Affiliations
Department of Medicine, Tufts University School of Medicine, Boston, Massachusetts
Division of General Medicine and Geriatrics, Baystate Medical Center, Springfield, Massachusetts
Email
Peter.Lindenauer@bhs.org
Given name(s)
Peter K.
Family name
Lindenauer
Degrees
MD, MSc

The Association of Frailty with Discharge Disposition for Hospitalized Community Dwelling Elderly Patients

Article Type
Changed
Thu, 03/15/2018 - 22:03

Frailty is a common geriatric syndrome characterized by decreased physiological reserves leading to increased vulnerability to stressors.1 Frail individuals are at increased risk of adverse health outcomes including falls, disability, hospitalization, and mortality.1 Discharge to skilled nursing facilities (SNFs) is also associated with adverse outcomes,2,3 but limited data exist on the utility of frailty in predicting discharge location in medical elders. We aimed to evaluate the association of frailty assessed by the Reported Edmonton Frailty Scale (REFS) with discharge disposition in hospitalized medical patients who were previously living in the community.

METHODS

We conducted a prospective study of community dwelling elders (≥65 years) hospitalized to the medical service from January 2014 to April 2016. Trained research assistants interviewed patients and/or caregivers on hospital day 1; the REFS was used to screen for frailty and the Mini-Cog assessment for cognitive impairment (supplementary Appendixes 1 and 2). The primary outcome was discharge disposition categorized as discharge to home (with or without home health services) or discharge to a postacute care (PAC) facility (SNF or inpatient rehabilitation). Multivariable Poisson regression analysis was used to estimate the relative risk of discharge to a PAC facility. Frailty was grouped into the following 3 categories: (1) not frail, (2) apparently vulnerable/mildly frail, and (3) moderately/severely frail.

RESULTS

Among the 775 patients screened, 272 declined to participate, were non-English speakers, were transferred from another facility, were admitted under observation status, had advanced dementia, or died during hospitalization. Five hundred and three medical patients were included: median age was 80 years (interquartile range 75-86 years); 54.1% were female and 82.9% were white. The most common comorbidities were hypertension (51.7%), diabetes (26.0%), and renal failure (26.0%). Of the included patients, 11.1% had a known diagnosis of dementia and 52.1% screened positive for cognitive impairment (Table).

Overall, 24.9% were not frail, 49.5% were apparently vulnerable/mildly frail, and 25.6% were moderately/severely frail. About two-thirds (64.8%) returned home (40.0% with home healthcare) and 35% were discharged to a PAC facility (97.1% of them to SNF). Compared with patients who were discharged home, those discharged to a PAC facility were older (≥85 years; 26.7% vs 40.1%) and more likely to have dementia (7.7% vs 17.5%) and be frail (apparently vulnerable/mild frailty = 48.5% vs 51.4%%, moderate/severe frailty = 19.9% vs 36.2%; P < .001). Median length of hospital stay was shorter in those returning home (4 vs 5 days, P < .001).

In the multivariate analysis, which was adjusted for demographics, comorbidities, and principal diagnosis, frailty was strongly associated with discharge to PAC facility (apparently vulnerable/mild frailty vs no frailty, relative ratio [RR] = 2.00; 95% confidence interval [CI], 1.28-3.27, and moderate/severe frailty vs no frailty; RR = 2.66, 95% CI, 1.67-4.43). When the frailty score was included as a continuous variable, 1 unit increase in the score was associated with a 12% higher risk for discharge to a PAC facility (RR = 1.12; 95% CI, 1.07-1.17).

DISCUSSION

In this analysis of over 500 community-dwelling elderly medical patients hospitalized at one large tertiary center, we found that almost half of the patients were frail and over one-third had a new discharge to a PAC facility. Frailty, as assessed by REFS, was strongly associated with discharge to a PAC facility after adjusting for possible confounders.

Frailty is increasingly recognized as a useful tool to risk stratify the highly heterogeneous population of elderly people.4 Previous studies reported that frailty was predictive of discharge to PAC facilities in geriatric trauma and burn injury patients.5,6 We found similar results in a population of elderly medical patients. A recent study showed that the Hospital Admission Risk Profile score comprising of age, modified Mini-Mental State Examination (MMSE), and functionality prior to admission was associated with discharge disposition in elderly patients admitted to a single geriatric unit in a rural hospital.7 Our study supports this finding by using a validated measure of frailty, the RFS, and does not include the lengthy MMSE.

Our study has several limitations. First, it a single-center study and results may not be generalizable; however, we included a large sample of patients with a variety of medical diagnoses. Second, the REFS is self-reported posing the risks of recall, respondent bias, and interview bias. We chose the REFS to assess frailty due to its practicality and ease of administration but also its completeness of assessing multiple important geriatric domains. Lastly, we did not collect the reason for discharge to PAC and it may have been a potential confounder.

In conclusion, our study demonstrates that frailty assessed by a practical validated scale, the REFS, is a strong predictor of a new discharge to PAC facilities in older medical patients. Accurate identification of elders at risk for discharge to PAC facilities provides the potential to counsel patients and families and plan for complex post discharge needs. Future studies should identify potential interventions targeting frail patients in which PAC is not obligatory, aiming to increase their chance of being discharged home.

 

 

Disclosure

Drs. Stefan and Ramdass had full access to all the data in the study. They take responsibility for the integrity of the data and the accuracy of the analysis. Drs. Stefan, Starr, Brennan, and Ramdass conceived the study. Ms. Liu and Dr. Pekow analyzed the data. Dr. Ramdass prepared the manuscript. Drs. Stefan, Brennan, Lindenauer, and Starr critically reviewed the manuscript for important intellectual content. A subset of the patients included in this study was part of a Health Resources and Services Administration funded Geri-Pal Transformation through Learning and Collaboration project awarded to Baystate Medical Center, grant number U1QHP28702 (PI: Maura J. Brennan). The investigators retained full independence in the conduct of this research. The authors have no conflicts of interest.

 

Files
References

1. Xue QL. The frailty syndrome: definition and natural history. Clin Geriatr Med. 2011;27(1):1-15. PubMed
2. Allen LA, Hernandez AF, Peterson ED, et al. Discharge to a skilled nursing facility and subsequent clinical outcomes among older patients hospitalized for heart failure. Circ Heart Fail. 2011;4(3):293-300. PubMed
3. Hakkarainen TW, Arbabi S, Willis M, et al. Outcomes of patients discharged to skilled nursing facilities after acute care hospitalizations. Ann Surg. 2016;263(2):280-285. PubMed
4. Rockwood K, Mitnitski A. Frailty in relation to the accumulation of deficits. J Gerontol A Biol Sci Med Sci. 2007;62(7):722-727. PubMed
5. Joseph B, Pandit V, Rhee Petal, et al. Predicting hospital discharge disposition in geriatric trauma patients: is frailty the answer? J Trauma Acute Care Surg. 2014;76(1):196-200. PubMed
6. Romanowski KS, Barsun, A, Pamlieri TL, Greenhalgh DG, Sen S. Frailty score on admission predicts outcomes in elderly burn injury. J Burn Care Res. 2015;36(1):1-6. PubMed
7. Liu SK, Montgomery J, Yan Y, et al. Association between hospital admission risk profile score and skilled nursing or acute rehabilitation facility discharges in hospitalized older adults. J Am Geriatr Soc. 2016;64(10):2095-2100. PubMed

Article PDF
Issue
Journal of Hospital Medicine 13(3)
Publications
Topics
Page Number
182-184. Published online first December 6, 2017
Sections
Files
Files
Article PDF
Article PDF

Frailty is a common geriatric syndrome characterized by decreased physiological reserves leading to increased vulnerability to stressors.1 Frail individuals are at increased risk of adverse health outcomes including falls, disability, hospitalization, and mortality.1 Discharge to skilled nursing facilities (SNFs) is also associated with adverse outcomes,2,3 but limited data exist on the utility of frailty in predicting discharge location in medical elders. We aimed to evaluate the association of frailty assessed by the Reported Edmonton Frailty Scale (REFS) with discharge disposition in hospitalized medical patients who were previously living in the community.

METHODS

We conducted a prospective study of community dwelling elders (≥65 years) hospitalized to the medical service from January 2014 to April 2016. Trained research assistants interviewed patients and/or caregivers on hospital day 1; the REFS was used to screen for frailty and the Mini-Cog assessment for cognitive impairment (supplementary Appendixes 1 and 2). The primary outcome was discharge disposition categorized as discharge to home (with or without home health services) or discharge to a postacute care (PAC) facility (SNF or inpatient rehabilitation). Multivariable Poisson regression analysis was used to estimate the relative risk of discharge to a PAC facility. Frailty was grouped into the following 3 categories: (1) not frail, (2) apparently vulnerable/mildly frail, and (3) moderately/severely frail.

RESULTS

Among the 775 patients screened, 272 declined to participate, were non-English speakers, were transferred from another facility, were admitted under observation status, had advanced dementia, or died during hospitalization. Five hundred and three medical patients were included: median age was 80 years (interquartile range 75-86 years); 54.1% were female and 82.9% were white. The most common comorbidities were hypertension (51.7%), diabetes (26.0%), and renal failure (26.0%). Of the included patients, 11.1% had a known diagnosis of dementia and 52.1% screened positive for cognitive impairment (Table).

Overall, 24.9% were not frail, 49.5% were apparently vulnerable/mildly frail, and 25.6% were moderately/severely frail. About two-thirds (64.8%) returned home (40.0% with home healthcare) and 35% were discharged to a PAC facility (97.1% of them to SNF). Compared with patients who were discharged home, those discharged to a PAC facility were older (≥85 years; 26.7% vs 40.1%) and more likely to have dementia (7.7% vs 17.5%) and be frail (apparently vulnerable/mild frailty = 48.5% vs 51.4%%, moderate/severe frailty = 19.9% vs 36.2%; P < .001). Median length of hospital stay was shorter in those returning home (4 vs 5 days, P < .001).

In the multivariate analysis, which was adjusted for demographics, comorbidities, and principal diagnosis, frailty was strongly associated with discharge to PAC facility (apparently vulnerable/mild frailty vs no frailty, relative ratio [RR] = 2.00; 95% confidence interval [CI], 1.28-3.27, and moderate/severe frailty vs no frailty; RR = 2.66, 95% CI, 1.67-4.43). When the frailty score was included as a continuous variable, 1 unit increase in the score was associated with a 12% higher risk for discharge to a PAC facility (RR = 1.12; 95% CI, 1.07-1.17).

DISCUSSION

In this analysis of over 500 community-dwelling elderly medical patients hospitalized at one large tertiary center, we found that almost half of the patients were frail and over one-third had a new discharge to a PAC facility. Frailty, as assessed by REFS, was strongly associated with discharge to a PAC facility after adjusting for possible confounders.

Frailty is increasingly recognized as a useful tool to risk stratify the highly heterogeneous population of elderly people.4 Previous studies reported that frailty was predictive of discharge to PAC facilities in geriatric trauma and burn injury patients.5,6 We found similar results in a population of elderly medical patients. A recent study showed that the Hospital Admission Risk Profile score comprising of age, modified Mini-Mental State Examination (MMSE), and functionality prior to admission was associated with discharge disposition in elderly patients admitted to a single geriatric unit in a rural hospital.7 Our study supports this finding by using a validated measure of frailty, the RFS, and does not include the lengthy MMSE.

Our study has several limitations. First, it a single-center study and results may not be generalizable; however, we included a large sample of patients with a variety of medical diagnoses. Second, the REFS is self-reported posing the risks of recall, respondent bias, and interview bias. We chose the REFS to assess frailty due to its practicality and ease of administration but also its completeness of assessing multiple important geriatric domains. Lastly, we did not collect the reason for discharge to PAC and it may have been a potential confounder.

In conclusion, our study demonstrates that frailty assessed by a practical validated scale, the REFS, is a strong predictor of a new discharge to PAC facilities in older medical patients. Accurate identification of elders at risk for discharge to PAC facilities provides the potential to counsel patients and families and plan for complex post discharge needs. Future studies should identify potential interventions targeting frail patients in which PAC is not obligatory, aiming to increase their chance of being discharged home.

 

 

Disclosure

Drs. Stefan and Ramdass had full access to all the data in the study. They take responsibility for the integrity of the data and the accuracy of the analysis. Drs. Stefan, Starr, Brennan, and Ramdass conceived the study. Ms. Liu and Dr. Pekow analyzed the data. Dr. Ramdass prepared the manuscript. Drs. Stefan, Brennan, Lindenauer, and Starr critically reviewed the manuscript for important intellectual content. A subset of the patients included in this study was part of a Health Resources and Services Administration funded Geri-Pal Transformation through Learning and Collaboration project awarded to Baystate Medical Center, grant number U1QHP28702 (PI: Maura J. Brennan). The investigators retained full independence in the conduct of this research. The authors have no conflicts of interest.

 

Frailty is a common geriatric syndrome characterized by decreased physiological reserves leading to increased vulnerability to stressors.1 Frail individuals are at increased risk of adverse health outcomes including falls, disability, hospitalization, and mortality.1 Discharge to skilled nursing facilities (SNFs) is also associated with adverse outcomes,2,3 but limited data exist on the utility of frailty in predicting discharge location in medical elders. We aimed to evaluate the association of frailty assessed by the Reported Edmonton Frailty Scale (REFS) with discharge disposition in hospitalized medical patients who were previously living in the community.

METHODS

We conducted a prospective study of community dwelling elders (≥65 years) hospitalized to the medical service from January 2014 to April 2016. Trained research assistants interviewed patients and/or caregivers on hospital day 1; the REFS was used to screen for frailty and the Mini-Cog assessment for cognitive impairment (supplementary Appendixes 1 and 2). The primary outcome was discharge disposition categorized as discharge to home (with or without home health services) or discharge to a postacute care (PAC) facility (SNF or inpatient rehabilitation). Multivariable Poisson regression analysis was used to estimate the relative risk of discharge to a PAC facility. Frailty was grouped into the following 3 categories: (1) not frail, (2) apparently vulnerable/mildly frail, and (3) moderately/severely frail.

RESULTS

Among the 775 patients screened, 272 declined to participate, were non-English speakers, were transferred from another facility, were admitted under observation status, had advanced dementia, or died during hospitalization. Five hundred and three medical patients were included: median age was 80 years (interquartile range 75-86 years); 54.1% were female and 82.9% were white. The most common comorbidities were hypertension (51.7%), diabetes (26.0%), and renal failure (26.0%). Of the included patients, 11.1% had a known diagnosis of dementia and 52.1% screened positive for cognitive impairment (Table).

Overall, 24.9% were not frail, 49.5% were apparently vulnerable/mildly frail, and 25.6% were moderately/severely frail. About two-thirds (64.8%) returned home (40.0% with home healthcare) and 35% were discharged to a PAC facility (97.1% of them to SNF). Compared with patients who were discharged home, those discharged to a PAC facility were older (≥85 years; 26.7% vs 40.1%) and more likely to have dementia (7.7% vs 17.5%) and be frail (apparently vulnerable/mild frailty = 48.5% vs 51.4%%, moderate/severe frailty = 19.9% vs 36.2%; P < .001). Median length of hospital stay was shorter in those returning home (4 vs 5 days, P < .001).

In the multivariate analysis, which was adjusted for demographics, comorbidities, and principal diagnosis, frailty was strongly associated with discharge to PAC facility (apparently vulnerable/mild frailty vs no frailty, relative ratio [RR] = 2.00; 95% confidence interval [CI], 1.28-3.27, and moderate/severe frailty vs no frailty; RR = 2.66, 95% CI, 1.67-4.43). When the frailty score was included as a continuous variable, 1 unit increase in the score was associated with a 12% higher risk for discharge to a PAC facility (RR = 1.12; 95% CI, 1.07-1.17).

DISCUSSION

In this analysis of over 500 community-dwelling elderly medical patients hospitalized at one large tertiary center, we found that almost half of the patients were frail and over one-third had a new discharge to a PAC facility. Frailty, as assessed by REFS, was strongly associated with discharge to a PAC facility after adjusting for possible confounders.

Frailty is increasingly recognized as a useful tool to risk stratify the highly heterogeneous population of elderly people.4 Previous studies reported that frailty was predictive of discharge to PAC facilities in geriatric trauma and burn injury patients.5,6 We found similar results in a population of elderly medical patients. A recent study showed that the Hospital Admission Risk Profile score comprising of age, modified Mini-Mental State Examination (MMSE), and functionality prior to admission was associated with discharge disposition in elderly patients admitted to a single geriatric unit in a rural hospital.7 Our study supports this finding by using a validated measure of frailty, the RFS, and does not include the lengthy MMSE.

Our study has several limitations. First, it a single-center study and results may not be generalizable; however, we included a large sample of patients with a variety of medical diagnoses. Second, the REFS is self-reported posing the risks of recall, respondent bias, and interview bias. We chose the REFS to assess frailty due to its practicality and ease of administration but also its completeness of assessing multiple important geriatric domains. Lastly, we did not collect the reason for discharge to PAC and it may have been a potential confounder.

In conclusion, our study demonstrates that frailty assessed by a practical validated scale, the REFS, is a strong predictor of a new discharge to PAC facilities in older medical patients. Accurate identification of elders at risk for discharge to PAC facilities provides the potential to counsel patients and families and plan for complex post discharge needs. Future studies should identify potential interventions targeting frail patients in which PAC is not obligatory, aiming to increase their chance of being discharged home.

 

 

Disclosure

Drs. Stefan and Ramdass had full access to all the data in the study. They take responsibility for the integrity of the data and the accuracy of the analysis. Drs. Stefan, Starr, Brennan, and Ramdass conceived the study. Ms. Liu and Dr. Pekow analyzed the data. Dr. Ramdass prepared the manuscript. Drs. Stefan, Brennan, Lindenauer, and Starr critically reviewed the manuscript for important intellectual content. A subset of the patients included in this study was part of a Health Resources and Services Administration funded Geri-Pal Transformation through Learning and Collaboration project awarded to Baystate Medical Center, grant number U1QHP28702 (PI: Maura J. Brennan). The investigators retained full independence in the conduct of this research. The authors have no conflicts of interest.

 

References

1. Xue QL. The frailty syndrome: definition and natural history. Clin Geriatr Med. 2011;27(1):1-15. PubMed
2. Allen LA, Hernandez AF, Peterson ED, et al. Discharge to a skilled nursing facility and subsequent clinical outcomes among older patients hospitalized for heart failure. Circ Heart Fail. 2011;4(3):293-300. PubMed
3. Hakkarainen TW, Arbabi S, Willis M, et al. Outcomes of patients discharged to skilled nursing facilities after acute care hospitalizations. Ann Surg. 2016;263(2):280-285. PubMed
4. Rockwood K, Mitnitski A. Frailty in relation to the accumulation of deficits. J Gerontol A Biol Sci Med Sci. 2007;62(7):722-727. PubMed
5. Joseph B, Pandit V, Rhee Petal, et al. Predicting hospital discharge disposition in geriatric trauma patients: is frailty the answer? J Trauma Acute Care Surg. 2014;76(1):196-200. PubMed
6. Romanowski KS, Barsun, A, Pamlieri TL, Greenhalgh DG, Sen S. Frailty score on admission predicts outcomes in elderly burn injury. J Burn Care Res. 2015;36(1):1-6. PubMed
7. Liu SK, Montgomery J, Yan Y, et al. Association between hospital admission risk profile score and skilled nursing or acute rehabilitation facility discharges in hospitalized older adults. J Am Geriatr Soc. 2016;64(10):2095-2100. PubMed

References

1. Xue QL. The frailty syndrome: definition and natural history. Clin Geriatr Med. 2011;27(1):1-15. PubMed
2. Allen LA, Hernandez AF, Peterson ED, et al. Discharge to a skilled nursing facility and subsequent clinical outcomes among older patients hospitalized for heart failure. Circ Heart Fail. 2011;4(3):293-300. PubMed
3. Hakkarainen TW, Arbabi S, Willis M, et al. Outcomes of patients discharged to skilled nursing facilities after acute care hospitalizations. Ann Surg. 2016;263(2):280-285. PubMed
4. Rockwood K, Mitnitski A. Frailty in relation to the accumulation of deficits. J Gerontol A Biol Sci Med Sci. 2007;62(7):722-727. PubMed
5. Joseph B, Pandit V, Rhee Petal, et al. Predicting hospital discharge disposition in geriatric trauma patients: is frailty the answer? J Trauma Acute Care Surg. 2014;76(1):196-200. PubMed
6. Romanowski KS, Barsun, A, Pamlieri TL, Greenhalgh DG, Sen S. Frailty score on admission predicts outcomes in elderly burn injury. J Burn Care Res. 2015;36(1):1-6. PubMed
7. Liu SK, Montgomery J, Yan Y, et al. Association between hospital admission risk profile score and skilled nursing or acute rehabilitation facility discharges in hospitalized older adults. J Am Geriatr Soc. 2016;64(10):2095-2100. PubMed

Issue
Journal of Hospital Medicine 13(3)
Issue
Journal of Hospital Medicine 13(3)
Page Number
182-184. Published online first December 6, 2017
Page Number
182-184. Published online first December 6, 2017
Publications
Publications
Topics
Article Type
Sections
Article Source

© 2018 Society of Hospital Medicine

Disallow All Ads
Correspondence Location
Sheryl K. Ramdass, MD, BMedSci, Department of Geriatrics, Baystate Medical Center, 759 Chestnut Street, Springfield, MA, 01199; Telephone: 413-629-8377; Fax #: 413-794-4054; E-mail: sherylkramdass@gmail.com
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Un-Gate On Date
Tue, 03/13/2018 - 06:00
Article PDF Media
Media Files

Keeping It Simple in Sepsis Measures

Article Type
Changed
Fri, 12/14/2018 - 07:43

I didn’t have time to write a short letter, so I wrote a long one instead.”

-Mark Twain

Sepsis is a logical target for quality measures. Specifically, sepsis represents the perfect storm of immense public health burden1-3 combined with unexplained practice4-6 and outcomes7 variation. Thus, it is not surprising that in October 2015, the Centers of Medicare and Medicaid Services (CMS) adopted a sepsis quality measure.8 More surprising were the complex contents of the CMS Sepsis Core Measure “SEP-1” quality measure.9 CMS had written a “long letter.”

The multiple processes targeted with the CMS SEP-1 quality measure can best be understood with a brief account of history. SEP-1 arose from the National Quality Forum’s (NQF) project #0500: “Severe Sepsis and Septic Shock: Management Bundle,” a measure based upon Rivers et al.’s10 single-center, randomized, controlled trial of early goal-directed therapy (EGDT) for severe sepsis. EGDT was an intervention that consisted of fluid resuscitation and hemodynamic management based upon fulfilling specific targets of central venous pressure, superior vena cava oxygen saturation (or lactic acid), and hemoglobin and mean arterial pressures.11 The large mortality benefits, physiological rationale, and algorithmic responses to a variety of abnormal clinical values provided an appealing protocol to critical care and emergency physicians trained to normalize measured values, as well as policy makers looking for quality measures. Observational studies consistently showed associations between adoption of guideline-based “sepsis bundles” and improved patient outcomes,12-14 setting the stage for the transition of NQF #0500 into SEP-1.

However, the transition from EGDT-based NQF #0500 to SEP-1 has been tumultuous. Soon after adoption of SEP-1, the consensus definitions of sepsis changed markedly. Sepsis went from being defined as the presence of infection with concomitant systemic inflammatory response syndrome (sepsis), organ dysfunction (severe sepsis), and/or shock,15 to being defined as a dysregulated response to infection resulting in life-threatening organ dysfunction (sepsis) and/or fluid-resistant hypotension requiring vasopressors and lactate greater than 2 mmol/L.16 As the study by Barbash et al.17in this issue clearly outlines, conflicting definitions of “sepsis” have left clinicians confused regarding whom the SEP-1 measure should apply. At the same time, results of 3 large, international, randomized trials investigating the efficacy of EGDT were published, providing strong evidence that EGDT did not provide improved patient outcomes over usual care.18 SEP-1 adapted with the evolving evidence base, adding putative “usual care” processes such as evaluation of skin and peripheral pulses, and use of dynamic measures of fluid responsiveness, as quality measures.19 However, as Barbash et al. also outline, the resulting process measure was incredibly complex, with potentially more than 50 data elements collected over 6 hours in the initial management of sepsis.

In addition to its unprecedented complexity, SEP-1 received criticism for the weak evidence base of its individual components. The general concepts behind SEP-1 are well-accepted tenets of sepsis management: rapid recognition, assessment and treatment of underlying infection, and institution of intravenous fluids and vasopressor support for septic shock. However, the “all or none” prescriptive nature of the SEP-1 bundle was based on a somewhat arbitrary set of measures and targets. For example, patients with septic shock must receive 30 cc/kg of intravenous fluids to be “SEP-1 compliant.” The value “30 cc/kg” was taken from the average volume of fluids reported in prior sepsis trials, essentially based on a very low level of evidence.20 The strict 30 cc/kg cutoff did not take into account that “the median isn’t the message”21 in fluid management: optimal resuscitation targets are unclear,22 and selecting the median as a target ignores the fact that 50% of patients enrolled in international trials of EGDT received less than 30 cc/kg of initial fluid resuscitation (the interquartile range was 16-42 cc/kg).18 Thus, most participants in trials upon which the SEP-1 fluid measure was based would ironically not have met the SEP-1 measure. Mandates for physical exam and physiological measures were based on similarly low levels of evidence.

Into this context, Barbash et al. use a representative sample of US hospitals to explore the opinions of hospital quality leaders regarding the SEP-1 measure. First, the qualitative methods used by Barbash et al. warrant some explanation. Much of biomedical research is characterized by hypothesis-driven, deductive reasoning: theories are tested using observations. In contrast, the methods of Barbash et al. use inductive reasoning: observations are used to develop theories within a systematic approach called “grounded theory” that explores common themes emerging from structured interviews.23 Inductive reasoning can later inform deductive reasoning, feeding theories into testable hypotheses. However, qualitative, inductive research is not meant to test hypotheses and is not subject to typical notions of “power and sample size” often expected of quantitative statistical analyses. Qualitative studies reach sufficient sample size when no further themes emerge, a situation called “thematic saturation”; the sample size here of 29 participants rests comfortably in the range of participants commonly needed for thematic saturation.23

Barbash et al. identified common themes in opinions of quality leaders regarding SEP-1. Namely, the complexity of SEP-1 necessitated a major resource investment into sepsis care and data collection. The major infrastructure investments needed to comply with SEP-1 also bred frustration regarding lack of perceived fairness around the “all or none” nature of the measure and raised multiple additional challenges including lack of clinician buy-in and resistance to protocolized care. Prior qualitative studies evaluating hospital quality leaders’ opinions on performance measures identified similar concerns about lack of “fairness,”24 but the implementation of SEP-1 has raised additional concern regarding the large burdens of instituting major infrastructure changes to monitor processes of care required to report on this measure. Despite the major challenges of responding to SEP-1, quality leaders expressed optimism that increased attention to sepsis would ultimately lead to better patient outcomes.

How might future sepsis quality measures achieve the adequate balance between focusing attention on improving care processes for high-impact diseases, without imposing additional burdens on the healthcare system? Lessons from Barbash et al. help us move forward. First, rather than taxing hospitals with administratively complex process measures, initial attempts at quality measures should start simply. Policy makers should consider moving forward into new areas of quality measurement in 2 ways: (1) pursue 1 or 2 processes with strong etiological links to important patient outcomes (eg, timely antibiotics in septic shock),25-28 and/or (2) use risk-adjusted outcomes and allow individual hospitals to adopt processes that improve local patient outcomes. Evidence suggests that the introduction of a quality measure may result in improved outcomes regardless of adoption of specific target processes,29 although results are mixed.30,31 In either case, complex “all or none” measures based upon weak evidence run a high risk of inciting clinician resentment and paradoxically perpetuating poor quality by increasing healthcare costs (decreased efficiency) without gains in safety, effectiveness, timeliness, or equity.32 It has been estimated that hospitals spend on average $2 million to implement SEP-1,33 with unclear return on the investment. The experience of SEP-1 is a reminder that, as evidence evolves, quality measures must adapt lest they become irrelevant. However, it is also a reminder that quality measures should not sit precariously on the edge of evidence. Withdrawal of process-based measures due to a changing evidence landscape breeds mistrust and impairs future attempts to improve quality.

Sepsis quality measures face additional challenges. If recent experience with interpretation of sepsis definitions can serve as a guide, variable uptake of newer sepsis definitions between/across hospitals will impair the ability to risk-adjust outcome measures and increase bias in identifying outlier hospitals.34 In addition, recent studies have already raised skepticism regarding the effectiveness of individual SEP-1 bundle components, confirming suspicions that the 30 cc/kg fluid bolus is not a magic quality target. Rather, the effectiveness of prior sepsis bundles has likely been driven by improved time to antibiotics, a process unstudied in sepsis trials, but driven by increased attention to the importance of early sepsis recognition and timely management.28 Timeliness of antibiotics can act as an effect modifier for more complex sepsis therapies, with quicker time to antibiotics associated with reversal of previously described effectiveness of activated protein C,35 and EGDT.28

Sepsis has a legacy in which improving simple processes (ie, time to antibiotics) obviates the need for more complex interventions (eg, activated protein C, EGDT). To the extent that CMS remains committed to using process-based measures of quality, those focused on sepsis are likely to be most effective when pared down to the simplest and strongest evidence base—improved recognition36 and timely antibiotics (for patients with infection-induced organ dysfunction and shock). Taking the time to start simply may best serve our current patients and preserve stakeholder buy-in for quality initiatives likely to benefit our future patients.

 

 

Disclosure

Dr. Lindenauer reports that he received support from the Centers for Medicare and Medicaid Services to develop and maintain hospital outcome measures for pneumonia and COPD. Dr. Lindenauer is supported by grant K24HL132008 from the National Heart, Lung, and Blood Institute. Dr. Walkey was supported by grants K01-HL116768 and R01-HL139751 from the National Heart, Lung, and Blood Institute.

References

1. Elixhauser A, Friedman B, Stranges E. Septicemia in U.S. Hospitals, 2009. HCUP. Statistical Brief #122. Rockville MD: Agency for Healthcare Research and Quality; 2011; p 1-13. PubMed
2. Liu V, Lei X, Prescott HC, Kipnis P, Iwashyna TJ, Escobar GJ. Hospital readmission and healthcare utilization following sepsis in community settings. J Hosp Med. 2014;9(8):502-507. PubMed
3. Liu V, Escobar GJ, Greene JD, et al. Hospital deaths in patients with sepsis from 2 independent cohorts. JAMA. 2014;312(1):90-92. PubMed
4. Peltan ID, Mitchell KH, Rudd KE, et al. Physician Variation in Time to Antimicrobial Treatment for Septic Patients Presenting to the Emergency Department. Crit Care Med. 2017;45(6):1011-1018. PubMed
5. Marik PE, Linde-Zwirble WT, Bittner EA, Sahatjian J, Hansell D. Fluid administration in severe sepsis and septic shock, patterns and outcomes: an analysis of a large national database. Intensive Care Med. 2017;43(5):625-632. PubMed
6. Lagu T, Rothberg MB, Nathanson BH, Pekow PS, Steingrub JS, Lindenauer PK. Variation in the care of septic shock: the impact of patient and hospital characteristics. J Crit Care. 2012;27(4):329-336. PubMed
7. Wang HE, Donnelly JP, Shapiro NI, Hohmann SF, Levitan EB. Hospital variations in severe sepsis mortality. Am J Med Qual. 2015;30(4):328-336. PubMed
8. Centers for Medicare & Medicaid Services. CMS Measures Inventory. https://www.cms.gov/Medicare/Quality-Initiatives-Patient-Assessment-Instruments/QualityMeasures/CMS-Measures-Inventory.html. Accessed June 8, 2017.
9. QualityNet. Specifications Manual, Version 5.0b, Section 2.2. Severe Sepsis and Septic Shock. https://www.qualitynet.org/dcs/ContentServer?c=Page&pagename=QnetPublic%2FPage%2FQnetTier4&cid=1228774725171. Accessed June 8, 2017.
10. National Quality Forum. 0500 Severe sepsis and septic shock management bundle. http://www.qualityforum.org. Accessed June 8, 2017.
11. Rivers E, Nguyen B, Havstad S, et al. Early Goal-Directed Therapy in the Treatment of Severe Sepsis and Septic Shock. N Engl J Med. 2001;345:1368-1377. PubMed
12. Levy MM, Dellinger RP, Townsend SR, et al. The Surviving Sepsis Campaign: results of an international guideline-based performance improvement program targeting severe sepsis. Crit Care Med. 2010;38(2):367-374. PubMed
13. Levy MM, Artigas A, Phillips GS, et al. Outcomes of the Surviving Sepsis Campaign in intensive care units in the USA and Europe: a prospective cohort study. Lancet Infect Dis. 2012;12(12):919-924. PubMed
14. Ferrer R, Artigas A, Levy MM, et al. Improvement in process of care and outcome after a multicenter severe sepsis educational program in Spain. JAMA. 2008;299(19):2294-2303PubMed
15. Bone RC, Balk RA, Cerra FB, et al. Definitions for sepsis and organ failure and guidelines for the use of innovative therapies in sepsis. The ACCP/SCCM Consensus Conference Committee. American College of Chest Physicians/Society of Critical Care Medicine. Chest. 1992;101(6):1644-1655. PubMed
16. Singer M, Deutschman CS, Seymour CW, et al. The Third International Consensus Definitions for Sepsis and Septic Shock (Sepsis-3). JAMA. 2016;315(8):801-810. PubMed
17. Barbash IJ, Rak KJ, Kuza CC, Kahn JM. Hospital Perceptions of Medicare’s Sepsis Quality Reporting Initiative. J Hosp Med. 2017;12(12):963-967. 
18. The PRISM Investigators. Early, Goal-Directed Therapy for Septic Shock — A Patient-Level Meta-Analysis. N Engl J Med. 2017;376:2223-2234PubMed
19. National Quality Forum. NQF Revises Sepsis Measure. http://www.qualityforum.org/NQF_Revises_Sepsis_Measure.aspx. Accessed June 8, 2017.
20. Rhodes A, Evans LE, Alhazzani W, et al. Surviving Sepsis Campaign: International Guidelines for Management of Sepsis and Septic Shock: 2016. Intensive Care Med. 2017;43(3):304-377. PubMed
21. Gould SJ. The median isn’t the message. Discover. 1985;6:40-42. PubMed
22. Hernandez G, Teboul JL. Fourth Surviving Sepsis Campaign’s hemodynamic recommendations: a step forward or a return to chaos? Crit Care. 2017;21(1):133. PubMed
23. Fugard AJ, Potts HW. Supporting thinking on sample sizes for thematic analyses. Int J Soc Res Methodol. 2015;18(6):669-684. 
24. Goff SL, Lagu T, Pekow PS, et al. A qualitative analysis of hospital leaders’ opinions about publicly reported measures of health care quality. Jt Comm J Qual Patient Saf. 2015;41(4):169-176. PubMed
25. Kumar A, Haery C, Paladugu B, et al. The duration of hypotension before the initiation of antibiotic treatment is a critical determinant of survival in a murine model of Escherichia coli septic shock: association with serum lactate and inflammatory cytokine levels. J Infect Dis. 2006;193(2):251-258.
 PubMed
26. Liu VX, Fielding-Singh V, Greene JD, et al. The Timing of Early Antibiotics and Hospital Mortality in Sepsis. Am J Respir Crit Care Med. 2017. [Epub ahead of print]. PubMed
27. Seymour CW, Gesten F, Prescott HC, et al. Time to Treatment and Mortality during Mandated Emergency Care for Sepsis. N Engl J Med. 2017;376:2235-2244PubMed
28. Kalil AC, Johnson DW, Lisco SJ, Sun J. Early Goal-Directed Therapy for Sepsis: A Novel Solution for Discordant Survival Outcomes in Clinical Trials. Crit Care Med. 2017;45(4):607-614. PubMed
29. Tu JV, Donovan LR, Lee DS, et al. Effectiveness of public report cards for improving the quality of cardiac care: the EFFECT study: a randomized trial. JAMA. 2009;302(21):2330-2337PubMed
30. Joynt KE, Blumenthal DM, Orav EJ, Resnic FS, Jha AK. Association of public reporting for percutaneous coronary intervention with utilization and outcomes among Medicare beneficiaries with acute myocardial infarction. JAMA. 2012;308(14):1460-1468. PubMed
31. Osborne NH, Nicholas LH, Ryan AM, Thumma JR, Dimick JB. Association of hospital participation in a quality reporting program with surgical outcomes and expenditures for Medicare beneficiaries. JAMA. 2015;313(5):496-504. PubMed
32. Institute of Medicine (US) Committee on Quality of Health Care in America. Crossing the Quality Chasm: A New Health System for the 21st Century. Washington (DC): National Academies Press (US); 2001. PubMed

 

 

33. 2015;12(11):1676-1684.Ann Am Thorac Soc36. Kramer RD, Cooke CR, Liu V, Miller RR 3rd, Iwashyna TJ. Variation in the Contents of Sepsis Bundles and Quality Measures. A Systematic Review. PubMed
34. 2012;40(11):2974-2981.Crit Care Med35. Rimmer E, Kumar A, Doucette S, et al. Activated protein C and septic shock: a propensity-matched cohort study*. PubMed
35. 2014;160(6):380-388.Ann Intern Med34. Rothberg MB, Pekow PS, Priya A, Lindenauer PK. Variation in diagnostic coding of patients with pneumonia and its association with hospital risk-standardized mortality rates: a cross-sectional analysis. PubMed
36. 2015;12(11):1597-1599. Ann Am Thorac Soc33. Wall MJ, Howell MD. Variation and Cost-effectiveness of Quality Measurement Programs. The Case of Sepsis Bundles. PubMed

Article PDF
Issue
Journal of Hospital Medicine 12(12)
Publications
Topics
Page Number
1019-1021
Sections
Article PDF
Article PDF

I didn’t have time to write a short letter, so I wrote a long one instead.”

