clinical history affect perception?Acad Radiol. 2006;13(3):402403.
  29. Berbaum KS, Franken EA. The effect of clinical history on chest radiograph interpretations in a PACS environment. Invest Radiol. 1991;26(5):512514.
  30. Korppi M, Kiekara O, Heiskanen‐Kosma T, Soimakallio S. Comparison of radiological findings and microbial aetiology of childhood pneumonia. Acta Paediatr. 1993;82(4):360363.
Issue
Journal of Hospital Medicine - 8(7)
Issue
Journal of Hospital Medicine - 8(7)
Page Number
359-364
Page Number
359-364
Publications
Publications
Article Type
Display Headline
Impact of clinical history on chest radiograph interpretation
Display Headline
Impact of clinical history on chest radiograph interpretation
Sections
Article Source

© 2012 Society of Hospital Medicine

Disallow All Ads
Correspondence Location
Address for correspondence and reprint requests: Samir S. Shah, MD, 3333 Burnet Avenue, ML 9016, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229. E-mail: samir.shah@cchmc.org
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Article PDF Media
Image
Disable zoom
Off
Media Files
Image
Disable zoom
Off

Discordant Antibiotics in Pediatric UTI

Article Type
Changed
Mon, 05/22/2017 - 18:33
Display Headline
Discordant antibiotic therapy and length of stay in children hospitalized for urinary tract infection

Urinary tract infections (UTIs) are one of the most common reasons for pediatric hospitalizations.1 Bacterial infections require prompt treatment with appropriate antimicrobial agents. Results from culture and susceptibility testing, however, are often unavailable until 48 hours after initial presentation. Therefore, the clinician must select antimicrobials empirically, basing decisions on likely pathogens and local resistance patterns.2 This decision is challenging because the effect of treatment delay on clinical outcomes is difficult to determine and resistance among uropathogens is increasing. Resistance rates have doubled over the past several years.3, 4 For common first‐line antibiotics, such as ampicillin and trimethoprim‐sulfamethoxazole, resistance rates for Escherichia coli, the most common uropathogen, exceed 25%.4, 5 While resistance to third‐generation cephalosporins remains low, rates in the United States have increased from <1% in 1999 to 4% in 2010. International data shows much higher resistance rates for cephalosporins in general.6, 7 This high prevalence of resistance may prompt the use of broad‐spectrum antibiotics for patients with UTI. For example, the use of third‐generation cephalosporins for UTI has doubled in recent years.3 Untreated, UTIs can lead to serious illness, but the consequences of inadequate initial antibiotic coverage are unknown.8, 9

Discordant antibiotic therapy, initial antibiotic therapy to which the causative bacterium is not susceptible, occurs in up to 9% of children hospitalized for UTI.10 However, there is reason to believe that discordant therapy may matter less for UTIs than for infections at other sites. First, in adults hospitalized with UTIs, discordant initial therapy did not affect the time to resolution of symptoms.11, 12 Second, most antibiotics used to treat UTIs are renally excreted and, thus, antibiotic concentrations at the site of infection are higher than can be achieved in the serum or cerebrospinal fluid.13 The Clinical and Laboratory Standard Institute has acknowledged that traditional susceptibility breakpoints may be too conservative for some non‐central nervous system infections; such as non‐central nervous system infections caused by Streptococcus pneumoniae.14

As resistance rates increase, more patients are likely to be treated with discordant therapy. Therefore, we sought to identify the clinical consequences of discordant antimicrobial therapy for patients hospitalized with a UTI.

METHODS

Design and Setting

We conducted a multicenter, retrospective cohort study. Data for this study were originally collected for a study that determined the accuracy of individual and combined International Classification of Diseases, Ninth Revision (ICD‐9) discharge diagnosis codes for children with laboratory tests for a UTI, in order to develop national quality measures for children hospitalized with UTIs.15 The institutional review board for each hospital (Seattle Children's Hospital, Seattle, WA; Monroe Carell Jr Children's Hospital at Vanderbilt, Nashville, TN; Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Children's Mercy Hospital, Kansas City, MO; Children's Hospital of Philadelphia, Philadelphia, PA) approved the study.

Data Sources

Data were obtained from the Pediatric Health Information System (PHIS) and medical records for patients at the 5 participating hospitals. PHIS contains clinical and billing data from hospitalized children at 43 freestanding children's hospitals. Data quality and coding reliability are assured through a joint effort between the Children's Hospital Association (Shawnee Mission, KS) and participating hospitals.16 PHIS was used to identify participants based on presence of discharge diagnosis code and laboratory tests indicating possible UTI, patient demographics, antibiotic administration date, and utilization of hospital resources (length of stay [LOS], laboratory testing).

Medical records for each participant were reviewed to obtain laboratory and clinical information such as past medical history (including vesicoureteral reflux [VUR], abnormal genitourinary [GU] anatomy, use of prophylactic antibiotic), culture data, and fever data. Data were entered into a secured centrally housed web‐based data collection system. To assure consistency of chart review, all investigators responsible for data collection underwent training. In addition, 2 pilot medical record reviews were performed, followed by group discussion, to reach consensus on questions, preselected answers, interpretation of medical record data, and parameters for free text data entry.

Subjects

The initial cohort included 460 hospitalized patients, aged 3 days to 18 years of age, discharged from participating hospitals between July 1, 2008 and June 30, 2009 with a positive urine culture at any time during hospitalization.15 We excluded patients under 3 days of age because patients this young are more likely to have been transferred from the birthing hospital for a complication related to birth or a congenital anomaly. For this secondary analysis of patients from a prior study, our target population included patients admitted for management of UTI.15 We excluded patients with a negative initial urine culture (n = 59) or if their initial urine culture did not meet definition of laboratory‐confirmed UTI, defined as urine culture with >50,000 colony‐forming units (CFU) with an abnormal urinalysis (UA) (n = 77).1, 1719 An abnormal UA was defined by presence of white blood cells, leukocyte esterase, bacteria, and/or nitrites. For our cohort, all cultures with >50,000 CFU also had an abnormal urinalysis. We excluded 19 patients with cultures classified as 10,000100,000 CFU because we could not confirm that the CFU was >50,000. We excluded 30 patients with urine cultures classified as normal or mixed flora, positive for a mixture of organisms not further identified, or if results were unavailable. Additionally, coagulase‐negative Staphylococcus species (n = 8) were excluded, as these are typically considered contaminants in the setting of urine cultures.2 Patients likely to have received antibiotics prior to admission, or develop a UTI after admission, were identified and removed from the cohort if they had a urine culture performed more than 1 day before, or 2 days after, admission (n = 35). Cultures without resistance testing to the initial antibiotic selection were also excluded (n = 16).

Main Outcome Measures

The primary outcome measure was hospital LOS. Time to fever resolution was a secondary outcome measure. Fever was defined as temperature 38C. Fever duration was defined as number of hours until resolution of fever; only patients with fever at admission were included in this subanalysis.

Main Exposure

The main exposure was initial antibiotic therapy. Patients were classified into 3 groups according to initial antibiotic selection: those receiving 1) concordant; 2) discordant; or 3) delayed initial therapy. Concordance was defined as in vitro susceptibility to the initial antibiotic or class of antibiotic. If the uropathogen was sensitive to a narrow‐spectrum antibiotic (eg, first‐generation cephalosporin), but was not tested against a more broad‐spectrum antibiotic of the same class (eg, third‐generation cephalosporin), concordance was based on the sensitivity to the narrow‐spectrum antibiotic. If the uropathogen was sensitive to a broad‐spectrum antibiotic (eg, third‐generation cephalosporin), concordance to a more narrow‐spectrum antibiotic was not assumed. Discordance was defined as laboratory confirmation of in vitro resistance, or intermediate sensitivity of the pathogen to the initial antibiotic or class of antibiotics. Patients were considered to have a delay in antibiotic therapy if they did not receive antibiotics on the day of, or day after, collection of UA and culture. Patients with more than 1 uropathogen identified in a single culture were classified as discordant if any of the organisms was discordant to the initial antibiotic; they were classified as concordant if all organisms were concordant to the initial antibiotic. Antibiotic susceptibility was not tested in some cases (n = 16).

Initial antibiotic was defined as the antibiotic(s) billed on the same day or day after the UA was billed. If the patient had the UA completed on the day prior to admission, we used the antibiotic administered on the day of admission as the initial antibiotic.

Covariates

Covariates were selected a priori to include patient characteristics likely to affect patient outcomes; all were included in the final analysis. These were age, race, sex, insurance, disposition, prophylactic antibiotic use for any reason (VUR, oncologic process, etc), presence of a chronic care condition, and presence of VUR or GU anatomic abnormality. Age, race, sex, and insurance were obtained from PHIS. Medical record review was used to determine prophylactic antibiotic use, and presence of VUR or GU abnormalities (eg, posterior urethral valves). Chronic care conditions were defined using a previously reported method.20

Data Analysis

Continuous variables were described using median and interquartile range (IQR). Categorical variables were described using frequencies. Multivariable analyses were used to determine the independent association of discordant antibiotic therapy and the outcomes of interest. Poisson regression was used to fit the skewed LOS distribution. The effect of antibiotic concordance or discordance on LOS was determined for all patients in our sample, as well as for those with a urine culture positive for a single identified organism. We used the KruskalWallis test statistic to determine the association between duration of fever and discordant antibiotic therapy, given that duration of fever is a continuous variable. Generalized estimating equations accounted for clustering by hospital and the variability that exists between hospitals.

RESULTS

Of the initial 460 cases with positive urine culture growth at any time during admission, 216 met inclusion criteria for a laboratory‐confirmed UTI from urine culture completed at admission. The median age was 2.46 years (IQR: 0.27,8.89). In the study population, 25.0% were male, 31.0% were receiving prophylactic antibiotics, 13.0% had any grade of VUR, and 16.7% had abnormal GU anatomy (Table 1). A total of 82.4% of patients were treated with concordant initial therapy, 10.2% with discordant initial therapy, and 7.4% received delayed initial antibiotic therapy. There were no significant differences between the groups for any of the covariates. Discordant antibiotic cases ranged from 4.9% to 21.7% across hospitals.