-Mark Twain

Sepsis is a logical target for quality measures. Specifically, sepsis represents the perfect storm of immense public health burden1-3 combined with unexplained practice4-6 and outcomes7 variation. Thus, it is not surprising that in October 2015, the Centers of Medicare and Medicaid Services (CMS) adopted a sepsis quality measure.8 More surprising were the complex contents of the CMS Sepsis Core Measure “SEP-1” quality measure.9 CMS had written a “long letter.”

The multiple processes targeted with the CMS SEP-1 quality measure can best be understood with a brief account of history. SEP-1 arose from the National Quality Forum’s (NQF) project #0500: “Severe Sepsis and Septic Shock: Management Bundle,” a measure based upon Rivers et al.’s10 single-center, randomized, controlled trial of early goal-directed therapy (EGDT) for severe sepsis. EGDT was an intervention that consisted of fluid resuscitation and hemodynamic management based upon fulfilling specific targets of central venous pressure, superior vena cava oxygen saturation (or lactic acid), and hemoglobin and mean arterial pressures.11 The large mortality benefits, physiological rationale, and algorithmic responses to a variety of abnormal clinical values provided an appealing protocol to critical care and emergency physicians trained to normalize measured values, as well as policy makers looking for quality measures. Observational studies consistently showed associations between adoption of guideline-based “sepsis bundles” and improved patient outcomes,12-14 setting the stage for the transition of NQF #0500 into SEP-1.

However, the transition from EGDT-based NQF #0500 to SEP-1 has been tumultuous. Soon after adoption of SEP-1, the consensus definitions of sepsis changed markedly. Sepsis went from being defined as the presence of infection with concomitant systemic inflammatory response syndrome (sepsis), organ dysfunction (severe sepsis), and/or shock,15 to being defined as a dysregulated response to infection resulting in life-threatening organ dysfunction (sepsis) and/or fluid-resistant hypotension requiring vasopressors and lactate greater than 2 mmol/L.16 As the study by Barbash et al.17in this issue clearly outlines, conflicting definitions of “sepsis” have left clinicians confused regarding whom the SEP-1 measure should apply. At the same time, results of 3 large, international, randomized trials investigating the efficacy of EGDT were published, providing strong evidence that EGDT did not provide improved patient outcomes over usual care.18 SEP-1 adapted with the evolving evidence base, adding putative “usual care” processes such as evaluation of skin and peripheral pulses, and use of dynamic measures of fluid responsiveness, as quality measures.19 However, as Barbash et al. also outline, the resulting process measure was incredibly complex, with potentially more than 50 data elements collected over 6 hours in the initial management of sepsis.

In addition to its unprecedented complexity, SEP-1 received criticism for the weak evidence base of its individual components. The general concepts behind SEP-1 are well-accepted tenets of sepsis management: rapid recognition, assessment and treatment of underlying infection, and institution of intravenous fluids and vasopressor support for septic shock. However, the “all or none” prescriptive nature of the SEP-1 bundle was based on a somewhat arbitrary set of measures and targets. For example, patients with septic shock must receive 30 cc/kg of intravenous fluids to be “SEP-1 compliant.” The value “30 cc/kg” was taken from the average volume of fluids reported in prior sepsis trials, essentially based on a very low level of evidence.20 The strict 30 cc/kg cutoff did not take into account that “the median isn’t the message”21 in fluid management: optimal resuscitation targets are unclear,22 and selecting the median as a target ignores the fact that 50% of patients enrolled in international trials of EGDT received less than 30 cc/kg of initial fluid resuscitation (the interquartile range was 16-42 cc/kg).18 Thus, most participants in trials upon which the SEP-1 fluid measure was based would ironically not have met the SEP-1 measure. Mandates for physical exam and physiological measures were based on similarly low levels of evidence.

Into this context, Barbash et al. use a representative sample of US hospitals to explore the opinions of hospital quality leaders regarding the SEP-1 measure. First, the qualitative methods used by Barbash et al. warrant some explanation. Much of biomedical research is characterized by hypothesis-driven, deductive reasoning: theories are tested using observations. In contrast, the methods of Barbash et al. use inductive reasoning: observations are used to develop theories within a systematic approach called “grounded theory” that explores common themes emerging from structured interviews.23 Inductive reasoning can later inform deductive reasoning, feeding theories into testable hypotheses. However, qualitative, inductive research is not meant to test hypotheses and is not subject to typical notions of “power and sample size” often expected of quantitative statistical analyses. Qualitative studies reach sufficient sample size when no further themes emerge, a situation called “thematic saturation”; the sample size here of 29 participants rests comfortably in the range of participants commonly needed for thematic saturation.23

Barbash et al. identified common themes in opinions of quality leaders regarding SEP-1. Namely, the complexity of SEP-1 necessitated a major resource investment into sepsis care and data collection. The major infrastructure investments needed to comply with SEP-1 also bred frustration regarding lack of perceived fairness around the “all or none” nature of the measure and raised multiple additional challenges including lack of clinician buy-in and resistance to protocolized care. Prior qualitative studies evaluating hospital quality leaders’ opinions on performance measures identified similar concerns about lack of “fairness,”24 but the implementation of SEP-1 has raised additional concern regarding the large burdens of instituting major infrastructure changes to monitor processes of care required to report on this measure. Despite the major challenges of responding to SEP-1, quality leaders expressed optimism that increased attention to sepsis would ultimately lead to better patient outcomes.

How might future sepsis quality measures achieve the adequate balance between focusing attention on improving care processes for high-impact diseases, without imposing additional burdens on the healthcare system? Lessons from Barbash et al. help us move forward. First, rather than taxing hospitals with administratively complex process measures, initial attempts at quality measures should start simply. Policy makers should consider moving forward into new areas of quality measurement in 2 ways: (1) pursue 1 or 2 processes with strong etiological links to important patient outcomes (eg, timely antibiotics in septic shock),25-28 and/or (2) use risk-adjusted outcomes and allow individual hospitals to adopt processes that improve local patient outcomes. Evidence suggests that the introduction of a quality measure may result in improved outcomes regardless of adoption of specific target processes,29 although results are mixed.30,31 In either case, complex “all or none” measures based upon weak evidence run a high risk of inciting clinician resentment and paradoxically perpetuating poor quality by increasing healthcare costs (decreased efficiency) without gains in safety, effectiveness, timeliness, or equity.32 It has been estimated that hospitals spend on average $2 million to implement SEP-1,33 with unclear return on the investment. The experience of SEP-1 is a reminder that, as evidence evolves, quality measures must adapt lest they become irrelevant. However, it is also a reminder that quality measures should not sit precariously on the edge of evidence. Withdrawal of process-based measures due to a changing evidence landscape breeds mistrust and impairs future attempts to improve quality.

Sepsis quality measures face additional challenges. If recent experience with interpretation of sepsis definitions can serve as a guide, variable uptake of newer sepsis definitions between/across hospitals will impair the ability to risk-adjust outcome measures and increase bias in identifying outlier hospitals.34 In addition, recent studies have already raised skepticism regarding the effectiveness of individual SEP-1 bundle components, confirming suspicions that the 30 cc/kg fluid bolus is not a magic quality target. Rather, the effectiveness of prior sepsis bundles has likely been driven by improved time to antibiotics, a process unstudied in sepsis trials, but driven by increased attention to the importance of early sepsis recognition and timely management.28 Timeliness of antibiotics can act as an effect modifier for more complex sepsis therapies, with quicker time to antibiotics associated with reversal of previously described effectiveness of activated protein C,35 and EGDT.28

Sepsis has a legacy in which improving simple processes (ie, time to antibiotics) obviates the need for more complex interventions (eg, activated protein C, EGDT). To the extent that CMS remains committed to using process-based measures of quality, those focused on sepsis are likely to be most effective when pared down to the simplest and strongest evidence base—improved recognition36 and timely antibiotics (for patients with infection-induced organ dysfunction and shock). Taking the time to start simply may best serve our current patients and preserve stakeholder buy-in for quality initiatives likely to benefit our future patients.

 

 

Disclosure

Dr. Lindenauer reports that he received support from the Centers for Medicare and Medicaid Services to develop and maintain hospital outcome measures for pneumonia and COPD. Dr. Lindenauer is supported by grant K24HL132008 from the National Heart, Lung, and Blood Institute. Dr. Walkey was supported by grants K01-HL116768 and R01-HL139751 from the National Heart, Lung, and Blood Institute.

I didn’t have time to write a short letter, so I wrote a long one instead.”

-Mark Twain

Sepsis is a logical target for quality measures. Specifically, sepsis represents the perfect storm of immense public health burden1-3 combined with unexplained practice4-6 and outcomes7 variation. Thus, it is not surprising that in October 2015, the Centers of Medicare and Medicaid Services (CMS) adopted a sepsis quality measure.8 More surprising were the complex contents of the CMS Sepsis Core Measure “SEP-1” quality measure.9 CMS had written a “long letter.”

The multiple processes targeted with the CMS SEP-1 quality measure can best be understood with a brief account of history. SEP-1 arose from the National Quality Forum’s (NQF) project #0500: “Severe Sepsis and Septic Shock: Management Bundle,” a measure based upon Rivers et al.’s10 single-center, randomized, controlled trial of early goal-directed therapy (EGDT) for severe sepsis. EGDT was an intervention that consisted of fluid resuscitation and hemodynamic management based upon fulfilling specific targets of central venous pressure, superior vena cava oxygen saturation (or lactic acid), and hemoglobin and mean arterial pressures.11 The large mortality benefits, physiological rationale, and algorithmic responses to a variety of abnormal clinical values provided an appealing protocol to critical care and emergency physicians trained to normalize measured values, as well as policy makers looking for quality measures. Observational studies consistently showed associations between adoption of guideline-based “sepsis bundles” and improved patient outcomes,12-14 setting the stage for the transition of NQF #0500 into SEP-1.

However, the transition from EGDT-based NQF #0500 to SEP-1 has been tumultuous. Soon after adoption of SEP-1, the consensus definitions of sepsis changed markedly. Sepsis went from being defined as the presence of infection with concomitant systemic inflammatory response syndrome (sepsis), organ dysfunction (severe sepsis), and/or shock,15 to being defined as a dysregulated response to infection resulting in life-threatening organ dysfunction (sepsis) and/or fluid-resistant hypotension requiring vasopressors and lactate greater than 2 mmol/L.16 As the study by Barbash et al.17in this issue clearly outlines, conflicting definitions of “sepsis” have left clinicians confused regarding whom the SEP-1 measure should apply. At the same time, results of 3 large, international, randomized trials investigating the efficacy of EGDT were published, providing strong evidence that EGDT did not provide improved patient outcomes over usual care.18 SEP-1 adapted with the evolving evidence base, adding putative “usual care” processes such as evaluation of skin and peripheral pulses, and use of dynamic measures of fluid responsiveness, as quality measures.19 However, as Barbash et al. also outline, the resulting process measure was incredibly complex, with potentially more than 50 data elements collected over 6 hours in the initial management of sepsis.

In addition to its unprecedented complexity, SEP-1 received criticism for the weak evidence base of its individual components. The general concepts behind SEP-1 are well-accepted tenets of sepsis management: rapid recognition, assessment and treatment of underlying infection, and institution of intravenous fluids and vasopressor support for septic shock. However, the “all or none” prescriptive nature of the SEP-1 bundle was based on a somewhat arbitrary set of measures and targets. For example, patients with septic shock must receive 30 cc/kg of intravenous fluids to be “SEP-1 compliant.” The value “30 cc/kg” was taken from the average volume of fluids reported in prior sepsis trials, essentially based on a very low level of evidence.20 The strict 30 cc/kg cutoff did not take into account that “the median isn’t the message”21 in fluid management: optimal resuscitation targets are unclear,22 and selecting the median as a target ignores the fact that 50% of patients enrolled in international trials of EGDT received less than 30 cc/kg of initial fluid resuscitation (the interquartile range was 16-42 cc/kg).18 Thus, most participants in trials upon which the SEP-1 fluid measure was based would ironically not have met the SEP-1 measure. Mandates for physical exam and physiological measures were based on similarly low levels of evidence.

Into this context, Barbash et al. use a representative sample of US hospitals to explore the opinions of hospital quality leaders regarding the SEP-1 measure. First, the qualitative methods used by Barbash et al. warrant some explanation. Much of biomedical research is characterized by hypothesis-driven, deductive reasoning: theories are tested using observations. In contrast, the methods of Barbash et al. use inductive reasoning: observations are used to develop theories within a systematic approach called “grounded theory” that explores common themes emerging from structured interviews.23 Inductive reasoning can later inform deductive reasoning, feeding theories into testable hypotheses. However, qualitative, inductive research is not meant to test hypotheses and is not subject to typical notions of “power and sample size” often expected of quantitative statistical analyses. Qualitative studies reach sufficient sample size when no further themes emerge, a situation called “thematic saturation”; the sample size here of 29 participants rests comfortably in the range of participants commonly needed for thematic saturation.23

Barbash et al. identified common themes in opinions of quality leaders regarding SEP-1. Namely, the complexity of SEP-1 necessitated a major resource investment into sepsis care and data collection. The major infrastructure investments needed to comply with SEP-1 also bred frustration regarding lack of perceived fairness around the “all or none” nature of the measure and raised multiple additional challenges including lack of clinician buy-in and resistance to protocolized care. Prior qualitative studies evaluating hospital quality leaders’ opinions on performance measures identified similar concerns about lack of “fairness,”24 but the implementation of SEP-1 has raised additional concern regarding the large burdens of instituting major infrastructure changes to monitor processes of care required to report on this measure. Despite the major challenges of responding to SEP-1, quality leaders expressed optimism that increased attention to sepsis would ultimately lead to better patient outcomes.

How might future sepsis quality measures achieve the adequate balance between focusing attention on improving care processes for high-impact diseases, without imposing additional burdens on the healthcare system? Lessons from Barbash et al. help us move forward. First, rather than taxing hospitals with administratively complex process measures, initial attempts at quality measures should start simply. Policy makers should consider moving forward into new areas of quality measurement in 2 ways: (1) pursue 1 or 2 processes with strong etiological links to important patient outcomes (eg, timely antibiotics in septic shock),25-28 and/or (2) use risk-adjusted outcomes and allow individual hospitals to adopt processes that improve local patient outcomes. Evidence suggests that the introduction of a quality measure may result in improved outcomes regardless of adoption of specific target processes,29 although results are mixed.30,31 In either case, complex “all or none” measures based upon weak evidence run a high risk of inciting clinician resentment and paradoxically perpetuating poor quality by increasing healthcare costs (decreased efficiency) without gains in safety, effectiveness, timeliness, or equity.32 It has been estimated that hospitals spend on average $2 million to implement SEP-1,33 with unclear return on the investment. The experience of SEP-1 is a reminder that, as evidence evolves, quality measures must adapt lest they become irrelevant. However, it is also a reminder that quality measures should not sit precariously on the edge of evidence. Withdrawal of process-based measures due to a changing evidence landscape breeds mistrust and impairs future attempts to improve quality.

Sepsis quality measures face additional challenges. If recent experience with interpretation of sepsis definitions can serve as a guide, variable uptake of newer sepsis definitions between/across hospitals will impair the ability to risk-adjust outcome measures and increase bias in identifying outlier hospitals.34 In addition, recent studies have already raised skepticism regarding the effectiveness of individual SEP-1 bundle components, confirming suspicions that the 30 cc/kg fluid bolus is not a magic quality target. Rather, the effectiveness of prior sepsis bundles has likely been driven by improved time to antibiotics, a process unstudied in sepsis trials, but driven by increased attention to the importance of early sepsis recognition and timely management.28 Timeliness of antibiotics can act as an effect modifier for more complex sepsis therapies, with quicker time to antibiotics associated with reversal of previously described effectiveness of activated protein C,35 and EGDT.28

Sepsis has a legacy in which improving simple processes (ie, time to antibiotics) obviates the need for more complex interventions (eg, activated protein C, EGDT). To the extent that CMS remains committed to using process-based measures of quality, those focused on sepsis are likely to be most effective when pared down to the simplest and strongest evidence base—improved recognition36 and timely antibiotics (for patients with infection-induced organ dysfunction and shock). Taking the time to start simply may best serve our current patients and preserve stakeholder buy-in for quality initiatives likely to benefit our future patients.

 

 

Disclosure

Dr. Lindenauer reports that he received support from the Centers for Medicare and Medicaid Services to develop and maintain hospital outcome measures for pneumonia and COPD. Dr. Lindenauer is supported by grant K24HL132008 from the National Heart, Lung, and Blood Institute. Dr. Walkey was supported by grants K01-HL116768 and R01-HL139751 from the National Heart, Lung, and Blood Institute.

References

1. Elixhauser A, Friedman B, Stranges E. Septicemia in U.S. Hospitals, 2009. HCUP. Statistical Brief #122. Rockville MD: Agency for Healthcare Research and Quality; 2011; p 1-13. PubMed
2. Liu V, Lei X, Prescott HC, Kipnis P, Iwashyna TJ, Escobar GJ. Hospital readmission and healthcare utilization following sepsis in community settings. J Hosp Med. 2014;9(8):502-507. PubMed
3. Liu V, Escobar GJ, Greene JD, et al. Hospital deaths in patients with sepsis from 2 independent cohorts. JAMA. 2014;312(1):90-92. PubMed
4. Peltan ID, Mitchell KH, Rudd KE, et al. Physician Variation in Time to Antimicrobial Treatment for Septic Patients Presenting to the Emergency Department. Crit Care Med. 2017;45(6):1011-1018. PubMed
5. Marik PE, Linde-Zwirble WT, Bittner EA, Sahatjian J, Hansell D. Fluid administration in severe sepsis and septic shock, patterns and outcomes: an analysis of a large national database. Intensive Care Med. 2017;43(5):625-632. PubMed
6. Lagu T, Rothberg MB, Nathanson BH, Pekow PS, Steingrub JS, Lindenauer PK. Variation in the care of septic shock: the impact of patient and hospital characteristics. J Crit Care. 2012;27(4):329-336. PubMed
7. Wang HE, Donnelly JP, Shapiro NI, Hohmann SF, Levitan EB. Hospital variations in severe sepsis mortality. Am J Med Qual. 2015;30(4):328-336. PubMed
8. Centers for Medicare & Medicaid Services. CMS Measures Inventory. https://www.cms.gov/Medicare/Quality-Initiatives-Patient-Assessment-Instruments/QualityMeasures/CMS-Measures-Inventory.html. Accessed June 8, 2017.
9. QualityNet. Specifications Manual, Version 5.0b, Section 2.2. Severe Sepsis and Septic Shock. https://www.qualitynet.org/dcs/ContentServer?c=Page&pagename=QnetPublic%2FPage%2FQnetTier4&cid=1228774725171. Accessed June 8, 2017.
10. National Quality Forum. 0500 Severe sepsis and septic shock management bundle. http://www.qualityforum.org. Accessed June 8, 2017.
11. Rivers E, Nguyen B, Havstad S, et al. Early Goal-Directed Therapy in the Treatment of Severe Sepsis and Septic Shock. N Engl J Med. 2001;345:1368-1377. PubMed
12. Levy MM, Dellinger RP, Townsend SR, et al. The Surviving Sepsis Campaign: results of an international guideline-based performance improvement program targeting severe sepsis. Crit Care Med. 2010;38(2):367-374. PubMed
13. Levy MM, Artigas A, Phillips GS, et al. Outcomes of the Surviving Sepsis Campaign in intensive care units in the USA and Europe: a prospective cohort study. Lancet Infect Dis. 2012;12(12):919-924. PubMed
14. Ferrer R, Artigas A, Levy MM, et al. Improvement in process of care and outcome after a multicenter severe sepsis educational program in Spain. JAMA. 2008;299(19):2294-2303PubMed
15. Bone RC, Balk RA, Cerra FB, et al. Definitions for sepsis and organ failure and guidelines for the use of innovative therapies in sepsis. The ACCP/SCCM Consensus Conference Committee. American College of Chest Physicians/Society of Critical Care Medicine. Chest. 1992;101(6):1644-1655. PubMed
16. Singer M, Deutschman CS, Seymour CW, et al. The Third International Consensus Definitions for Sepsis and Septic Shock (Sepsis-3). JAMA. 2016;315(8):801-810. PubMed
17. Barbash IJ, Rak KJ, Kuza CC, Kahn JM. Hospital Perceptions of Medicare’s Sepsis Quality Reporting Initiative. J Hosp Med. 2017;12(12):963-967. 
18. The PRISM Investigators. Early, Goal-Directed Therapy for Septic Shock — A Patient-Level Meta-Analysis. N Engl J Med. 2017;376:2223-2234PubMed
19. National Quality Forum. NQF Revises Sepsis Measure. http://www.qualityforum.org/NQF_Revises_Sepsis_Measure.aspx. Accessed June 8, 2017.
20. Rhodes A, Evans LE, Alhazzani W, et al. Surviving Sepsis Campaign: International Guidelines for Management of Sepsis and Septic Shock: 2016. Intensive Care Med. 2017;43(3):304-377. PubMed
21. Gould SJ. The median isn’t the message. Discover. 1985;6:40-42. PubMed
22. Hernandez G, Teboul JL. Fourth Surviving Sepsis Campaign’s hemodynamic recommendations: a step forward or a return to chaos? Crit Care. 2017;21(1):133. PubMed
23. Fugard AJ, Potts HW. Supporting thinking on sample sizes for thematic analyses. Int J Soc Res Methodol. 2015;18(6):669-684. 
24. Goff SL, Lagu T, Pekow PS, et al. A qualitative analysis of hospital leaders’ opinions about publicly reported measures of health care quality. Jt Comm J Qual Patient Saf. 2015;41(4):169-176. PubMed
25. Kumar A, Haery C, Paladugu B, et al. The duration of hypotension before the initiation of antibiotic treatment is a critical determinant of survival in a murine model of Escherichia coli septic shock: association with serum lactate and inflammatory cytokine levels. J Infect Dis. 2006;193(2):251-258.
 PubMed
26. Liu VX, Fielding-Singh V, Greene JD, et al. The Timing of Early Antibiotics and Hospital Mortality in Sepsis. Am J Respir Crit Care Med. 2017. [Epub ahead of print]. PubMed
27. Seymour CW, Gesten F, Prescott HC, et al. Time to Treatment and Mortality during Mandated Emergency Care for Sepsis. N Engl J Med. 2017;376:2235-2244PubMed
28. Kalil AC, Johnson DW, Lisco SJ, Sun J. Early Goal-Directed Therapy for Sepsis: A Novel Solution for Discordant Survival Outcomes in Clinical Trials. Crit Care Med. 2017;45(4):607-614. PubMed
29. Tu JV, Donovan LR, Lee DS, et al. Effectiveness of public report cards for improving the quality of cardiac care: the EFFECT study: a randomized trial. JAMA. 2009;302(21):2330-2337PubMed
30. Joynt KE, Blumenthal DM, Orav EJ, Resnic FS, Jha AK. Association of public reporting for percutaneous coronary intervention with utilization and outcomes among Medicare beneficiaries with acute myocardial infarction. JAMA. 2012;308(14):1460-1468. PubMed
31. Osborne NH, Nicholas LH, Ryan AM, Thumma JR, Dimick JB. Association of hospital participation in a quality reporting program with surgical outcomes and expenditures for Medicare beneficiaries. JAMA. 2015;313(5):496-504. PubMed
32. Institute of Medicine (US) Committee on Quality of Health Care in America. Crossing the Quality Chasm: A New Health System for the 21st Century. Washington (DC): National Academies Press (US); 2001. PubMed

 

 

33. 2015;12(11):1676-1684.Ann Am Thorac Soc36. Kramer RD, Cooke CR, Liu V, Miller RR 3rd, Iwashyna TJ. Variation in the Contents of Sepsis Bundles and Quality Measures. A Systematic Review. PubMed
34. 2012;40(11):2974-2981.Crit Care Med35. Rimmer E, Kumar A, Doucette S, et al. Activated protein C and septic shock: a propensity-matched cohort study*. PubMed
35. 2014;160(6):380-388.Ann Intern Med34. Rothberg MB, Pekow PS, Priya A, Lindenauer PK. Variation in diagnostic coding of patients with pneumonia and its association with hospital risk-standardized mortality rates: a cross-sectional analysis. PubMed
36. 2015;12(11):1597-1599. Ann Am Thorac Soc33. Wall MJ, Howell MD. Variation and Cost-effectiveness of Quality Measurement Programs. The Case of Sepsis Bundles. PubMed

References

1. Elixhauser A, Friedman B, Stranges E. Septicemia in U.S. Hospitals, 2009. HCUP. Statistical Brief #122. Rockville MD: Agency for Healthcare Research and Quality; 2011; p 1-13. PubMed
2. Liu V, Lei X, Prescott HC, Kipnis P, Iwashyna TJ, Escobar GJ. Hospital readmission and healthcare utilization following sepsis in community settings. J Hosp Med. 2014;9(8):502-507. PubMed
3. Liu V, Escobar GJ, Greene JD, et al. Hospital deaths in patients with sepsis from 2 independent cohorts. JAMA. 2014;312(1):90-92. PubMed
4. Peltan ID, Mitchell KH, Rudd KE, et al. Physician Variation in Time to Antimicrobial Treatment for Septic Patients Presenting to the Emergency Department. Crit Care Med. 2017;45(6):1011-1018. PubMed
5. Marik PE, Linde-Zwirble WT, Bittner EA, Sahatjian J, Hansell D. Fluid administration in severe sepsis and septic shock, patterns and outcomes: an analysis of a large national database. Intensive Care Med. 2017;43(5):625-632. PubMed
6. Lagu T, Rothberg MB, Nathanson BH, Pekow PS, Steingrub JS, Lindenauer PK. Variation in the care of septic shock: the impact of patient and hospital characteristics. J Crit Care. 2012;27(4):329-336. PubMed
7. Wang HE, Donnelly JP, Shapiro NI, Hohmann SF, Levitan EB. Hospital variations in severe sepsis mortality. Am J Med Qual. 2015;30(4):328-336. PubMed
8. Centers for Medicare & Medicaid Services. CMS Measures Inventory. https://www.cms.gov/Medicare/Quality-Initiatives-Patient-Assessment-Instruments/QualityMeasures/CMS-Measures-Inventory.html. Accessed June 8, 2017.
9. QualityNet. Specifications Manual, Version 5.0b, Section 2.2. Severe Sepsis and Septic Shock. https://www.qualitynet.org/dcs/ContentServer?c=Page&pagename=QnetPublic%2FPage%2FQnetTier4&cid=1228774725171. Accessed June 8, 2017.
10. National Quality Forum. 0500 Severe sepsis and septic shock management bundle. http://www.qualityforum.org. Accessed June 8, 2017.
11. Rivers E, Nguyen B, Havstad S, et al. Early Goal-Directed Therapy in the Treatment of Severe Sepsis and Septic Shock. N Engl J Med. 2001;345:1368-1377. PubMed
12. Levy MM, Dellinger RP, Townsend SR, et al. The Surviving Sepsis Campaign: results of an international guideline-based performance improvement program targeting severe sepsis. Crit Care Med. 2010;38(2):367-374. PubMed
13. Levy MM, Artigas A, Phillips GS, et al. Outcomes of the Surviving Sepsis Campaign in intensive care units in the USA and Europe: a prospective cohort study. Lancet Infect Dis. 2012;12(12):919-924. PubMed
14. Ferrer R, Artigas A, Levy MM, et al. Improvement in process of care and outcome after a multicenter severe sepsis educational program in Spain. JAMA. 2008;299(19):2294-2303PubMed
15. Bone RC, Balk RA, Cerra FB, et al. Definitions for sepsis and organ failure and guidelines for the use of innovative therapies in sepsis. The ACCP/SCCM Consensus Conference Committee. American College of Chest Physicians/Society of Critical Care Medicine. Chest. 1992;101(6):1644-1655. PubMed
16. Singer M, Deutschman CS, Seymour CW, et al. The Third International Consensus Definitions for Sepsis and Septic Shock (Sepsis-3). JAMA. 2016;315(8):801-810. PubMed
17. Barbash IJ, Rak KJ, Kuza CC, Kahn JM. Hospital Perceptions of Medicare’s Sepsis Quality Reporting Initiative. J Hosp Med. 2017;12(12):963-967. 
18. The PRISM Investigators. Early, Goal-Directed Therapy for Septic Shock — A Patient-Level Meta-Analysis. N Engl J Med. 2017;376:2223-2234PubMed
19. National Quality Forum. NQF Revises Sepsis Measure. http://www.qualityforum.org/NQF_Revises_Sepsis_Measure.aspx. Accessed June 8, 2017.
20. Rhodes A, Evans LE, Alhazzani W, et al. Surviving Sepsis Campaign: International Guidelines for Management of Sepsis and Septic Shock: 2016. Intensive Care Med. 2017;43(3):304-377. PubMed
21. Gould SJ. The median isn’t the message. Discover. 1985;6:40-42. PubMed
22. Hernandez G, Teboul JL. Fourth Surviving Sepsis Campaign’s hemodynamic recommendations: a step forward or a return to chaos? Crit Care. 2017;21(1):133. PubMed
23. Fugard AJ, Potts HW. Supporting thinking on sample sizes for thematic analyses. Int J Soc Res Methodol. 2015;18(6):669-684. 
24. Goff SL, Lagu T, Pekow PS, et al. A qualitative analysis of hospital leaders’ opinions about publicly reported measures of health care quality. Jt Comm J Qual Patient Saf. 2015;41(4):169-176. PubMed
25. Kumar A, Haery C, Paladugu B, et al. The duration of hypotension before the initiation of antibiotic treatment is a critical determinant of survival in a murine model of Escherichia coli septic shock: association with serum lactate and inflammatory cytokine levels. J Infect Dis. 2006;193(2):251-258.
 PubMed
26. Liu VX, Fielding-Singh V, Greene JD, et al. The Timing of Early Antibiotics and Hospital Mortality in Sepsis. Am J Respir Crit Care Med. 2017. [Epub ahead of print]. PubMed
27. Seymour CW, Gesten F, Prescott HC, et al. Time to Treatment and Mortality during Mandated Emergency Care for Sepsis. N Engl J Med. 2017;376:2235-2244PubMed
28. Kalil AC, Johnson DW, Lisco SJ, Sun J. Early Goal-Directed Therapy for Sepsis: A Novel Solution for Discordant Survival Outcomes in Clinical Trials. Crit Care Med. 2017;45(4):607-614. PubMed
29. Tu JV, Donovan LR, Lee DS, et al. Effectiveness of public report cards for improving the quality of cardiac care: the EFFECT study: a randomized trial. JAMA. 2009;302(21):2330-2337PubMed
30. Joynt KE, Blumenthal DM, Orav EJ, Resnic FS, Jha AK. Association of public reporting for percutaneous coronary intervention with utilization and outcomes among Medicare beneficiaries with acute myocardial infarction. JAMA. 2012;308(14):1460-1468. PubMed
31. Osborne NH, Nicholas LH, Ryan AM, Thumma JR, Dimick JB. Association of hospital participation in a quality reporting program with surgical outcomes and expenditures for Medicare beneficiaries. JAMA. 2015;313(5):496-504. PubMed
32. Institute of Medicine (US) Committee on Quality of Health Care in America. Crossing the Quality Chasm: A New Health System for the 21st Century. Washington (DC): National Academies Press (US); 2001. PubMed

 

 

33. 2015;12(11):1676-1684.Ann Am Thorac Soc36. Kramer RD, Cooke CR, Liu V, Miller RR 3rd, Iwashyna TJ. Variation in the Contents of Sepsis Bundles and Quality Measures. A Systematic Review. PubMed
34. 2012;40(11):2974-2981.Crit Care Med35. Rimmer E, Kumar A, Doucette S, et al. Activated protein C and septic shock: a propensity-matched cohort study*. PubMed
35. 2014;160(6):380-388.Ann Intern Med34. Rothberg MB, Pekow PS, Priya A, Lindenauer PK. Variation in diagnostic coding of patients with pneumonia and its association with hospital risk-standardized mortality rates: a cross-sectional analysis. PubMed
36. 2015;12(11):1597-1599. Ann Am Thorac Soc33. Wall MJ, Howell MD. Variation and Cost-effectiveness of Quality Measurement Programs. The Case of Sepsis Bundles. PubMed

Issue
Journal of Hospital Medicine 12(12)
Issue
Journal of Hospital Medicine 12(12)
Page Number
1019-1021
Page Number
1019-1021
Publications
Publications
Topics
Article Type
Sections
Article Source

©2017 Society of Hospital Medicine

Disallow All Ads
Correspondence Location
Peter K. Lindenauer, MD, MSc, Institute for Healthcare Delivery and Population Science and Department of Medicine, University of Massachusetts Medical School-Baystate, 3601 Main Street, Springfield, MA, 01199; Telephone: 413-794-5987; Fax: 413-794-8866; E-mail: peter.lindenauer@baystatehealth.org
Content Gating
Open Access (article Unlocked/Open Access)
Alternative CME
Disqus Comments
Default
Use ProPublica
Article PDF Media

Treatment Trends and Outcomes in Healthcare-Associated Pneumonia

Article Type
Changed
Fri, 12/14/2018 - 07:45

Bacterial pneumonia remains an important cause of morbidity and mortality in the United States, and is the 8th leading cause of death with 55,227 deaths among adults annually.1 In 2005, the American Thoracic Society (ATS) and the Infectious Diseases Society of America (IDSA) collaborated to update guidelines for hospital-acquired pneumonia (HAP), ventilator-associated pneumonia, and healthcare-associated pneumonia (HCAP).2 This broad document outlines an evidence-based approach to diagnostic testing and antibiotic management based on the epidemiology and risk factors for these conditions. The guideline specifies the following criteria for HCAP: hospitalization in the past 90 days, residence in a skilled nursing facility (SNF), home infusion therapy, hemodialysis, home wound care, family members with multidrug resistant organisms (MDRO), and immunosuppressive diseases or medications, with the presumption that these patients are more likely to be harboring MDRO and should thus be treated empirically with broad-spectrum antibiotic therapy. Prior studies have shown that patients with HCAP have a more severe illness, are more likely to have MDRO, are more likely to be inadequately treated, and are at a higher risk for mortality than patients with community-acquired pneumonia (CAP).3,4

These guidelines are controversial, especially in regard to the recommendations to empirically treat broadly with 2 antibiotics targeting Pseudomonas species, whether patients with HCAP merit broader spectrum coverage than patients with CAP, and whether the criteria for defining HCAP are adequate to predict which patients are harboring MDRO. It has subsequently been proposed that HCAP is more related to CAP than to HAP, and a recent update to the guideline removed recommendations for treatment of HCAP and will be placing HCAP into the guidelines for CAP instead.5 We sought to investigate the degree of uptake of the ATS and IDSA guideline recommendations by physicians over time, and whether this led to a change in outcomes among patients who met the criteria for HCAP.