Study Population
 OverallConcordant*DiscordantDelayed AntibioticsP Value
  • NOTE: Values listed as number (percentage). Abbreviations: CCC, complex chronic condition; GU, genitourinary; VUR, vesicoureteral reflux.

  • In vitro susceptibility of uropathogen to initial antibiotic.

  • In vitro nonsusceptibility of uropathogen to initial antibiotic.

  • No antibiotics given on day of, or day after, urine culture collection.

N216178 (82.4)22 (10.2)16 (7.4) 
Gender     
Male54 (25.0)40 (22.5)8 (36.4)6 (37.5)0.18
Female162 (75.0)138 (77.5)14 (63.64)10 (62.5) 
Race     
Non‐Hispanic white136 (63.9)110 (62.5)15 (71.4)11 (68.8)0.83
Non‐Hispanic black28 (13.2)24 (13.6)2 (9.5)2 (12.5) 
Hispanic20 (9.4)16 (9.1)3 (14.3)1 (6.3) 
Asian10 (4.7)9 (5.1)1 (4.7)  
Other19 (8.9)17 (9.7) 2 (12.5) 
Payor     
Government97 (44.9)80 (44.9)11 (50.0)6 (37.5)0.58
Private70 (32.4)56 (31.5)6 (27.3)8 (50.0) 
Other49 (22.7)42 (23.6)5 (22.7)2 (12.5) 
Disposition     
Home204 (94.4)168 (94.4)21 (95.5)15 (93.8)0.99
Died1 (0.5)1 (0.6)   
Other11 (5.1)9 (5.1)1 (4.6)1 (6.3) 
Age     
3 d60 d40 (18.5)35 (19.7)3 (13.6)2 (12.5)0.53
61 d2 y62 (28.7)54 (30.3)4 (18.2)4 (25.0) 
3 y12 y75 (34.7)61 (34.3)8 (36.4)6 (37.5) 
13 y18 y39 (18.1)28 (15.7)7 (31.8)4 (25.0) 
Length of stay     
1 d5 d171 (79.2)147 (82.6)12 (54.6)12 (75.0)0.03
6 d10 d24 (11.1)17 (9.6)5 (22.7)2 (12.5) 
11 d15 d10 (4.6)5 (2.8)3 (13.6)2 (12.5) 
16 d+11 (5.1)9 (5.1)2 (9.1)0 
Complex chronic conditions
Any CCC94 (43.5)77 (43.3)12 (54.6)5 (31.3)0.35
Cardiovascular20 (9.3)19 (10.7) 1 (6.3)0.24
Neuromuscular34 (15.7)26 (14.6)7 (31.8)1 (6.3)0.06
Respiratory6 (2.8)6 (3.4)  0.52
Renal26 (12.0)21 (11.8)4 (18.2)1 (6.3)0.52
Gastrointestinal3 (1.4)3 (1.7)  0.72
Hematologic/ immunologic1 (0.5) 1 (4.6) 0.01
Metabolic8 (3.7)6 (3.4)1 (4.6)1 (6.3)0.82
Congenital or genetic15 (6.9)11 (6.2)3 (13.6)1 (6.3)0.43
Malignancy5 (2.3)3 (1.7)2 (9.1) 0.08
VUR28 (13.0)23 (12.9)3 (13.6)2 (12.5)0.99
Abnormal GU36 (16.7)31 (17.4)4 (18.2)1 (6.3)0.51
Prophylactic antibiotics67 (31.0)53 (29.8)10 (45.5)4 (25.0)0.28

The most common causative organisms were E. coli (65.7%) and Klebsiella spp (9.7%) (Table 2). The most common initial antibiotics were a third‐generation cephalosporin (39.1%), combination of ampicillin and a third‐ or fourth‐generation cephalosporin (16.7%), and combination of ampicillin with gentamicin (11.1%). A third‐generation cephalosporin was the initial antibiotic for 46.1% of the E. coli and 56.9% of Klebsiella spp UTIs. Resistance to third‐generation cephalosporins but carbapenem susceptibility was noted for 4.5% of E. coli and 7.7% of Klebsiella spp isolates. Patients with UTIs caused by Klebsiella spp, mixed organisms, and Enterobacter spp were more likely to receive discordant antibiotic therapy. Patients with Enterobacter spp and mixed‐organism UTIs were more likely to have delayed antibiotic therapy. Nineteen patients (8.8%) had positive blood cultures. Fifteen (6.9%) required intensive care unit (ICU) admission during hospitalization.

UTIs by Primary Culture Causative Organism
OrganismCasesConcordant* No. (%)Discordant No. (%)Delayed Antibiotics No. (%)
  • Abbreviations: UTI, urinary tract infection.

  • In vitro susceptibility of uropathogen to initial antibiotic.

  • In vitro nonsusceptibility of uropathogen to initial antibiotic.

  • No antibiotics given on day of, or after, urine culture collection.

E. coli142129 (90.8)3 (2.1)10 (7.0)
Klebsiella spp2114 (66.7)7 (33.3)0 (0)
Enterococcus spp129 (75.0)3 (25.0)0 (0)
Enterobacter spp105 (50.0)3 (30.0)2 (20.0)
Pseudomonas spp109 (90.0)1 (10.0)0 (0)
Other single organisms65 (83.3)0 (0)1 (16.7)
Other identified multiple organisms157 (46.7)5 (33.3)3 (20.0)

Unadjusted results are shown in Supporting Appendix 1, in the online version of this article. In the adjusted analysis, discordant antibiotic therapy was associated with a significantly longer LOS, compared with concordant therapy for all UTIs and for all UTIs caused by a single organism (Table 3). In adjusted analysis, discordant therapy was also associated with a 3.1 day (IQR: 2.0, 4.7) longer length of stay compared with concordant therapy for all E. coli UTIs.

Difference in LOS for Children With UTI Based on Empiric Antibiotic Therapy
BacteriaDifference in LOS (95% CI)*P Value
  • Abbreviations: CI, confidence interval; LOS, length of stay; UTI, urinary tract infection.

  • Models adjusted for age, sex, race, presence of vesicoureteral reflux (VUR), chronic care condition, abnormal genitourinary (GU) anatomy, prophylactic antibiotic use.

All organisms  
Concordant vs discordant1.8 (2.1, 1.5)<0.0001
Concordant vs delayed antibiotics1.4 (1.7, 1.1)0.01
Single organisms  
Concordant vs discordant1.9 (2.4, 1.5)<0.0001
Concordant vs delayed antibiotics1.2 (1.6, 1.2)0.37

Time to fever resolution was analyzed for patients with a documented fever at presentation for each treatment subgroup. One hundred thirty‐six patients were febrile at admission and 122 were febrile beyond the first recorded vital signs. Fever was present at admission in 60% of the concordant group and 55% of the discordant group (P = 0.6). The median duration of fever was 48 hours for the concordant group (n = 107; IQR: 24, 240) and 78 hours for the discordant group (n = 12; IQR: 48, 132). All patients were afebrile at discharge. Differences in fever duration between treatment groups were not statistically significant (P = 0.7).

DISCUSSION

Across 5 children's hospitals, 1 out of every 10 children hospitalized for UTI received discordant initial antibiotic therapy. Children receiving discordant antibiotic therapy had a 1.8 day longer LOS when compared with those on concordant therapy. However, there was no significant difference in time to fever resolution between the groups, suggesting that the increase in LOS was not explained by increased fever duration.

The overall rate of discordant therapy in this study is consistent with prior studies, as was the more common association of discordant therapy with non‐E. coli UTIs.10 According to the Kids' Inpatient Database 2009, there are 48,100 annual admissions for patients less than 20 years of age with a discharge diagnosis code of UTI in the United States.1 This suggests that nearly 4800 children with UTI could be affected by discordant therapy annually.

Children treated with discordant antibiotic therapy had a significantly longer LOS compared to those treated with concordant therapy. However, differences in time to fever resolution between the groups were not statistically significant. While resolution of fever may suggest clinical improvement and adequate empiric therapy, the lack of association with antibiotic concordance was not unexpected, since the relationship between fever resolution, clinical improvement, and LOS is complex and thus challenging to measure.21 These results support the notion that fever resolution alone may not be an adequate measure of clinical response.

It is possible that variability in discharge decision‐making may contribute to increased length of stay. Some clinicians may delay a patient's discharge until complete resolution of symptoms or knowledge of susceptibilities, while others may discharge patients that are still febrile and/or still receiving empiric antibiotics. Evidence‐based guidelines that address the appropriate time to discharge a patient with UTI are lacking. The American Academy of Pediatrics provides recommendations for use of parenteral antibiotics and hospital admission for patients with UTI, but does not address discharge decision‐making or patient management in the setting of discordant antibiotic therapy.2, 21

This study must be interpreted in the context of several limitations. First, our primary and secondary outcomes, LOS and fever duration, were surrogate measures for clinical response. We were not able to measure all clinical factors that may contribute to LOS, such as the patient's ability to tolerate oral fluids and antibiotics. Also, there may have been too few patients to detect a clinically important difference in fever duration between the concordant and discordant groups, especially for individual organisms. Although we did find a significant difference in LOS between patients treated with concordant compared with discordant therapy, there may be residual confounding from unobserved differences. This confounding, in conjunction with the small sample size, may cause us to underestimate the magnitude of the difference in LOS resulting from discordant therapy. Second, short‐term outcomes such as ICU admission were not investigated in this study; however, the proportion of patients admitted to the ICU in our population was quite small, precluding its use as a meaningful outcome measure. Third, the potential benefits to patients who were not exposed to unnecessary antibiotics, or harm to those that were exposed, could not be measured. Finally, our study was obtained using data from 5 free‐standing tertiary care pediatric facilities, thereby limiting its generalizability to other settings. Still, our rates of prophylactic antibiotic use, VUR, and GU abnormalities are similar to others reported in tertiary care children's hospitals, and we accounted for these covariates in our model.2225

As the frequency of infections caused by resistant bacteria increase, so will the number of patients receiving discordant antibiotics for UTI, compounding the challenge of empiric antimicrobial selection. Further research is needed to better understand how discordant initial antibiotic therapy contributes to LOS and whether it is associated with adverse short‐ and long‐term clinical outcomes. Such research could also aid in weighing the risk of broader‐spectrum prescribing on antimicrobial resistance patterns. While we identified an association between discordant initial antibiotic therapy and LOS, we were unable to determine the ideal empiric antibiotic therapy for patients hospitalized with UTI. Further investigation is needed to inform local and national practice guidelines for empiric antibiotic selection in patients with UTIs. This may also be an opportunity to decrease discordant empiric antibiotic selection, perhaps through use of antibiograms that stratify patients based on known factors, to lead to more specific initial therapy.

CONCLUSIONS

This study demonstrates that discordant antibiotic selection for UTI at admission is associated with longer hospital stay, but not fever duration. The full clinical consequences of discordant therapy, and the effects on length of stay, need to be better understood. Our findings, taken in combination with careful consideration of patient characteristics and prior history, may provide an opportunity to improve the hospital care for patients with UTIs.

Acknowledgements

Disclosure: Nothing to report.

Files
References
  1. HCUP Kids' Inpatient Database (KID). Healthcare Cost and Utilization Project (HCUP). Rockville, MD: Agency for Healthcare Research and Quality; 2006 and 2009. Available at: http://www.hcup‐us.ahrq.gov/kidoverview.jsp.
  2. Subcommitee on Urinary Tract Infection, Steering Committee on Quality Improvement and Management. Urinary tract infection: clinical practice guideline for the diagnosis and management of the initial UTI in febrile infants and children 2 to 24 months. Pediatrics. 2011;128(3)595–610. doi: 10.1542/peds.2011–1330. Available at: http://pediatrics.aappublications.org/content/128/3/595.full.html.
  3. Copp HL, Shapiro DJ, Hersh AL. National ambulatory antibiotic prescribing patterns for pediatric urinary tract infection, 1998–2007. Pediatrics. 2011;127(6):10271033.
  4. Paschke AA, Zaoutis T, Conway PH, Xie D, Keren R. Previous antimicrobial exposure is associated with drug‐resistant urinary tract infections in children. Pediatrics. 2010;125(4):664672.
  5. CDC. National Antimicrobial Resistance Monitoring System for Enteric Bacteria (NARMS): Human Isolates Final Report. Atlanta, GA: US Department of Health and Human Services, CDC; 2009.
  6. Mohammad‐Jafari H, Saffar MJ, Nemate I, Saffar H, Khalilian AR. Increasing antibiotic resistance among uropathogens isolated during years 2006–2009: impact on the empirical management. Int Braz J Urol. 2012;38(1):2532.
  7. Network ETS. 3rd Generation Cephalosporin‐Resistant Escherichia coli. 2010. Available at: http://www.cddep.org/ResistanceMap/bug‐drug/EC‐CS. Accessed May 14, 2012.
  8. Shaikh N, Ewing AL, Bhatnagar S, Hoberman A. Risk of renal scarring in children with a first urinary tract infection: a systematic review. Pediatrics. 2010;126(6):10841091.
  9. Hoberman A, Wald ER. Treatment of urinary tract infections. Pediatr Infect Dis J. 1999;18(11):10201021.
  10. Marcus N, Ashkenazi S, Yaari A, Samra Z, Livni G. Non‐Escherichia coli versus Escherichia coli community‐acquired urinary tract infections in children hospitalized in a tertiary center: relative frequency, risk factors, antimicrobial resistance and outcome. Pediatr Infect Dis J. 2005;24(7):581585.
  11. Ramos‐Martinez A, Alonso‐Moralejo R, Ortega‐Mercader P, Sanchez‐Romero I, Millan‐Santos I, Romero‐Pizarro Y. Prognosis of urinary tract infections with discordant antibiotic treatment [in Spanish]. Rev Clin Esp. 2010;210(11):545549.
  12. Velasco Arribas M, Rubio Cirilo L, Casas Martin A, et al. Appropriateness of empiric antibiotic therapy in urinary tract infection in emergency room [in Spanish]. Rev Clin Esp. 2010;210(1):1116.
  13. Long SS, Pickering LK, Prober CG. Principles and Practice of Pediatric Infectious Diseases. 3rd ed. New York, NY: Churchill Livingstone/Elsevier; 2009.
  14. National Committee for Clinical Laboratory Standards. Performance Standards for Antimicrobial Susceptibility Testing; Twelfth Informational Supplement.Vol M100‐S12. Wayne, PA: NCCLS; 2002.
  15. Tieder JS, Hall M, Auger KA, et al. Accuracy of administrative billing codes to detect urinary tract infection hospitalizations. Pediatrics. 2011;128(2):323330.
  16. Mongelluzzo J, Mohamad Z, Ten Have TR, Shah SS. Corticosteroids and mortality in children with bacterial meningitis. JAMA. 2008;299(17):20482055.
  17. Hoberman A, Wald ER, Penchansky L, Reynolds EA, Young S. Enhanced urinalysis as a screening test for urinary tract infection. Pediatrics. 1993;91(6):11961199.
  18. Hoberman A, Wald ER, Reynolds EA, Penchansky L, Charron M. Pyuria and bacteriuria in urine specimens obtained by catheter from young children with fever. J Pediatr. 1994;124(4):513519.
  19. Zorc JJ, Levine DA, Platt SL, et al. Clinical and demographic factors associated with urinary tract infection in young febrile infants. Pediatrics. 2005;116(3):644648.
  20. Feudtner C, Hays RM, Haynes G, Geyer JR, Neff JM, Koepsell TD. Deaths attributed to pediatric complex chronic conditions: national trends and implications for supportive care services. Pediatrics. 2001;107(6):E99.
  21. Committee on Quality Improvement. Subcommittee on Urinary Tract Infection. Practice parameter: the diagnosis, treatment, and evaluation of the initial urinary tract infection in febrile infants and young children. Pediatrics. 1999;103:843852.
  22. Fanos V, Cataldi L. Antibiotics or surgery for vesicoureteric reflux in children. Lancet. 2004;364(9446):17201722.
  23. Chesney RW, Carpenter MA, Moxey‐Mims M, et al. Randomized intervention for children with vesicoureteral reflux (RIVUR): background commentary of RIVUR investigators. Pediatrics. 2008;122(suppl 5):S233S239.
  24. Brady PW, Conway PH, Goudie A. Length of intravenous antibiotic therapy and treatment failure in infants with urinary tract infections. Pediatrics. 2010;126(2):196203.
  25. Hannula A, Venhola M, Renko M, Pokka T, Huttunen NP, Uhari M. Vesicoureteral reflux in children with suspected and proven urinary tract infection. Pediatr Nephrol. 2010;25(8):14631469.
Article PDF
Issue
Journal of Hospital Medicine - 7(8)
Publications
Page Number
622-627
Sections
Files
Files
Article PDF
Article PDF

Urinary tract infections (UTIs) are one of the most common reasons for pediatric hospitalizations.1 Bacterial infections require prompt treatment with appropriate antimicrobial agents. Results from culture and susceptibility testing, however, are often unavailable until 48 hours after initial presentation. Therefore, the clinician must select antimicrobials empirically, basing decisions on likely pathogens and local resistance patterns.2 This decision is challenging because the effect of treatment delay on clinical outcomes is difficult to determine and resistance among uropathogens is increasing. Resistance rates have doubled over the past several years.3, 4 For common first‐line antibiotics, such as ampicillin and trimethoprim‐sulfamethoxazole, resistance rates for Escherichia coli, the most common uropathogen, exceed 25%.4, 5 While resistance to third‐generation cephalosporins remains low, rates in the United States have increased from <1% in 1999 to 4% in 2010. International data shows much higher resistance rates for cephalosporins in general.6, 7 This high prevalence of resistance may prompt the use of broad‐spectrum antibiotics for patients with UTI. For example, the use of third‐generation cephalosporins for UTI has doubled in recent years.3 Untreated, UTIs can lead to serious illness, but the consequences of inadequate initial antibiotic coverage are unknown.8, 9

Discordant antibiotic therapy, initial antibiotic therapy to which the causative bacterium is not susceptible, occurs in up to 9% of children hospitalized for UTI.10 However, there is reason to believe that discordant therapy may matter less for UTIs than for infections at other sites. First, in adults hospitalized with UTIs, discordant initial therapy did not affect the time to resolution of symptoms.11, 12 Second, most antibiotics used to treat UTIs are renally excreted and, thus, antibiotic concentrations at the site of infection are higher than can be achieved in the serum or cerebrospinal fluid.13 The Clinical and Laboratory Standard Institute has acknowledged that traditional susceptibility breakpoints may be too conservative for some non‐central nervous system infections; such as non‐central nervous system infections caused by Streptococcus pneumoniae.14

As resistance rates increase, more patients are likely to be treated with discordant therapy. Therefore, we sought to identify the clinical consequences of discordant antimicrobial therapy for patients hospitalized with a UTI.

METHODS

Design and Setting

We conducted a multicenter, retrospective cohort study. Data for this study were originally collected for a study that determined the accuracy of individual and combined International Classification of Diseases, Ninth Revision (ICD‐9) discharge diagnosis codes for children with laboratory tests for a UTI, in order to develop national quality measures for children hospitalized with UTIs.15 The institutional review board for each hospital (Seattle Children's Hospital, Seattle, WA; Monroe Carell Jr Children's Hospital at Vanderbilt, Nashville, TN; Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Children's Mercy Hospital, Kansas City, MO; Children's Hospital of Philadelphia, Philadelphia, PA) approved the study.