METHODS

Setting and Patients

We identified patients discharged between July 1, 2007, and November 30, 2011, from 488 US hospitals that participated in the Premier database (Premier Inc., Charlotte, North Carolina), an inpatient database developed for measuring quality and healthcare utilization. The database is frequently used for healthcare research and has been described previously.6 Member hospitals are in all regions of the US and are generally reflective of US hospitals. This database contains multiple data elements, including sociodemographic information, International Classification of Diseases, 9th Revision-Clinical Modification (ICD-9-CM) diagnosis and procedure codes, hospital and physician information, source of admission, and discharge status. It also includes a date-stamped log of all billed items and services, including diagnostic tests, medications, and other treatments. Because the data do not contain identifiable information, the institutional review board at our medical center determined that this study did not constitute human subjects research.

We included all patients aged ≥18 years with a principal diagnosis of pneumonia or with a secondary diagnosis of pneumonia paired with a principal diagnosis of respiratory failure, acute respiratory distress syndrome, respiratory arrest, sepsis, or influenza. Patients were excluded if they were transferred to or from another acute care institution, had a length of stay of 1 day or less, had cystic fibrosis, did not have a chest radiograph, or did not receive antibiotics within 48 hours of admission.

For each patient, we extracted age, gender, principal diagnosis, comorbidities, and the specialty of the attending physician. Comorbidities were identified from ICD-9-CM secondary diagnosis codes and Diagnosis Related Groups by using Healthcare Cost and Utilization Project Comorbidity Software, version 3.1, based on the work of Elixhauser (Agency for Healthcare Research and Quality, Rockville, Maryland).7 In order to ensure that patients had HCAP, we required the presence of ≥1 HCAP criteria, including hospitalization in the past 90 days, hemodialysis, admission from an SNF, or immune suppression (which was derived from either a secondary diagnosis for neutropenia, hematological malignancy, organ transplant, acquired immunodeficiency virus, or receiving immunosuppressant drugs or corticosteroids [equivalent to ≥20 mg/day of prednisone]).

 

 

Definitions of Guideline-Concordant and Discordant Antibiotic Therapy

The ATS and IDSA guidelines recommended the following antibiotic combinations for HCAP: an antipseudomonal cephalosporin or carbapenem or a beta-lactam/lactamase inhibitor, plus an antipseudomonal quinolone or aminoglycoside, plus an antibiotic with activity versus methicillin resistant Staphylococcus aureus (MRSA), such as vancomycin or linezolid. Based on these guidelines, we defined the receipt of fully guideline-concordant antibiotics as 2 recommended antibiotics for Pseudomonas species plus 1 for MRSA administered by the second day of admission. Partially guideline-concordant antibiotics were defined as 1 recommended antibiotic for Pseudomonas species plus 1 for MRSA by the second day of hospitalization. Guideline-discordant antibiotics were defined as all other combinations.

Statistical Analysis

Descriptive statistics on patient characteristics are presented as frequency, proportions for categorical factors, and median with interquartile range (IQR) for continuous variables for the full cohort and by treatment group, defined as fully or partially guideline-concordant antibiotic therapy or discordant therapy. Hospital rates of fully guideline-concordant treatment are presented overall and by hospital characteristics. The association of hospital characteristics with rates of fully guideline-concordant therapy were assessed by using 1-way analysis of variance tests.

To assess trends across hospitals for the association between the use of guideline-concordant therapy and mortality, progression to respiratory failure as measured by the late initiation of invasive mechanical ventilation (day 3 or later), and the length of stay among survivors, we divided the 4.5-year study period into 9 intervals of 6 months each; 292 hospitals that submitted data for all 9 time points were examined in this analysis. Based on the distribution of length of stay in the first time period, we created an indicator variable for extended length of stay with length of stay at or above the 75th percentile, defined as extended. For each hospital at each 6-month interval, we then computed risk-standardized guideline-concordant treatment (RS-treatment) rates and risk-standardized in-hospital outcome rates similar to methods used by the Centers for Medicare and Medicaid Services for public reporting.8 For each hospital at each time interval, we estimated a predicted rate of guideline-concordant treatment as the sum of predicted probabilities of guideline-concordant treatment from patient factors and the random intercept for the hospital in which they were admitted. We then calculated the expected rate of guideline-concordant treatment as the sum of expected probabilities of treatment received from patient factors only. RS-treatment was then calculated as the ratio of predicted to expected rates multiplied by the overall unadjusted mean treatment rate from all patients.9 We repeated the same modeling strategy to calculate risk-standardized outcome (RS-outcome) rates for each hospital across all time points. All models were adjusted for patient demographics and comorbidities. Similar models using administrative data have moderate discrimination for mortality.10

We then fit mixed-effects linear models with random hospital intercept and slope across time for the RS-treatment and outcome rates, respectively. From these models, we estimated the mean slope for RS-treatment and for RS-outcome over time. In addition, we estimated a slope or trend over time for each hospital for treatment and for outcome and evaluated the correlation between the treatment and outcome trends.

All analyses were performed using the Statistical Analysis System version 9.4 (SAS Institute Inc., Cary, NC) and STATA release 13 (StataCorp, LLC, College Station, Texas).

RESULTS

Of 1,601,064 patients with a diagnosis of pneumonia in our dataset, 436,483 patients met our inclusion criteria, and of those, 149,963 (34.4%) met at least 1 HCAP criterion and were included as our study cohort (supplementary Figure). Among the study cohort, the median age was 73 years (IQR, 61-83), 51.4% of patients were female, 69.6% of patients were white, and a majority of patients (76.2 %) were covered by Medicare. HCAP categories included hospitalization in the past 90 days (63.1%), hemodialysis (12.8%), admission from a SNF (23.6%), and immunosuppression (28.9%). One-quarter of the patients were treated in the intensive care unit (ICU) by day 2 of their hospitalization. The most common comorbidities were hypertension (65.1%), chronic obstructive pulmonary disease (47.3%), anemia (40.9%), diabetes (36.6%), and congestive heart failure (35.7%). Pneumonia was the principal diagnosis in 61.9% of patients, and sepsis was the principal diagnosis in 29.3% of patients. The unadjusted median length of stay was 6 days, the median cost was $10,049, and the in-hospital mortality was 11.1%. Patients who received fully or partially guideline-concordant antibiotics were younger on average and had a higher combined comorbidity score, and they were more likely to have been admitted to the ICU and to have received vasopressor medications and mechanical ventilation. They also had higher unadjusted mortality, longer lengths of stay, and higher costs (see supplemental Table 1 for more details).

 

 

The Table shows the antibiotics received by patients. Overall, 19.6% of patients received fully guideline-concordant treatment, 21.7% received partially guideline-concordant treatment, and the remaining 58.9% received guideline-discordant antibiotics. Among the guideline-discordant patients, 81.5% were treated according to CAP guidelines instead. Next, we examined the degree to which guideline-concordant antibiotics were prescribed at the hospital level. Figure 1 shows the distribution of hospital rates of administering at least partially guideline-concordant therapy. Rates range from 0% to 87.1%, with a median of 36.4%. Hospital-level characteristics associated with administering higher rates of at least partially guideline-concordant antibiotics included larger size, urban location, and being a teaching institution (supplementary Table 2). Overall, physician adherence to guideline-recommended empiric antibiotic therapy slowly increased over the 4-year study period with no indication of a plateau (Figure 2, top line).

Next, we examined the outcomes associated with the administration of guideline-concordant antibiotics at the hospital level. Among the 488 hospitals, there were 292 hospitals for which we had data over the entire study period, which included 121,600 patients. Among these patients, 49,445 (40.7%) received guideline-concordant antibiotics and 72,155 (59.3%) received guideline-discordant antibiotics. On average, the rate of guideline concordance increased by 2.2% per 6-month interval, while mortality fell by 0.24% per interval. After adjustment for patient demographics and comorbidities at the hospital level, there was no significant correlation between increases in concordant antibiotic prescribing rates and hospital mortality (Pearson correlation = −0.064; P = 0.28), progression to respiratory failure (ie, late initiation of intermittent mandatory ventilation; Pearson correlation = 0.084; P = 0.15), or extended length of stay among survivors (Pearson correlation = 0.10; P = 0.08; Figure 2).

DISCUSSION

In this large, retrospective cohort study, we found that there was a substantial gap between the empiric antibiotics recommended by the ATS and IDSA guidelines and the empiric antibiotics that patients actually received. Over the study period, we saw an increased adherence to guidelines, in spite of growing evidence that HCAP risk factors do not adequately predict which patients are at risk for infection with an MDRO.11 We used this change in antibiotic prescribing behavior over time to determine if there was a clinical impact on patient outcomes and found that at the hospital level, there were no improvements in mortality, excess length of stay, or progression to respiratory failure despite a doubling in guideline-concordant antibiotic use.

At least 2 other large studies have assessed the association between guideline-concordant therapy and outcomes in HCAP.12,13 Both found that guideline-concordant therapy was associated with increased mortality, despite propensity matching. Both were conducted at the individual patient level by using administrative data, and results were likely affected by unmeasured clinical confounders, with sicker patients being more likely to receive guideline-concordant therapy. Our focus on the outcomes at the hospital level avoids this selection bias because the overall severity of illness of patients at any given hospital would not be expected to change over the study period, while physician uptake of antibiotic prescribing guidelines would be expected to increase over time. Determining the correlation between increases in guideline adherence and changes in patient outcome may offer a better assessment of the impact of guideline adherence. In this regard, our results are similar to those achieved by 1 quality improvement collaborative that was aimed at increasing guideline concordant therapy in ICUs. Despite an increase in guideline concordance from 33% to 47% of patients, they found no change in overall mortality.14

There were several limitations to our study. We did not have access to microbiologic data, so we were unable to determine which patients had MDRO infection or determine antibiotic-pathogen matching. However, the treating physicians in our study population presumably did not have access to this data at the time of treatment either because the time period we examined was within the first 48 hours of hospitalization, the interval during which cultures are incubating and the patients are being treated empirically. In addition, there may have been HCAP patients that we failed to identify, such as patients who were admitted in the past 90 days to a hospital that does not submit data to Premier. However, it is unlikely that prescribing for such patients should differ systematically from what we observed. While the database draws from 488 hospitals nationwide, it is possible that practices may be different at facilities that are not contained within the Premier database, such as Veterans Administration Hospitals. Similarly, we did not have readings for chest x-rays; hence, there could be some patients in the dataset who did not have pneumonia. However, we tried to overcome this by including only those patients with a principal diagnosis of pneumonia or sepsis with a secondary pneumonia diagnosis, a chest x-ray, and antibiotics administered within the first 48 hours of admission.

There are likely several reasons why so few HCAP patients in our study received guideline-concordant antibiotics. A lack of knowledge about the ATS and IDSA guidelines may have impacted the physicians in our study population. El-Solh et al.15 surveyed physicians about the ATS-IDSA guidelines 4 years after publication and found that only 45% were familiar with the document. We found that the rate of prescribing at least partially guideline-concordant antibiotics rose steadily over time, supporting the idea that the newness of the guidelines was 1 barrier. Additionally, prior studies have shown that many physicians may not agree with or choose to follow guidelines, with only 20% of physicians indicating that guidelines have a major impact on their clinical decision making,16 and the majority do not choose HCAP guideline-concordant antibiotics when tested.17 Alternatively, clinicians may not follow the guidelines because of a belief that the HCAP criteria do not adequately indicate patients who are at risk for MDRO. Previous studies have demonstrated the relative inability of HCAP risk factors to predict patients who harbor MDRO18 and suggest that better tools such as clinical scoring systems, which include not only the traditional HCAP risk factors but also prior exposure to antibiotics, prior culture data, and a cumulative assessment of both intrinsic and extrinsic factors, could more accurately predict MDRO and lead to a more judicious use of broad-spectrum antimicrobial agents.19-25 Indeed, these collective findings have led the authors of the recently updated guidelines to remove HCAP as a clinical entity from the hospital-acquired or ventilator-associated pneumonia guidelines and place them instead in the upcoming updated guidelines on the management of CAP.5 Of these 3 explanations, the lack of familiarity fits best with our observation that guideline-concordant therapy increased steadily over time with no evidence of reaching a plateau. Ironically, as consensus was building that HCAP is a poor marker for MDROs, routine empiric treatment with vancomycin and piperacillin-tazobactam (“vanco and zosyn”) have become routine in many hospitals. Additional studies are needed to know if this trend has stabilized or reversed.

 

 

CONCLUSIONS

In conclusion, clinicians in our large, nationally representative sample treated the majority of HCAP patients as though they had CAP. Although there was an increase in the administration of guideline-concordant therapy over time, this increase was not associated with improved outcomes. This study supports the growing consensus that HCAP criteria do not accurately predict which patients benefit from broad-spectrum antibiotics for pneumonia, and most patients fare well with antibiotics targeting common community-acquired organisms.

Disclosure

 This work was supported by grant # R01HS018723 from the Agency for Healthcare Research and Quality. Dr. Lagu is also supported by the National Heart, Lung, and Blood Institute of the National Institutes of Health under award number K01HL114745. Dr. Lindenauer is supported by grant K24HL132008 from the National Heart, Lung, and Blood Institute. The funding agency had no role in the data acquisition, analysis, or manuscript preparation for this study. Drs. Haessler and Rothberg had full access to all the data in the study and take responsibility for the integrity of the data and the accuracy of the data analysis. Drs. Haessler, Lagu, Lindenauer, Skiest, Zilberberg, Higgins, and Rothberg conceived of the study and analyzed and interpreted the data. Dr. Lindenauer acquired the data. Dr. Pekow and Ms. Priya carried out the statistical analyses. Dr. Haessler drafted the manuscript. All authors critically reviewed the manuscript for accuracy and integrity. All authors certify no potential conflicts of interest. Preliminary results from this study were presented in oral and poster format at IDWeek in 2012 and 2013.

Files
References

1. Kochanek KD, Murphy SL, Xu JQ, Tejada-Vera B. Deaths: Final data for 2014. National vital statistics reports; vol 65 no 4. Hyattsville, MD: National Center for Health Statistics. 2016. PubMed
2. American Thoracic Society, Infectious Diseases Society of America. Guidelines for the Management of Adults with Hospital-acquired, Ventilator-associated, and Healthcare-associated Pneumonia. Am J Respir Crit Care Med. 2005;171(4):388-416. PubMed
3. Zilberberg MD, Shorr A. Healthcare-associated pneumonia: the state of the evidence to date. Curr Opin Pulm Med. 2011;17(3):142-147. PubMed
4. Kollef MK, Shorr A, Tabak YP, Gupta V, Liu LZ, Johannes RS. Epidemiology and Outcomes of Health-care-associated pneumonia. Chest. 2005;128(6):3854-3862. PubMed
5. Kalil AC, Metersky ML, Klompas M, et al. Management of Adults With Hospital-acquired and Ventilator-associated Pneumonia: 2016 Clinical Practice Guidelines by the Infectious Diseases Society of America and the American Thoracic Society. Clin Infect Dis. 2016;63(5):575-582. PubMed
6. Lindenauer PK, Pekow PS, Lahti MC, Lee Y, Benjamin EM, Rothberg MB. Association of corticosteroid dose and route of administration with risk of treatment failure in acute exacerbation of chronic obstructive pulmonary disease. JAMA. 2010;303(23):2359-2367. PubMed
7. Elixhauser A, Steiner C, Harris DR, Coffey RM. Comorbidity measures for use with administrative data. Med Care. 1998;36(1):8-27. PubMed
8. Centers for Medicare & Medicaid Services. Frequently asked questions (FAQs): Implementation and maintenance of CMS mortality measures for AMI & HF. 2007. https://www.cms.gov/Medicare/Quality-Initiatives-Patient-Assessment-Instruments/HospitalQualityInits/downloads/HospitalMortalityAboutAMI_HF.pdf. Accessed November 1, 2016.
9. Normand SL, Shahian DM. Statistical and Clinical Aspects of Hospital Outcomes Profiling. Stat Sci. 2007;22(2):206-226. 
10. Rothberg MB, Pekow PS, Priya A, et al. Using highly detailed administrative data to predict pneumonia mortality. PLoS One. 2014;9(1):e87382. PubMed
11. Jones BE, Jones MM, Huttner B, et al. Trends in antibiotic use and nosocomial pathogens in hospitalized veterans with pneumonia at 128 medical centers, 2006-2010. Clin Infect Dis. 2015;61(9):1403-1410. PubMed
12. Attridge RT, Frei CR, Restrepo MI, et al. Guideline-concordant therapy and outcomes in healthcare-associated pneumonia. Eur Respir J. 2011;38(4):878-887. PubMed
13. Rothberg MB, Zilberberg MD, Pekow PS, et al. Association of Guideline-based Antimicrobial Therapy and Outcomes in Healthcare-Associated Pneumonia. J Antimicrob Chemother. 2015;70(5):1573-1579. PubMed
14. Kett DH, Cano E, Quartin AA, et al. Improving Medicine through Pathway Assessment of Critical Therapy of Hospital-Acquired Pneumonia (IMPACT-HAP) Investigators. Implementation of guidelines for management of possible multidrug-resistant pneumonia in intensive care: an observational, multicentre cohort study. Lancet Infect Dis. 2011;11(3):181-189. PubMed
15. El-Solh AA, Alhajhusain A, Saliba RG, Drinka P. Physicians’ Attitudes Toward Guidelines for the Treatment of Hospitalized Nursing-Home -Acquired Pneumonia. J Am Med Dir Assoc. 2011;12(4):270-276. PubMed
16. Tunis S, Hayward R, Wilson M, et al. Internists’ Attitudes about Clinical Practice Guidelines. Ann Intern Med. 1994;120(11):956-963. PubMed
17. Seymann GB, Di Francesco L, Sharpe B, et al. The HCAP Gap: Differences between Self-Reported Practice Patterns and Published Guidelines for Health Care-Associated Pneumonia. Clin Infect Dis. 2009;49(12):1868-1874. PubMed
18. Chalmers JD, Rother C, Salih W, Ewig S. Healthcare associated pneumonia does not accurately identify potentially resistant pathogens: a systematic review and meta-analysis. Clin Infect Dis. 2014;58(3):330-339. PubMed
19. Shorr A, Zilberberg MD, Reichley R, et al. Validation of a Clinical Score for Assessing the Risk of Resistant Pathogens in Patients with Pneumonia Presenting to the Emergency Department. Clin Infect Dis. 2012;54(2):193-198. PubMed
20. Aliberti S, Pasquale MD, Zanaboni AM, et al. Stratifying Risk Factors for Multidrug-Resistant Pathogens in Hospitalized Patients Coming from the Community with Pneumonia. Clin Infect Dis. 2012;54(4):470-478. PubMed
21. Schreiber MP, Chan CM, Shorr AF. Resistant Pathogens in Nonnosocomial Pneumonia and Respiratory Failure: Is it Time to Refine the Definition of Health-care-Associated Pneumonia? Chest. 2010;137(6):1283-1288. PubMed
22. Madaras-Kelly KJ, Remington RE, Fan VS, Sloan KL. Predicting antibiotic resistance to community-acquired pneumonia antibiotics in culture-positive patients with healthcare-associated pneumonia. J Hosp Med. 2012;7(3):195-202. PubMed
23. Shindo Y, Ito R, Kobayashi D, et al. Risk factors for drug-resistant pathogens in community-acquired and healthcare-associated pneumonia. Am J Respir Crit Care Med. 2013;188(8):985-995. PubMed
24. Metersky ML, Frei CR, Mortensen EM. Predictors of Pseudomonas and methicillin-resistant Staphylococcus aureus in hospitalized patients with healthcare-associated pneumonia. Respirology. 2016;21(1):157-163. PubMed
25. Webb BJ, Dascomb K, Stenehjem E, Dean N. Predicting risk of drug-resistant organisms in pneumonia: moving beyond the HCAP model. Respir Med. 2015;109(1):1-10. PubMed

Article PDF
Issue
Journal of Hospital Medicine 12(11)
Publications
Topics
Page Number
886-891
Sections
Files
Files
Article PDF
Article PDF

Bacterial pneumonia remains an important cause of morbidity and mortality in the United States, and is the 8th leading cause of death with 55,227 deaths among adults annually.1 In 2005, the American Thoracic Society (ATS) and the Infectious Diseases Society of America (IDSA) collaborated to update guidelines for hospital-acquired pneumonia (HAP), ventilator-associated pneumonia, and healthcare-associated pneumonia (HCAP).2 This broad document outlines an evidence-based approach to diagnostic testing and antibiotic management based on the epidemiology and risk factors for these conditions. The guideline specifies the following criteria for HCAP: hospitalization in the past 90 days, residence in a skilled nursing facility (SNF), home infusion therapy, hemodialysis, home wound care, family members with multidrug resistant organisms (MDRO), and immunosuppressive diseases or medications, with the presumption that these patients are more likely to be harboring MDRO and should thus be treated empirically with broad-spectrum antibiotic therapy. Prior studies have shown that patients with HCAP have a more severe illness, are more likely to have MDRO, are more likely to be inadequately treated, and are at a higher risk for mortality than patients with community-acquired pneumonia (CAP).3,4

These guidelines are controversial, especially in regard to the recommendations to empirically treat broadly with 2 antibiotics targeting Pseudomonas species, whether patients with HCAP merit broader spectrum coverage than patients with CAP, and whether the criteria for defining HCAP are adequate to predict which patients are harboring MDRO. It has subsequently been proposed that HCAP is more related to CAP than to HAP, and a recent update to the guideline removed recommendations for treatment of HCAP and will be placing HCAP into the guidelines for CAP instead.5 We sought to investigate the degree of uptake of the ATS and IDSA guideline recommendations by physicians over time, and whether this led to a change in outcomes among patients who met the criteria for HCAP.

METHODS

Setting and Patients

We identified patients discharged between July 1, 2007, and November 30, 2011, from 488 US hospitals that participated in the Premier database (Premier Inc., Charlotte, North Carolina), an inpatient database developed for measuring quality and healthcare utilization. The database is frequently used for healthcare research and has been described previously.6 Member hospitals are in all regions of the US and are generally reflective of US hospitals. This database contains multiple data elements, including sociodemographic information, International Classification of Diseases, 9th Revision-Clinical Modification (ICD-9-CM) diagnosis and procedure codes, hospital and physician information, source of admission, and discharge status. It also includes a date-stamped log of all billed items and services, including diagnostic tests, medications, and other treatments. Because the data do not contain identifiable information, the institutional review board at our medical center determined that this study did not constitute human subjects research.

We included all patients aged ≥18 years with a principal diagnosis of pneumonia or with a secondary diagnosis of pneumonia paired with a principal diagnosis of respiratory failure, acute respiratory distress syndrome, respiratory arrest, sepsis, or influenza. Patients were excluded if they were transferred to or from another acute care institution, had a length of stay of 1 day or less, had cystic fibrosis, did not have a chest radiograph, or did not receive antibiotics within 48 hours of admission.

For each patient, we extracted age, gender, principal diagnosis, comorbidities, and the specialty of the attending physician. Comorbidities were identified from ICD-9-CM secondary diagnosis codes and Diagnosis Related Groups by using Healthcare Cost and Utilization Project Comorbidity Software, version 3.1, based on the work of Elixhauser (Agency for Healthcare Research and Quality, Rockville, Maryland).7 In order to ensure that patients had HCAP, we required the presence of ≥1 HCAP criteria, including hospitalization in the past 90 days, hemodialysis, admission from an SNF, or immune suppression (which was derived from either a secondary diagnosis for neutropenia, hematological malignancy, organ transplant, acquired immunodeficiency virus, or receiving immunosuppressant drugs or corticosteroids [equivalent to ≥20 mg/day of prednisone]).

 

 

Definitions of Guideline-Concordant and Discordant Antibiotic Therapy

The ATS and IDSA guidelines recommended the following antibiotic combinations for HCAP: an antipseudomonal cephalosporin or carbapenem or a beta-lactam/lactamase inhibitor, plus an antipseudomonal quinolone or aminoglycoside, plus an antibiotic with activity versus methicillin resistant Staphylococcus aureus (MRSA), such as vancomycin or linezolid. Based on these guidelines, we defined the receipt of fully guideline-concordant antibiotics as 2 recommended antibiotics for Pseudomonas species plus 1 for MRSA administered by the second day of admission. Partially guideline-concordant antibiotics were defined as 1 recommended antibiotic for Pseudomonas species plus 1 for MRSA by the second day of hospitalization. Guideline-discordant antibiotics were defined as all other combinations.

Statistical Analysis

Descriptive statistics on patient characteristics are presented as frequency, proportions for categorical factors, and median with interquartile range (IQR) for continuous variables for the full cohort and by treatment group, defined as fully or partially guideline-concordant antibiotic therapy or discordant therapy. Hospital rates of fully guideline-concordant treatment are presented overall and by hospital characteristics. The association of hospital characteristics with rates of fully guideline-concordant therapy were assessed by using 1-way analysis of variance tests.

To assess trends across hospitals for the association between the use of guideline-concordant therapy and mortality, progression to respiratory failure as measured by the late initiation of invasive mechanical ventilation (day 3 or later), and the length of stay among survivors, we divided the 4.5-year study period into 9 intervals of 6 months each; 292 hospitals that submitted data for all 9 time points were examined in this analysis. Based on the distribution of length of stay in the first time period, we created an indicator variable for extended length of stay with length of stay at or above the 75th percentile, defined as extended. For each hospital at each 6-month interval, we then computed risk-standardized guideline-concordant treatment (RS-treatment) rates and risk-standardized in-hospital outcome rates similar to methods used by the Centers for Medicare and Medicaid Services for public reporting.8 For each hospital at each time interval, we estimated a predicted rate of guideline-concordant treatment as the sum of predicted probabilities of guideline-concordant treatment from patient factors and the random intercept for the hospital in which they were admitted. We then calculated the expected rate of guideline-concordant treatment as the sum of expected probabilities of treatment received from patient factors only. RS-treatment was then calculated as the ratio of predicted to expected rates multiplied by the overall unadjusted mean treatment rate from all patients.9 We repeated the same modeling strategy to calculate risk-standardized outcome (RS-outcome) rates for each hospital across all time points. All models were adjusted for patient demographics and comorbidities. Similar models using administrative data have moderate discrimination for mortality.10

We then fit mixed-effects linear models with random hospital intercept and slope across time for the RS-treatment and outcome rates, respectively. From these models, we estimated the mean slope for RS-treatment and for RS-outcome over time. In addition, we estimated a slope or trend over time for each hospital for treatment and for outcome and evaluated the correlation between the treatment and outcome trends.

All analyses were performed using the Statistical Analysis System version 9.4 (SAS Institute Inc., Cary, NC) and STATA release 13 (StataCorp, LLC, College Station, Texas).

RESULTS

Of 1,601,064 patients with a diagnosis of pneumonia in our dataset, 436,483 patients met our inclusion criteria, and of those, 149,963 (34.4%) met at least 1 HCAP criterion and were included as our study cohort (supplementary Figure). Among the study cohort, the median age was 73 years (IQR, 61-83), 51.4% of patients were female, 69.6% of patients were white, and a majority of patients (76.2 %) were covered by Medicare. HCAP categories included hospitalization in the past 90 days (63.1%), hemodialysis (12.8%), admission from a SNF (23.6%), and immunosuppression (28.9%). One-quarter of the patients were treated in the intensive care unit (ICU) by day 2 of their hospitalization. The most common comorbidities were hypertension (65.1%), chronic obstructive pulmonary disease (47.3%), anemia (40.9%), diabetes (36.6%), and congestive heart failure (35.7%). Pneumonia was the principal diagnosis in 61.9% of patients, and sepsis was the principal diagnosis in 29.3% of patients. The unadjusted median length of stay was 6 days, the median cost was $10,049, and the in-hospital mortality was 11.1%. Patients who received fully or partially guideline-concordant antibiotics were younger on average and had a higher combined comorbidity score, and they were more likely to have been admitted to the ICU and to have received vasopressor medications and mechanical ventilation. They also had higher unadjusted mortality, longer lengths of stay, and higher costs (see supplemental Table 1 for more details).

 

 

The Table shows the antibiotics received by patients. Overall, 19.6% of patients received fully guideline-concordant treatment, 21.7% received partially guideline-concordant treatment, and the remaining 58.9% received guideline-discordant antibiotics. Among the guideline-discordant patients, 81.5% were treated according to CAP guidelines instead. Next, we examined the degree to which guideline-concordant antibiotics were prescribed at the hospital level. Figure 1 shows the distribution of hospital rates of administering at least partially guideline-concordant therapy. Rates range from 0% to 87.1%, with a median of 36.4%. Hospital-level characteristics associated with administering higher rates of at least partially guideline-concordant antibiotics included larger size, urban location, and being a teaching institution (supplementary Table 2). Overall, physician adherence to guideline-recommended empiric antibiotic therapy slowly increased over the 4-year study period with no indication of a plateau (Figure 2, top line).

Next, we examined the outcomes associated with the administration of guideline-concordant antibiotics at the hospital level. Among the 488 hospitals, there were 292 hospitals for which we had data over the entire study period, which included 121,600 patients. Among these patients, 49,445 (40.7%) received guideline-concordant antibiotics and 72,155 (59.3%) received guideline-discordant antibiotics. On average, the rate of guideline concordance increased by 2.2% per 6-month interval, while mortality fell by 0.24% per interval. After adjustment for patient demographics and comorbidities at the hospital level, there was no significant correlation between increases in concordant antibiotic prescribing rates and hospital mortality (Pearson correlation = −0.064; P = 0.28), progression to respiratory failure (ie, late initiation of intermittent mandatory ventilation; Pearson correlation = 0.084; P = 0.15), or extended length of stay among survivors (Pearson correlation = 0.10; P = 0.08; Figure 2).

DISCUSSION

In this large, retrospective cohort study, we found that there was a substantial gap between the empiric antibiotics recommended by the ATS and IDSA guidelines and the empiric antibiotics that patients actually received. Over the study period, we saw an increased adherence to guidelines, in spite of growing evidence that HCAP risk factors do not adequately predict which patients are at risk for infection with an MDRO.11 We used this change in antibiotic prescribing behavior over time to determine if there was a clinical impact on patient outcomes and found that at the hospital level, there were no improvements in mortality, excess length of stay, or progression to respiratory failure despite a doubling in guideline-concordant antibiotic use.