Data Sources

Data were obtained from the Pediatric Health Information System (PHIS) and medical records for patients at the 5 participating hospitals. PHIS contains clinical and billing data from hospitalized children at 43 freestanding children's hospitals. Data quality and coding reliability are assured through a joint effort between the Children's Hospital Association (Shawnee Mission, KS) and participating hospitals.16 PHIS was used to identify participants based on presence of discharge diagnosis code and laboratory tests indicating possible UTI, patient demographics, antibiotic administration date, and utilization of hospital resources (length of stay [LOS], laboratory testing).

Medical records for each participant were reviewed to obtain laboratory and clinical information such as past medical history (including vesicoureteral reflux [VUR], abnormal genitourinary [GU] anatomy, use of prophylactic antibiotic), culture data, and fever data. Data were entered into a secured centrally housed web‐based data collection system. To assure consistency of chart review, all investigators responsible for data collection underwent training. In addition, 2 pilot medical record reviews were performed, followed by group discussion, to reach consensus on questions, preselected answers, interpretation of medical record data, and parameters for free text data entry.

Subjects

The initial cohort included 460 hospitalized patients, aged 3 days to 18 years of age, discharged from participating hospitals between July 1, 2008 and June 30, 2009 with a positive urine culture at any time during hospitalization.15 We excluded patients under 3 days of age because patients this young are more likely to have been transferred from the birthing hospital for a complication related to birth or a congenital anomaly. For this secondary analysis of patients from a prior study, our target population included patients admitted for management of UTI.15 We excluded patients with a negative initial urine culture (n = 59) or if their initial urine culture did not meet definition of laboratory‐confirmed UTI, defined as urine culture with >50,000 colony‐forming units (CFU) with an abnormal urinalysis (UA) (n = 77).1, 1719 An abnormal UA was defined by presence of white blood cells, leukocyte esterase, bacteria, and/or nitrites. For our cohort, all cultures with >50,000 CFU also had an abnormal urinalysis. We excluded 19 patients with cultures classified as 10,000100,000 CFU because we could not confirm that the CFU was >50,000. We excluded 30 patients with urine cultures classified as normal or mixed flora, positive for a mixture of organisms not further identified, or if results were unavailable. Additionally, coagulase‐negative Staphylococcus species (n = 8) were excluded, as these are typically considered contaminants in the setting of urine cultures.2 Patients likely to have received antibiotics prior to admission, or develop a UTI after admission, were identified and removed from the cohort if they had a urine culture performed more than 1 day before, or 2 days after, admission (n = 35). Cultures without resistance testing to the initial antibiotic selection were also excluded (n = 16).

Main Outcome Measures

The primary outcome measure was hospital LOS. Time to fever resolution was a secondary outcome measure. Fever was defined as temperature 38C. Fever duration was defined as number of hours until resolution of fever; only patients with fever at admission were included in this subanalysis.

Main Exposure

The main exposure was initial antibiotic therapy. Patients were classified into 3 groups according to initial antibiotic selection: those receiving 1) concordant; 2) discordant; or 3) delayed initial therapy. Concordance was defined as in vitro susceptibility to the initial antibiotic or class of antibiotic. If the uropathogen was sensitive to a narrow‐spectrum antibiotic (eg, first‐generation cephalosporin), but was not tested against a more broad‐spectrum antibiotic of the same class (eg, third‐generation cephalosporin), concordance was based on the sensitivity to the narrow‐spectrum antibiotic. If the uropathogen was sensitive to a broad‐spectrum antibiotic (eg, third‐generation cephalosporin), concordance to a more narrow‐spectrum antibiotic was not assumed. Discordance was defined as laboratory confirmation of in vitro resistance, or intermediate sensitivity of the pathogen to the initial antibiotic or class of antibiotics. Patients were considered to have a delay in antibiotic therapy if they did not receive antibiotics on the day of, or day after, collection of UA and culture. Patients with more than 1 uropathogen identified in a single culture were classified as discordant if any of the organisms was discordant to the initial antibiotic; they were classified as concordant if all organisms were concordant to the initial antibiotic. Antibiotic susceptibility was not tested in some cases (n = 16).

Initial antibiotic was defined as the antibiotic(s) billed on the same day or day after the UA was billed. If the patient had the UA completed on the day prior to admission, we used the antibiotic administered on the day of admission as the initial antibiotic.

Covariates

Covariates were selected a priori to include patient characteristics likely to affect patient outcomes; all were included in the final analysis. These were age, race, sex, insurance, disposition, prophylactic antibiotic use for any reason (VUR, oncologic process, etc), presence of a chronic care condition, and presence of VUR or GU anatomic abnormality. Age, race, sex, and insurance were obtained from PHIS. Medical record review was used to determine prophylactic antibiotic use, and presence of VUR or GU abnormalities (eg, posterior urethral valves). Chronic care conditions were defined using a previously reported method.20

Data Analysis

Continuous variables were described using median and interquartile range (IQR). Categorical variables were described using frequencies. Multivariable analyses were used to determine the independent association of discordant antibiotic therapy and the outcomes of interest. Poisson regression was used to fit the skewed LOS distribution. The effect of antibiotic concordance or discordance on LOS was determined for all patients in our sample, as well as for those with a urine culture positive for a single identified organism. We used the KruskalWallis test statistic to determine the association between duration of fever and discordant antibiotic therapy, given that duration of fever is a continuous variable. Generalized estimating equations accounted for clustering by hospital and the variability that exists between hospitals.

RESULTS

Of the initial 460 cases with positive urine culture growth at any time during admission, 216 met inclusion criteria for a laboratory‐confirmed UTI from urine culture completed at admission. The median age was 2.46 years (IQR: 0.27,8.89). In the study population, 25.0% were male, 31.0% were receiving prophylactic antibiotics, 13.0% had any grade of VUR, and 16.7% had abnormal GU anatomy (Table 1). A total of 82.4% of patients were treated with concordant initial therapy, 10.2% with discordant initial therapy, and 7.4% received delayed initial antibiotic therapy. There were no significant differences between the groups for any of the covariates. Discordant antibiotic cases ranged from 4.9% to 21.7% across hospitals.

Study Population
 OverallConcordant*DiscordantDelayed AntibioticsP Value
  • NOTE: Values listed as number (percentage). Abbreviations: CCC, complex chronic condition; GU, genitourinary; VUR, vesicoureteral reflux.

  • In vitro susceptibility of uropathogen to initial antibiotic.

  • In vitro nonsusceptibility of uropathogen to initial antibiotic.

  • No antibiotics given on day of, or day after, urine culture collection.

N216178 (82.4)22 (10.2)16 (7.4) 
Gender     
Male54 (25.0)40 (22.5)8 (36.4)6 (37.5)0.18
Female162 (75.0)138 (77.5)14 (63.64)10 (62.5) 
Race     
Non‐Hispanic white136 (63.9)110 (62.5)15 (71.4)11 (68.8)0.83
Non‐Hispanic black28 (13.2)24 (13.6)2 (9.5)2 (12.5) 
Hispanic20 (9.4)16 (9.1)3 (14.3)1 (6.3) 
Asian10 (4.7)9 (5.1)1 (4.7)  
Other19 (8.9)17 (9.7) 2 (12.5) 
Payor     
Government97 (44.9)80 (44.9)11 (50.0)6 (37.5)0.58
Private70 (32.4)56 (31.5)6 (27.3)8 (50.0) 
Other49 (22.7)42 (23.6)5 (22.7)2 (12.5) 
Disposition     
Home204 (94.4)168 (94.4)21 (95.5)15 (93.8)0.99
Died1 (0.5)1 (0.6)   
Other11 (5.1)9 (5.1)1 (4.6)1 (6.3) 
Age     
3 d60 d40 (18.5)35 (19.7)3 (13.6)2 (12.5)0.53
61 d2 y62 (28.7)54 (30.3)4 (18.2)4 (25.0) 
3 y12 y75 (34.7)61 (34.3)8 (36.4)6 (37.5) 
13 y18 y39 (18.1)28 (15.7)7 (31.8)4 (25.0) 
Length of stay     
1 d5 d171 (79.2)147 (82.6)12 (54.6)12 (75.0)0.03
6 d10 d24 (11.1)17 (9.6)5 (22.7)2 (12.5) 
11 d15 d10 (4.6)5 (2.8)3 (13.6)2 (12.5) 
16 d+11 (5.1)9 (5.1)2 (9.1)0 
Complex chronic conditions
Any CCC94 (43.5)77 (43.3)12 (54.6)5 (31.3)0.35
Cardiovascular20 (9.3)19 (10.7) 1 (6.3)0.24
Neuromuscular34 (15.7)26 (14.6)7 (31.8)1 (6.3)0.06
Respiratory6 (2.8)6 (3.4)  0.52
Renal26 (12.0)21 (11.8)4 (18.2)1 (6.3)0.52
Gastrointestinal3 (1.4)3 (1.7)  0.72
Hematologic/ immunologic1 (0.5) 1 (4.6) 0.01
Metabolic8 (3.7)6 (3.4)1 (4.6)1 (6.3)0.82
Congenital or genetic15 (6.9)11 (6.2)3 (13.6)1 (6.3)0.43
Malignancy5 (2.3)3 (1.7)2 (9.1) 0.08
VUR28 (13.0)23 (12.9)3 (13.6)2 (12.5)0.99
Abnormal GU36 (16.7)31 (17.4)4 (18.2)1 (6.3)0.51
Prophylactic antibiotics67 (31.0)53 (29.8)10 (45.5)4 (25.0)0.28

The most common causative organisms were E. coli (65.7%) and Klebsiella spp (9.7%) (Table 2). The most common initial antibiotics were a third‐generation cephalosporin (39.1%), combination of ampicillin and a third‐ or fourth‐generation cephalosporin (16.7%), and combination of ampicillin with gentamicin (11.1%). A third‐generation cephalosporin was the initial antibiotic for 46.1% of the E. coli and 56.9% of Klebsiella spp UTIs. Resistance to third‐generation cephalosporins but carbapenem susceptibility was noted for 4.5% of E. coli and 7.7% of Klebsiella spp isolates. Patients with UTIs caused by Klebsiella spp, mixed organisms, and Enterobacter spp were more likely to receive discordant antibiotic therapy. Patients with Enterobacter spp and mixed‐organism UTIs were more likely to have delayed antibiotic therapy. Nineteen patients (8.8%) had positive blood cultures. Fifteen (6.9%) required intensive care unit (ICU) admission during hospitalization.