At least 2 other large studies have assessed the association between guideline-concordant therapy and outcomes in HCAP.12,13 Both found that guideline-concordant therapy was associated with increased mortality, despite propensity matching. Both were conducted at the individual patient level by using administrative data, and results were likely affected by unmeasured clinical confounders, with sicker patients being more likely to receive guideline-concordant therapy. Our focus on the outcomes at the hospital level avoids this selection bias because the overall severity of illness of patients at any given hospital would not be expected to change over the study period, while physician uptake of antibiotic prescribing guidelines would be expected to increase over time. Determining the correlation between increases in guideline adherence and changes in patient outcome may offer a better assessment of the impact of guideline adherence. In this regard, our results are similar to those achieved by 1 quality improvement collaborative that was aimed at increasing guideline concordant therapy in ICUs. Despite an increase in guideline concordance from 33% to 47% of patients, they found no change in overall mortality.14

There were several limitations to our study. We did not have access to microbiologic data, so we were unable to determine which patients had MDRO infection or determine antibiotic-pathogen matching. However, the treating physicians in our study population presumably did not have access to this data at the time of treatment either because the time period we examined was within the first 48 hours of hospitalization, the interval during which cultures are incubating and the patients are being treated empirically. In addition, there may have been HCAP patients that we failed to identify, such as patients who were admitted in the past 90 days to a hospital that does not submit data to Premier. However, it is unlikely that prescribing for such patients should differ systematically from what we observed. While the database draws from 488 hospitals nationwide, it is possible that practices may be different at facilities that are not contained within the Premier database, such as Veterans Administration Hospitals. Similarly, we did not have readings for chest x-rays; hence, there could be some patients in the dataset who did not have pneumonia. However, we tried to overcome this by including only those patients with a principal diagnosis of pneumonia or sepsis with a secondary pneumonia diagnosis, a chest x-ray, and antibiotics administered within the first 48 hours of admission.

There are likely several reasons why so few HCAP patients in our study received guideline-concordant antibiotics. A lack of knowledge about the ATS and IDSA guidelines may have impacted the physicians in our study population. El-Solh et al.15 surveyed physicians about the ATS-IDSA guidelines 4 years after publication and found that only 45% were familiar with the document. We found that the rate of prescribing at least partially guideline-concordant antibiotics rose steadily over time, supporting the idea that the newness of the guidelines was 1 barrier. Additionally, prior studies have shown that many physicians may not agree with or choose to follow guidelines, with only 20% of physicians indicating that guidelines have a major impact on their clinical decision making,16 and the majority do not choose HCAP guideline-concordant antibiotics when tested.17 Alternatively, clinicians may not follow the guidelines because of a belief that the HCAP criteria do not adequately indicate patients who are at risk for MDRO. Previous studies have demonstrated the relative inability of HCAP risk factors to predict patients who harbor MDRO18 and suggest that better tools such as clinical scoring systems, which include not only the traditional HCAP risk factors but also prior exposure to antibiotics, prior culture data, and a cumulative assessment of both intrinsic and extrinsic factors, could more accurately predict MDRO and lead to a more judicious use of broad-spectrum antimicrobial agents.19-25 Indeed, these collective findings have led the authors of the recently updated guidelines to remove HCAP as a clinical entity from the hospital-acquired or ventilator-associated pneumonia guidelines and place them instead in the upcoming updated guidelines on the management of CAP.5 Of these 3 explanations, the lack of familiarity fits best with our observation that guideline-concordant therapy increased steadily over time with no evidence of reaching a plateau. Ironically, as consensus was building that HCAP is a poor marker for MDROs, routine empiric treatment with vancomycin and piperacillin-tazobactam (“vanco and zosyn”) have become routine in many hospitals. Additional studies are needed to know if this trend has stabilized or reversed.

 

 

CONCLUSIONS

In conclusion, clinicians in our large, nationally representative sample treated the majority of HCAP patients as though they had CAP. Although there was an increase in the administration of guideline-concordant therapy over time, this increase was not associated with improved outcomes. This study supports the growing consensus that HCAP criteria do not accurately predict which patients benefit from broad-spectrum antibiotics for pneumonia, and most patients fare well with antibiotics targeting common community-acquired organisms.

Disclosure

 This work was supported by grant # R01HS018723 from the Agency for Healthcare Research and Quality. Dr. Lagu is also supported by the National Heart, Lung, and Blood Institute of the National Institutes of Health under award number K01HL114745. Dr. Lindenauer is supported by grant K24HL132008 from the National Heart, Lung, and Blood Institute. The funding agency had no role in the data acquisition, analysis, or manuscript preparation for this study. Drs. Haessler and Rothberg had full access to all the data in the study and take responsibility for the integrity of the data and the accuracy of the data analysis. Drs. Haessler, Lagu, Lindenauer, Skiest, Zilberberg, Higgins, and Rothberg conceived of the study and analyzed and interpreted the data. Dr. Lindenauer acquired the data. Dr. Pekow and Ms. Priya carried out the statistical analyses. Dr. Haessler drafted the manuscript. All authors critically reviewed the manuscript for accuracy and integrity. All authors certify no potential conflicts of interest. Preliminary results from this study were presented in oral and poster format at IDWeek in 2012 and 2013.

Bacterial pneumonia remains an important cause of morbidity and mortality in the United States, and is the 8th leading cause of death with 55,227 deaths among adults annually.1 In 2005, the American Thoracic Society (ATS) and the Infectious Diseases Society of America (IDSA) collaborated to update guidelines for hospital-acquired pneumonia (HAP), ventilator-associated pneumonia, and healthcare-associated pneumonia (HCAP).2 This broad document outlines an evidence-based approach to diagnostic testing and antibiotic management based on the epidemiology and risk factors for these conditions. The guideline specifies the following criteria for HCAP: hospitalization in the past 90 days, residence in a skilled nursing facility (SNF), home infusion therapy, hemodialysis, home wound care, family members with multidrug resistant organisms (MDRO), and immunosuppressive diseases or medications, with the presumption that these patients are more likely to be harboring MDRO and should thus be treated empirically with broad-spectrum antibiotic therapy. Prior studies have shown that patients with HCAP have a more severe illness, are more likely to have MDRO, are more likely to be inadequately treated, and are at a higher risk for mortality than patients with community-acquired pneumonia (CAP).3,4

These guidelines are controversial, especially in regard to the recommendations to empirically treat broadly with 2 antibiotics targeting Pseudomonas species, whether patients with HCAP merit broader spectrum coverage than patients with CAP, and whether the criteria for defining HCAP are adequate to predict which patients are harboring MDRO. It has subsequently been proposed that HCAP is more related to CAP than to HAP, and a recent update to the guideline removed recommendations for treatment of HCAP and will be placing HCAP into the guidelines for CAP instead.5 We sought to investigate the degree of uptake of the ATS and IDSA guideline recommendations by physicians over time, and whether this led to a change in outcomes among patients who met the criteria for HCAP.

METHODS

Setting and Patients

We identified patients discharged between July 1, 2007, and November 30, 2011, from 488 US hospitals that participated in the Premier database (Premier Inc., Charlotte, North Carolina), an inpatient database developed for measuring quality and healthcare utilization. The database is frequently used for healthcare research and has been described previously.6 Member hospitals are in all regions of the US and are generally reflective of US hospitals. This database contains multiple data elements, including sociodemographic information, International Classification of Diseases, 9th Revision-Clinical Modification (ICD-9-CM) diagnosis and procedure codes, hospital and physician information, source of admission, and discharge status. It also includes a date-stamped log of all billed items and services, including diagnostic tests, medications, and other treatments. Because the data do not contain identifiable information, the institutional review board at our medical center determined that this study did not constitute human subjects research.

We included all patients aged ≥18 years with a principal diagnosis of pneumonia or with a secondary diagnosis of pneumonia paired with a principal diagnosis of respiratory failure, acute respiratory distress syndrome, respiratory arrest, sepsis, or influenza. Patients were excluded if they were transferred to or from another acute care institution, had a length of stay of 1 day or less, had cystic fibrosis, did not have a chest radiograph, or did not receive antibiotics within 48 hours of admission.

For each patient, we extracted age, gender, principal diagnosis, comorbidities, and the specialty of the attending physician. Comorbidities were identified from ICD-9-CM secondary diagnosis codes and Diagnosis Related Groups by using Healthcare Cost and Utilization Project Comorbidity Software, version 3.1, based on the work of Elixhauser (Agency for Healthcare Research and Quality, Rockville, Maryland).7 In order to ensure that patients had HCAP, we required the presence of ≥1 HCAP criteria, including hospitalization in the past 90 days, hemodialysis, admission from an SNF, or immune suppression (which was derived from either a secondary diagnosis for neutropenia, hematological malignancy, organ transplant, acquired immunodeficiency virus, or receiving immunosuppressant drugs or corticosteroids [equivalent to ≥20 mg/day of prednisone]).

 

 

Definitions of Guideline-Concordant and Discordant Antibiotic Therapy

The ATS and IDSA guidelines recommended the following antibiotic combinations for HCAP: an antipseudomonal cephalosporin or carbapenem or a beta-lactam/lactamase inhibitor, plus an antipseudomonal quinolone or aminoglycoside, plus an antibiotic with activity versus methicillin resistant Staphylococcus aureus (MRSA), such as vancomycin or linezolid. Based on these guidelines, we defined the receipt of fully guideline-concordant antibiotics as 2 recommended antibiotics for Pseudomonas species plus 1 for MRSA administered by the second day of admission. Partially guideline-concordant antibiotics were defined as 1 recommended antibiotic for Pseudomonas species plus 1 for MRSA by the second day of hospitalization. Guideline-discordant antibiotics were defined as all other combinations.

Statistical Analysis

Descriptive statistics on patient characteristics are presented as frequency, proportions for categorical factors, and median with interquartile range (IQR) for continuous variables for the full cohort and by treatment group, defined as fully or partially guideline-concordant antibiotic therapy or discordant therapy. Hospital rates of fully guideline-concordant treatment are presented overall and by hospital characteristics. The association of hospital characteristics with rates of fully guideline-concordant therapy were assessed by using 1-way analysis of variance tests.

To assess trends across hospitals for the association between the use of guideline-concordant therapy and mortality, progression to respiratory failure as measured by the late initiation of invasive mechanical ventilation (day 3 or later), and the length of stay among survivors, we divided the 4.5-year study period into 9 intervals of 6 months each; 292 hospitals that submitted data for all 9 time points were examined in this analysis. Based on the distribution of length of stay in the first time period, we created an indicator variable for extended length of stay with length of stay at or above the 75th percentile, defined as extended. For each hospital at each 6-month interval, we then computed risk-standardized guideline-concordant treatment (RS-treatment) rates and risk-standardized in-hospital outcome rates similar to methods used by the Centers for Medicare and Medicaid Services for public reporting.8 For each hospital at each time interval, we estimated a predicted rate of guideline-concordant treatment as the sum of predicted probabilities of guideline-concordant treatment from patient factors and the random intercept for the hospital in which they were admitted. We then calculated the expected rate of guideline-concordant treatment as the sum of expected probabilities of treatment received from patient factors only. RS-treatment was then calculated as the ratio of predicted to expected rates multiplied by the overall unadjusted mean treatment rate from all patients.9 We repeated the same modeling strategy to calculate risk-standardized outcome (RS-outcome) rates for each hospital across all time points. All models were adjusted for patient demographics and comorbidities. Similar models using administrative data have moderate discrimination for mortality.10

We then fit mixed-effects linear models with random hospital intercept and slope across time for the RS-treatment and outcome rates, respectively. From these models, we estimated the mean slope for RS-treatment and for RS-outcome over time. In addition, we estimated a slope or trend over time for each hospital for treatment and for outcome and evaluated the correlation between the treatment and outcome trends.

All analyses were performed using the Statistical Analysis System version 9.4 (SAS Institute Inc., Cary, NC) and STATA release 13 (StataCorp, LLC, College Station, Texas).

RESULTS

Of 1,601,064 patients with a diagnosis of pneumonia in our dataset, 436,483 patients met our inclusion criteria, and of those, 149,963 (34.4%) met at least 1 HCAP criterion and were included as our study cohort (supplementary Figure). Among the study cohort, the median age was 73 years (IQR, 61-83), 51.4% of patients were female, 69.6% of patients were white, and a majority of patients (76.2 %) were covered by Medicare. HCAP categories included hospitalization in the past 90 days (63.1%), hemodialysis (12.8%), admission from a SNF (23.6%), and immunosuppression (28.9%). One-quarter of the patients were treated in the intensive care unit (ICU) by day 2 of their hospitalization. The most common comorbidities were hypertension (65.1%), chronic obstructive pulmonary disease (47.3%), anemia (40.9%), diabetes (36.6%), and congestive heart failure (35.7%). Pneumonia was the principal diagnosis in 61.9% of patients, and sepsis was the principal diagnosis in 29.3% of patients. The unadjusted median length of stay was 6 days, the median cost was $10,049, and the in-hospital mortality was 11.1%. Patients who received fully or partially guideline-concordant antibiotics were younger on average and had a higher combined comorbidity score, and they were more likely to have been admitted to the ICU and to have received vasopressor medications and mechanical ventilation. They also had higher unadjusted mortality, longer lengths of stay, and higher costs (see supplemental Table 1 for more details).

 

 

The Table shows the antibiotics received by patients. Overall, 19.6% of patients received fully guideline-concordant treatment, 21.7% received partially guideline-concordant treatment, and the remaining 58.9% received guideline-discordant antibiotics. Among the guideline-discordant patients, 81.5% were treated according to CAP guidelines instead. Next, we examined the degree to which guideline-concordant antibiotics were prescribed at the hospital level. Figure 1 shows the distribution of hospital rates of administering at least partially guideline-concordant therapy. Rates range from 0% to 87.1%, with a median of 36.4%. Hospital-level characteristics associated with administering higher rates of at least partially guideline-concordant antibiotics included larger size, urban location, and being a teaching institution (supplementary Table 2). Overall, physician adherence to guideline-recommended empiric antibiotic therapy slowly increased over the 4-year study period with no indication of a plateau (Figure 2, top line).

Next, we examined the outcomes associated with the administration of guideline-concordant antibiotics at the hospital level. Among the 488 hospitals, there were 292 hospitals for which we had data over the entire study period, which included 121,600 patients. Among these patients, 49,445 (40.7%) received guideline-concordant antibiotics and 72,155 (59.3%) received guideline-discordant antibiotics. On average, the rate of guideline concordance increased by 2.2% per 6-month interval, while mortality fell by 0.24% per interval. After adjustment for patient demographics and comorbidities at the hospital level, there was no significant correlation between increases in concordant antibiotic prescribing rates and hospital mortality (Pearson correlation = −0.064; P = 0.28), progression to respiratory failure (ie, late initiation of intermittent mandatory ventilation; Pearson correlation = 0.084; P = 0.15), or extended length of stay among survivors (Pearson correlation = 0.10; P = 0.08; Figure 2).

DISCUSSION

In this large, retrospective cohort study, we found that there was a substantial gap between the empiric antibiotics recommended by the ATS and IDSA guidelines and the empiric antibiotics that patients actually received. Over the study period, we saw an increased adherence to guidelines, in spite of growing evidence that HCAP risk factors do not adequately predict which patients are at risk for infection with an MDRO.11 We used this change in antibiotic prescribing behavior over time to determine if there was a clinical impact on patient outcomes and found that at the hospital level, there were no improvements in mortality, excess length of stay, or progression to respiratory failure despite a doubling in guideline-concordant antibiotic use.

At least 2 other large studies have assessed the association between guideline-concordant therapy and outcomes in HCAP.12,13 Both found that guideline-concordant therapy was associated with increased mortality, despite propensity matching. Both were conducted at the individual patient level by using administrative data, and results were likely affected by unmeasured clinical confounders, with sicker patients being more likely to receive guideline-concordant therapy. Our focus on the outcomes at the hospital level avoids this selection bias because the overall severity of illness of patients at any given hospital would not be expected to change over the study period, while physician uptake of antibiotic prescribing guidelines would be expected to increase over time. Determining the correlation between increases in guideline adherence and changes in patient outcome may offer a better assessment of the impact of guideline adherence. In this regard, our results are similar to those achieved by 1 quality improvement collaborative that was aimed at increasing guideline concordant therapy in ICUs. Despite an increase in guideline concordance from 33% to 47% of patients, they found no change in overall mortality.14

There were several limitations to our study. We did not have access to microbiologic data, so we were unable to determine which patients had MDRO infection or determine antibiotic-pathogen matching. However, the treating physicians in our study population presumably did not have access to this data at the time of treatment either because the time period we examined was within the first 48 hours of hospitalization, the interval during which cultures are incubating and the patients are being treated empirically. In addition, there may have been HCAP patients that we failed to identify, such as patients who were admitted in the past 90 days to a hospital that does not submit data to Premier. However, it is unlikely that prescribing for such patients should differ systematically from what we observed. While the database draws from 488 hospitals nationwide, it is possible that practices may be different at facilities that are not contained within the Premier database, such as Veterans Administration Hospitals. Similarly, we did not have readings for chest x-rays; hence, there could be some patients in the dataset who did not have pneumonia. However, we tried to overcome this by including only those patients with a principal diagnosis of pneumonia or sepsis with a secondary pneumonia diagnosis, a chest x-ray, and antibiotics administered within the first 48 hours of admission.

There are likely several reasons why so few HCAP patients in our study received guideline-concordant antibiotics. A lack of knowledge about the ATS and IDSA guidelines may have impacted the physicians in our study population. El-Solh et al.15 surveyed physicians about the ATS-IDSA guidelines 4 years after publication and found that only 45% were familiar with the document. We found that the rate of prescribing at least partially guideline-concordant antibiotics rose steadily over time, supporting the idea that the newness of the guidelines was 1 barrier. Additionally, prior studies have shown that many physicians may not agree with or choose to follow guidelines, with only 20% of physicians indicating that guidelines have a major impact on their clinical decision making,16 and the majority do not choose HCAP guideline-concordant antibiotics when tested.17 Alternatively, clinicians may not follow the guidelines because of a belief that the HCAP criteria do not adequately indicate patients who are at risk for MDRO. Previous studies have demonstrated the relative inability of HCAP risk factors to predict patients who harbor MDRO18 and suggest that better tools such as clinical scoring systems, which include not only the traditional HCAP risk factors but also prior exposure to antibiotics, prior culture data, and a cumulative assessment of both intrinsic and extrinsic factors, could more accurately predict MDRO and lead to a more judicious use of broad-spectrum antimicrobial agents.19-25 Indeed, these collective findings have led the authors of the recently updated guidelines to remove HCAP as a clinical entity from the hospital-acquired or ventilator-associated pneumonia guidelines and place them instead in the upcoming updated guidelines on the management of CAP.5 Of these 3 explanations, the lack of familiarity fits best with our observation that guideline-concordant therapy increased steadily over time with no evidence of reaching a plateau. Ironically, as consensus was building that HCAP is a poor marker for MDROs, routine empiric treatment with vancomycin and piperacillin-tazobactam (“vanco and zosyn”) have become routine in many hospitals. Additional studies are needed to know if this trend has stabilized or reversed.

 

 

CONCLUSIONS

In conclusion, clinicians in our large, nationally representative sample treated the majority of HCAP patients as though they had CAP. Although there was an increase in the administration of guideline-concordant therapy over time, this increase was not associated with improved outcomes. This study supports the growing consensus that HCAP criteria do not accurately predict which patients benefit from broad-spectrum antibiotics for pneumonia, and most patients fare well with antibiotics targeting common community-acquired organisms.

Disclosure

 This work was supported by grant # R01HS018723 from the Agency for Healthcare Research and Quality. Dr. Lagu is also supported by the National Heart, Lung, and Blood Institute of the National Institutes of Health under award number K01HL114745. Dr. Lindenauer is supported by grant K24HL132008 from the National Heart, Lung, and Blood Institute. The funding agency had no role in the data acquisition, analysis, or manuscript preparation for this study. Drs. Haessler and Rothberg had full access to all the data in the study and take responsibility for the integrity of the data and the accuracy of the data analysis. Drs. Haessler, Lagu, Lindenauer, Skiest, Zilberberg, Higgins, and Rothberg conceived of the study and analyzed and interpreted the data. Dr. Lindenauer acquired the data. Dr. Pekow and Ms. Priya carried out the statistical analyses. Dr. Haessler drafted the manuscript. All authors critically reviewed the manuscript for accuracy and integrity. All authors certify no potential conflicts of interest. Preliminary results from this study were presented in oral and poster format at IDWeek in 2012 and 2013.

References

1. Kochanek KD, Murphy SL, Xu JQ, Tejada-Vera B. Deaths: Final data for 2014. National vital statistics reports; vol 65 no 4. Hyattsville, MD: National Center for Health Statistics. 2016. PubMed
2. American Thoracic Society, Infectious Diseases Society of America. Guidelines for the Management of Adults with Hospital-acquired, Ventilator-associated, and Healthcare-associated Pneumonia. Am J Respir Crit Care Med. 2005;171(4):388-416. PubMed
3. Zilberberg MD, Shorr A. Healthcare-associated pneumonia: the state of the evidence to date. Curr Opin Pulm Med. 2011;17(3):142-147. PubMed
4. Kollef MK, Shorr A, Tabak YP, Gupta V, Liu LZ, Johannes RS. Epidemiology and Outcomes of Health-care-associated pneumonia. Chest. 2005;128(6):3854-3862. PubMed
5. Kalil AC, Metersky ML, Klompas M, et al. Management of Adults With Hospital-acquired and Ventilator-associated Pneumonia: 2016 Clinical Practice Guidelines by the Infectious Diseases Society of America and the American Thoracic Society. Clin Infect Dis. 2016;63(5):575-582. PubMed
6. Lindenauer PK, Pekow PS, Lahti MC, Lee Y, Benjamin EM, Rothberg MB. Association of corticosteroid dose and route of administration with risk of treatment failure in acute exacerbation of chronic obstructive pulmonary disease. JAMA. 2010;303(23):2359-2367. PubMed
7. Elixhauser A, Steiner C, Harris DR, Coffey RM. Comorbidity measures for use with administrative data. Med Care. 1998;36(1):8-27. PubMed
8. Centers for Medicare & Medicaid Services. Frequently asked questions (FAQs): Implementation and maintenance of CMS mortality measures for AMI & HF. 2007. https://www.cms.gov/Medicare/Quality-Initiatives-Patient-Assessment-Instruments/HospitalQualityInits/downloads/HospitalMortalityAboutAMI_HF.pdf. Accessed November 1, 2016.
9. Normand SL, Shahian DM. Statistical and Clinical Aspects of Hospital Outcomes Profiling. Stat Sci. 2007;22(2):206-226. 
10. Rothberg MB, Pekow PS, Priya A, et al. Using highly detailed administrative data to predict pneumonia mortality. PLoS One. 2014;9(1):e87382. PubMed
11. Jones BE, Jones MM, Huttner B, et al. Trends in antibiotic use and nosocomial pathogens in hospitalized veterans with pneumonia at 128 medical centers, 2006-2010. Clin Infect Dis. 2015;61(9):1403-1410. PubMed
12. Attridge RT, Frei CR, Restrepo MI, et al. Guideline-concordant therapy and outcomes in healthcare-associated pneumonia. Eur Respir J. 2011;38(4):878-887. PubMed
13. Rothberg MB, Zilberberg MD, Pekow PS, et al. Association of Guideline-based Antimicrobial Therapy and Outcomes in Healthcare-Associated Pneumonia. J Antimicrob Chemother. 2015;70(5):1573-1579. PubMed
14. Kett DH, Cano E, Quartin AA, et al. Improving Medicine through Pathway Assessment of Critical Therapy of Hospital-Acquired Pneumonia (IMPACT-HAP) Investigators. Implementation of guidelines for management of possible multidrug-resistant pneumonia in intensive care: an observational, multicentre cohort study. Lancet Infect Dis. 2011;11(3):181-189. PubMed
15. El-Solh AA, Alhajhusain A, Saliba RG, Drinka P. Physicians’ Attitudes Toward Guidelines for the Treatment of Hospitalized Nursing-Home -Acquired Pneumonia. J Am Med Dir Assoc. 2011;12(4):270-276. PubMed
16. Tunis S, Hayward R, Wilson M, et al. Internists’ Attitudes about Clinical Practice Guidelines. Ann Intern Med. 1994;120(11):956-963. PubMed
17. Seymann GB, Di Francesco L, Sharpe B, et al. The HCAP Gap: Differences between Self-Reported Practice Patterns and Published Guidelines for Health Care-Associated Pneumonia. Clin Infect Dis. 2009;49(12):1868-1874. PubMed
18. Chalmers JD, Rother C, Salih W, Ewig S. Healthcare associated pneumonia does not accurately identify potentially resistant pathogens: a systematic review and meta-analysis. Clin Infect Dis. 2014;58(3):330-339. PubMed
19. Shorr A, Zilberberg MD, Reichley R, et al. Validation of a Clinical Score for Assessing the Risk of Resistant Pathogens in Patients with Pneumonia Presenting to the Emergency Department. Clin Infect Dis. 2012;54(2):193-198. PubMed
20. Aliberti S, Pasquale MD, Zanaboni AM, et al. Stratifying Risk Factors for Multidrug-Resistant Pathogens in Hospitalized Patients Coming from the Community with Pneumonia. Clin Infect Dis. 2012;54(4):470-478. PubMed
21. Schreiber MP, Chan CM, Shorr AF. Resistant Pathogens in Nonnosocomial Pneumonia and Respiratory Failure: Is it Time to Refine the Definition of Health-care-Associated Pneumonia? Chest. 2010;137(6):1283-1288. PubMed
22. Madaras-Kelly KJ, Remington RE, Fan VS, Sloan KL. Predicting antibiotic resistance to community-acquired pneumonia antibiotics in culture-positive patients with healthcare-associated pneumonia. J Hosp Med. 2012;7(3):195-202. PubMed
23. Shindo Y, Ito R, Kobayashi D, et al. Risk factors for drug-resistant pathogens in community-acquired and healthcare-associated pneumonia. Am J Respir Crit Care Med. 2013;188(8):985-995. PubMed
24. Metersky ML, Frei CR, Mortensen EM. Predictors of Pseudomonas and methicillin-resistant Staphylococcus aureus in hospitalized patients with healthcare-associated pneumonia. Respirology. 2016;21(1):157-163. PubMed
25. Webb BJ, Dascomb K, Stenehjem E, Dean N. Predicting risk of drug-resistant organisms in pneumonia: moving beyond the HCAP model. Respir Med. 2015;109(1):1-10. PubMed

References

1. Kochanek KD, Murphy SL, Xu JQ, Tejada-Vera B. Deaths: Final data for 2014. National vital statistics reports; vol 65 no 4. Hyattsville, MD: National Center for Health Statistics. 2016. PubMed
2. American Thoracic Society, Infectious Diseases Society of America. Guidelines for the Management of Adults with Hospital-acquired, Ventilator-associated, and Healthcare-associated Pneumonia. Am J Respir Crit Care Med. 2005;171(4):388-416. PubMed
3. Zilberberg MD, Shorr A. Healthcare-associated pneumonia: the state of the evidence to date. Curr Opin Pulm Med. 2011;17(3):142-147. PubMed
4. Kollef MK, Shorr A, Tabak YP, Gupta V, Liu LZ, Johannes RS. Epidemiology and Outcomes of Health-care-associated pneumonia. Chest. 2005;128(6):3854-3862. PubMed
5. Kalil AC, Metersky ML, Klompas M, et al. Management of Adults With Hospital-acquired and Ventilator-associated Pneumonia: 2016 Clinical Practice Guidelines by the Infectious Diseases Society of America and the American Thoracic Society. Clin Infect Dis. 2016;63(5):575-582. PubMed
6. Lindenauer PK, Pekow PS, Lahti MC, Lee Y, Benjamin EM, Rothberg MB. Association of corticosteroid dose and route of administration with risk of treatment failure in acute exacerbation of chronic obstructive pulmonary disease. JAMA. 2010;303(23):2359-2367. PubMed
7. Elixhauser A, Steiner C, Harris DR, Coffey RM. Comorbidity measures for use with administrative data. Med Care. 1998;36(1):8-27. PubMed
8. Centers for Medicare & Medicaid Services. Frequently asked questions (FAQs): Implementation and maintenance of CMS mortality measures for AMI & HF. 2007. https://www.cms.gov/Medicare/Quality-Initiatives-Patient-Assessment-Instruments/HospitalQualityInits/downloads/HospitalMortalityAboutAMI_HF.pdf. Accessed November 1, 2016.
9. Normand SL, Shahian DM. Statistical and Clinical Aspects of Hospital Outcomes Profiling. Stat Sci. 2007;22(2):206-226. 
10. Rothberg MB, Pekow PS, Priya A, et al. Using highly detailed administrative data to predict pneumonia mortality. PLoS One. 2014;9(1):e87382. PubMed
11. Jones BE, Jones MM, Huttner B, et al. Trends in antibiotic use and nosocomial pathogens in hospitalized veterans with pneumonia at 128 medical centers, 2006-2010. Clin Infect Dis. 2015;61(9):1403-1410. PubMed
12. Attridge RT, Frei CR, Restrepo MI, et al. Guideline-concordant therapy and outcomes in healthcare-associated pneumonia. Eur Respir J. 2011;38(4):878-887. PubMed
13. Rothberg MB, Zilberberg MD, Pekow PS, et al. Association of Guideline-based Antimicrobial Therapy and Outcomes in Healthcare-Associated Pneumonia. J Antimicrob Chemother. 2015;70(5):1573-1579. PubMed
14. Kett DH, Cano E, Quartin AA, et al. Improving Medicine through Pathway Assessment of Critical Therapy of Hospital-Acquired Pneumonia (IMPACT-HAP) Investigators. Implementation of guidelines for management of possible multidrug-resistant pneumonia in intensive care: an observational, multicentre cohort study. Lancet Infect Dis. 2011;11(3):181-189. PubMed
15. El-Solh AA, Alhajhusain A, Saliba RG, Drinka P. Physicians’ Attitudes Toward Guidelines for the Treatment of Hospitalized Nursing-Home -Acquired Pneumonia. J Am Med Dir Assoc. 2011;12(4):270-276. PubMed
16. Tunis S, Hayward R, Wilson M, et al. Internists’ Attitudes about Clinical Practice Guidelines. Ann Intern Med. 1994;120(11):956-963. PubMed
17. Seymann GB, Di Francesco L, Sharpe B, et al. The HCAP Gap: Differences between Self-Reported Practice Patterns and Published Guidelines for Health Care-Associated Pneumonia. Clin Infect Dis. 2009;49(12):1868-1874. PubMed
18. Chalmers JD, Rother C, Salih W, Ewig S. Healthcare associated pneumonia does not accurately identify potentially resistant pathogens: a systematic review and meta-analysis. Clin Infect Dis. 2014;58(3):330-339. PubMed
19. Shorr A, Zilberberg MD, Reichley R, et al. Validation of a Clinical Score for Assessing the Risk of Resistant Pathogens in Patients with Pneumonia Presenting to the Emergency Department. Clin Infect Dis. 2012;54(2):193-198. PubMed
20. Aliberti S, Pasquale MD, Zanaboni AM, et al. Stratifying Risk Factors for Multidrug-Resistant Pathogens in Hospitalized Patients Coming from the Community with Pneumonia. Clin Infect Dis. 2012;54(4):470-478. PubMed
21. Schreiber MP, Chan CM, Shorr AF. Resistant Pathogens in Nonnosocomial Pneumonia and Respiratory Failure: Is it Time to Refine the Definition of Health-care-Associated Pneumonia? Chest. 2010;137(6):1283-1288. PubMed
22. Madaras-Kelly KJ, Remington RE, Fan VS, Sloan KL. Predicting antibiotic resistance to community-acquired pneumonia antibiotics in culture-positive patients with healthcare-associated pneumonia. J Hosp Med. 2012;7(3):195-202. PubMed
23. Shindo Y, Ito R, Kobayashi D, et al. Risk factors for drug-resistant pathogens in community-acquired and healthcare-associated pneumonia. Am J Respir Crit Care Med. 2013;188(8):985-995. PubMed
24. Metersky ML, Frei CR, Mortensen EM. Predictors of Pseudomonas and methicillin-resistant Staphylococcus aureus in hospitalized patients with healthcare-associated pneumonia. Respirology. 2016;21(1):157-163. PubMed
25. Webb BJ, Dascomb K, Stenehjem E, Dean N. Predicting risk of drug-resistant organisms in pneumonia: moving beyond the HCAP model. Respir Med. 2015;109(1):1-10. PubMed

Issue
Journal of Hospital Medicine 12(11)
Issue
Journal of Hospital Medicine 12(11)
Page Number
886-891
Page Number
886-891
Publications
Publications
Topics
Article Type
Sections
Article Source

© 2017 Society of Hospital Medicine

Disallow All Ads
Correspondence Location
Sarah Haessler, MD, Assistant Professor, Tufts University School of Medicine, Infectious Diseases Division, Baystate Medical Center, 759 Chestnut Street, Springfield, MA 01199; Telephone: 413-794-5376; Fax: 413-794-4199; E-mail: Sarah.Haessler@baystatehealth.org
Content Gating
Open Access (article Unlocked/Open Access)
Alternative CME
Disqus Comments
Default
Use ProPublica
Article PDF Media
Media Files

Pediatric Hospitalization Epidemiology

Article Type
Changed
Mon, 01/02/2017 - 19:34
Display Headline
Epidemiology of pediatric hospitalizations at general hospitals and freestanding children's hospitals in the United States

Improvement in the quality of hospital care in the United States is a national priority, both to advance patient safety and because our expenditures exceed any other nation's, but our health outcomes lag behind.[1, 2] Healthcare spending for children is growing at a faster rate than any other age group, with hospital care accounting for more than 40% of pediatric healthcare expenditures.[3] Inpatient healthcare comprises a greater proportion of healthcare costs for children than for adults, yet we have limited knowledge about where this care is provided.[4]

There is substantial variability in the settings in which children are hospitalized. Children may be hospitalized in freestanding children's hospitals, where all services are designed for children and which operate independently of adult‐focused institutions. They may also be hospitalized in general hospitals where care may be provided in a general inpatient bed, on a dedicated pediatric ward, or in a children's hospital nested within a hospital, which may have specialized nursing and physician care but often shares other resources such as laboratory and radiology with the primarily adult‐focused institution. Medical students and residents may be trained in all of these settings. We know little about how these hospital types differ with respect to patient populations, disease volumes, and resource utilization, and this knowledge is important to inform clinical programs, implementation research, and quality improvement (QI) priorities. To this end, we aimed to describe the volume and characteristics of pediatric hospitalizations at acute care general hospitals and freestanding children's hospitals in the United States.

METHODS

Study Design and Eligibility

The data source for this analysis was the Healthcare Cost and Utilization Project's (HCUP) 2012 Kids' Inpatient Database (KID). We conducted a cross‐sectional study of hospitalizations in children and adolescents less than 18 years of age, excluding in‐hospital births and hospitalizations for pregnancy and delivery (identified using All Patient Refined‐Diagnostic Related Groups [APR‐DRGs]).[5] Neonatal hospitalizations not representing in‐hospital births but resulting from transfers or new admissions were retained. Because the dataset does not contain identifiable information, the institutional review board at Baystate Medical Center determined that our study did not constitute human subjects research.

The KID is released every 3 years and is the only publicly available, nationally representative database developed to study pediatric hospitalizations, including an 80% sample of noninborn pediatric discharges from all community, nonrehabilitation hospitals from 44 participating states.[6] Short‐term rehabilitation hospitals, long‐term nonacute care hospitals, psychiatric hospitals, and alcoholism/chemical dependency treatment facilities are excluded. The KID contains information on all patients, regardless of payer, and provides discharge weights to calculate national estimates.[6] It contains both hospital‐level and patient‐level variables, including demographic characteristics, charges, and other clinical and resource use data available from discharge abstracts. Beginning in 2012, freestanding children's hospitals (FCHs) are assigned to a separate stratum in the KID, with data from the Children's Hospital Association used by HCUP to verify the American Hospital Association's (AHA) list of FCHs.[6] Hospitals that are not FCHs were categorized as general hospitals (GHs). We were interested in examining patterns of care at acute care hospitals and not specialty hospitals; unlike previous years, the KID 2012 does not include a specialty hospital identifier.[6] Therefore, as a proxy for specialty hospital status, we excluded hospitals that had 2% hospitalizations for 12 common medical conditions (pneumonia, asthma, bronchiolitis, cellulitis, dehydration, urinary tract infection, neonatal hyperbilirubinemia, fever, upper respiratory infection, infectious gastroenteritis, unspecified viral infection, and croup). These medical conditions were the 12 most common reasons for medical hospitalizations identified using Keren's pediatric diagnosis code grouper,[7] excluding chronic diseases, and represented 26.2% of all pediatric hospitalizations. This 2% threshold was developed empirically, based on visual analysis of the distribution of cases across hospitals and was limited to hospitals with total pediatric volumes >25/year, allowing for stable case‐mix estimates.

Descriptor Variables

Hospital level characteristics included US Census region; teaching status classified in the KID based on results of the AHA Annual Survey; urban/rural location; hospital ownership, classified as public, private nonprofit and private investor‐owned; and total volume of pediatric hospitalizations, in deciles.[6] At the patient level, we examined age, gender, race/ethnicity, expected primary payer, and median household income (in quartiles) for patient's zip code. Medical complexity was categorized as (1) nonchronic disease, (2) complex chronic disease, or (3) noncomplex chronic disease, using the previously validated Pediatric Medical Complexity Algorithm (PMCA) based on International Classification of Diseases, 9th Revision, Clinical Modification (ICD‐9‐CM) codes.[8] Disease severity was classified based on APR‐DRG severity of illness coding, which classifies illnesses severity as minor, moderate, major, or extreme.[9]

We examined the following characteristics of the hospitalizations: (1) length of hospital stay (LOS) measured in calendar days; (2) high‐turnover hospitalization defined as LOS less than 2 days[10, 11, 12]; (3) long LOS, defined as greater than 4 days, equivalent to LOS greater than the 75th percentile; (4) neonatal versus non‐neonatal hospitalization, identified using APR‐DRGs; (5) admission type categorized as elective and nonelective; (6) admission source, categorized as transfer from another acute care hospital, admission from the emergency department, or direct admission; (7) discharge status, categorized as routine discharge, transfer to another hospital or healthcare facility, and discharge against medical advice; and (8) total hospital costs, calculated by applying the cost‐to‐charge ratios available in the KID to total hospital charges.

Reasons for hospitalization were categorized using the pediatric diagnosis code grouper by Keren, which uses ICD‐9‐CM codes to group common and costly principal diagnoses into distinct conditions (eg, pneumonia, idiopathic scoliosis), excluding children who have ICD‐9‐CM principal procedure codes unlikely related to their principal diagnosis (for example, appendectomy for a child with a principal diagnosis of pneumonia).[7] This pediatric grouper classifies diagnoses as medical, surgical, or medical‐surgical based on whether <20% (medical), >80% (surgical) or between 20% and 80% (medical‐surgical) of encounters for the condition had an ICD‐9‐CM principal procedure code for a surgery related to that condition. We further characterized medical hospitalizations as either medical or mental health hospitalizations.

Statistical Analysis

We categorized each discharge record as a hospitalization at a GH or an FCH. We then calculated patient‐level summary statistics, applying weights to calculate national estimates with an associated standard deviation (SD). We assessed differences in characteristics of hospitalizations at GHs and FCHs using Rao‐Scott 2 tests for categorical variables and Wald F tests for continuous variables.[6] We identified the most common reasons for hospitalization, including those responsible for at least 2% of all medical or surgical hospitalizations and at least 0.5% of medical hospitalizations for mental health diagnoses, given the lower prevalence of these conditions and our desire to include mental health diagnoses in our analysis. For these common conditions, we calculated the proportion of condition‐specific hospitalizations and aggregate hospital costs at GHs and FCHs. We also determined the number of hospitalizations at each hospital and calculated the median and interquartile range for the number of hospitalizations for each of these conditions according to hospital type, assessing for differences using Kruskal‐Wallis tests. Finally, we identified the most common and costly conditions at GHs and FCHs by ranking frequency and aggregate costs for each condition according to hospital type, limited to the 20 most costly and/or prevalent pediatric diagnoses. Because we used a novel method to identify specialty hospitals in this dataset, we repeated these analyses using all hospitals classified as a GH and FCH as a sensitivity analysis.

RESULTS

Overall, 3866 hospitals were categorized as a GH, whereas 70 hospitals were categorized as FCHs. Following exclusion of specialty hospitals, 3758 GHs and 50 FCHs were retained in this study. The geographic distribution of hospitals was similar, but although GHs included those in both urban and rural regions, all FCHs were located in urban regions (Table 1).

Characteristics of General Hospitals and Freestanding Children's Hospitals
General Hospitals, n = 3,758 Children's Hospitals, n = 50
Hospital characteristics n % n % P Value
  • NOTE: Abbreviations: IQR, interquartile range.

Geographic region
Northeast 458 12.2 4 8.0 0.50
Midwest 1,209 32.2 15 30.0
South 1,335 35.6 17 34.0
West 753 20.1 14 28.0
Location and teaching status
Rural 1,524 40.6 0 0 <0.0001
Urban nonteaching 1,506 40.1 7 14.0
Urban teaching 725 19.3 43 86.0
Hospital ownership
Government, nonfederal 741 19.7 0 0 <0.0001
Private, nonprofit 2,364 63.0 48 96.0
Private, investor‐owned 650 17.3 2 4.0
Volume of pediatric hospitalizations (deciles)
<185 hospitalizations/year (<8th decile) 2,664 71.0 0 0 <0.0001
186375 hospitalizations/year (8th decile) 378 10.1 2 4.0
376996 hospitalizations/year (9th decile) 380 10.1 1 2.0
>986 hospitalizations/year (10th decile) 333 8.9 47 94.0
Volume of pediatric hospitalizations, median [IQR] 56 [14240] 12,001 [5,83815,448] <0.0001

A total of 1,407,822 (SD 50,456) hospitalizations occurred at GHs, representing 71.7% of pediatric hospitalizations, whereas 554,458 (SD 45,046) hospitalizations occurred at FCHs. Hospitalizations at GHs accounted for 63.6% of days in hospital and 50.0% of pediatric inpatient healthcare costs. Eighty percent of the GHs had total pediatric patient volumes of less than 375 hospitalizations yearly; 11.1% of pediatric hospitalizations occurred at these lower‐volume centers. At FCHs, the median volume of pediatric hospitalizations was 12,001 (interquartile range [IQR]: 583815,448). A total of 36 GHs had pediatric hospitalization volumes in this IQR.

The median age for pediatric patients was slightly higher at GHs, whereas gender, race/ethnicity, primary payer, and median household income for zip code did not differ significantly between hospital types (Table 2). Medical complexity differed between hospital types: children with complex chronic diseases represented 20.2% of hospitalizations at GHs and 35.6% of hospitalizations at FCHs. Severity of illness differed between hospital types, with fewer hospitalizations categorized at the highest level of severity at GHs than FCHs. There were no significant differences between hospital types with respect to the proportion of hospitalizations categorized as neonatal hospitalizations or as elective hospitalizations. The median LOS was shorter at GHs than FCHs. Approximately 1 in 5 children hospitalized at GHs had LOS greater than 4 days, whereas almost 30% of children hospitalized at FCHs had LOS of this duration.

Patient Characteristics and Characteristics of Hospitalizations at General Hospitals and Freestanding Children's Hospitals
Patient Characteristics

General Hospitals,1,407,822 (50,456), 71.7%

Children's Hospitals,554,458 (45,046), 28.3%

P Value
n (SD Weighted Frequency) (%) n (SD Weighted Frequency) %
  • NOTE: Abbreviations: APR‐DRG, All Patient Refined Diagnosis‐Related Group; ED, emergency department; IQR, interquartile range; SD, standard deviation. *Race/ethnicity data missing for approximately 8% of discharge records.[8] Includes in‐hospital death, discharge destination unknown.

Age, y, median [IQR] 3.6 [011.7] 3.4 [010.8] 0.001
Gender (% female) 644,250 (23,089) 45.8 254,505 (20,688) 45.9 0.50
Race*
White 668,876 (27,741) 47.5 233,930 (26,349) 42.2 0.05
Black 231,586 (12,890) 16.5 80,568 (11,739) 14.5
Hispanic 279,021 (16,843) 19.8 12,1425 (21,183) 21.9
Other 133,062 (8,572) 9.5 41,190 (6,394) 7.4
Insurance status
Public 740,033 (28,675) 52.6 284,795 (25,324) 51.4 0.90
Private 563,562 (21,930) 40.0 224,042 (21,613) 40.4
Uninsured 37,265 (1,445) 2.7 16,355 (3,804) 3.0
No charge/other/unknown 66,962 (5,807) 4.8 29,266 (6,789) 5.3
Median household income for zip code, quartiles
<$38,999 457,139 (19,725) 33.3 164,831 (17,016) 30.1 0.07
$39,000$47,999 347,229 (14,104) 25.3 125,105 (10,712) 22.9
$48,000$62,999 304,795 (13,427) 22.2 134,915 (13,999) 24.7
>$63,000 263,171 (15,418) 19.2 122,164 (16,279) 22.3
Medical complexity
Nonchronic disease 717,009 (21,807) 50.9 211,089 (17,023) 38.1 <0.001
Noncomplex chronic disease 406,070 (14,951) 28.8 146,077 (12,442) 26.4
Complex chronic disease 284,742 (17,111) 20.2 197,292 (18,236) 35.6
APR‐DRG severity of illness
1 (lowest severity) 730,134 (23,162) 51.9 217,202 (18,433) 39.2 <0.001
2 486,748 (18,395) 34.6 202,931 (16,864) 36.6
3 146,921 (8,432) 10.4 100,566 (9,041) 18.1
4 (highest severity) 41,749 (3,002) 3.0 33,340 (3,199) 6.0
Hospitalization characteristics
Neonatal hospitalization 98,512 (3,336) 7.0 39,584 (4,274) 7.1 0.84
Admission type
Elective 255,774 (12,285) 18.3 109,854 (13,061) 19.8 0.05
Length of stay, d, (median [IQR]) 1.8 (0.01) [0.8‐3.6] 2.2 (0.06) [1.1‐4.7] <0.001
High turnover hospitalizations 416,790 (14,995) 29.6 130,441 (12,405) 23.5 <0.001
Length of stay >4 days 298,315 (14,421) 21.2 161,804 (14,354) 29.2 <0.001
Admission source
Transfer from another acute care hospital 154,058 (10,067) 10.9 82,118 (8,952) 14.8 0.05
Direct admission 550,123 (21,954) 39.1 211,117 (20,203) 38.1
Admission from ED 703,641 (26,155) 50.0 261,223 (28,708) 47.1
Discharge status
Routine 1,296,638 (46,012) 92.1 519,785 (42,613) 93.8 <0.01
Transfer to another hospital or healthcare facility 56,115 (1,922) 4.0 13,035 (1,437) 2.4
Discharge against medical advice 2,792 (181) 0.2 382 (70) 0.1
Other 52,276 (4,223) 3.7 21,256 (4,501) 3.8

The most common pediatric medical, mental health, and surgical conditions are shown in Figure 1, together representing 32% of pediatric hospitalizations during the study period. For these medical conditions, 77.9% of hospitalizations occurred at GHs, ranging from 52.6% of chemotherapy hospitalizations to 89.0% of hospitalizations for neonatal hyperbilirubinemia. Sixty‐two percent of total hospital costs for these conditions were incurred at GHs. For the common mental health hospitalizations, 86% of hospitalizations occurred at GHs. The majority of hospitalizations and aggregate hospital costs for common surgical conditions also occurred at GHs.

Figure 1
Share of national pediatric hospitalizations and aggregate costs in general and freestanding children's hospitals, by condition, for common medical, mental health and surgical diagnoses. (n = national estimates of number of hospitalizations and associated total hospital costs at general hospitals and children's hospitals).

Whereas pneumonia, asthma, and bronchiolitis were the most common reasons for hospitalization at both GHs and FCHs, the most costly conditions differed (see Supporting Table 1 in the online version of this article). At GHs, these respiratory diseases were responsible for the highest condition‐specific total hospital costs. At FCHs, the highest aggregate costs were due to respiratory distress syndrome and chemotherapy. Congenital heart diseases, including hypoplastic left heart syndrome, transposition of the great vessels, tetralogy of Fallot, endocardial cushion defects, coarctation of the aorta and ventricular septal defects accounted for 6 of the 20 most costly conditions at FCHs.

Figure 2 illustrates the volume of hospitalizations, per hospital, at GHs and FCHs for the most common medical hospitalizations. The median number of hospitalizations, per hospital, was consistently significantly lower at GHs than at FCHs (all P values <0.001). Similar results for surgical and mental health hospitalizations are shown as Supporting Figures 1 and 2 in the online version of this article. In our sensitivity analyses that included all hospitals classified as GH and FCH, all results were essentially unchanged.

Figure 2
Box and whisker plots illustrating median volume of hospitalizations per hospital and associated interquartile range for common medical condition at general hospitals and freestanding children's hospitals (n = number of hospitals represented).

Recognizing the wide range of pediatric volumes at GHs (Table 1) and our inability to differentiate children's hospitals nested within GHs from GHs with pediatric beds, we examined differences in patient and hospitalization characteristics at GHs with volumes 5838 hospitalizations (the 25th percentile for FCH volume) and GHs with pediatric volumes <5838/year (see Supporting Table 2 in the online version of this article). We also compared patient and hospitalization characteristics at FCHs and the higher‐volume GHs. A total of 36 GHs had pediatric volumes 5838, with hospitalizations at these sites together accounting for 15.4% of all pediatric hospitalizations. Characteristics of patients hospitalized at these higher‐volume GHs were similar to patients hospitalized at FCHs, but they had significantly lower disease severity, fewer neonatal hospitalizations, shorter LOS, and more high‐turnover hospitalizations than patients hospitalized at FCHs. We also observed several differences between children hospitalized at higher‐ and lower‐volume GHs (see Supporting Table 2 in the online version of this article). Children hospitalized at the lower‐volume GHs were more likely to have public health insurance and less likely to have complex chronic diseases, although overall, 39.0% of all hospitalizations for children with complex chronic diseases occurred at these lower‐volume GHs. Compared to children hospitalized at higher‐volume GHs, children hospitalized at the lower‐volume hospitals had significantly lower disease severity, shorter LOS, more direct admissions, and a greater proportion of routine discharges.

DISCUSSION

Of the 2 million pediatric hospitalizations in the United States in 2012, more than 70% occurred at GHs. We observed considerable heterogeneity in pediatric volumes across GHs, with 11% of pediatric hospitalizations occurring at hospitals with pediatric volumes of <375 hospitalizations annually, whereas 15% of pediatric hospitalizations occurred at GHs with volumes similar to those observed at FCHs. The remaining pediatric hospitalizations at GHs occurred at centers with intermediate volumes. The most common reasons for hospitalization were similar at GHs and FCHs, but the most costly conditions differed substantially. These findings have important implications for pediatric clinical care programs, research, and QI efforts.

Our finding that more than 70% of pediatric hospitalizations occurred at GHs speaks to the importance of quality measurement at these hospitals, whereas low per‐hospital pediatric volumes at the majority of GHs makes such measurement particularly challenging. Several previous studies have illustrated that volumes of pediatric hospitalizations are too small to detect meaningful differences in quality between hospitals using established condition‐specific metrics.[13, 14, 15] Our finding that more than 10% of pediatric hospitalizations occurred at GHs with pediatric volumes <375 year supports previous research suggesting that cross‐cutting, all‐condition quality metrics, composite measures, and/or multihospital reporting networks may be needed to enable quality measurement at these sites. In addition, the heterogeneity in patient volumes and characteristics across GHs raise questions about the applicability of quality metrics developed and validated at FCHs to the many GH settings. Field‐testing quality measures to ensure their validity at diverse GHs, particularly those with patient volumes and infrastructure different from FCHs, will be important to meaningful pediatric quality measurement.

Our results illustrating differences in the most common and costly conditions at GHs and FCHs have further implications for prioritization and implementation of research and QI efforts. Implementation research and QI efforts focused on cardiac and neurosurgical procedures, as well as neonatal intensive care, may have considerable impact on cost and quality at FCHs. At GHs, research and QI efforts focused on common conditions are needed to increase our knowledge of contextually relevant barriers to and facilitators of high‐quality pediatric care. This, however, can be made more difficult by small sample sizes, limited resources, and infrastructure, and competing priorities in adult‐focused GH settings.[16, 17, 18] Multihospital learning collaboratives and partnerships between FCHs and GHs can begin to address these challenges, but their success is contingent upon national advocacy and funding to support pediatric research and quality measures at GHs.

One of the most notable differences in the characteristics of pediatric hospitalizations at GHs and FCHs was the proportion of hospitalizations attributable to children with medical complexity (CMC); more than one‐third of hospitalizations at FCHs were for CMC compared to 1 in 5 at GHs. These findings align with the results of several previous studies describing the substantial resource utilization attributed to CMC, and with growing research, innovation, and quality metrics focused on improving both inpatient and outpatient care for these vulnerable children.[19, 20, 21, 22] Structured complex care programs, developed to improve care coordination and healthcare quality for CMC, are common at FCHs, and have been associated with decreased resource utilization and improved outcomes.[23, 24, 25] Notably, however, more than half of all hospitalizations for CMC, exceeding 250,000 annually, occurred at GHs, and almost 40% of hospitalizations for CMC occurred at the lower‐volume GHs. These findings speak to the importance of translating effective and innovative programs of care for CMC to GHs as resources allow, accompanied by robust evaluations of their effectiveness. Lower patient volume at most GHs, however, may be a barrier to dedicated CMC programs. As a result, decentralized community‐based programs of care for CMC, linking primary care programs with regional and tertiary care hospitals, warrant further consideration.[26, 27, 28]

This analysis should be interpreted in light of several limitations. First, we were unable to distinguish between GHs with scant pediatric‐specific resources from those with a large volume of dedicated pediatric resources, such as children's hospitals nested within GHs. We did identify 36 GHs with pediatric volumes similar to those observed at FCHs (see Supporting Table 2 in the online version of this article); patient and hospitalization characteristics at these higher‐volume GHs were similar in many ways to children hospitalized at FCHs. Several of these higher‐volume GHs may have considerable resources dedicated to the care of children, including subspecialty care, and may represent children's hospitals nested within GHs. Because nested children's hospitals are included in the GH categorization, our results may have underestimated the proportion of children cared for at children's hospitals. Further work is needed to identify the health systems challenges and opportunities that may be unique to these institutions. Second, because the 2012 KID does not include a specialty hospital indicator, we developed a proxy method for identifying these hospitals, which may have resulted in some misclassification. We are reassured that the results of our analyses did not change substantively when we included all hospitals. Similarly, although we are reassured that the number of hospitals classified in our analysis as acute care FCHs aligns, approximately, with the number of hospitals classified as such by the Children's Hospital Association, we were unable to assess the validity of this variable within the KID. Third, the KID does not link records at the patient level, so we are unable to report the number of unique children included in this analysis. In addition, the KID includes only inpatient stays with exclusion of observation status stays; potential differences between GH and FCH in the use of observation status could have biased our findings. Fifth, we used the PMCA to identify CMC; although this algorithm has been shown to have excellent sensitivity in identifying children with chronic diseases, using up to 3 years of Medicaid claims data, the sensitivity using the KID, where only 1 inpatient stay is available for assessment, is unknown.[8, 29] Similarly, use of Keren's pediatric diagnosis grouper to classify reasons for hospitalization may have resulted in misclassification, though there are few other nonproprietary pediatric‐specific diagnostic groupers available.

In 2012, more than 70% of pediatric hospitalizations occurred at GHs in the United States. The considerably higher pediatric volumes at FCHs makes these institutions well suited for research, innovation, and the development and application of disease‐specific QI initiatives. Recognizing that the majority of pediatric hospitalizations occurred at GHs, there is a clear need for implementation research, program development, and quality metrics that align with the characteristics of hospitalizations at these centers. National support for research and quality improvement that reflects the diverse hospital settings where children receive their hospital care is critical to further our nation's goal of improving hospital quality for children.

Disclosures

Dr. Leyenaar was supported by grant number K08HS024133 from the Agency for Healthcare Research and Quality. The content is solely the responsibility of the authors and does not necessarily represent the official views of the Agency for Healthcare Research and Quality. The authors have no conflicts of interest relevant to this article to disclose.

Files
References
  1. Davis K, Stremikis K, Squires D, Schoen C. Mirror, Mirror on the wall: how the performance of the US health care system compares internationally. The Commonwealth Fund. Available at: http://www.commonwealthfund.org/publications/fund‐reports/2014/jun/mirror‐mirror. Published June 16, 2014. Accessed August 26, 2015.
  2. Fairbrother G, Guttmann A, Klein JD, Simpson LA, Thomas P, Kempe A. Higher cost, but poorer outcomes: the US health disadvantage and implications for pediatrics. Pediatrics. 2015;135(6):961964.
  3. Lassman D, Hartman M, Washington B, Andrews K, Catlin A. US health spending trends by age and gender: selected years 2002–10. Health Aff (Millwood). 2014;33(5):815822.
  4. Moore B, Levit K, Elixhauser A. Costs for hospital stays in the United States, 2012. Healthcare Cost and Utilization Project 181. Available at: http://www.hcup‐us.ahrq.gov/reports/statbriefs/sb181‐Hospital‐Costs‐United‐States‐2012.pdf. Published October 2014. Accessed September 2015.
  5. Averill RF, Goldfield N, Hughes JS, et al. All Patient Refined Diagnosis Related Groups: Methodology Overview. 3M Health Information Systems. Available at: https://www.hcup‐us.ahrq.gov/db/nation/nis/APR‐DRGsV20MethodologyOverviewandBibliography.pdf. Accessed February 8, 2016.
  6. Agency for Healthcare Research and Quality. Healthcare Cost and Utilization Project. Introduction to the HCUP Kids' Inpatient Database (KID) 2012. Available at: https://www.hcup‐us.ahrq.gov/db/nation/kid/kid_2012_introduction.jsp. Published Issued July 2014. Accessed February 8, 2016.
  7. Keren R. Prioritization of comparative effectiveness research topics in hospital pediatrics. Arch Pediatr Adolesc Med. 2012;166(12):1155.
  8. Simon TD, Cawthon ML, Stanford S, et al. Pediatric medical complexity algorithm: a new method to stratify children by medical complexity. Pediatrics. 2014;133(6):e1647e1654.
  9. Averill RF, Goldfield N, Hughes JS, et al. 3M APR DRG Classification System. 3M Health Information Systems. Available at: http://www.hcup‐us.ahrq.gov/db/nation/nis/v261_aprdrg_meth_ovrview.pdf. Accessed August 7, 2015.
  10. Macy ML, Stanley RM, Lozon MM, Sasson C, Gebremariam A, Davis MM. Trends in high‐turnover stays among children hospitalized in the United States, 1993–2003. Pediatrics. 2009;123(3):9961002.
  11. Macy ML, Stanley RM, Sasson C, Gebremariam A DM. High turnover stays for pediatric asthma in the United States. Med Care. 2010;48(9):827833.
  12. Leyenaar JK, Shieh M, Lagu T, Pekow PS, Lindenauer PK. Variation and outcomes associated with direct admission among children with pneumonia in the United States. JAMA Pediatr. 2014;168(9):829836.
  13. Berry JG, Zaslavsky AM, Toomey SL, et al. Recognizing differences in hospital quality performance for pediatric inpatient care. Pediatrics. 2015;136(2):251262.
  14. Bardach NS, Chien AT, Dudley RA. Small numbers limit the use of the inpatient pediatric quality indicators for hospital comparison. Acad Pediatr. 2010;10(4):266273.
  15. Feudtner C, Berry JG, Parry G, et al. Statistical uncertainty of mortality rates and rankings for children's hospitals. Pediatrics. 2011;128(4):e966e972.
  16. Leyenaar JK, Capra LA, O'Brien ER, Leslie LK, Mackie TI. Determinants of career satisfaction among pediatric hospitalists: a qualitative exploration. Acad Pediatr. 2014;14(4):361368.
  17. Simon TD, Starmer AJ, Conway PH, et al. Quality improvement research in pediatric hospital medicine and the role of the Pediatric Research in Inpatient Settings (PRIS) network. Acad Pediatr. 2013;13(6 suppl):S54S60.
  18. Miller M. Roles for children's hospitals in pediatric collaborative improvement networks. Pediatrics. 2013;131(suppl 4):S215S218.
  19. Cohen E, Kuo DZ, Agrawal R, et al. Children with medical complexity: an emerging population for clinical and research initiatives. Pediatrics. 2011;127(3):529538.
  20. Simon TD, Berry J, Feudtner C, et al. Children with complex chronic conditions in inpatient hospital settings in the United States. Pediatrics. 2010;126(4):647655.
  21. Cohen E, Berry JG, Camacho X, Anderson G, Wodchis W, Guttmann A. Patterns and costs of health care use of children with medical complexity. Pediatrics. 2012;130(6):e1463e1470.
  22. Berry JG, Hall DE, Kuo DZ, Hall M, Kueser J, Kaplan W. Hospital utilization and characteristics of patients experiencing recurrent readmissions within children's hospitals. JAMA. 2011;305(7):682690.
  23. Berry JG, Agrawal R, Kuo DZ, et al. Characteristics of hospitalizations for patients who use a structured clinical care program for children with medical complexity. J Pediatr. 2011;159(2):284290.
  24. Cohen E, Jovcevska V, Kuo D, Mahant S. Hospital‐based comprehensive care programs for children with special health care needs: a systematic review. Arch Pediatr Adolesc Med. 2011;165(6):554561.
  25. Gordon J, Colby H, Bartelt T, Jablonski D, Krauthoefer ML, Havens P. A tertiary care–primary care partnership model for medically complex and fragile children and youth with special health care needs. Arch Pediatr Adolesc Med. 2007;161(10):937944.
  26. Cohen E, Lacombe‐Duncan A, Spalding K, et al. Integrated complex care coordination for children with medical complexity: a mixed‐methods evaluation of tertiary care‐community collaboration. BMC Health Serv Res. 2012;12:366.
  27. Lerner CF, Kelly RB, Hamilton LJ, Klitzner TS. Medical transport of children with complex chronic conditions. Emerg Med Int. 2012;2012:837020.
  28. Stiles AD, Tayloe DT, Wegner SE. Comanagement of medically complex children by subspecialists, generalists, and care coordinators. Pediatrics. 2014;134(2):203205.
  29. Berry JG, Hall M, Cohen E, O'Neill M, Feudtner C. Ways to identify children with medical complexity and the importance of why. J Pediatr. 2015;167(2):229237.
Article PDF
Issue
Journal of Hospital Medicine - 11(11)
Publications
Page Number
743-749
Sections
Files
Files
Article PDF
Article PDF

Improvement in the quality of hospital care in the United States is a national priority, both to advance patient safety and because our expenditures exceed any other nation's, but our health outcomes lag behind.[1, 2] Healthcare spending for children is growing at a faster rate than any other age group, with hospital care accounting for more than 40% of pediatric healthcare expenditures.[3] Inpatient healthcare comprises a greater proportion of healthcare costs for children than for adults, yet we have limited knowledge about where this care is provided.[4]

There is substantial variability in the settings in which children are hospitalized. Children may be hospitalized in freestanding children's hospitals, where all services are designed for children and which operate independently of adult‐focused institutions. They may also be hospitalized in general hospitals where care may be provided in a general inpatient bed, on a dedicated pediatric ward, or in a children's hospital nested within a hospital, which may have specialized nursing and physician care but often shares other resources such as laboratory and radiology with the primarily adult‐focused institution. Medical students and residents may be trained in all of these settings. We know little about how these hospital types differ with respect to patient populations, disease volumes, and resource utilization, and this knowledge is important to inform clinical programs, implementation research, and quality improvement (QI) priorities. To this end, we aimed to describe the volume and characteristics of pediatric hospitalizations at acute care general hospitals and freestanding children's hospitals in the United States.

METHODS

Study Design and Eligibility

The data source for this analysis was the Healthcare Cost and Utilization Project's (HCUP) 2012 Kids' Inpatient Database (KID). We conducted a cross‐sectional study of hospitalizations in children and adolescents less than 18 years of age, excluding in‐hospital births and hospitalizations for pregnancy and delivery (identified using All Patient Refined‐Diagnostic Related Groups [APR‐DRGs]).[5] Neonatal hospitalizations not representing in‐hospital births but resulting from transfers or new admissions were retained. Because the dataset does not contain identifiable information, the institutional review board at Baystate Medical Center determined that our study did not constitute human subjects research.

The KID is released every 3 years and is the only publicly available, nationally representative database developed to study pediatric hospitalizations, including an 80% sample of noninborn pediatric discharges from all community, nonrehabilitation hospitals from 44 participating states.[6] Short‐term rehabilitation hospitals, long‐term nonacute care hospitals, psychiatric hospitals, and alcoholism/chemical dependency treatment facilities are excluded. The KID contains information on all patients, regardless of payer, and provides discharge weights to calculate national estimates.[6] It contains both hospital‐level and patient‐level variables, including demographic characteristics, charges, and other clinical and resource use data available from discharge abstracts. Beginning in 2012, freestanding children's hospitals (FCHs) are assigned to a separate stratum in the KID, with data from the Children's Hospital Association used by HCUP to verify the American Hospital Association's (AHA) list of FCHs.[6] Hospitals that are not FCHs were categorized as general hospitals (GHs). We were interested in examining patterns of care at acute care hospitals and not specialty hospitals; unlike previous years, the KID 2012 does not include a specialty hospital identifier.[6] Therefore, as a proxy for specialty hospital status, we excluded hospitals that had 2% hospitalizations for 12 common medical conditions (pneumonia, asthma, bronchiolitis, cellulitis, dehydration, urinary tract infection, neonatal hyperbilirubinemia, fever, upper respiratory infection, infectious gastroenteritis, unspecified viral infection, and croup). These medical conditions were the 12 most common reasons for medical hospitalizations identified using Keren's pediatric diagnosis code grouper,[7] excluding chronic diseases, and represented 26.2% of all pediatric hospitalizations. This 2% threshold was developed empirically, based on visual analysis of the distribution of cases across hospitals and was limited to hospitals with total pediatric volumes >25/year, allowing for stable case‐mix estimates.

Descriptor Variables

Hospital level characteristics included US Census region; teaching status classified in the KID based on results of the AHA Annual Survey; urban/rural location; hospital ownership, classified as public, private nonprofit and private investor‐owned; and total volume of pediatric hospitalizations, in deciles.[6] At the patient level, we examined age, gender, race/ethnicity, expected primary payer, and median household income (in quartiles) for patient's zip code. Medical complexity was categorized as (1) nonchronic disease, (2) complex chronic disease, or (3) noncomplex chronic disease, using the previously validated Pediatric Medical Complexity Algorithm (PMCA) based on International Classification of Diseases, 9th Revision, Clinical Modification (ICD‐9‐CM) codes.[8] Disease severity was classified based on APR‐DRG severity of illness coding, which classifies illnesses severity as minor, moderate, major, or extreme.[9]

We examined the following characteristics of the hospitalizations: (1) length of hospital stay (LOS) measured in calendar days; (2) high‐turnover hospitalization defined as LOS less than 2 days[10, 11, 12]; (3) long LOS, defined as greater than 4 days, equivalent to LOS greater than the 75th percentile; (4) neonatal versus non‐neonatal hospitalization, identified using APR‐DRGs; (5) admission type categorized as elective and nonelective; (6) admission source, categorized as transfer from another acute care hospital, admission from the emergency department, or direct admission; (7) discharge status, categorized as routine discharge, transfer to another hospital or healthcare facility, and discharge against medical advice; and (8) total hospital costs, calculated by applying the cost‐to‐charge ratios available in the KID to total hospital charges.

Reasons for hospitalization were categorized using the pediatric diagnosis code grouper by Keren, which uses ICD‐9‐CM codes to group common and costly principal diagnoses into distinct conditions (eg, pneumonia, idiopathic scoliosis), excluding children who have ICD‐9‐CM principal procedure codes unlikely related to their principal diagnosis (for example, appendectomy for a child with a principal diagnosis of pneumonia).[7] This pediatric grouper classifies diagnoses as medical, surgical, or medical‐surgical based on whether <20% (medical), >80% (surgical) or between 20% and 80% (medical‐surgical) of encounters for the condition had an ICD‐9‐CM principal procedure code for a surgery related to that condition. We further characterized medical hospitalizations as either medical or mental health hospitalizations.