UTIs by Primary Culture Causative Organism
OrganismCasesConcordant* No. (%)Discordant No. (%)Delayed Antibiotics No. (%)
  • Abbreviations: UTI, urinary tract infection.

  • In vitro susceptibility of uropathogen to initial antibiotic.

  • In vitro nonsusceptibility of uropathogen to initial antibiotic.

  • No antibiotics given on day of, or after, urine culture collection.

E. coli142129 (90.8)3 (2.1)10 (7.0)
Klebsiella spp2114 (66.7)7 (33.3)0 (0)
Enterococcus spp129 (75.0)3 (25.0)0 (0)
Enterobacter spp105 (50.0)3 (30.0)2 (20.0)
Pseudomonas spp109 (90.0)1 (10.0)0 (0)
Other single organisms65 (83.3)0 (0)1 (16.7)
Other identified multiple organisms157 (46.7)5 (33.3)3 (20.0)

Unadjusted results are shown in Supporting Appendix 1, in the online version of this article. In the adjusted analysis, discordant antibiotic therapy was associated with a significantly longer LOS, compared with concordant therapy for all UTIs and for all UTIs caused by a single organism (Table 3). In adjusted analysis, discordant therapy was also associated with a 3.1 day (IQR: 2.0, 4.7) longer length of stay compared with concordant therapy for all E. coli UTIs.

Difference in LOS for Children With UTI Based on Empiric Antibiotic Therapy
BacteriaDifference in LOS (95% CI)*P Value
  • Abbreviations: CI, confidence interval; LOS, length of stay; UTI, urinary tract infection.

  • Models adjusted for age, sex, race, presence of vesicoureteral reflux (VUR), chronic care condition, abnormal genitourinary (GU) anatomy, prophylactic antibiotic use.

All organisms  
Concordant vs discordant1.8 (2.1, 1.5)<0.0001
Concordant vs delayed antibiotics1.4 (1.7, 1.1)0.01
Single organisms  
Concordant vs discordant1.9 (2.4, 1.5)<0.0001
Concordant vs delayed antibiotics1.2 (1.6, 1.2)0.37

Time to fever resolution was analyzed for patients with a documented fever at presentation for each treatment subgroup. One hundred thirty‐six patients were febrile at admission and 122 were febrile beyond the first recorded vital signs. Fever was present at admission in 60% of the concordant group and 55% of the discordant group (P = 0.6). The median duration of fever was 48 hours for the concordant group (n = 107; IQR: 24, 240) and 78 hours for the discordant group (n = 12; IQR: 48, 132). All patients were afebrile at discharge. Differences in fever duration between treatment groups were not statistically significant (P = 0.7).

DISCUSSION

Across 5 children's hospitals, 1 out of every 10 children hospitalized for UTI received discordant initial antibiotic therapy. Children receiving discordant antibiotic therapy had a 1.8 day longer LOS when compared with those on concordant therapy. However, there was no significant difference in time to fever resolution between the groups, suggesting that the increase in LOS was not explained by increased fever duration.

The overall rate of discordant therapy in this study is consistent with prior studies, as was the more common association of discordant therapy with non‐E. coli UTIs.10 According to the Kids' Inpatient Database 2009, there are 48,100 annual admissions for patients less than 20 years of age with a discharge diagnosis code of UTI in the United States.1 This suggests that nearly 4800 children with UTI could be affected by discordant therapy annually.

Children treated with discordant antibiotic therapy had a significantly longer LOS compared to those treated with concordant therapy. However, differences in time to fever resolution between the groups were not statistically significant. While resolution of fever may suggest clinical improvement and adequate empiric therapy, the lack of association with antibiotic concordance was not unexpected, since the relationship between fever resolution, clinical improvement, and LOS is complex and thus challenging to measure.21 These results support the notion that fever resolution alone may not be an adequate measure of clinical response.

It is possible that variability in discharge decision‐making may contribute to increased length of stay. Some clinicians may delay a patient's discharge until complete resolution of symptoms or knowledge of susceptibilities, while others may discharge patients that are still febrile and/or still receiving empiric antibiotics. Evidence‐based guidelines that address the appropriate time to discharge a patient with UTI are lacking. The American Academy of Pediatrics provides recommendations for use of parenteral antibiotics and hospital admission for patients with UTI, but does not address discharge decision‐making or patient management in the setting of discordant antibiotic therapy.2, 21

This study must be interpreted in the context of several limitations. First, our primary and secondary outcomes, LOS and fever duration, were surrogate measures for clinical response. We were not able to measure all clinical factors that may contribute to LOS, such as the patient's ability to tolerate oral fluids and antibiotics. Also, there may have been too few patients to detect a clinically important difference in fever duration between the concordant and discordant groups, especially for individual organisms. Although we did find a significant difference in LOS between patients treated with concordant compared with discordant therapy, there may be residual confounding from unobserved differences. This confounding, in conjunction with the small sample size, may cause us to underestimate the magnitude of the difference in LOS resulting from discordant therapy. Second, short‐term outcomes such as ICU admission were not investigated in this study; however, the proportion of patients admitted to the ICU in our population was quite small, precluding its use as a meaningful outcome measure. Third, the potential benefits to patients who were not exposed to unnecessary antibiotics, or harm to those that were exposed, could not be measured. Finally, our study was obtained using data from 5 free‐standing tertiary care pediatric facilities, thereby limiting its generalizability to other settings. Still, our rates of prophylactic antibiotic use, VUR, and GU abnormalities are similar to others reported in tertiary care children's hospitals, and we accounted for these covariates in our model.2225

As the frequency of infections caused by resistant bacteria increase, so will the number of patients receiving discordant antibiotics for UTI, compounding the challenge of empiric antimicrobial selection. Further research is needed to better understand how discordant initial antibiotic therapy contributes to LOS and whether it is associated with adverse short‐ and long‐term clinical outcomes. Such research could also aid in weighing the risk of broader‐spectrum prescribing on antimicrobial resistance patterns. While we identified an association between discordant initial antibiotic therapy and LOS, we were unable to determine the ideal empiric antibiotic therapy for patients hospitalized with UTI. Further investigation is needed to inform local and national practice guidelines for empiric antibiotic selection in patients with UTIs. This may also be an opportunity to decrease discordant empiric antibiotic selection, perhaps through use of antibiograms that stratify patients based on known factors, to lead to more specific initial therapy.

CONCLUSIONS

This study demonstrates that discordant antibiotic selection for UTI at admission is associated with longer hospital stay, but not fever duration. The full clinical consequences of discordant therapy, and the effects on length of stay, need to be better understood. Our findings, taken in combination with careful consideration of patient characteristics and prior history, may provide an opportunity to improve the hospital care for patients with UTIs.

Acknowledgements

Disclosure: Nothing to report.

Urinary tract infections (UTIs) are one of the most common reasons for pediatric hospitalizations.1 Bacterial infections require prompt treatment with appropriate antimicrobial agents. Results from culture and susceptibility testing, however, are often unavailable until 48 hours after initial presentation. Therefore, the clinician must select antimicrobials empirically, basing decisions on likely pathogens and local resistance patterns.2 This decision is challenging because the effect of treatment delay on clinical outcomes is difficult to determine and resistance among uropathogens is increasing. Resistance rates have doubled over the past several years.3, 4 For common first‐line antibiotics, such as ampicillin and trimethoprim‐sulfamethoxazole, resistance rates for Escherichia coli, the most common uropathogen, exceed 25%.4, 5 While resistance to third‐generation cephalosporins remains low, rates in the United States have increased from <1% in 1999 to 4% in 2010. International data shows much higher resistance rates for cephalosporins in general.6, 7 This high prevalence of resistance may prompt the use of broad‐spectrum antibiotics for patients with UTI. For example, the use of third‐generation cephalosporins for UTI has doubled in recent years.3 Untreated, UTIs can lead to serious illness, but the consequences of inadequate initial antibiotic coverage are unknown.8, 9

Discordant antibiotic therapy, initial antibiotic therapy to which the causative bacterium is not susceptible, occurs in up to 9% of children hospitalized for UTI.10 However, there is reason to believe that discordant therapy may matter less for UTIs than for infections at other sites. First, in adults hospitalized with UTIs, discordant initial therapy did not affect the time to resolution of symptoms.11, 12 Second, most antibiotics used to treat UTIs are renally excreted and, thus, antibiotic concentrations at the site of infection are higher than can be achieved in the serum or cerebrospinal fluid.13 The Clinical and Laboratory Standard Institute has acknowledged that traditional susceptibility breakpoints may be too conservative for some non‐central nervous system infections; such as non‐central nervous system infections caused by Streptococcus pneumoniae.14

As resistance rates increase, more patients are likely to be treated with discordant therapy. Therefore, we sought to identify the clinical consequences of discordant antimicrobial therapy for patients hospitalized with a UTI.

METHODS

Design and Setting

We conducted a multicenter, retrospective cohort study. Data for this study were originally collected for a study that determined the accuracy of individual and combined International Classification of Diseases, Ninth Revision (ICD‐9) discharge diagnosis codes for children with laboratory tests for a UTI, in order to develop national quality measures for children hospitalized with UTIs.15 The institutional review board for each hospital (Seattle Children's Hospital, Seattle, WA; Monroe Carell Jr Children's Hospital at Vanderbilt, Nashville, TN; Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Children's Mercy Hospital, Kansas City, MO; Children's Hospital of Philadelphia, Philadelphia, PA) approved the study.

Data Sources

Data were obtained from the Pediatric Health Information System (PHIS) and medical records for patients at the 5 participating hospitals. PHIS contains clinical and billing data from hospitalized children at 43 freestanding children's hospitals. Data quality and coding reliability are assured through a joint effort between the Children's Hospital Association (Shawnee Mission, KS) and participating hospitals.16 PHIS was used to identify participants based on presence of discharge diagnosis code and laboratory tests indicating possible UTI, patient demographics, antibiotic administration date, and utilization of hospital resources (length of stay [LOS], laboratory testing).

Medical records for each participant were reviewed to obtain laboratory and clinical information such as past medical history (including vesicoureteral reflux [VUR], abnormal genitourinary [GU] anatomy, use of prophylactic antibiotic), culture data, and fever data. Data were entered into a secured centrally housed web‐based data collection system. To assure consistency of chart review, all investigators responsible for data collection underwent training. In addition, 2 pilot medical record reviews were performed, followed by group discussion, to reach consensus on questions, preselected answers, interpretation of medical record data, and parameters for free text data entry.

Subjects

The initial cohort included 460 hospitalized patients, aged 3 days to 18 years of age, discharged from participating hospitals between July 1, 2008 and June 30, 2009 with a positive urine culture at any time during hospitalization.15 We excluded patients under 3 days of age because patients this young are more likely to have been transferred from the birthing hospital for a complication related to birth or a congenital anomaly. For this secondary analysis of patients from a prior study, our target population included patients admitted for management of UTI.15 We excluded patients with a negative initial urine culture (n = 59) or if their initial urine culture did not meet definition of laboratory‐confirmed UTI, defined as urine culture with >50,000 colony‐forming units (CFU) with an abnormal urinalysis (UA) (n = 77).1, 1719 An abnormal UA was defined by presence of white blood cells, leukocyte esterase, bacteria, and/or nitrites. For our cohort, all cultures with >50,000 CFU also had an abnormal urinalysis. We excluded 19 patients with cultures classified as 10,000100,000 CFU because we could not confirm that the CFU was >50,000. We excluded 30 patients with urine cultures classified as normal or mixed flora, positive for a mixture of organisms not further identified, or if results were unavailable. Additionally, coagulase‐negative Staphylococcus species (n = 8) were excluded, as these are typically considered contaminants in the setting of urine cultures.2 Patients likely to have received antibiotics prior to admission, or develop a UTI after admission, were identified and removed from the cohort if they had a urine culture performed more than 1 day before, or 2 days after, admission (n = 35). Cultures without resistance testing to the initial antibiotic selection were also excluded (n = 16).

Main Outcome Measures

The primary outcome measure was hospital LOS. Time to fever resolution was a secondary outcome measure. Fever was defined as temperature 38C. Fever duration was defined as number of hours until resolution of fever; only patients with fever at admission were included in this subanalysis.

Main Exposure

The main exposure was initial antibiotic therapy. Patients were classified into 3 groups according to initial antibiotic selection: those receiving 1) concordant; 2) discordant; or 3) delayed initial therapy. Concordance was defined as in vitro susceptibility to the initial antibiotic or class of antibiotic. If the uropathogen was sensitive to a narrow‐spectrum antibiotic (eg, first‐generation cephalosporin), but was not tested against a more broad‐spectrum antibiotic of the same class (eg, third‐generation cephalosporin), concordance was based on the sensitivity to the narrow‐spectrum antibiotic. If the uropathogen was sensitive to a broad‐spectrum antibiotic (eg, third‐generation cephalosporin), concordance to a more narrow‐spectrum antibiotic was not assumed. Discordance was defined as laboratory confirmation of in vitro resistance, or intermediate sensitivity of the pathogen to the initial antibiotic or class of antibiotics. Patients were considered to have a delay in antibiotic therapy if they did not receive antibiotics on the day of, or day after, collection of UA and culture. Patients with more than 1 uropathogen identified in a single culture were classified as discordant if any of the organisms was discordant to the initial antibiotic; they were classified as concordant if all organisms were concordant to the initial antibiotic. Antibiotic susceptibility was not tested in some cases (n = 16).

Initial antibiotic was defined as the antibiotic(s) billed on the same day or day after the UA was billed. If the patient had the UA completed on the day prior to admission, we used the antibiotic administered on the day of admission as the initial antibiotic.

Covariates

Covariates were selected a priori to include patient characteristics likely to affect patient outcomes; all were included in the final analysis. These were age, race, sex, insurance, disposition, prophylactic antibiotic use for any reason (VUR, oncologic process, etc), presence of a chronic care condition, and presence of VUR or GU anatomic abnormality. Age, race, sex, and insurance were obtained from PHIS. Medical record review was used to determine prophylactic antibiotic use, and presence of VUR or GU abnormalities (eg, posterior urethral valves). Chronic care conditions were defined using a previously reported method.20

Data Analysis

Continuous variables were described using median and interquartile range (IQR). Categorical variables were described using frequencies. Multivariable analyses were used to determine the independent association of discordant antibiotic therapy and the outcomes of interest. Poisson regression was used to fit the skewed LOS distribution. The effect of antibiotic concordance or discordance on LOS was determined for all patients in our sample, as well as for those with a urine culture positive for a single identified organism. We used the KruskalWallis test statistic to determine the association between duration of fever and discordant antibiotic therapy, given that duration of fever is a continuous variable. Generalized estimating equations accounted for clustering by hospital and the variability that exists between hospitals.

RESULTS

Of the initial 460 cases with positive urine culture growth at any time during admission, 216 met inclusion criteria for a laboratory‐confirmed UTI from urine culture completed at admission. The median age was 2.46 years (IQR: 0.27,8.89). In the study population, 25.0% were male, 31.0% were receiving prophylactic antibiotics, 13.0% had any grade of VUR, and 16.7% had abnormal GU anatomy (Table 1). A total of 82.4% of patients were treated with concordant initial therapy, 10.2% with discordant initial therapy, and 7.4% received delayed initial antibiotic therapy. There were no significant differences between the groups for any of the covariates. Discordant antibiotic cases ranged from 4.9% to 21.7% across hospitals.

Study Population
 OverallConcordant*DiscordantDelayed AntibioticsP Value
  • NOTE: Values listed as number (percentage). Abbreviations: CCC, complex chronic condition; GU, genitourinary; VUR, vesicoureteral reflux.

  • In vitro susceptibility of uropathogen to initial antibiotic.

  • In vitro nonsusceptibility of uropathogen to initial antibiotic.

  • No antibiotics given on day of, or day after, urine culture collection.

N216178 (82.4)22 (10.2)16 (7.4) 
Gender     
Male54 (25.0)40 (22.5)8 (36.4)6 (37.5)0.18
Female162 (75.0)138 (77.5)14 (63.64)10 (62.5) 
Race     
Non‐Hispanic white136 (63.9)110 (62.5)15 (71.4)11 (68.8)0.83
Non‐Hispanic black28 (13.2)24 (13.6)2 (9.5)2 (12.5) 
Hispanic20 (9.4)16 (9.1)3 (14.3)1 (6.3) 
Asian10 (4.7)9 (5.1)1 (4.7)  
Other19 (8.9)17 (9.7) 2 (12.5) 
Payor     
Government97 (44.9)80 (44.9)11 (50.0)6 (37.5)0.58
Private70 (32.4)56 (31.5)6 (27.3)8 (50.0) 
Other49 (22.7)42 (23.6)5 (22.7)2 (12.5) 
Disposition     
Home204 (94.4)168 (94.4)21 (95.5)15 (93.8)0.99
Died1 (0.5)1 (0.6)   
Other11 (5.1)9 (5.1)1 (4.6)1 (6.3) 
Age     
3 d60 d40 (18.5)35 (19.7)3 (13.6)2 (12.5)0.53
61 d2 y62 (28.7)54 (30.3)4 (18.2)4 (25.0) 
3 y12 y75 (34.7)61 (34.3)8 (36.4)6 (37.5) 
13 y18 y39 (18.1)28 (15.7)7 (31.8)4 (25.0) 
Length of stay     
1 d5 d171 (79.2)147 (82.6)12 (54.6)12 (75.0)0.03
6 d10 d24 (11.1)17 (9.6)5 (22.7)2 (12.5) 
11 d15 d10 (4.6)5 (2.8)3 (13.6)2 (12.5) 
16 d+11 (5.1)9 (5.1)2 (9.1)0 
Complex chronic conditions
Any CCC94 (43.5)77 (43.3)12 (54.6)5 (31.3)0.35
Cardiovascular20 (9.3)19 (10.7) 1 (6.3)0.24
Neuromuscular34 (15.7)26 (14.6)7 (31.8)1 (6.3)0.06
Respiratory6 (2.8)6 (3.4)  0.52
Renal26 (12.0)21 (11.8)4 (18.2)1 (6.3)0.52
Gastrointestinal3 (1.4)3 (1.7)  0.72
Hematologic/ immunologic1 (0.5) 1 (4.6) 0.01
Metabolic8 (3.7)6 (3.4)1 (4.6)1 (6.3)0.82
Congenital or genetic15 (6.9)11 (6.2)3 (13.6)1 (6.3)0.43
Malignancy5 (2.3)3 (1.7)2 (9.1) 0.08
VUR28 (13.0)23 (12.9)3 (13.6)2 (12.5)0.99
Abnormal GU36 (16.7)31 (17.4)4 (18.2)1 (6.3)0.51
Prophylactic antibiotics67 (31.0)53 (29.8)10 (45.5)4 (25.0)0.28

The most common causative organisms were E. coli (65.7%) and Klebsiella spp (9.7%) (Table 2). The most common initial antibiotics were a third‐generation cephalosporin (39.1%), combination of ampicillin and a third‐ or fourth‐generation cephalosporin (16.7%), and combination of ampicillin with gentamicin (11.1%). A third‐generation cephalosporin was the initial antibiotic for 46.1% of the E. coli and 56.9% of Klebsiella spp UTIs. Resistance to third‐generation cephalosporins but carbapenem susceptibility was noted for 4.5% of E. coli and 7.7% of Klebsiella spp isolates. Patients with UTIs caused by Klebsiella spp, mixed organisms, and Enterobacter spp were more likely to receive discordant antibiotic therapy. Patients with Enterobacter spp and mixed‐organism UTIs were more likely to have delayed antibiotic therapy. Nineteen patients (8.8%) had positive blood cultures. Fifteen (6.9%) required intensive care unit (ICU) admission during hospitalization.