Statistical Analysis

We categorized each discharge record as a hospitalization at a GH or an FCH. We then calculated patient‐level summary statistics, applying weights to calculate national estimates with an associated standard deviation (SD). We assessed differences in characteristics of hospitalizations at GHs and FCHs using Rao‐Scott 2 tests for categorical variables and Wald F tests for continuous variables.[6] We identified the most common reasons for hospitalization, including those responsible for at least 2% of all medical or surgical hospitalizations and at least 0.5% of medical hospitalizations for mental health diagnoses, given the lower prevalence of these conditions and our desire to include mental health diagnoses in our analysis. For these common conditions, we calculated the proportion of condition‐specific hospitalizations and aggregate hospital costs at GHs and FCHs. We also determined the number of hospitalizations at each hospital and calculated the median and interquartile range for the number of hospitalizations for each of these conditions according to hospital type, assessing for differences using Kruskal‐Wallis tests. Finally, we identified the most common and costly conditions at GHs and FCHs by ranking frequency and aggregate costs for each condition according to hospital type, limited to the 20 most costly and/or prevalent pediatric diagnoses. Because we used a novel method to identify specialty hospitals in this dataset, we repeated these analyses using all hospitals classified as a GH and FCH as a sensitivity analysis.

RESULTS

Overall, 3866 hospitals were categorized as a GH, whereas 70 hospitals were categorized as FCHs. Following exclusion of specialty hospitals, 3758 GHs and 50 FCHs were retained in this study. The geographic distribution of hospitals was similar, but although GHs included those in both urban and rural regions, all FCHs were located in urban regions (Table 1).

Characteristics of General Hospitals and Freestanding Children's Hospitals
General Hospitals, n = 3,758 Children's Hospitals, n = 50
Hospital characteristics n % n % P Value
  • NOTE: Abbreviations: IQR, interquartile range.

Geographic region
Northeast 458 12.2 4 8.0 0.50
Midwest 1,209 32.2 15 30.0
South 1,335 35.6 17 34.0
West 753 20.1 14 28.0
Location and teaching status
Rural 1,524 40.6 0 0 <0.0001
Urban nonteaching 1,506 40.1 7 14.0
Urban teaching 725 19.3 43 86.0
Hospital ownership
Government, nonfederal 741 19.7 0 0 <0.0001
Private, nonprofit 2,364 63.0 48 96.0
Private, investor‐owned 650 17.3 2 4.0
Volume of pediatric hospitalizations (deciles)
<185 hospitalizations/year (<8th decile) 2,664 71.0 0 0 <0.0001
186375 hospitalizations/year (8th decile) 378 10.1 2 4.0
376996 hospitalizations/year (9th decile) 380 10.1 1 2.0
>986 hospitalizations/year (10th decile) 333 8.9 47 94.0
Volume of pediatric hospitalizations, median [IQR] 56 [14240] 12,001 [5,83815,448] <0.0001

A total of 1,407,822 (SD 50,456) hospitalizations occurred at GHs, representing 71.7% of pediatric hospitalizations, whereas 554,458 (SD 45,046) hospitalizations occurred at FCHs. Hospitalizations at GHs accounted for 63.6% of days in hospital and 50.0% of pediatric inpatient healthcare costs. Eighty percent of the GHs had total pediatric patient volumes of less than 375 hospitalizations yearly; 11.1% of pediatric hospitalizations occurred at these lower‐volume centers. At FCHs, the median volume of pediatric hospitalizations was 12,001 (interquartile range [IQR]: 583815,448). A total of 36 GHs had pediatric hospitalization volumes in this IQR.

The median age for pediatric patients was slightly higher at GHs, whereas gender, race/ethnicity, primary payer, and median household income for zip code did not differ significantly between hospital types (Table 2). Medical complexity differed between hospital types: children with complex chronic diseases represented 20.2% of hospitalizations at GHs and 35.6% of hospitalizations at FCHs. Severity of illness differed between hospital types, with fewer hospitalizations categorized at the highest level of severity at GHs than FCHs. There were no significant differences between hospital types with respect to the proportion of hospitalizations categorized as neonatal hospitalizations or as elective hospitalizations. The median LOS was shorter at GHs than FCHs. Approximately 1 in 5 children hospitalized at GHs had LOS greater than 4 days, whereas almost 30% of children hospitalized at FCHs had LOS of this duration.

Patient Characteristics and Characteristics of Hospitalizations at General Hospitals and Freestanding Children's Hospitals
Patient Characteristics

General Hospitals,1,407,822 (50,456), 71.7%

Children's Hospitals,554,458 (45,046), 28.3%

P Value
n (SD Weighted Frequency) (%) n (SD Weighted Frequency) %
  • NOTE: Abbreviations: APR‐DRG, All Patient Refined Diagnosis‐Related Group; ED, emergency department; IQR, interquartile range; SD, standard deviation. *Race/ethnicity data missing for approximately 8% of discharge records.[8] Includes in‐hospital death, discharge destination unknown.

Age, y, median [IQR] 3.6 [011.7] 3.4 [010.8] 0.001
Gender (% female) 644,250 (23,089) 45.8 254,505 (20,688) 45.9 0.50
Race*
White 668,876 (27,741) 47.5 233,930 (26,349) 42.2 0.05
Black 231,586 (12,890) 16.5 80,568 (11,739) 14.5
Hispanic 279,021 (16,843) 19.8 12,1425 (21,183) 21.9
Other 133,062 (8,572) 9.5 41,190 (6,394) 7.4
Insurance status
Public 740,033 (28,675) 52.6 284,795 (25,324) 51.4 0.90
Private 563,562 (21,930) 40.0 224,042 (21,613) 40.4
Uninsured 37,265 (1,445) 2.7 16,355 (3,804) 3.0
No charge/other/unknown 66,962 (5,807) 4.8 29,266 (6,789) 5.3
Median household income for zip code, quartiles
<$38,999 457,139 (19,725) 33.3 164,831 (17,016) 30.1 0.07
$39,000$47,999 347,229 (14,104) 25.3 125,105 (10,712) 22.9
$48,000$62,999 304,795 (13,427) 22.2 134,915 (13,999) 24.7
>$63,000 263,171 (15,418) 19.2 122,164 (16,279) 22.3
Medical complexity
Nonchronic disease 717,009 (21,807) 50.9 211,089 (17,023) 38.1 <0.001
Noncomplex chronic disease 406,070 (14,951) 28.8 146,077 (12,442) 26.4
Complex chronic disease 284,742 (17,111) 20.2 197,292 (18,236) 35.6
APR‐DRG severity of illness
1 (lowest severity) 730,134 (23,162) 51.9 217,202 (18,433) 39.2 <0.001
2 486,748 (18,395) 34.6 202,931 (16,864) 36.6
3 146,921 (8,432) 10.4 100,566 (9,041) 18.1
4 (highest severity) 41,749 (3,002) 3.0 33,340 (3,199) 6.0
Hospitalization characteristics
Neonatal hospitalization 98,512 (3,336) 7.0 39,584 (4,274) 7.1 0.84
Admission type
Elective 255,774 (12,285) 18.3 109,854 (13,061) 19.8 0.05
Length of stay, d, (median [IQR]) 1.8 (0.01) [0.8‐3.6] 2.2 (0.06) [1.1‐4.7] <0.001
High turnover hospitalizations 416,790 (14,995) 29.6 130,441 (12,405) 23.5 <0.001
Length of stay >4 days 298,315 (14,421) 21.2 161,804 (14,354) 29.2 <0.001
Admission source
Transfer from another acute care hospital 154,058 (10,067) 10.9 82,118 (8,952) 14.8 0.05
Direct admission 550,123 (21,954) 39.1 211,117 (20,203) 38.1
Admission from ED 703,641 (26,155) 50.0 261,223 (28,708) 47.1
Discharge status
Routine 1,296,638 (46,012) 92.1 519,785 (42,613) 93.8 <0.01
Transfer to another hospital or healthcare facility 56,115 (1,922) 4.0 13,035 (1,437) 2.4
Discharge against medical advice 2,792 (181) 0.2 382 (70) 0.1
Other 52,276 (4,223) 3.7 21,256 (4,501) 3.8

The most common pediatric medical, mental health, and surgical conditions are shown in Figure 1, together representing 32% of pediatric hospitalizations during the study period. For these medical conditions, 77.9% of hospitalizations occurred at GHs, ranging from 52.6% of chemotherapy hospitalizations to 89.0% of hospitalizations for neonatal hyperbilirubinemia. Sixty‐two percent of total hospital costs for these conditions were incurred at GHs. For the common mental health hospitalizations, 86% of hospitalizations occurred at GHs. The majority of hospitalizations and aggregate hospital costs for common surgical conditions also occurred at GHs.

Figure 1
Share of national pediatric hospitalizations and aggregate costs in general and freestanding children's hospitals, by condition, for common medical, mental health and surgical diagnoses. (n = national estimates of number of hospitalizations and associated total hospital costs at general hospitals and children's hospitals).

Whereas pneumonia, asthma, and bronchiolitis were the most common reasons for hospitalization at both GHs and FCHs, the most costly conditions differed (see Supporting Table 1 in the online version of this article). At GHs, these respiratory diseases were responsible for the highest condition‐specific total hospital costs. At FCHs, the highest aggregate costs were due to respiratory distress syndrome and chemotherapy. Congenital heart diseases, including hypoplastic left heart syndrome, transposition of the great vessels, tetralogy of Fallot, endocardial cushion defects, coarctation of the aorta and ventricular septal defects accounted for 6 of the 20 most costly conditions at FCHs.

Figure 2 illustrates the volume of hospitalizations, per hospital, at GHs and FCHs for the most common medical hospitalizations. The median number of hospitalizations, per hospital, was consistently significantly lower at GHs than at FCHs (all P values <0.001). Similar results for surgical and mental health hospitalizations are shown as Supporting Figures 1 and 2 in the online version of this article. In our sensitivity analyses that included all hospitals classified as GH and FCH, all results were essentially unchanged.

Figure 2
Box and whisker plots illustrating median volume of hospitalizations per hospital and associated interquartile range for common medical condition at general hospitals and freestanding children's hospitals (n = number of hospitals represented).

Recognizing the wide range of pediatric volumes at GHs (Table 1) and our inability to differentiate children's hospitals nested within GHs from GHs with pediatric beds, we examined differences in patient and hospitalization characteristics at GHs with volumes 5838 hospitalizations (the 25th percentile for FCH volume) and GHs with pediatric volumes <5838/year (see Supporting Table 2 in the online version of this article). We also compared patient and hospitalization characteristics at FCHs and the higher‐volume GHs. A total of 36 GHs had pediatric volumes 5838, with hospitalizations at these sites together accounting for 15.4% of all pediatric hospitalizations. Characteristics of patients hospitalized at these higher‐volume GHs were similar to patients hospitalized at FCHs, but they had significantly lower disease severity, fewer neonatal hospitalizations, shorter LOS, and more high‐turnover hospitalizations than patients hospitalized at FCHs. We also observed several differences between children hospitalized at higher‐ and lower‐volume GHs (see Supporting Table 2 in the online version of this article). Children hospitalized at the lower‐volume GHs were more likely to have public health insurance and less likely to have complex chronic diseases, although overall, 39.0% of all hospitalizations for children with complex chronic diseases occurred at these lower‐volume GHs. Compared to children hospitalized at higher‐volume GHs, children hospitalized at the lower‐volume hospitals had significantly lower disease severity, shorter LOS, more direct admissions, and a greater proportion of routine discharges.

DISCUSSION

Of the 2 million pediatric hospitalizations in the United States in 2012, more than 70% occurred at GHs. We observed considerable heterogeneity in pediatric volumes across GHs, with 11% of pediatric hospitalizations occurring at hospitals with pediatric volumes of <375 hospitalizations annually, whereas 15% of pediatric hospitalizations occurred at GHs with volumes similar to those observed at FCHs. The remaining pediatric hospitalizations at GHs occurred at centers with intermediate volumes. The most common reasons for hospitalization were similar at GHs and FCHs, but the most costly conditions differed substantially. These findings have important implications for pediatric clinical care programs, research, and QI efforts.

Our finding that more than 70% of pediatric hospitalizations occurred at GHs speaks to the importance of quality measurement at these hospitals, whereas low per‐hospital pediatric volumes at the majority of GHs makes such measurement particularly challenging. Several previous studies have illustrated that volumes of pediatric hospitalizations are too small to detect meaningful differences in quality between hospitals using established condition‐specific metrics.[13, 14, 15] Our finding that more than 10% of pediatric hospitalizations occurred at GHs with pediatric volumes <375 year supports previous research suggesting that cross‐cutting, all‐condition quality metrics, composite measures, and/or multihospital reporting networks may be needed to enable quality measurement at these sites. In addition, the heterogeneity in patient volumes and characteristics across GHs raise questions about the applicability of quality metrics developed and validated at FCHs to the many GH settings. Field‐testing quality measures to ensure their validity at diverse GHs, particularly those with patient volumes and infrastructure different from FCHs, will be important to meaningful pediatric quality measurement.

Our results illustrating differences in the most common and costly conditions at GHs and FCHs have further implications for prioritization and implementation of research and QI efforts. Implementation research and QI efforts focused on cardiac and neurosurgical procedures, as well as neonatal intensive care, may have considerable impact on cost and quality at FCHs. At GHs, research and QI efforts focused on common conditions are needed to increase our knowledge of contextually relevant barriers to and facilitators of high‐quality pediatric care. This, however, can be made more difficult by small sample sizes, limited resources, and infrastructure, and competing priorities in adult‐focused GH settings.[16, 17, 18] Multihospital learning collaboratives and partnerships between FCHs and GHs can begin to address these challenges, but their success is contingent upon national advocacy and funding to support pediatric research and quality measures at GHs.

One of the most notable differences in the characteristics of pediatric hospitalizations at GHs and FCHs was the proportion of hospitalizations attributable to children with medical complexity (CMC); more than one‐third of hospitalizations at FCHs were for CMC compared to 1 in 5 at GHs. These findings align with the results of several previous studies describing the substantial resource utilization attributed to CMC, and with growing research, innovation, and quality metrics focused on improving both inpatient and outpatient care for these vulnerable children.[19, 20, 21, 22] Structured complex care programs, developed to improve care coordination and healthcare quality for CMC, are common at FCHs, and have been associated with decreased resource utilization and improved outcomes.[23, 24, 25] Notably, however, more than half of all hospitalizations for CMC, exceeding 250,000 annually, occurred at GHs, and almost 40% of hospitalizations for CMC occurred at the lower‐volume GHs. These findings speak to the importance of translating effective and innovative programs of care for CMC to GHs as resources allow, accompanied by robust evaluations of their effectiveness. Lower patient volume at most GHs, however, may be a barrier to dedicated CMC programs. As a result, decentralized community‐based programs of care for CMC, linking primary care programs with regional and tertiary care hospitals, warrant further consideration.[26, 27, 28]

This analysis should be interpreted in light of several limitations. First, we were unable to distinguish between GHs with scant pediatric‐specific resources from those with a large volume of dedicated pediatric resources, such as children's hospitals nested within GHs. We did identify 36 GHs with pediatric volumes similar to those observed at FCHs (see Supporting Table 2 in the online version of this article); patient and hospitalization characteristics at these higher‐volume GHs were similar in many ways to children hospitalized at FCHs. Several of these higher‐volume GHs may have considerable resources dedicated to the care of children, including subspecialty care, and may represent children's hospitals nested within GHs. Because nested children's hospitals are included in the GH categorization, our results may have underestimated the proportion of children cared for at children's hospitals. Further work is needed to identify the health systems challenges and opportunities that may be unique to these institutions. Second, because the 2012 KID does not include a specialty hospital indicator, we developed a proxy method for identifying these hospitals, which may have resulted in some misclassification. We are reassured that the results of our analyses did not change substantively when we included all hospitals. Similarly, although we are reassured that the number of hospitals classified in our analysis as acute care FCHs aligns, approximately, with the number of hospitals classified as such by the Children's Hospital Association, we were unable to assess the validity of this variable within the KID. Third, the KID does not link records at the patient level, so we are unable to report the number of unique children included in this analysis. In addition, the KID includes only inpatient stays with exclusion of observation status stays; potential differences between GH and FCH in the use of observation status could have biased our findings. Fifth, we used the PMCA to identify CMC; although this algorithm has been shown to have excellent sensitivity in identifying children with chronic diseases, using up to 3 years of Medicaid claims data, the sensitivity using the KID, where only 1 inpatient stay is available for assessment, is unknown.[8, 29] Similarly, use of Keren's pediatric diagnosis grouper to classify reasons for hospitalization may have resulted in misclassification, though there are few other nonproprietary pediatric‐specific diagnostic groupers available.

In 2012, more than 70% of pediatric hospitalizations occurred at GHs in the United States. The considerably higher pediatric volumes at FCHs makes these institutions well suited for research, innovation, and the development and application of disease‐specific QI initiatives. Recognizing that the majority of pediatric hospitalizations occurred at GHs, there is a clear need for implementation research, program development, and quality metrics that align with the characteristics of hospitalizations at these centers. National support for research and quality improvement that reflects the diverse hospital settings where children receive their hospital care is critical to further our nation's goal of improving hospital quality for children.

Disclosures

Dr. Leyenaar was supported by grant number K08HS024133 from the Agency for Healthcare Research and Quality. The content is solely the responsibility of the authors and does not necessarily represent the official views of the Agency for Healthcare Research and Quality. The authors have no conflicts of interest relevant to this article to disclose.

Improvement in the quality of hospital care in the United States is a national priority, both to advance patient safety and because our expenditures exceed any other nation's, but our health outcomes lag behind.[1, 2] Healthcare spending for children is growing at a faster rate than any other age group, with hospital care accounting for more than 40% of pediatric healthcare expenditures.[3] Inpatient healthcare comprises a greater proportion of healthcare costs for children than for adults, yet we have limited knowledge about where this care is provided.[4]

There is substantial variability in the settings in which children are hospitalized. Children may be hospitalized in freestanding children's hospitals, where all services are designed for children and which operate independently of adult‐focused institutions. They may also be hospitalized in general hospitals where care may be provided in a general inpatient bed, on a dedicated pediatric ward, or in a children's hospital nested within a hospital, which may have specialized nursing and physician care but often shares other resources such as laboratory and radiology with the primarily adult‐focused institution. Medical students and residents may be trained in all of these settings. We know little about how these hospital types differ with respect to patient populations, disease volumes, and resource utilization, and this knowledge is important to inform clinical programs, implementation research, and quality improvement (QI) priorities. To this end, we aimed to describe the volume and characteristics of pediatric hospitalizations at acute care general hospitals and freestanding children's hospitals in the United States.

METHODS

Study Design and Eligibility

The data source for this analysis was the Healthcare Cost and Utilization Project's (HCUP) 2012 Kids' Inpatient Database (KID). We conducted a cross‐sectional study of hospitalizations in children and adolescents less than 18 years of age, excluding in‐hospital births and hospitalizations for pregnancy and delivery (identified using All Patient Refined‐Diagnostic Related Groups [APR‐DRGs]).[5] Neonatal hospitalizations not representing in‐hospital births but resulting from transfers or new admissions were retained. Because the dataset does not contain identifiable information, the institutional review board at Baystate Medical Center determined that our study did not constitute human subjects research.

The KID is released every 3 years and is the only publicly available, nationally representative database developed to study pediatric hospitalizations, including an 80% sample of noninborn pediatric discharges from all community, nonrehabilitation hospitals from 44 participating states.[6] Short‐term rehabilitation hospitals, long‐term nonacute care hospitals, psychiatric hospitals, and alcoholism/chemical dependency treatment facilities are excluded. The KID contains information on all patients, regardless of payer, and provides discharge weights to calculate national estimates.[6] It contains both hospital‐level and patient‐level variables, including demographic characteristics, charges, and other clinical and resource use data available from discharge abstracts. Beginning in 2012, freestanding children's hospitals (FCHs) are assigned to a separate stratum in the KID, with data from the Children's Hospital Association used by HCUP to verify the American Hospital Association's (AHA) list of FCHs.[6] Hospitals that are not FCHs were categorized as general hospitals (GHs). We were interested in examining patterns of care at acute care hospitals and not specialty hospitals; unlike previous years, the KID 2012 does not include a specialty hospital identifier.[6] Therefore, as a proxy for specialty hospital status, we excluded hospitals that had 2% hospitalizations for 12 common medical conditions (pneumonia, asthma, bronchiolitis, cellulitis, dehydration, urinary tract infection, neonatal hyperbilirubinemia, fever, upper respiratory infection, infectious gastroenteritis, unspecified viral infection, and croup). These medical conditions were the 12 most common reasons for medical hospitalizations identified using Keren's pediatric diagnosis code grouper,[7] excluding chronic diseases, and represented 26.2% of all pediatric hospitalizations. This 2% threshold was developed empirically, based on visual analysis of the distribution of cases across hospitals and was limited to hospitals with total pediatric volumes >25/year, allowing for stable case‐mix estimates.

Descriptor Variables

Hospital level characteristics included US Census region; teaching status classified in the KID based on results of the AHA Annual Survey; urban/rural location; hospital ownership, classified as public, private nonprofit and private investor‐owned; and total volume of pediatric hospitalizations, in deciles.[6] At the patient level, we examined age, gender, race/ethnicity, expected primary payer, and median household income (in quartiles) for patient's zip code. Medical complexity was categorized as (1) nonchronic disease, (2) complex chronic disease, or (3) noncomplex chronic disease, using the previously validated Pediatric Medical Complexity Algorithm (PMCA) based on International Classification of Diseases, 9th Revision, Clinical Modification (ICD‐9‐CM) codes.[8] Disease severity was classified based on APR‐DRG severity of illness coding, which classifies illnesses severity as minor, moderate, major, or extreme.[9]

We examined the following characteristics of the hospitalizations: (1) length of hospital stay (LOS) measured in calendar days; (2) high‐turnover hospitalization defined as LOS less than 2 days[10, 11, 12]; (3) long LOS, defined as greater than 4 days, equivalent to LOS greater than the 75th percentile; (4) neonatal versus non‐neonatal hospitalization, identified using APR‐DRGs; (5) admission type categorized as elective and nonelective; (6) admission source, categorized as transfer from another acute care hospital, admission from the emergency department, or direct admission; (7) discharge status, categorized as routine discharge, transfer to another hospital or healthcare facility, and discharge against medical advice; and (8) total hospital costs, calculated by applying the cost‐to‐charge ratios available in the KID to total hospital charges.

Reasons for hospitalization were categorized using the pediatric diagnosis code grouper by Keren, which uses ICD‐9‐CM codes to group common and costly principal diagnoses into distinct conditions (eg, pneumonia, idiopathic scoliosis), excluding children who have ICD‐9‐CM principal procedure codes unlikely related to their principal diagnosis (for example, appendectomy for a child with a principal diagnosis of pneumonia).[7] This pediatric grouper classifies diagnoses as medical, surgical, or medical‐surgical based on whether <20% (medical), >80% (surgical) or between 20% and 80% (medical‐surgical) of encounters for the condition had an ICD‐9‐CM principal procedure code for a surgery related to that condition. We further characterized medical hospitalizations as either medical or mental health hospitalizations.

Statistical Analysis

We categorized each discharge record as a hospitalization at a GH or an FCH. We then calculated patient‐level summary statistics, applying weights to calculate national estimates with an associated standard deviation (SD). We assessed differences in characteristics of hospitalizations at GHs and FCHs using Rao‐Scott 2 tests for categorical variables and Wald F tests for continuous variables.[6] We identified the most common reasons for hospitalization, including those responsible for at least 2% of all medical or surgical hospitalizations and at least 0.5% of medical hospitalizations for mental health diagnoses, given the lower prevalence of these conditions and our desire to include mental health diagnoses in our analysis. For these common conditions, we calculated the proportion of condition‐specific hospitalizations and aggregate hospital costs at GHs and FCHs. We also determined the number of hospitalizations at each hospital and calculated the median and interquartile range for the number of hospitalizations for each of these conditions according to hospital type, assessing for differences using Kruskal‐Wallis tests. Finally, we identified the most common and costly conditions at GHs and FCHs by ranking frequency and aggregate costs for each condition according to hospital type, limited to the 20 most costly and/or prevalent pediatric diagnoses. Because we used a novel method to identify specialty hospitals in this dataset, we repeated these analyses using all hospitals classified as a GH and FCH as a sensitivity analysis.

RESULTS

Overall, 3866 hospitals were categorized as a GH, whereas 70 hospitals were categorized as FCHs. Following exclusion of specialty hospitals, 3758 GHs and 50 FCHs were retained in this study. The geographic distribution of hospitals was similar, but although GHs included those in both urban and rural regions, all FCHs were located in urban regions (Table 1).

Characteristics of General Hospitals and Freestanding Children's Hospitals
General Hospitals, n = 3,758 Children's Hospitals, n = 50
Hospital characteristics n % n % P Value
  • NOTE: Abbreviations: IQR, interquartile range.

Geographic region
Northeast 458 12.2 4 8.0 0.50
Midwest 1,209 32.2 15 30.0
South 1,335 35.6 17 34.0
West 753 20.1 14 28.0
Location and teaching status
Rural 1,524 40.6 0 0 <0.0001
Urban nonteaching 1,506 40.1 7 14.0
Urban teaching 725 19.3 43 86.0
Hospital ownership
Government, nonfederal 741 19.7 0 0 <0.0001
Private, nonprofit 2,364 63.0 48 96.0
Private, investor‐owned 650 17.3 2 4.0
Volume of pediatric hospitalizations (deciles)
<185 hospitalizations/year (<8th decile) 2,664 71.0 0 0 <0.0001
186375 hospitalizations/year (8th decile) 378 10.1 2 4.0
376996 hospitalizations/year (9th decile) 380 10.1 1 2.0
>986 hospitalizations/year (10th decile) 333 8.9 47 94.0
Volume of pediatric hospitalizations, median [IQR] 56 [14240] 12,001 [5,83815,448] <0.0001

A total of 1,407,822 (SD 50,456) hospitalizations occurred at GHs, representing 71.7% of pediatric hospitalizations, whereas 554,458 (SD 45,046) hospitalizations occurred at FCHs. Hospitalizations at GHs accounted for 63.6% of days in hospital and 50.0% of pediatric inpatient healthcare costs. Eighty percent of the GHs had total pediatric patient volumes of less than 375 hospitalizations yearly; 11.1% of pediatric hospitalizations occurred at these lower‐volume centers. At FCHs, the median volume of pediatric hospitalizations was 12,001 (interquartile range [IQR]: 583815,448). A total of 36 GHs had pediatric hospitalization volumes in this IQR.

The median age for pediatric patients was slightly higher at GHs, whereas gender, race/ethnicity, primary payer, and median household income for zip code did not differ significantly between hospital types (Table 2). Medical complexity differed between hospital types: children with complex chronic diseases represented 20.2% of hospitalizations at GHs and 35.6% of hospitalizations at FCHs. Severity of illness differed between hospital types, with fewer hospitalizations categorized at the highest level of severity at GHs than FCHs. There were no significant differences between hospital types with respect to the proportion of hospitalizations categorized as neonatal hospitalizations or as elective hospitalizations. The median LOS was shorter at GHs than FCHs. Approximately 1 in 5 children hospitalized at GHs had LOS greater than 4 days, whereas almost 30% of children hospitalized at FCHs had LOS of this duration.

Patient Characteristics and Characteristics of Hospitalizations at General Hospitals and Freestanding Children's Hospitals
Patient Characteristics

General Hospitals,1,407,822 (50,456), 71.7%

Children's Hospitals,554,458 (45,046), 28.3%

P Value
n (SD Weighted Frequency) (%) n (SD Weighted Frequency) %
  • NOTE: Abbreviations: APR‐DRG, All Patient Refined Diagnosis‐Related Group; ED, emergency department; IQR, interquartile range; SD, standard deviation. *Race/ethnicity data missing for approximately 8% of discharge records.[8] Includes in‐hospital death, discharge destination unknown.

Age, y, median [IQR] 3.6 [011.7] 3.4 [010.8] 0.001
Gender (% female) 644,250 (23,089) 45.8 254,505 (20,688) 45.9 0.50
Race*
White 668,876 (27,741) 47.5 233,930 (26,349) 42.2 0.05
Black 231,586 (12,890) 16.5 80,568 (11,739) 14.5
Hispanic 279,021 (16,843) 19.8 12,1425 (21,183) 21.9
Other 133,062 (8,572) 9.5 41,190 (6,394) 7.4
Insurance status
Public 740,033 (28,675) 52.6 284,795 (25,324) 51.4 0.90
Private 563,562 (21,930) 40.0 224,042 (21,613) 40.4
Uninsured 37,265 (1,445) 2.7 16,355 (3,804) 3.0
No charge/other/unknown 66,962 (5,807) 4.8 29,266 (6,789) 5.3
Median household income for zip code, quartiles
<$38,999 457,139 (19,725) 33.3 164,831 (17,016) 30.1 0.07
$39,000$47,999 347,229 (14,104) 25.3 125,105 (10,712) 22.9
$48,000$62,999 304,795 (13,427) 22.2 134,915 (13,999) 24.7
>$63,000 263,171 (15,418) 19.2 122,164 (16,279) 22.3
Medical complexity
Nonchronic disease 717,009 (21,807) 50.9 211,089 (17,023) 38.1 <0.001
Noncomplex chronic disease 406,070 (14,951) 28.8 146,077 (12,442) 26.4
Complex chronic disease 284,742 (17,111) 20.2 197,292 (18,236) 35.6
APR‐DRG severity of illness
1 (lowest severity) 730,134 (23,162) 51.9 217,202 (18,433) 39.2 <0.001
2 486,748 (18,395) 34.6 202,931 (16,864) 36.6
3 146,921 (8,432) 10.4 100,566 (9,041) 18.1
4 (highest severity) 41,749 (3,002) 3.0 33,340 (3,199) 6.0
Hospitalization characteristics
Neonatal hospitalization 98,512 (3,336) 7.0 39,584 (4,274) 7.1 0.84
Admission type
Elective 255,774 (12,285) 18.3 109,854 (13,061) 19.8 0.05
Length of stay, d, (median [IQR]) 1.8 (0.01) [0.8‐3.6] 2.2 (0.06) [1.1‐4.7] <0.001
High turnover hospitalizations 416,790 (14,995) 29.6 130,441 (12,405) 23.5 <0.001
Length of stay >4 days 298,315 (14,421) 21.2 161,804 (14,354) 29.2 <0.001
Admission source
Transfer from another acute care hospital 154,058 (10,067) 10.9 82,118 (8,952) 14.8 0.05
Direct admission 550,123 (21,954) 39.1 211,117 (20,203) 38.1
Admission from ED 703,641 (26,155) 50.0 261,223 (28,708) 47.1
Discharge status
Routine 1,296,638 (46,012) 92.1 519,785 (42,613) 93.8 <0.01
Transfer to another hospital or healthcare facility 56,115 (1,922) 4.0 13,035 (1,437) 2.4
Discharge against medical advice 2,792 (181) 0.2 382 (70) 0.1
Other 52,276 (4,223) 3.7 21,256 (4,501) 3.8

The most common pediatric medical, mental health, and surgical conditions are shown in Figure 1, together representing 32% of pediatric hospitalizations during the study period. For these medical conditions, 77.9% of hospitalizations occurred at GHs, ranging from 52.6% of chemotherapy hospitalizations to 89.0% of hospitalizations for neonatal hyperbilirubinemia. Sixty‐two percent of total hospital costs for these conditions were incurred at GHs. For the common mental health hospitalizations, 86% of hospitalizations occurred at GHs. The majority of hospitalizations and aggregate hospital costs for common surgical conditions also occurred at GHs.

Figure 1
Share of national pediatric hospitalizations and aggregate costs in general and freestanding children's hospitals, by condition, for common medical, mental health and surgical diagnoses. (n = national estimates of number of hospitalizations and associated total hospital costs at general hospitals and children's hospitals).

Whereas pneumonia, asthma, and bronchiolitis were the most common reasons for hospitalization at both GHs and FCHs, the most costly conditions differed (see Supporting Table 1 in the online version of this article). At GHs, these respiratory diseases were responsible for the highest condition‐specific total hospital costs. At FCHs, the highest aggregate costs were due to respiratory distress syndrome and chemotherapy. Congenital heart diseases, including hypoplastic left heart syndrome, transposition of the great vessels, tetralogy of Fallot, endocardial cushion defects, coarctation of the aorta and ventricular septal defects accounted for 6 of the 20 most costly conditions at FCHs.

Figure 2 illustrates the volume of hospitalizations, per hospital, at GHs and FCHs for the most common medical hospitalizations. The median number of hospitalizations, per hospital, was consistently significantly lower at GHs than at FCHs (all P values <0.001). Similar results for surgical and mental health hospitalizations are shown as Supporting Figures 1 and 2 in the online version of this article. In our sensitivity analyses that included all hospitals classified as GH and FCH, all results were essentially unchanged.

Figure 2
Box and whisker plots illustrating median volume of hospitalizations per hospital and associated interquartile range for common medical condition at general hospitals and freestanding children's hospitals (n = number of hospitals represented).

Recognizing the wide range of pediatric volumes at GHs (Table 1) and our inability to differentiate children's hospitals nested within GHs from GHs with pediatric beds, we examined differences in patient and hospitalization characteristics at GHs with volumes 5838 hospitalizations (the 25th percentile for FCH volume) and GHs with pediatric volumes <5838/year (see Supporting Table 2 in the online version of this article). We also compared patient and hospitalization characteristics at FCHs and the higher‐volume GHs. A total of 36 GHs had pediatric volumes 5838, with hospitalizations at these sites together accounting for 15.4% of all pediatric hospitalizations. Characteristics of patients hospitalized at these higher‐volume GHs were similar to patients hospitalized at FCHs, but they had significantly lower disease severity, fewer neonatal hospitalizations, shorter LOS, and more high‐turnover hospitalizations than patients hospitalized at FCHs. We also observed several differences between children hospitalized at higher‐ and lower‐volume GHs (see Supporting Table 2 in the online version of this article). Children hospitalized at the lower‐volume GHs were more likely to have public health insurance and less likely to have complex chronic diseases, although overall, 39.0% of all hospitalizations for children with complex chronic diseases occurred at these lower‐volume GHs. Compared to children hospitalized at higher‐volume GHs, children hospitalized at the lower‐volume hospitals had significantly lower disease severity, shorter LOS, more direct admissions, and a greater proportion of routine discharges.