UTIs by Primary Culture Causative Organism
OrganismCasesConcordant* No. (%)Discordant No. (%)Delayed Antibiotics No. (%)
  • Abbreviations: UTI, urinary tract infection.

  • In vitro susceptibility of uropathogen to initial antibiotic.

  • In vitro nonsusceptibility of uropathogen to initial antibiotic.

  • No antibiotics given on day of, or after, urine culture collection.

E. coli142129 (90.8)3 (2.1)10 (7.0)
Klebsiella spp2114 (66.7)7 (33.3)0 (0)
Enterococcus spp129 (75.0)3 (25.0)0 (0)
Enterobacter spp105 (50.0)3 (30.0)2 (20.0)
Pseudomonas spp109 (90.0)1 (10.0)0 (0)
Other single organisms65 (83.3)0 (0)1 (16.7)
Other identified multiple organisms157 (46.7)5 (33.3)3 (20.0)

Unadjusted results are shown in Supporting Appendix 1, in the online version of this article. In the adjusted analysis, discordant antibiotic therapy was associated with a significantly longer LOS, compared with concordant therapy for all UTIs and for all UTIs caused by a single organism (Table 3). In adjusted analysis, discordant therapy was also associated with a 3.1 day (IQR: 2.0, 4.7) longer length of stay compared with concordant therapy for all E. coli UTIs.

Difference in LOS for Children With UTI Based on Empiric Antibiotic Therapy
BacteriaDifference in LOS (95% CI)*P Value
  • Abbreviations: CI, confidence interval; LOS, length of stay; UTI, urinary tract infection.

  • Models adjusted for age, sex, race, presence of vesicoureteral reflux (VUR), chronic care condition, abnormal genitourinary (GU) anatomy, prophylactic antibiotic use.

All organisms  
Concordant vs discordant1.8 (2.1, 1.5)<0.0001
Concordant vs delayed antibiotics1.4 (1.7, 1.1)0.01
Single organisms  
Concordant vs discordant1.9 (2.4, 1.5)<0.0001
Concordant vs delayed antibiotics1.2 (1.6, 1.2)0.37

Time to fever resolution was analyzed for patients with a documented fever at presentation for each treatment subgroup. One hundred thirty‐six patients were febrile at admission and 122 were febrile beyond the first recorded vital signs. Fever was present at admission in 60% of the concordant group and 55% of the discordant group (P = 0.6). The median duration of fever was 48 hours for the concordant group (n = 107; IQR: 24, 240) and 78 hours for the discordant group (n = 12; IQR: 48, 132). All patients were afebrile at discharge. Differences in fever duration between treatment groups were not statistically significant (P = 0.7).

DISCUSSION

Across 5 children's hospitals, 1 out of every 10 children hospitalized for UTI received discordant initial antibiotic therapy. Children receiving discordant antibiotic therapy had a 1.8 day longer LOS when compared with those on concordant therapy. However, there was no significant difference in time to fever resolution between the groups, suggesting that the increase in LOS was not explained by increased fever duration.

The overall rate of discordant therapy in this study is consistent with prior studies, as was the more common association of discordant therapy with non‐E. coli UTIs.10 According to the Kids' Inpatient Database 2009, there are 48,100 annual admissions for patients less than 20 years of age with a discharge diagnosis code of UTI in the United States.1 This suggests that nearly 4800 children with UTI could be affected by discordant therapy annually.

Children treated with discordant antibiotic therapy had a significantly longer LOS compared to those treated with concordant therapy. However, differences in time to fever resolution between the groups were not statistically significant. While resolution of fever may suggest clinical improvement and adequate empiric therapy, the lack of association with antibiotic concordance was not unexpected, since the relationship between fever resolution, clinical improvement, and LOS is complex and thus challenging to measure.21 These results support the notion that fever resolution alone may not be an adequate measure of clinical response.

It is possible that variability in discharge decision‐making may contribute to increased length of stay. Some clinicians may delay a patient's discharge until complete resolution of symptoms or knowledge of susceptibilities, while others may discharge patients that are still febrile and/or still receiving empiric antibiotics. Evidence‐based guidelines that address the appropriate time to discharge a patient with UTI are lacking. The American Academy of Pediatrics provides recommendations for use of parenteral antibiotics and hospital admission for patients with UTI, but does not address discharge decision‐making or patient management in the setting of discordant antibiotic therapy.2, 21

This study must be interpreted in the context of several limitations. First, our primary and secondary outcomes, LOS and fever duration, were surrogate measures for clinical response. We were not able to measure all clinical factors that may contribute to LOS, such as the patient's ability to tolerate oral fluids and antibiotics. Also, there may have been too few patients to detect a clinically important difference in fever duration between the concordant and discordant groups, especially for individual organisms. Although we did find a significant difference in LOS between patients treated with concordant compared with discordant therapy, there may be residual confounding from unobserved differences. This confounding, in conjunction with the small sample size, may cause us to underestimate the magnitude of the difference in LOS resulting from discordant therapy. Second, short‐term outcomes such as ICU admission were not investigated in this study; however, the proportion of patients admitted to the ICU in our population was quite small, precluding its use as a meaningful outcome measure. Third, the potential benefits to patients who were not exposed to unnecessary antibiotics, or harm to those that were exposed, could not be measured. Finally, our study was obtained using data from 5 free‐standing tertiary care pediatric facilities, thereby limiting its generalizability to other settings. Still, our rates of prophylactic antibiotic use, VUR, and GU abnormalities are similar to others reported in tertiary care children's hospitals, and we accounted for these covariates in our model.2225

As the frequency of infections caused by resistant bacteria increase, so will the number of patients receiving discordant antibiotics for UTI, compounding the challenge of empiric antimicrobial selection. Further research is needed to better understand how discordant initial antibiotic therapy contributes to LOS and whether it is associated with adverse short‐ and long‐term clinical outcomes. Such research could also aid in weighing the risk of broader‐spectrum prescribing on antimicrobial resistance patterns. While we identified an association between discordant initial antibiotic therapy and LOS, we were unable to determine the ideal empiric antibiotic therapy for patients hospitalized with UTI. Further investigation is needed to inform local and national practice guidelines for empiric antibiotic selection in patients with UTIs. This may also be an opportunity to decrease discordant empiric antibiotic selection, perhaps through use of antibiograms that stratify patients based on known factors, to lead to more specific initial therapy.

CONCLUSIONS

This study demonstrates that discordant antibiotic selection for UTI at admission is associated with longer hospital stay, but not fever duration. The full clinical consequences of discordant therapy, and the effects on length of stay, need to be better understood. Our findings, taken in combination with careful consideration of patient characteristics and prior history, may provide an opportunity to improve the hospital care for patients with UTIs.

Acknowledgements

Disclosure: Nothing to report.