DISCUSSION

Of the 2 million pediatric hospitalizations in the United States in 2012, more than 70% occurred at GHs. We observed considerable heterogeneity in pediatric volumes across GHs, with 11% of pediatric hospitalizations occurring at hospitals with pediatric volumes of <375 hospitalizations annually, whereas 15% of pediatric hospitalizations occurred at GHs with volumes similar to those observed at FCHs. The remaining pediatric hospitalizations at GHs occurred at centers with intermediate volumes. The most common reasons for hospitalization were similar at GHs and FCHs, but the most costly conditions differed substantially. These findings have important implications for pediatric clinical care programs, research, and QI efforts.

Our finding that more than 70% of pediatric hospitalizations occurred at GHs speaks to the importance of quality measurement at these hospitals, whereas low per‐hospital pediatric volumes at the majority of GHs makes such measurement particularly challenging. Several previous studies have illustrated that volumes of pediatric hospitalizations are too small to detect meaningful differences in quality between hospitals using established condition‐specific metrics.[13, 14, 15] Our finding that more than 10% of pediatric hospitalizations occurred at GHs with pediatric volumes <375 year supports previous research suggesting that cross‐cutting, all‐condition quality metrics, composite measures, and/or multihospital reporting networks may be needed to enable quality measurement at these sites. In addition, the heterogeneity in patient volumes and characteristics across GHs raise questions about the applicability of quality metrics developed and validated at FCHs to the many GH settings. Field‐testing quality measures to ensure their validity at diverse GHs, particularly those with patient volumes and infrastructure different from FCHs, will be important to meaningful pediatric quality measurement.

Our results illustrating differences in the most common and costly conditions at GHs and FCHs have further implications for prioritization and implementation of research and QI efforts. Implementation research and QI efforts focused on cardiac and neurosurgical procedures, as well as neonatal intensive care, may have considerable impact on cost and quality at FCHs. At GHs, research and QI efforts focused on common conditions are needed to increase our knowledge of contextually relevant barriers to and facilitators of high‐quality pediatric care. This, however, can be made more difficult by small sample sizes, limited resources, and infrastructure, and competing priorities in adult‐focused GH settings.[16, 17, 18] Multihospital learning collaboratives and partnerships between FCHs and GHs can begin to address these challenges, but their success is contingent upon national advocacy and funding to support pediatric research and quality measures at GHs.

One of the most notable differences in the characteristics of pediatric hospitalizations at GHs and FCHs was the proportion of hospitalizations attributable to children with medical complexity (CMC); more than one‐third of hospitalizations at FCHs were for CMC compared to 1 in 5 at GHs. These findings align with the results of several previous studies describing the substantial resource utilization attributed to CMC, and with growing research, innovation, and quality metrics focused on improving both inpatient and outpatient care for these vulnerable children.[19, 20, 21, 22] Structured complex care programs, developed to improve care coordination and healthcare quality for CMC, are common at FCHs, and have been associated with decreased resource utilization and improved outcomes.[23, 24, 25] Notably, however, more than half of all hospitalizations for CMC, exceeding 250,000 annually, occurred at GHs, and almost 40% of hospitalizations for CMC occurred at the lower‐volume GHs. These findings speak to the importance of translating effective and innovative programs of care for CMC to GHs as resources allow, accompanied by robust evaluations of their effectiveness. Lower patient volume at most GHs, however, may be a barrier to dedicated CMC programs. As a result, decentralized community‐based programs of care for CMC, linking primary care programs with regional and tertiary care hospitals, warrant further consideration.[26, 27, 28]

This analysis should be interpreted in light of several limitations. First, we were unable to distinguish between GHs with scant pediatric‐specific resources from those with a large volume of dedicated pediatric resources, such as children's hospitals nested within GHs. We did identify 36 GHs with pediatric volumes similar to those observed at FCHs (see Supporting Table 2 in the online version of this article); patient and hospitalization characteristics at these higher‐volume GHs were similar in many ways to children hospitalized at FCHs. Several of these higher‐volume GHs may have considerable resources dedicated to the care of children, including subspecialty care, and may represent children's hospitals nested within GHs. Because nested children's hospitals are included in the GH categorization, our results may have underestimated the proportion of children cared for at children's hospitals. Further work is needed to identify the health systems challenges and opportunities that may be unique to these institutions. Second, because the 2012 KID does not include a specialty hospital indicator, we developed a proxy method for identifying these hospitals, which may have resulted in some misclassification. We are reassured that the results of our analyses did not change substantively when we included all hospitals. Similarly, although we are reassured that the number of hospitals classified in our analysis as acute care FCHs aligns, approximately, with the number of hospitals classified as such by the Children's Hospital Association, we were unable to assess the validity of this variable within the KID. Third, the KID does not link records at the patient level, so we are unable to report the number of unique children included in this analysis. In addition, the KID includes only inpatient stays with exclusion of observation status stays; potential differences between GH and FCH in the use of observation status could have biased our findings. Fifth, we used the PMCA to identify CMC; although this algorithm has been shown to have excellent sensitivity in identifying children with chronic diseases, using up to 3 years of Medicaid claims data, the sensitivity using the KID, where only 1 inpatient stay is available for assessment, is unknown.[8, 29] Similarly, use of Keren's pediatric diagnosis grouper to classify reasons for hospitalization may have resulted in misclassification, though there are few other nonproprietary pediatric‐specific diagnostic groupers available.

In 2012, more than 70% of pediatric hospitalizations occurred at GHs in the United States. The considerably higher pediatric volumes at FCHs makes these institutions well suited for research, innovation, and the development and application of disease‐specific QI initiatives. Recognizing that the majority of pediatric hospitalizations occurred at GHs, there is a clear need for implementation research, program development, and quality metrics that align with the characteristics of hospitalizations at these centers. National support for research and quality improvement that reflects the diverse hospital settings where children receive their hospital care is critical to further our nation's goal of improving hospital quality for children.

Disclosures

Dr. Leyenaar was supported by grant number K08HS024133 from the Agency for Healthcare Research and Quality. The content is solely the responsibility of the authors and does not necessarily represent the official views of the Agency for Healthcare Research and Quality. The authors have no conflicts of interest relevant to this article to disclose.

References
  1. Davis K, Stremikis K, Squires D, Schoen C. Mirror, Mirror on the wall: how the performance of the US health care system compares internationally. The Commonwealth Fund. Available at: http://www.commonwealthfund.org/publications/fund‐reports/2014/jun/mirror‐mirror. Published June 16, 2014. Accessed August 26, 2015.
  2. Fairbrother G, Guttmann A, Klein JD, Simpson LA, Thomas P, Kempe A. Higher cost, but poorer outcomes: the US health disadvantage and implications for pediatrics. Pediatrics. 2015;135(6):961964.
  3. Lassman D, Hartman M, Washington B, Andrews K, Catlin A. US health spending trends by age and gender: selected years 2002–10. Health Aff (Millwood). 2014;33(5):815822.
  4. Moore B, Levit K, Elixhauser A. Costs for hospital stays in the United States, 2012. Healthcare Cost and Utilization Project 181. Available at: http://www.hcup‐us.ahrq.gov/reports/statbriefs/sb181‐Hospital‐Costs‐United‐States‐2012.pdf. Published October 2014. Accessed September 2015.
  5. Averill RF, Goldfield N, Hughes JS, et al. All Patient Refined Diagnosis Related Groups: Methodology Overview. 3M Health Information Systems. Available at: https://www.hcup‐us.ahrq.gov/db/nation/nis/APR‐DRGsV20MethodologyOverviewandBibliography.pdf. Accessed February 8, 2016.
  6. Agency for Healthcare Research and Quality. Healthcare Cost and Utilization Project. Introduction to the HCUP Kids' Inpatient Database (KID) 2012. Available at: https://www.hcup‐us.ahrq.gov/db/nation/kid/kid_2012_introduction.jsp. Published Issued July 2014. Accessed February 8, 2016.
  7. Keren R. Prioritization of comparative effectiveness research topics in hospital pediatrics. Arch Pediatr Adolesc Med. 2012;166(12):1155.
  8. Simon TD, Cawthon ML, Stanford S, et al. Pediatric medical complexity algorithm: a new method to stratify children by medical complexity. Pediatrics. 2014;133(6):e1647e1654.
  9. Averill RF, Goldfield N, Hughes JS, et al. 3M APR DRG Classification System. 3M Health Information Systems. Available at: http://www.hcup‐us.ahrq.gov/db/nation/nis/v261_aprdrg_meth_ovrview.pdf. Accessed August 7, 2015.
  10. Macy ML, Stanley RM, Lozon MM, Sasson C, Gebremariam A, Davis MM. Trends in high‐turnover stays among children hospitalized in the United States, 1993–2003. Pediatrics. 2009;123(3):9961002.
  11. Macy ML, Stanley RM, Sasson C, Gebremariam A DM. High turnover stays for pediatric asthma in the United States. Med Care. 2010;48(9):827833.
  12. Leyenaar JK, Shieh M, Lagu T, Pekow PS, Lindenauer PK. Variation and outcomes associated with direct admission among children with pneumonia in the United States. JAMA Pediatr. 2014;168(9):829836.
  13. Berry JG, Zaslavsky AM, Toomey SL, et al. Recognizing differences in hospital quality performance for pediatric inpatient care. Pediatrics. 2015;136(2):251262.
  14. Bardach NS, Chien AT, Dudley RA. Small numbers limit the use of the inpatient pediatric quality indicators for hospital comparison. Acad Pediatr. 2010;10(4):266273.
  15. Feudtner C, Berry JG, Parry G, et al. Statistical uncertainty of mortality rates and rankings for children's hospitals. Pediatrics. 2011;128(4):e966e972.
  16. Leyenaar JK, Capra LA, O'Brien ER, Leslie LK, Mackie TI. Determinants of career satisfaction among pediatric hospitalists: a qualitative exploration. Acad Pediatr. 2014;14(4):361368.
  17. Simon TD, Starmer AJ, Conway PH, et al. Quality improvement research in pediatric hospital medicine and the role of the Pediatric Research in Inpatient Settings (PRIS) network. Acad Pediatr. 2013;13(6 suppl):S54S60.
  18. Miller M. Roles for children's hospitals in pediatric collaborative improvement networks. Pediatrics. 2013;131(suppl 4):S215S218.
  19. Cohen E, Kuo DZ, Agrawal R, et al. Children with medical complexity: an emerging population for clinical and research initiatives. Pediatrics. 2011;127(3):529538.
  20. Simon TD, Berry J, Feudtner C, et al. Children with complex chronic conditions in inpatient hospital settings in the United States. Pediatrics. 2010;126(4):647655.
  21. Cohen E, Berry JG, Camacho X, Anderson G, Wodchis W, Guttmann A. Patterns and costs of health care use of children with medical complexity. Pediatrics. 2012;130(6):e1463e1470.
  22. Berry JG, Hall DE, Kuo DZ, Hall M, Kueser J, Kaplan W. Hospital utilization and characteristics of patients experiencing recurrent readmissions within children's hospitals. JAMA. 2011;305(7):682690.
  23. Berry JG, Agrawal R, Kuo DZ, et al. Characteristics of hospitalizations for patients who use a structured clinical care program for children with medical complexity. J Pediatr. 2011;159(2):284290.
  24. Cohen E, Jovcevska V, Kuo D, Mahant S. Hospital‐based comprehensive care programs for children with special health care needs: a systematic review. Arch Pediatr Adolesc Med. 2011;165(6):554561.
  25. Gordon J, Colby H, Bartelt T, Jablonski D, Krauthoefer ML, Havens P. A tertiary care–primary care partnership model for medically complex and fragile children and youth with special health care needs. Arch Pediatr Adolesc Med. 2007;161(10):937944.
  26. Cohen E, Lacombe‐Duncan A, Spalding K, et al. Integrated complex care coordination for children with medical complexity: a mixed‐methods evaluation of tertiary care‐community collaboration. BMC Health Serv Res. 2012;12:366.
  27. Lerner CF, Kelly RB, Hamilton LJ, Klitzner TS. Medical transport of children with complex chronic conditions. Emerg Med Int. 2012;2012:837020.
  28. Stiles AD, Tayloe DT, Wegner SE. Comanagement of medically complex children by subspecialists, generalists, and care coordinators. Pediatrics. 2014;134(2):203205.
  29. Berry JG, Hall M, Cohen E, O'Neill M, Feudtner C. Ways to identify children with medical complexity and the importance of why. J Pediatr. 2015;167(2):229237.
References
  1. Davis K, Stremikis K, Squires D, Schoen C. Mirror, Mirror on the wall: how the performance of the US health care system compares internationally. The Commonwealth Fund. Available at: http://www.commonwealthfund.org/publications/fund‐reports/2014/jun/mirror‐mirror. Published June 16, 2014. Accessed August 26, 2015.
  2. Fairbrother G, Guttmann A, Klein JD, Simpson LA, Thomas P, Kempe A. Higher cost, but poorer outcomes: the US health disadvantage and implications for pediatrics. Pediatrics. 2015;135(6):961964.
  3. Lassman D, Hartman M, Washington B, Andrews K, Catlin A. US health spending trends by age and gender: selected years 2002–10. Health Aff (Millwood). 2014;33(5):815822.
  4. Moore B, Levit K, Elixhauser A. Costs for hospital stays in the United States, 2012. Healthcare Cost and Utilization Project 181. Available at: http://www.hcup‐us.ahrq.gov/reports/statbriefs/sb181‐Hospital‐Costs‐United‐States‐2012.pdf. Published October 2014. Accessed September 2015.
  5. Averill RF, Goldfield N, Hughes JS, et al. All Patient Refined Diagnosis Related Groups: Methodology Overview. 3M Health Information Systems. Available at: https://www.hcup‐us.ahrq.gov/db/nation/nis/APR‐DRGsV20MethodologyOverviewandBibliography.pdf. Accessed February 8, 2016.
  6. Agency for Healthcare Research and Quality. Healthcare Cost and Utilization Project. Introduction to the HCUP Kids' Inpatient Database (KID) 2012. Available at: https://www.hcup‐us.ahrq.gov/db/nation/kid/kid_2012_introduction.jsp. Published Issued July 2014. Accessed February 8, 2016.
  7. Keren R. Prioritization of comparative effectiveness research topics in hospital pediatrics. Arch Pediatr Adolesc Med. 2012;166(12):1155.
  8. Simon TD, Cawthon ML, Stanford S, et al. Pediatric medical complexity algorithm: a new method to stratify children by medical complexity. Pediatrics. 2014;133(6):e1647e1654.
  9. Averill RF, Goldfield N, Hughes JS, et al. 3M APR DRG Classification System. 3M Health Information Systems. Available at: http://www.hcup‐us.ahrq.gov/db/nation/nis/v261_aprdrg_meth_ovrview.pdf. Accessed August 7, 2015.
  10. Macy ML, Stanley RM, Lozon MM, Sasson C, Gebremariam A, Davis MM. Trends in high‐turnover stays among children hospitalized in the United States, 1993–2003. Pediatrics. 2009;123(3):9961002.
  11. Macy ML, Stanley RM, Sasson C, Gebremariam A DM. High turnover stays for pediatric asthma in the United States. Med Care. 2010;48(9):827833.
  12. Leyenaar JK, Shieh M, Lagu T, Pekow PS, Lindenauer PK. Variation and outcomes associated with direct admission among children with pneumonia in the United States. JAMA Pediatr. 2014;168(9):829836.
  13. Berry JG, Zaslavsky AM, Toomey SL, et al. Recognizing differences in hospital quality performance for pediatric inpatient care. Pediatrics. 2015;136(2):251262.
  14. Bardach NS, Chien AT, Dudley RA. Small numbers limit the use of the inpatient pediatric quality indicators for hospital comparison. Acad Pediatr. 2010;10(4):266273.
  15. Feudtner C, Berry JG, Parry G, et al. Statistical uncertainty of mortality rates and rankings for children's hospitals. Pediatrics. 2011;128(4):e966e972.
  16. Leyenaar JK, Capra LA, O'Brien ER, Leslie LK, Mackie TI. Determinants of career satisfaction among pediatric hospitalists: a qualitative exploration. Acad Pediatr. 2014;14(4):361368.
  17. Simon TD, Starmer AJ, Conway PH, et al. Quality improvement research in pediatric hospital medicine and the role of the Pediatric Research in Inpatient Settings (PRIS) network. Acad Pediatr. 2013;13(6 suppl):S54S60.
  18. Miller M. Roles for children's hospitals in pediatric collaborative improvement networks. Pediatrics. 2013;131(suppl 4):S215S218.
  19. Cohen E, Kuo DZ, Agrawal R, et al. Children with medical complexity: an emerging population for clinical and research initiatives. Pediatrics. 2011;127(3):529538.
  20. Simon TD, Berry J, Feudtner C, et al. Children with complex chronic conditions in inpatient hospital settings in the United States. Pediatrics. 2010;126(4):647655.
  21. Cohen E, Berry JG, Camacho X, Anderson G, Wodchis W, Guttmann A. Patterns and costs of health care use of children with medical complexity. Pediatrics. 2012;130(6):e1463e1470.
  22. Berry JG, Hall DE, Kuo DZ, Hall M, Kueser J, Kaplan W. Hospital utilization and characteristics of patients experiencing recurrent readmissions within children's hospitals. JAMA. 2011;305(7):682690.
  23. Berry JG, Agrawal R, Kuo DZ, et al. Characteristics of hospitalizations for patients who use a structured clinical care program for children with medical complexity. J Pediatr. 2011;159(2):284290.
  24. Cohen E, Jovcevska V, Kuo D, Mahant S. Hospital‐based comprehensive care programs for children with special health care needs: a systematic review. Arch Pediatr Adolesc Med. 2011;165(6):554561.
  25. Gordon J, Colby H, Bartelt T, Jablonski D, Krauthoefer ML, Havens P. A tertiary care–primary care partnership model for medically complex and fragile children and youth with special health care needs. Arch Pediatr Adolesc Med. 2007;161(10):937944.
  26. Cohen E, Lacombe‐Duncan A, Spalding K, et al. Integrated complex care coordination for children with medical complexity: a mixed‐methods evaluation of tertiary care‐community collaboration. BMC Health Serv Res. 2012;12:366.
  27. Lerner CF, Kelly RB, Hamilton LJ, Klitzner TS. Medical transport of children with complex chronic conditions. Emerg Med Int. 2012;2012:837020.
  28. Stiles AD, Tayloe DT, Wegner SE. Comanagement of medically complex children by subspecialists, generalists, and care coordinators. Pediatrics. 2014;134(2):203205.
  29. Berry JG, Hall M, Cohen E, O'Neill M, Feudtner C. Ways to identify children with medical complexity and the importance of why. J Pediatr. 2015;167(2):229237.
Issue
Journal of Hospital Medicine - 11(11)
Issue
Journal of Hospital Medicine - 11(11)
Page Number
743-749
Page Number
743-749
Publications
Publications
Article Type
Display Headline
Epidemiology of pediatric hospitalizations at general hospitals and freestanding children's hospitals in the United States
Display Headline
Epidemiology of pediatric hospitalizations at general hospitals and freestanding children's hospitals in the United States
Sections
Article Source
© 2016 Society of Hospital Medicine
Disallow All Ads
Correspondence Location
Address for correspondence and reprint requests: JoAnna Leyenaar, MD, Division of Pediatric Hospital Medicine, Department of Pediatrics, Tufts University School of Medicine, 800 Washington Street, Boston, MA 02111; Telephone: 617‐636‐8821; Fax: 617‐636‐8391; E‐mail: jleyenaar@post.harvard.edu
Content Gating
Gated (full article locked unless allowed per User)
Gating Strategy
First Peek Free
Article PDF Media
Media Files

PICC Use in Adults With Pneumonia

Article Type
Changed
Mon, 01/02/2017 - 19:34
Display Headline
Variation in prevalence and patterns of peripherally inserted central catheter use in adults hospitalized with pneumonia

Pneumonia is the most common cause of unplanned hospitalization in the United States.[1] Despite its clinical toll, the management of this disease has evolved markedly. Expanding vaccination programs, efforts to improve timeliness of antibiotic therapy, and improved processes of care are but a few developments that have improved outcomes for patients afflicted with this illness.[2, 3]

Use of peripherally inserted central catheters (PICCs) is an example of a modern development in the management of patients with pneumonia.[4, 5, 6, 7] PICCs provide many of the benefits associated with central venous catheters (CVCs) including reliable venous access for delivery of antibiotics, phlebotomy, and invasive hemodynamic monitoring. However, as they are placed in veins of the upper extremity, PICCs bypass insertion risks (eg, injury to the carotid vessels or pneumothorax) associated with placement of traditional CVCs.[8] Because they offer durable venous access, PICCs also facilitate care transitions while continuing intravenous antimicrobial therapy in patients with pneumonia.

However, accumulating evidence also suggests that PICCs are associated with important complications, including central lineassociated bloodstream infectionand venous thromboembolism.[9, 10] Furthermore, knowledge gaps in clinicians regarding indications for appropriate use and management of complications associated with PICCs have been recognized.[10, 11] These elements are problematic because reports of unjustified and inappropriate PICC use are growing in the literature.[12, 13] Such concerns have prompted a number of policy calls to improve PICC use, including Choosing Wisely recommendations by various professional societies.[14, 15]

As little is known about the prevalence or patterns of PICC use in adults hospitalized with pneumonia, we conducted a retrospective cohort study using data from a large network of US hospitals.

METHODS

Setting and Participants

We included patients from hospitals that participated in Premier's inpatient dataset, a large, fee‐supported, multipayer administrative database that has been used extensively in health services research to measure quality of care and comparative effectiveness of interventions.[16] Participating hospitals represent all regions of the United States and include teaching and nonteaching facilities in rural and urban locations. In addition to variables found in the uniform billing form, the Premier inpatient database also includes a date‐stamped list of charges for procedures conducted during hospitalization such as PICC placement. As PICC‐specific data are not available in most nationally representative datasets, Premier offers unique insights into utilization, timing, and factors associated with use of PICCs in hospitalized settings.

We included adult patients aged 18 years who were (1) admitted with a principal diagnosis of pneumonia present on admission, or secondary diagnosis of pneumonia if paired with a principal diagnosis of sepsis, respiratory failure, or influenza; (2) received at least 1 day of antibiotics between July 1, 2007 and November 30, 2011, and (3) underwent chest x‐ray or computed tomography (CT) at the time of admission. International Classification of Disease, 9th Revision, Clinical Modification (ICD‐9‐CM) codes were used for patient selection. Patients who were not admitted (eg, observation cases), had cystic fibrosis, or marked as pneumonia not present on admission were excluded. For patients who had more than 1 hospitalization during the study period, a single admission was randomly selected for inclusion.

Patient, Physician, and Hospital Data

For all patients, age, gender, marital status, insurance, race, and ethnicity were captured. Using software provided by the Healthcare Costs and Utilization Project, we categorized information on 29 comorbid conditions and computed a combined comorbidity score as described by Gagne et al.[17] Patients were considered to have healthcare‐associated pneumonia (HCAP) if they were: (1) admitted from a skilled nursing or a long‐term care facility, (2) hospitalized in the previous 90 days, (3) on dialysis, or (4) receiving immunosuppressing medications (eg, chemotherapy or steroids equivalent to at least 20 mg of prednisone per day) at the time of admission. Information on specialty of the admitting physician and hospital characteristics (eg, size, location, teaching status) were sourced through Premier data.

Receipt of PICCs and Related Therapies

Among eligible adult patients hospitalized with pneumonia, we identified patients who received a PICC at any time during hospitalization via PICC‐specific billing codes. Non‐PICC devices (eg, midlines, Hickman catheters) were not included. For all insertions, we assessed day of PICC placement relative to admission date. Data on type of PICC (eg, power‐injection capable, antibiotic coating) or PICC characteristics (size, number of lumens) were not available. We used billing codes to assess use of invasive or noninvasive ventilation, vasopressors, and administration of pneumonia‐specific antibiotics (eg, ‐lactams, macrolides, fluoroquinolones). Early exposure was defined when a billing code appeared within 2 days of hospital admission.

Outcomes of Interest

The primary outcome of interest was receipt of a PICC. Additionally, we assessed factors associated with PICC placement and variation in risk‐standardized rates of PICC use between hospitals.

Statistical Analyses

Patient and hospital characteristics were summarized using frequencies for categorical variables and medians with interquartile ranges for continuous variables. We examined association of individual patient and hospital characteristics with use of PICCs using generalized estimating equations models with a logit link for categorical variables and identity link for continuous variables, accounting for patient clustering within hospitals.

Characteristics of the Study Population
Characteristic Total, No. (%) No PICC, No. (%) PICC, No. (%) P Value*
  • NOTE: Abbreviations: CAP, community‐acquired pneumonia; GEE, generalized estimating equations; HCAP, healthcare‐associated pneumonia; ICU, intensive care unit; LMWH, low‐molecular‐weight heparin; MRSA, methicillin‐resistant Staphylococcus aureus; PICC, peripherally inserted central catheter; VTE, venous thromboembolism. *P value from GEE models that account for clustering within the hospital. Includes: discharged/transferred to cancer center/children's hospital, discharged/transferred to federal hospital; discharged/transferred to swing bed, discharged/transferred to long‐term care facility, discharged/transferred to psychiatric hospital, discharged/transferred to assisted living, discharged/transferred to other health institution not in list.

545,250 (100) 503,401 (92.3) 41,849 (7.7)
Demographics
Age, median (Q1Q3), y 71 (5782) 72 (5782) 69 (5780) <0.001
Gender <0.001
Male 256,448 (47.0) 237,232 (47.1) 19,216 (45.9)
Female 288,802 (53.0) 266,169 (52.9) 22,633 (54.1)
Race/ethnicity <0.001
White 377,255 (69.2) 346,689 (68.9) 30,566 (73.0)
Black 63,345 (11.6) 58,407 (11.6) 4,938 (11.8)
Hispanic 22,855 (4.2) 21,716 (4.3) 1,139 (2.7)
Other 81,795 (15.0) 76,589 (15.2) 5,206 (12.4)
Admitting specialty <0.001
Internal medicine 236,859 (43.4) 218,689 (43.4) 18,170 (43.4)
Hospital medicine 116,499 (21.4) 107,671 (21.4) 8,828 (21.1)
Family practice 80,388 (14.7) 75,482 (15.0) 4,906 (11.7)
Critical care and pulmonary 35,670 (6.5) 30,529 (6.1) 41,849 (12.3)
Geriatrics 4,812 (0.9) 4,098 (0.8) 714 (1.7)
Other 71,022 (13.0) 66,932 (13.3) 4,090 (9.8)
Insurance <0.001
Medicare 370,303 (67.9) 341,379 (67.8) 28,924 (69.1)
Medicaid 45,505 (8.3) 41,100 (8.2) 4,405 (10.5)
Managed care 69,984 (12.8) 65,280 (13.0) 4,704 (11.2)
Commercialindemnity 20,672 (3.8) 19,251 (3.8) 1,421 (3.4)
Other 38,786 (7.1) 36,391 (7.2) 2,395 (5.7)
Comorbidities
Gagne combined comorbidity score, median (Q1Q3) 2 (15) 2 (14) 4 (26) <0.001
Hypertension 332,347 (60.9) 306,964 (61.0) 25,383 (60.7) 0.13
Chronic pulmonary disease 255,403 (46.8) 234,619 (46.6) 20,784 (49.7) <0.001
Diabetes 171,247 (31.4) 155,540 (30.9) 15,707 (37.5) <0.001
Congestive heart failure 146,492 (26.9) 131,041 (26.0) 15,451 (36.9) <0.001
Atrial fibrillation 108,405 (19.9) 97,124 (19.3) 11,281 (27.0) <0.001
Renal failure 104,404 (19.1) 94,277 (18.7) 10,127 (24.2) <0.001
Nicotine replacement therapy/tobacco use 89,938 (16.5) 83,247 (16.5) 6,691 (16.0) <0.001
Obesity 60,242 (11.0) 53,268 (10.6) 6,974 (16.7) <0.001
Coagulopathy 41,717 (7.6) 35,371 (7.0) 6,346 (15.2) <0.001
Prior stroke (1 year) 26,787 (4.9) 24,046 (4.78) 2,741 (6.55) <0.001
Metastatic cancer 21,868 (4.0) 20,244 (4.0) 1,624 (3.9) 0.16
Solid tumor w/out metastasis 21,083 (3.9) 19,380 (3.8) 1,703 (4.1) 0.12
Prior VTE (1 year) 19,090 (3.5) 16,906 (3.4) 2,184 (5.2) <0.001
Chronic liver disease 16,273 (3.0) 14,207 (2.8) 2,066 (4.9) <0.001
Prior bacteremia (1 year) 4,106 (0.7) 3,584 (0.7) 522 (1.2) <0.001
Nephrotic syndrome 671 (0.1) 607 (0.1) 64 (0.2) 0.03
Morbidity markers
Type of pneumonia <0.001
CAP 376,370 (69.1) 352,900 (70.1) 23,830 (56.9)
HCAP 168,520 (30.9) 150,501 (29.9) 18,019 (43.1)
Sepsis present on admission 114,578 (21.0) 96,467 (19.2) 18,111 (43.3) <0.001
Non‐invasive ventilation 47,913(8.8) 40,599 (8.1) 7,314 (17.5) <0.001
Invasive mechanical ventilation 56,179 (10.3) 44,228 (8.8) 11,951 (28.6) <0.001
ICU status 97,703 (17.9) 80,380 (16.0) 17,323 (41.4) <0.001
Vasopressor use 48,353 (8.9) 38,030 (7.6) 10,323 (24.7) <0.001
Antibiotic/medication use
Anti‐MRSA agent (vancomycin) 146,068 (26.8) 123,327 (24.5) 22,741 (54.3) <0.001
Third‐generation cephalosporin 250,782 (46.0) 235,556 (46.8) 15,226 (36.4) <0.001
Anti‐Pseudomonal cephalosporin 41,798 (7.7) 36,982 (7.3) 4,816 (11.5) <0.001
Anti‐Pseudomonal ‐lactam 122,215 (22.4) 105,741 (21.0) 16,474 (39.4) <0.001
Fluroquinolone 288,051 (52.8) 267,131 (53.1) 20,920 (50.0) <0.001
Macrolide 223,737 (41.0) 210,954 (41.9) 12,783 (30.5) <0.001
Aminoglycoside 15,415 (2.8) 12,661 (2.5) 2,754 (6.6) <0.001
Oral steroids 44,486 (8.2) 41,586 (8.3) 2,900 (6.9) <0.001
Intravenous steroids 146,308 (26.8) 133,920 (26.6) 12,388 (29.6) <0.001
VTE prophylaxis with LMWH 190,735 (35.0) 174,612 (34.7) 16,123 (38.5) 0.01
Discharge disposition
Home 282,146 (51.7) 272,604(54.1) 9,542 (22.8) <0.001
Home with home health 71,977 (13.2) 65,289 (13.0) 6,688 (16.0) <0.001
Skilled nursing facility 111,541 (20.5) 97,113 (19.3) 14,428 (34.5) <0.001
Hospice 20,428 (3.7) 17,902 (3.6) 2,526 (6.0) <0.001
Expired 47,733 (8.7) 40,768 (8.1) 6,965 (16.6) <0.001
Other 11,425 (2.1) 9,725 (1.9) 1,700 (4.1) <0.001

We then developed a multivariable hierarchical generalized linear model (HGLM) for PICC placement with a random effect for hospital. In this model, we included patient demographics, comorbidities, sepsis on admission, type of pneumonia (eg, HCAP vs community‐associated pneumonia [CAP]), admitting physician specialty, and indicators for early receipt of specific treatments such as guideline‐recommended antibiotics, vasopressors, ventilation (invasive or noninvasive), and pneumatic compression devices for prophylaxis of deep vein thrombosis.

To understand and estimate between‐hospital variation in PICC use, we calculated risk‐standardized rates of PICC use (RSPICC) across hospitals using HGLM methods. These methods are also employed by the Centers for Medicare and Medicaid Services to calculate risk‐standardized measures for public reporting.[18] Because hospital rates of PICC use were highly skewed (21.2% [n = 105] of hospitals had no patients with PICCs), we restricted this model to the 343 hospitals that had at least 5 patients with a PICC to obtain stable estimates. For each hospital, we estimated a predicted rate of PICC use (pPICC) as the sum of predicted probabilities of PICC receipt from patient factors and the random intercept for hospital in which they were admitted. We then calculated an expected rate of PICC use (ePICC) per hospital as the sum of expected probabilities of PICC receipt from patient factors only. RSPICC for each hospital was then computed as the product of the overall unadjusted mean PICC rate (PICC) from all patients and the ratio of the predicted to expected PICC rate (uPICC*[pPICC/ePICC]).[19] Kruskal‐Wallis tests were used to evaluate the association between hospital characteristics with RSPICC rates. To evaluate the impact of the hospital in variation in PICC use, we assessed the change in likelihood ratio of a hierarchical model with hospital random effects compared to a logistic regression model with patient factors only. In addition, we estimated the intraclass correlation (ICC) to assess the proportion of variation in PICC use associated with the hospital, and the median odds ratio (MOR) from the hierarchical model. The MOR is the median of a set of odds ratios comparing 2 patients with the same set of characteristics treated at 2 randomly selected hospitals.[20, 21, 22] All analyses were performed using the Statistical Analysis System version 9.3 (SAS Institute, Inc., Cary, NC) and Stata 13 (StataCorp Inc., College Station, TX).

Ethical and Regulatory Oversight

Permission to conduct this study was obtained from the institutional review board at Baystate Medical Center, Springfield, Massachusetts. The study did not qualify as human subjects research and made use of fully deidentified data.