References
  1. HCUP Kids' Inpatient Database (KID). Healthcare Cost and Utilization Project (HCUP). Rockville, MD: Agency for Healthcare Research and Quality; 2006 and 2009. Available at: http://www.hcup‐us.ahrq.gov/kidoverview.jsp.
  2. Subcommitee on Urinary Tract Infection, Steering Committee on Quality Improvement and Management. Urinary tract infection: clinical practice guideline for the diagnosis and management of the initial UTI in febrile infants and children 2 to 24 months. Pediatrics. 2011;128(3)595–610. doi: 10.1542/peds.2011–1330. Available at: http://pediatrics.aappublications.org/content/128/3/595.full.html.
  3. Copp HL, Shapiro DJ, Hersh AL. National ambulatory antibiotic prescribing patterns for pediatric urinary tract infection, 1998–2007. Pediatrics. 2011;127(6):10271033.
  4. Paschke AA, Zaoutis T, Conway PH, Xie D, Keren R. Previous antimicrobial exposure is associated with drug‐resistant urinary tract infections in children. Pediatrics. 2010;125(4):664672.
  5. CDC. National Antimicrobial Resistance Monitoring System for Enteric Bacteria (NARMS): Human Isolates Final Report. Atlanta, GA: US Department of Health and Human Services, CDC; 2009.
  6. Mohammad‐Jafari H, Saffar MJ, Nemate I, Saffar H, Khalilian AR. Increasing antibiotic resistance among uropathogens isolated during years 2006–2009: impact on the empirical management. Int Braz J Urol. 2012;38(1):2532.
  7. Network ETS. 3rd Generation Cephalosporin‐Resistant Escherichia coli. 2010. Available at: http://www.cddep.org/ResistanceMap/bug‐drug/EC‐CS. Accessed May 14, 2012.
  8. Shaikh N, Ewing AL, Bhatnagar S, Hoberman A. Risk of renal scarring in children with a first urinary tract infection: a systematic review. Pediatrics. 2010;126(6):10841091.
  9. Hoberman A, Wald ER. Treatment of urinary tract infections. Pediatr Infect Dis J. 1999;18(11):10201021.
  10. Marcus N, Ashkenazi S, Yaari A, Samra Z, Livni G. Non‐Escherichia coli versus Escherichia coli community‐acquired urinary tract infections in children hospitalized in a tertiary center: relative frequency, risk factors, antimicrobial resistance and outcome. Pediatr Infect Dis J. 2005;24(7):581585.
  11. Ramos‐Martinez A, Alonso‐Moralejo R, Ortega‐Mercader P, Sanchez‐Romero I, Millan‐Santos I, Romero‐Pizarro Y. Prognosis of urinary tract infections with discordant antibiotic treatment [in Spanish]. Rev Clin Esp. 2010;210(11):545549.
  12. Velasco Arribas M, Rubio Cirilo L, Casas Martin A, et al. Appropriateness of empiric antibiotic therapy in urinary tract infection in emergency room [in Spanish]. Rev Clin Esp. 2010;210(1):1116.
  13. Long SS, Pickering LK, Prober CG. Principles and Practice of Pediatric Infectious Diseases. 3rd ed. New York, NY: Churchill Livingstone/Elsevier; 2009.
  14. National Committee for Clinical Laboratory Standards. Performance Standards for Antimicrobial Susceptibility Testing; Twelfth Informational Supplement.Vol M100‐S12. Wayne, PA: NCCLS; 2002.
  15. Tieder JS, Hall M, Auger KA, et al. Accuracy of administrative billing codes to detect urinary tract infection hospitalizations. Pediatrics. 2011;128(2):323330.
  16. Mongelluzzo J, Mohamad Z, Ten Have TR, Shah SS. Corticosteroids and mortality in children with bacterial meningitis. JAMA. 2008;299(17):20482055.
  17. Hoberman A, Wald ER, Penchansky L, Reynolds EA, Young S. Enhanced urinalysis as a screening test for urinary tract infection. Pediatrics. 1993;91(6):11961199.
  18. Hoberman A, Wald ER, Reynolds EA, Penchansky L, Charron M. Pyuria and bacteriuria in urine specimens obtained by catheter from young children with fever. J Pediatr. 1994;124(4):513519.
  19. Zorc JJ, Levine DA, Platt SL, et al. Clinical and demographic factors associated with urinary tract infection in young febrile infants. Pediatrics. 2005;116(3):644648.
  20. Feudtner C, Hays RM, Haynes G, Geyer JR, Neff JM, Koepsell TD. Deaths attributed to pediatric complex chronic conditions: national trends and implications for supportive care services. Pediatrics. 2001;107(6):E99.
  21. Committee on Quality Improvement. Subcommittee on Urinary Tract Infection. Practice parameter: the diagnosis, treatment, and evaluation of the initial urinary tract infection in febrile infants and young children. Pediatrics. 1999;103:843852.
  22. Fanos V, Cataldi L. Antibiotics or surgery for vesicoureteric reflux in children. Lancet. 2004;364(9446):17201722.
  23. Chesney RW, Carpenter MA, Moxey‐Mims M, et al. Randomized intervention for children with vesicoureteral reflux (RIVUR): background commentary of RIVUR investigators. Pediatrics. 2008;122(suppl 5):S233S239.
  24. Brady PW, Conway PH, Goudie A. Length of intravenous antibiotic therapy and treatment failure in infants with urinary tract infections. Pediatrics. 2010;126(2):196203.
  25. Hannula A, Venhola M, Renko M, Pokka T, Huttunen NP, Uhari M. Vesicoureteral reflux in children with suspected and proven urinary tract infection. Pediatr Nephrol. 2010;25(8):14631469.
References
  1. HCUP Kids' Inpatient Database (KID). Healthcare Cost and Utilization Project (HCUP). Rockville, MD: Agency for Healthcare Research and Quality; 2006 and 2009. Available at: http://www.hcup‐us.ahrq.gov/kidoverview.jsp.
  2. Subcommitee on Urinary Tract Infection, Steering Committee on Quality Improvement and Management. Urinary tract infection: clinical practice guideline for the diagnosis and management of the initial UTI in febrile infants and children 2 to 24 months. Pediatrics. 2011;128(3)595–610. doi: 10.1542/peds.2011–1330. Available at: http://pediatrics.aappublications.org/content/128/3/595.full.html.
  3. Copp HL, Shapiro DJ, Hersh AL. National ambulatory antibiotic prescribing patterns for pediatric urinary tract infection, 1998–2007. Pediatrics. 2011;127(6):10271033.
  4. Paschke AA, Zaoutis T, Conway PH, Xie D, Keren R. Previous antimicrobial exposure is associated with drug‐resistant urinary tract infections in children. Pediatrics. 2010;125(4):664672.
  5. CDC. National Antimicrobial Resistance Monitoring System for Enteric Bacteria (NARMS): Human Isolates Final Report. Atlanta, GA: US Department of Health and Human Services, CDC; 2009.
  6. Mohammad‐Jafari H, Saffar MJ, Nemate I, Saffar H, Khalilian AR. Increasing antibiotic resistance among uropathogens isolated during years 2006–2009: impact on the empirical management. Int Braz J Urol. 2012;38(1):2532.
  7. Network ETS. 3rd Generation Cephalosporin‐Resistant Escherichia coli. 2010. Available at: http://www.cddep.org/ResistanceMap/bug‐drug/EC‐CS. Accessed May 14, 2012.
  8. Shaikh N, Ewing AL, Bhatnagar S, Hoberman A. Risk of renal scarring in children with a first urinary tract infection: a systematic review. Pediatrics. 2010;126(6):10841091.
  9. Hoberman A, Wald ER. Treatment of urinary tract infections. Pediatr Infect Dis J. 1999;18(11):10201021.
  10. Marcus N, Ashkenazi S, Yaari A, Samra Z, Livni G. Non‐Escherichia coli versus Escherichia coli community‐acquired urinary tract infections in children hospitalized in a tertiary center: relative frequency, risk factors, antimicrobial resistance and outcome. Pediatr Infect Dis J. 2005;24(7):581585.
  11. Ramos‐Martinez A, Alonso‐Moralejo R, Ortega‐Mercader P, Sanchez‐Romero I, Millan‐Santos I, Romero‐Pizarro Y. Prognosis of urinary tract infections with discordant antibiotic treatment [in Spanish]. Rev Clin Esp. 2010;210(11):545549.
  12. Velasco Arribas M, Rubio Cirilo L, Casas Martin A, et al. Appropriateness of empiric antibiotic therapy in urinary tract infection in emergency room [in Spanish]. Rev Clin Esp. 2010;210(1):1116.
  13. Long SS, Pickering LK, Prober CG. Principles and Practice of Pediatric Infectious Diseases. 3rd ed. New York, NY: Churchill Livingstone/Elsevier; 2009.
  14. National Committee for Clinical Laboratory Standards. Performance Standards for Antimicrobial Susceptibility Testing; Twelfth Informational Supplement.Vol M100‐S12. Wayne, PA: NCCLS; 2002.
  15. Tieder JS, Hall M, Auger KA, et al. Accuracy of administrative billing codes to detect urinary tract infection hospitalizations. Pediatrics. 2011;128(2):323330.
  16. Mongelluzzo J, Mohamad Z, Ten Have TR, Shah SS. Corticosteroids and mortality in children with bacterial meningitis. JAMA. 2008;299(17):20482055.
  17. Hoberman A, Wald ER, Penchansky L, Reynolds EA, Young S. Enhanced urinalysis as a screening test for urinary tract infection. Pediatrics. 1993;91(6):11961199.
  18. Hoberman A, Wald ER, Reynolds EA, Penchansky L, Charron M. Pyuria and bacteriuria in urine specimens obtained by catheter from young children with fever. J Pediatr. 1994;124(4):513519.
  19. Zorc JJ, Levine DA, Platt SL, et al. Clinical and demographic factors associated with urinary tract infection in young febrile infants. Pediatrics. 2005;116(3):644648.
  20. Feudtner C, Hays RM, Haynes G, Geyer JR, Neff JM, Koepsell TD. Deaths attributed to pediatric complex chronic conditions: national trends and implications for supportive care services. Pediatrics. 2001;107(6):E99.
  21. Committee on Quality Improvement. Subcommittee on Urinary Tract Infection. Practice parameter: the diagnosis, treatment, and evaluation of the initial urinary tract infection in febrile infants and young children. Pediatrics. 1999;103:843852.
  22. Fanos V, Cataldi L. Antibiotics or surgery for vesicoureteric reflux in children. Lancet. 2004;364(9446):17201722.
  23. Chesney RW, Carpenter MA, Moxey‐Mims M, et al. Randomized intervention for children with vesicoureteral reflux (RIVUR): background commentary of RIVUR investigators. Pediatrics. 2008;122(suppl 5):S233S239.
  24. Brady PW, Conway PH, Goudie A. Length of intravenous antibiotic therapy and treatment failure in infants with urinary tract infections. Pediatrics. 2010;126(2):196203.
  25. Hannula A, Venhola M, Renko M, Pokka T, Huttunen NP, Uhari M. Vesicoureteral reflux in children with suspected and proven urinary tract infection. Pediatr Nephrol. 2010;25(8):14631469.
Issue
Journal of Hospital Medicine - 7(8)
Issue
Journal of Hospital Medicine - 7(8)
Page Number
622-627
Page Number
622-627
Publications
Publications
Article Type
Display Headline
Discordant antibiotic therapy and length of stay in children hospitalized for urinary tract infection
Display Headline
Discordant antibiotic therapy and length of stay in children hospitalized for urinary tract infection
Sections
Article Source

Copyright © 2012 Society of Hospital Medicine

Disallow All Ads
Correspondence Location
3333 Burnet Ave, 5th Floor, Kasota Bldg, MLC 9016, Cincinnati, OH 45229‐3039
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Article PDF Media
Image
Disable zoom
Off
Media Files
Image
Disable zoom
Off