RESULTS

Between July 2007 and November 2011, 634,285 admissions representing 545,250 unique patients from 495 hospitals met eligibility criteria and were included in the study (Figure 1). Included patients had a median age of 71 years (interquartile range [IQR]: 5782), and 53.0% were female. Most patients were Caucasian (69.2%), unmarried (51.6%), and insured by Medicare (67.9%). Patients were admitted to the hospital by internal medicine providers (43.4%), hospitalists (21.4%), and family practice providers (14.7%); notably, critical care and pulmonary medicine providers admitted 6.5% of patients. The median Gagne comorbidity score was 2 (IQR: 15). Hypertension, chronic obstructive pulmonary disease, diabetes, and congestive heart failure were among the most common comorbidities observed (Table 1).

Figure 1
Study flow diagram. Abbreviations: CT, computed tomography; DRG, diagnosis‐related group; MS, missing; PICC, peripherally inserted central catheter; PN, pneumonia; POA, present on admission.

Among eligible patients, 41,849 (7.7%) received a PICC during hospitalization. Approximately a quarter of all patients who received PICCs did so by hospital day 2; 90% underwent insertion by hospital day 11 (mean = 5.4 days, median = 4 days). Patients who received PICCs were younger (median IQR: 69 years, 5780 years) but otherwise demographically similar to those that did not receive PICCs (median IQR: 72 years, 5782 years). Compared to other specialties, patients admitted by critical care/pulmonary providers were twice as likely to receive PICCs (12.3% vs 6.1%, P < .001). Patients who received PICCs had higher comorbidity scores than those who did not (median Gagne comorbidity score 4 vs 2, P < 0.001) and were more likely to be diagnosed with HCAP (43.1% vs 29.9%, P < 0.001) than CAP (56.9% vs 70.1%, P < 0.001).

PICC recipients were also more likely to receive intensive care unit (ICU) level of care (41.4% vs 16%, P < 0.001) and both noninvasive (17.5% vs 8.1%, P < 0.001) and invasive ventilation (28.6% vs 8.8%, P < 0.001) upon admission. Vasopressor use was also significantly more frequent in patients who received PICCs (24.7% vs 7.6%, P < 0.001) compared to those who did not receive these devices. Patients with PICCs were more often discharged to skilled nursing facilities (34.5% vs 19.3%) than those without PICCs.

Characteristics Associated With PICC Use Following Multivariable Modeling

Using HGLM with a random hospital effect, multiple patient characteristics were associated with PICC use (Table 2). Patients 65 years of age were less likely to receive a PICC compared to younger patients (odds ratio [OR]: 0.81, 95% confidence interval [CI]: 0.79‐0.84). Weight loss (OR: 2.03, 95% CI: 1.97‐2.10), sepsis on admission (OR: 1.80, 95% CI: 1.75‐1.85), and ICU status on hospital day 1 or 2 (OR: 1.70, 95% CI: 1.64‐1.75) represented 3 factors most strongly associated with PICC use.

Patient Factors Associated With PICC Use
Patient Characteristic Odds Ratio 95% Confidence Intervals
  • NOTE: Abbreviations: CAP, community‐associated pneumonia; DVT, deep vein thrombosis; FP, family practice; HCAP, healthcare‐associated pneumonia; IM, internal medicine; LMWH, low‐molecular‐weight heparin; MRSA, methicillin‐resistant Staphylococcus aureus; PICC, peripherally inserted central catheter; POA, present on admission; VTE, venous thromboembolism.

Age group (>66 vs 65 years) 0.82 0.790.84
Race/ethnicity
Other 1.02 0.971.06
Black 0.99 0.951.03
Hispanic 0.82 0.760.88
White Referent
Marital status
Other/missing 1.07 1.011.14
Single 1.02 1.001.05
Married Referent
Insurance payor
Other 0.85 0.800.89
Medicaid 1.13 1.081.18
Managed care 0.95 0.910.99
Commercialindemnity 0.93 0.871.00
Medicare Referent
Admitting physician specialty
Pulmonary/critical care medicine 1.18 1.131.24
Family practice 1.01 0.971.05
Geriatric medicine (FP and IM) 1.85 1.662.05
Hospitalist 0.94 0.910.98
Other specialties 1.02 0.971.06
Internal medicine Referent
Comorbidities
Congestive heart failure 1.27 1.241.31
Valvular disease 1.11 1.071.15
Pulmonary circulation disorders 1.37 1.321.42
Peripheral vascular disease 1.09 1.051.13
Hypertension 0.94 0.920.97
Paralysis 1.59 1.511.67
Other neurological disorders 1.20 1.161.23
Chronic lung disease 1.10 1.071.12
Diabetes 1.13 1.101.16
Hypothyroidism 1.03 1.001.06
Liver disease 1.16 1.101.23
Ulcer 1.86 1.153.02
Lymphoma 0.88 0.810.96
Metastatic cancer 0.75 0.710.80
Solid tumor without metastasis 0.93 0.880.98
Arthritis 1.22 1.161.28
Obesity 1.47 1.421.52
Weight loss 2.03 1.972.10
Blood loss 1.69 1.551.85
Deficiency anemias 1.40 1.371.44
Alcohol abuse 1.19 1.131.26
Drug abuse 1.31 1.231.39
Psychoses 1.16 1.111.21
Depression 1.10 1.061.13
Renal failure 0.96 0.930.98
Type of pneumonia
HCAP 1.03 1.011.06
CAP Referent
Sepsis (POA) 1.80 1.751.85
Antibiotic exposure
Anti‐MRSA agent 1.72 1.671.76
Anti‐Pseudomonal carbapenem 1.37 1.311.44
Non‐Pseudomonal carbapenem 1.48 1.331.66
Third‐generation cephalosporin 1.04 1.011.07
Anti‐Pseudomonal cephalosporin 1.25 1.201.30
Anti‐Pseudomonal ‐lactam 1.27 1.231.31
Aztreonam 1.31 1.231.40
Non‐Pseudomonal ‐lactam 1.36 1.231.50
‐lactam 1.55 1.261.90
Respiratory quinolone 0.90 0.870.92
Macrolide 0.85 0.820.88
Doxycycline 0.94 0.871.01
Aminoglycoside 1.21 1.141.27
Vasopressors 1.06 1.031.10
Noninvasive ventilation 1.29 1.251.34
Invasive ventilation 1.66 1.611.72
Intensive care unit on admission 1.70 1.641.75
Atrial fibrillation 1.26 1.221.29
Upper extremity chronic DVT 1.61 1.132.28
Nicotine replacement therapy/tobacco abuse 0.91 0.880.94
Aspirin 0.94 0.920.97
Warfarin 0.90 0.860.94
LMWH, prophylactic dose 1.10 1.081.13
LMWH, treatment dose 1.22 1.161.29
Intravenous steroids 1.05 1.021.08
Bacteremia (prior year) 1.14 1.021.27
VTE (prior year) 1.11 1.061.18
Pneumatic compression device 1.25 1.081.45
Invasive ventilation (prior year) 1.17 1.111.24
Irritable bowel disease 1.19 1.051.36

Therapy with potent parenteral antimicrobials including antimethicillin‐resistant Staphylococcus aureus agents (OR: 1.72, 95% CI: 1.67‐1.76), antipseudomonal ‐lactamases (OR: 1.27, 95% CI: 1.23‐1.31), and carbapenems (OR: 1.37, 95% CI: 1.31‐1.44) were significantly associated with PICC use. Conversely, use of macrolides (OR: 0.85, 95% CI: 0.82‐0.88) or respiratory fluoroquinolones (OR: 0.90, 95% CI: 0.87‐0.92) were associated with lower likelihood of PICC use. After adjusting for antimicrobial therapy, HCAP was only slightly more likely to result in PICC use than CAP (OR: 1.03, 95% CI: 1.01‐1.06). Compared to internal medicine providers, admission by geriatricians and critical care/pulmonary specialists was associated with greater likelihood of PICC use (OR: 1.85, 95% CI: 1.66‐2.05 and OR: 1.18, 95% CI: =1.13‐1.24, respectively). Admission by hospitalists was associated with a modestly lower likelihood of PICC placement (OR: 0.94, 95% CI: 0.91‐0.98).

Hospital Level Variation in PICC Use

To ensure stable estimates of hospital PICC use, we excluded 152 facilities (31%): 10% had no patients with PICCs and 21% had <5 patients who received a PICC. Therefore, RSPICC was estimated for 343 of 495 facilities (69%) (Figure 2). In these facilities, RSPICC varied from 0.3% to 41.7%. Hospital RSPICC was significantly associated with hospital location (median 11.9% vs 7.8% for urban vs rural hospitals respectively, P = 0.05). RSPICCs were also greater among hospitals in Southern (11.3%), Western (12.7%), and Midwest (12.0%) regions of the nation compared to those in the Northeast (8.4%) (P = 0.02) (Table 3).

Association Between Hospital Characteristics and Risk‐Standardized Rate of PICC Use*
Hospital Characteristic (No.) Median (IQR), % P Value
  • NOTE: Abbreviations: IQR, interquartile range; PICC, peripherally inserted central catheter.*Numbers indicate the percentage of patients with a PICC in each category, accounting for risk associated with PICC receipt. To ensure stable estimates, 152 facilities (31%) were excluded, as 10% had no patients with PICCs and 21% had <5 patients who received a PICC. Kruskal‐Wallis test.

Bed size 0.12
200 beds (106) 9.1 (4.816.3)
201 beds (237) 11.6 (5.817.6)
Rural/urban 0.05
Urban (275) 11.9 (5.517.4)
Rural (68) 7.8 (5.014.0)
Region 0.02
Northeast (50) 8.4 (3.913.0)
Midwest (69) 12.0 (5.817.4)
West (57) 12.7 (7.617.0)
South (167) 11.3 (4.817.8)
Teaching status 0.77
Nonteaching (246) 10.9 (5.017.4)
Teaching (97) 12.0 (5.816.9)
Figure 2
Observed vs risk‐standardized rate of peripherally inserted central catheter (PICC) use across 343 US hospitals (restricted to sites where >5 patients received PICCs). Horizontal axis represents rate of PICC use, whereas vertical axis represents number of hospitals. The dark shaded bars represents the observed rate of PICC use, whereas the nonshaded bars reflect risk‐standardized rate of PICC use.

A likelihood ratio test comparing the hierarchical model to a logistic model with patient factors only was highly significant (P < 0.001), indicating that the hospital where the patient was treated had a major impact on receipt of PICC after accounting for patient factors. The MOR was 2.71, which is a larger effect than we found for any of the individual patient characteristics. The proportion of variance explained by hospitals was 25% (95% CI: 22%‐28%), as measured by the ICC.

DISCUSSION

In this study of 545,250 adults hospitalized with pneumonia, we found that approximately 8% of patients received a PICC. Patients who received PICCs had more comorbidities, were more frequently diagnosed with HCAP, and were more often admitted to the ICU, where they experienced greater rates of mechanical ventilation, noninvasive ventilation, and vasopressor use compared to those who did not receive a PICC. Additionally, risk‐adjusted rates of PICC use varied as much as 10‐fold across institutions. In fact, almost 70% of the total variation in rates of PICC use remained unexplained by hospital or patient characteristics. Although use of PICCs is often clinically nuanced in ways that are difficult to capture in large datasets (eg, difficult venous access or inability to tolerate oral medications), the substantial variation of PICC use observed suggests that physician and institutional practice styles are the major determinants of PICC placement during a hospitalization for pneumonia. Because PICCs are associated with serious complications, and evidence regarding discretionary use is accumulating, a research agenda examining reasons for such use and related outcomes appears necessary.

The placement of PICCs has grown substantially in hospitalized patients all over the world.[23, 24] Although originally developed for total parenteral nutrition in surgical patients,[25] contemporary reports of PICC use in critical illness,[26] diseases such as cystic fibrosis,[27] and even pregnancy[28] are now common. Although PICCs are clinically invaluable in many of these conditions, growing use of these devices has led to the realization that benefits may be offset by complications.[9, 10, 29, 30] Additionally, recent data suggest that not all PICCs may be used for appropriate reasons. For instance, in a decade‐long study at a tertiary care center, changes in patterns of PICC use including shortened dwell times, multiple insertions in a single patient, and unclear indications for use were reported.[11] In another study at an academic medical center, a substantial proportion of PICCs were found to be idle or unjustified.[12] It comes as little surprise, then, that a recent multicenter study found that 1 out of every 5 clinicians did not even know that their patient had a PICC.[29] Although calls to improve PICC use in the hospital setting have emerged, strategies to do so are limited by data that emanate from single‐center reports or retrospective designs. No other studies reporting use of PICCs across US hospitals for any clinical condition currently exist.[31]

We found that patients with weight loss, those with greater combined comorbidity scores, and those who were critically ill or diagnosed with sepsis were more likely to receive PICCs than others. These observations suggest that PICC use may reflect underlying severity of illness, as advanced care such as ventilator support was often associated with PICC use. Additionally, discharge to a skilled nursing facility was frequently associated with PICC placement, a finding consistent with a recent study evaluating the use of PICCs in these settings.[32] However, a substantial proportion of PICC use remained unexplained by available patient or hospital factors. Although our study was not specifically designed to examine this question, a possible reason may relate to unmeasured institutional factors that influence the propensity to use a PICC, recently termed as PICC culture.[33] For example, it is plausible that hospitals with nursing‐led PICC teams or interventional radiology (such as teaching hospitals) are more likely to use PICCs than those without such operators. This hypothesis may explain why urban, larger, and teaching hospitals exhibited higher rates of PICC use. Conversely, providers may have an affinity toward PICC use that is predicated not just by operator availability, but also local hospital norms. Understanding why some facilities use PICCs at higher rates than others and implications of such variation with respect to patient safety, cost, and outcomes is important. Study designs that use mixed‐methods approaches or seek to qualitatively understand reasons behind PICC use are likely to be valuable in this enquiry.

Our study has limitations. First, we used an administrative dataset and ICD‐9‐CM codes rather than clinical data from medical records to identify cases of pneumonia or comorbidities. Our estimates of PICC use across hospitals thus may not fully account for differences in severity of illness, and it is possible that patients needed a PICC for reasons that we could not observe. However, the substantial variation observed in rates of PICC use across hospitals is unlikely to be explained by differences in patient severity of illness, documentation, or coding practices. Second, as PICC removal codes were not available, we are unable to comment on how often hospitalized pneumonia patients were discharged with PICCs or received antimicrobial therapy beyond their inpatient stay. Third, although we observed that a number of patient and hospital factors were associated with PICC receipt, our study was not designed to determine the reasons underlying these patterns.

These limitations aside, our study has important strengths. To our knowledge, this is the first study to report utilization and outcomes associated with PICC use among those hospitalized with pneumonia across the United States. The inclusion of a large number of patients receiving care in diverse facilities lends a high degree of external validity to our findings. Second, we used advanced modeling to identify factors associated with PICC use in hospitalized patients with pneumonia, producing innovative and novel findings. Third, our study is the first to show the existence of substantial variation in rates of PICC use across US hospitals within the single disease state of pneumonia. Understanding the drivers of this variability is important as it may inform future studies, policies, and practices to improve PICC use in hospitalized patients.

In conclusion, we found that PICC use in patients hospitalized with pneumonia is common and highly variable. Future studies examining the contextual factors behind PICC use and their association with outcomes are needed to facilitate efforts to standardize PICC use across hospitals.

Disclosures

Dr. Chopra is supported by a career development award (1‐K08‐HS022835‐01) from the Agency of Healthcare Research and Quality. The authors report no conflicts of interest.

Files
References
  1. Elixhauser A, Owens P. Reasons for being admitted to the hospital through the emergency department, 2003. Healthcare Cost and Utilization Project Statistical Brief 2. Rockville, MD: Agency for Healthcare Research and Quality. Available at: www.hcup‐us.ahrq.gov/reports/statbriefs/sb2.pdf. Published February 2006. Accessed June 27, 2014.
  2. Suter LG, Li SX, Grady JN, et al. National patterns of risk‐standardized mortality and readmission after hospitalization for acute myocardial infarction, heart failure, and pneumonia: update on publicly reported outcomes measures based on the 2013 release. J Gen Intern Med. 2014;29(10):13331340.
  3. Lee JS, Nsa W, Hausmann LR, et al. Quality of care for elderly patients hospitalized for pneumonia in the United States, 2006 to 2010. JAMA Intern Med. 2014;174(11):18061814.
  4. Masoorli S, Angeles T. PICC lines: the latest home care challenge. RN. 1990;53(1):4451.
  5. Lam S, Scannell R, Roessler D, Smith MA. Peripherally inserted central catheters in an acute‐care hospital. Arch Intern Med. 1994;154(16):18331837.
  6. Goodwin ML, Carlson I. The peripherally inserted central catheter: a retrospective look at three years of insertions. J Intraven Nurs. 1993;16(2):92103.
  7. Ng PK, Ault MJ, Ellrodt AG, Maldonado L. Peripherally inserted central catheters in general medicine. Mayo Clin Proc. 1997;72(3):225233.
  8. Funk D, Gray J, Plourde PJ. Two‐year trends of peripherally inserted central catheter‐line complications at a tertiary‐care hospital: role of nursing expertise. Infect Control Hosp Epidemiol. 2001;22(6):377379.
  9. Chopra V, Ratz D, Kuhn L, Lopus T, Chenoweth C, Krein S. PICC‐associated bloodstream infections: prevalence, patterns, and predictors. Am J Med. 2014;127(4):319328.
  10. Chopra V, O'Horo JC, Rogers MA, Maki DG, Safdar N. The risk of bloodstream infection associated with peripherally inserted central catheters compared with central venous catheters in adults: a systematic review and meta‐analysis. Infect Control Hosp Epidemiol. 2013;34(9):908918.
  11. Gibson C, Connolly BL, Moineddin R, Mahant S, Filipescu D, Amaral JG. Peripherally inserted central catheters: use at a tertiary care pediatric center. J Vasc Interv Radiol. 2013;24(9):13231331.
  12. Tejedor SC, Tong D, Stein J, et al. Temporary central venous catheter utilization patterns in a large tertiary care center: tracking the “idle central venous catheter”. Infect Control Hosp Epidemiol. 2012;33(1):5057.
  13. Tiwari MM, Hermsen ED, Charlton ME, Anderson JR, Rupp ME. Inappropriate intravascular device use: a prospective study. Journal Hosp Infect. 2011;78(2):128132.
  14. McMahon LF, Beyth RJ, Burger A, et al. Enhancing patient‐centered care: SGIM and choosing wisely. J Gen Intern Med. 2014;29(3):432433.
  15. Williams AW, Dwyer AC, Eddy AA, et al. Critical and honest conversations: the evidence behind the “Choosing Wisely” campaign recommendations by the American Society of Nephrology. Clin J Am Soc Nephrol. 2012;7(10):16641672.
  16. Rothberg MB, Pekow PS, Priya A, et al. Using highly detailed administrative data to predict pneumonia mortality. PLoS One. 2014;9(1):e87382.
  17. Gagne JJ, Glynn RJ, Avorn J, Levin R, Schneeweiss S. A combined comorbidity score predicted mortality in elderly patients better than existing scores. J Clin Epidemiol. 2011;64(7):749759.
  18. Sjoding MW, Prescott HC, Wunsch H, Iwashyna TJ, Cooke CR. Hospitals with the highest intensive care utilization provide lower quality pneumonia care to the elderly. Crit Care Med. 2015;43(6):11781186.
  19. Normand SL, Shahian DM. Statistical and clinical aspects of hospital outcomes profiling. Stat Sci. 2007;22(2):206226.
  20. Larsen K, Merlo J. Appropriate assessment of neighborhood effects on individual health: integrating random and fixed effects in multilevel logistic regression. Am J Epidemiol. 2005;161(1):8188.
  21. Larsen K, Petersen JH, Budtz‐Jorgensen E, Endahl L. Interpreting parameters in the logistic regression model with random effects. Biometrics. 2000;56(3):909914.
  22. Sanagou M, Wolfe R, Forbes A, Reid CM. Hospital‐level associations with 30‐day patient mortality after cardiac surgery: a tutorial on the application and interpretation of marginal and multilevel logistic regression. BMC Med Res Methodol. 2012;12:28.
  23. Lisova K, Paulinova V, Zemanova K, Hromadkova J. Experiences of the first PICC team in the Czech Republic. Br J Nurs. 2015;24(suppl 2):S4S10.
  24. Konstantinou EA, Stafylarakis E, Kapritsou M, et al. Greece reports prototype intervention with first peripherally inserted central catheter: case report and literature review. J Vasc Nurs. 2012;30(3):8893.
  25. Hoshal VL Total intravenous nutrition with peripherally inserted silicone elastomer central venous catheters. Arch Surg. 1975;110(5):644646.
  26. Cotogni P, Pittiruti M. Focus on peripherally inserted central catheters in critically ill patients. World J Crit Care Med. 2014;3(4):8094.
  27. Mermis JD, Strom JC, Greenwood JP, et al. Quality improvement initiative to reduce deep vein thrombosis associated with peripherally inserted central catheters in adults with cystic fibrosis. Ann Am Thorac Soc. 2014;11(9):14041410.
  28. Cape AV, Mogensen KM, Robinson MK, Carusi DA. Peripherally Inserted central catheter (PICC) complications during pregnancy. JPEN J Parenter Enteral Nutr. 2013;38(5):595601.
  29. Chopra V, Govindan S, Kuhn L, et al. Do clinicians know which of their patients have central venous catheters?: a multicenter observational study. Ann Intern Med. 2014;161(8):562567.
  30. Chopra V, Anand S, Hickner A, et al. Risk of venous thromboembolism associated with peripherally inserted central catheters: a systematic review and meta‐analysis. Lancet. 2013;382(9889):311325.
  31. Chopra V, Flanders SA, Saint S. The problem with peripherally inserted central catheters. JAMA. 2012;308(15):15271528.
  32. Chopra V, Montoya A, Joshi D, et al. Peripherally inserted central catheter use in skilled nursing facilities: a pilot study. J Am Geriatr Soc. 2015;63(9):18941899.
  33. McGill RL, Tsukahara T, Bhardwaj R, Kapetanos AT, Marcus RJ. Inpatient venous access practices: PICC culture and the kidney patient. J Vasc Access. 2015;16(3):206210.
Article PDF
Issue
Journal of Hospital Medicine - 11(8)
Publications
Page Number
568-575
Sections
Files
Files
Article PDF
Article PDF

Pneumonia is the most common cause of unplanned hospitalization in the United States.[1] Despite its clinical toll, the management of this disease has evolved markedly. Expanding vaccination programs, efforts to improve timeliness of antibiotic therapy, and improved processes of care are but a few developments that have improved outcomes for patients afflicted with this illness.[2, 3]

Use of peripherally inserted central catheters (PICCs) is an example of a modern development in the management of patients with pneumonia.[4, 5, 6, 7] PICCs provide many of the benefits associated with central venous catheters (CVCs) including reliable venous access for delivery of antibiotics, phlebotomy, and invasive hemodynamic monitoring. However, as they are placed in veins of the upper extremity, PICCs bypass insertion risks (eg, injury to the carotid vessels or pneumothorax) associated with placement of traditional CVCs.[8] Because they offer durable venous access, PICCs also facilitate care transitions while continuing intravenous antimicrobial therapy in patients with pneumonia.

However, accumulating evidence also suggests that PICCs are associated with important complications, including central lineassociated bloodstream infectionand venous thromboembolism.[9, 10] Furthermore, knowledge gaps in clinicians regarding indications for appropriate use and management of complications associated with PICCs have been recognized.[10, 11] These elements are problematic because reports of unjustified and inappropriate PICC use are growing in the literature.[12, 13] Such concerns have prompted a number of policy calls to improve PICC use, including Choosing Wisely recommendations by various professional societies.[14, 15]

As little is known about the prevalence or patterns of PICC use in adults hospitalized with pneumonia, we conducted a retrospective cohort study using data from a large network of US hospitals.

METHODS

Setting and Participants

We included patients from hospitals that participated in Premier's inpatient dataset, a large, fee‐supported, multipayer administrative database that has been used extensively in health services research to measure quality of care and comparative effectiveness of interventions.[16] Participating hospitals represent all regions of the United States and include teaching and nonteaching facilities in rural and urban locations. In addition to variables found in the uniform billing form, the Premier inpatient database also includes a date‐stamped list of charges for procedures conducted during hospitalization such as PICC placement. As PICC‐specific data are not available in most nationally representative datasets, Premier offers unique insights into utilization, timing, and factors associated with use of PICCs in hospitalized settings.

We included adult patients aged 18 years who were (1) admitted with a principal diagnosis of pneumonia present on admission, or secondary diagnosis of pneumonia if paired with a principal diagnosis of sepsis, respiratory failure, or influenza; (2) received at least 1 day of antibiotics between July 1, 2007 and November 30, 2011, and (3) underwent chest x‐ray or computed tomography (CT) at the time of admission. International Classification of Disease, 9th Revision, Clinical Modification (ICD‐9‐CM) codes were used for patient selection. Patients who were not admitted (eg, observation cases), had cystic fibrosis, or marked as pneumonia not present on admission were excluded. For patients who had more than 1 hospitalization during the study period, a single admission was randomly selected for inclusion.

Patient, Physician, and Hospital Data

For all patients, age, gender, marital status, insurance, race, and ethnicity were captured. Using software provided by the Healthcare Costs and Utilization Project, we categorized information on 29 comorbid conditions and computed a combined comorbidity score as described by Gagne et al.[17] Patients were considered to have healthcare‐associated pneumonia (HCAP) if they were: (1) admitted from a skilled nursing or a long‐term care facility, (2) hospitalized in the previous 90 days, (3) on dialysis, or (4) receiving immunosuppressing medications (eg, chemotherapy or steroids equivalent to at least 20 mg of prednisone per day) at the time of admission. Information on specialty of the admitting physician and hospital characteristics (eg, size, location, teaching status) were sourced through Premier data.

Receipt of PICCs and Related Therapies

Among eligible adult patients hospitalized with pneumonia, we identified patients who received a PICC at any time during hospitalization via PICC‐specific billing codes. Non‐PICC devices (eg, midlines, Hickman catheters) were not included. For all insertions, we assessed day of PICC placement relative to admission date. Data on type of PICC (eg, power‐injection capable, antibiotic coating) or PICC characteristics (size, number of lumens) were not available. We used billing codes to assess use of invasive or noninvasive ventilation, vasopressors, and administration of pneumonia‐specific antibiotics (eg, ‐lactams, macrolides, fluoroquinolones). Early exposure was defined when a billing code appeared within 2 days of hospital admission.

Outcomes of Interest

The primary outcome of interest was receipt of a PICC. Additionally, we assessed factors associated with PICC placement and variation in risk‐standardized rates of PICC use between hospitals.

Statistical Analyses

Patient and hospital characteristics were summarized using frequencies for categorical variables and medians with interquartile ranges for continuous variables. We examined association of individual patient and hospital characteristics with use of PICCs using generalized estimating equations models with a logit link for categorical variables and identity link for continuous variables, accounting for patient clustering within hospitals.

Characteristics of the Study Population
Characteristic Total, No. (%) No PICC, No. (%) PICC, No. (%) P Value*
  • NOTE: Abbreviations: CAP, community‐acquired pneumonia; GEE, generalized estimating equations; HCAP, healthcare‐associated pneumonia; ICU, intensive care unit; LMWH, low‐molecular‐weight heparin; MRSA, methicillin‐resistant Staphylococcus aureus; PICC, peripherally inserted central catheter; VTE, venous thromboembolism. *P value from GEE models that account for clustering within the hospital. Includes: discharged/transferred to cancer center/children's hospital, discharged/transferred to federal hospital; discharged/transferred to swing bed, discharged/transferred to long‐term care facility, discharged/transferred to psychiatric hospital, discharged/transferred to assisted living, discharged/transferred to other health institution not in list.

545,250 (100) 503,401 (92.3) 41,849 (7.7)
Demographics
Age, median (Q1Q3), y 71 (5782) 72 (5782) 69 (5780) <0.001
Gender <0.001
Male 256,448 (47.0) 237,232 (47.1) 19,216 (45.9)
Female 288,802 (53.0) 266,169 (52.9) 22,633 (54.1)
Race/ethnicity <0.001
White 377,255 (69.2) 346,689 (68.9) 30,566 (73.0)
Black 63,345 (11.6) 58,407 (11.6) 4,938 (11.8)
Hispanic 22,855 (4.2) 21,716 (4.3) 1,139 (2.7)
Other 81,795 (15.0) 76,589 (15.2) 5,206 (12.4)
Admitting specialty <0.001
Internal medicine 236,859 (43.4) 218,689 (43.4) 18,170 (43.4)
Hospital medicine 116,499 (21.4) 107,671 (21.4) 8,828 (21.1)
Family practice 80,388 (14.7) 75,482 (15.0) 4,906 (11.7)
Critical care and pulmonary 35,670 (6.5) 30,529 (6.1) 41,849 (12.3)
Geriatrics 4,812 (0.9) 4,098 (0.8) 714 (1.7)
Other 71,022 (13.0) 66,932 (13.3) 4,090 (9.8)
Insurance <0.001
Medicare 370,303 (67.9) 341,379 (67.8) 28,924 (69.1)
Medicaid 45,505 (8.3) 41,100 (8.2) 4,405 (10.5)
Managed care 69,984 (12.8) 65,280 (13.0) 4,704 (11.2)
Commercialindemnity 20,672 (3.8) 19,251 (3.8) 1,421 (3.4)
Other 38,786 (7.1) 36,391 (7.2) 2,395 (5.7)
Comorbidities
Gagne combined comorbidity score, median (Q1Q3) 2 (15) 2 (14) 4 (26) <0.001
Hypertension 332,347 (60.9) 306,964 (61.0) 25,383 (60.7) 0.13
Chronic pulmonary disease 255,403 (46.8) 234,619 (46.6) 20,784 (49.7) <0.001
Diabetes 171,247 (31.4) 155,540 (30.9) 15,707 (37.5) <0.001
Congestive heart failure 146,492 (26.9) 131,041 (26.0) 15,451 (36.9) <0.001
Atrial fibrillation 108,405 (19.9) 97,124 (19.3) 11,281 (27.0) <0.001
Renal failure 104,404 (19.1) 94,277 (18.7) 10,127 (24.2) <0.001
Nicotine replacement therapy/tobacco use 89,938 (16.5) 83,247 (16.5) 6,691 (16.0) <0.001
Obesity 60,242 (11.0) 53,268 (10.6) 6,974 (16.7) <0.001
Coagulopathy 41,717 (7.6) 35,371 (7.0) 6,346 (15.2) <0.001
Prior stroke (1 year) 26,787 (4.9) 24,046 (4.78) 2,741 (6.55) <0.001
Metastatic cancer 21,868 (4.0) 20,244 (4.0) 1,624 (3.9) 0.16
Solid tumor w/out metastasis 21,083 (3.9) 19,380 (3.8) 1,703 (4.1) 0.12
Prior VTE (1 year) 19,090 (3.5) 16,906 (3.4) 2,184 (5.2) <0.001
Chronic liver disease 16,273 (3.0) 14,207 (2.8) 2,066 (4.9) <0.001
Prior bacteremia (1 year) 4,106 (0.7) 3,584 (0.7) 522 (1.2) <0.001
Nephrotic syndrome 671 (0.1) 607 (0.1) 64 (0.2) 0.03
Morbidity markers
Type of pneumonia <0.001
CAP 376,370 (69.1) 352,900 (70.1) 23,830 (56.9)
HCAP 168,520 (30.9) 150,501 (29.9) 18,019 (43.1)
Sepsis present on admission 114,578 (21.0) 96,467 (19.2) 18,111 (43.3) <0.001
Non‐invasive ventilation 47,913(8.8) 40,599 (8.1) 7,314 (17.5) <0.001
Invasive mechanical ventilation 56,179 (10.3) 44,228 (8.8) 11,951 (28.6) <0.001
ICU status 97,703 (17.9) 80,380 (16.0) 17,323 (41.4) <0.001
Vasopressor use 48,353 (8.9) 38,030 (7.6) 10,323 (24.7) <0.001
Antibiotic/medication use
Anti‐MRSA agent (vancomycin) 146,068 (26.8) 123,327 (24.5) 22,741 (54.3) <0.001
Third‐generation cephalosporin 250,782 (46.0) 235,556 (46.8) 15,226 (36.4) <0.001
Anti‐Pseudomonal cephalosporin 41,798 (7.7) 36,982 (7.3) 4,816 (11.5) <0.001
Anti‐Pseudomonal ‐lactam 122,215 (22.4) 105,741 (21.0) 16,474 (39.4) <0.001
Fluroquinolone 288,051 (52.8) 267,131 (53.1) 20,920 (50.0) <0.001
Macrolide 223,737 (41.0) 210,954 (41.9) 12,783 (30.5) <0.001
Aminoglycoside 15,415 (2.8) 12,661 (2.5) 2,754 (6.6) <0.001
Oral steroids 44,486 (8.2) 41,586 (8.3) 2,900 (6.9) <0.001
Intravenous steroids 146,308 (26.8) 133,920 (26.6) 12,388 (29.6) <0.001
VTE prophylaxis with LMWH 190,735 (35.0) 174,612 (34.7) 16,123 (38.5) 0.01
Discharge disposition
Home 282,146 (51.7) 272,604(54.1) 9,542 (22.8) <0.001
Home with home health 71,977 (13.2) 65,289 (13.0) 6,688 (16.0) <0.001
Skilled nursing facility 111,541 (20.5) 97,113 (19.3) 14,428 (34.5) <0.001
Hospice 20,428 (3.7) 17,902 (3.6) 2,526 (6.0) <0.001
Expired 47,733 (8.7) 40,768 (8.1) 6,965 (16.6) <0.001
Other 11,425 (2.1) 9,725 (1.9) 1,700 (4.1) <0.001

We then developed a multivariable hierarchical generalized linear model (HGLM) for PICC placement with a random effect for hospital. In this model, we included patient demographics, comorbidities, sepsis on admission, type of pneumonia (eg, HCAP vs community‐associated pneumonia [CAP]), admitting physician specialty, and indicators for early receipt of specific treatments such as guideline‐recommended antibiotics, vasopressors, ventilation (invasive or noninvasive), and pneumatic compression devices for prophylaxis of deep vein thrombosis.

To understand and estimate between‐hospital variation in PICC use, we calculated risk‐standardized rates of PICC use (RSPICC) across hospitals using HGLM methods. These methods are also employed by the Centers for Medicare and Medicaid Services to calculate risk‐standardized measures for public reporting.[18] Because hospital rates of PICC use were highly skewed (21.2% [n = 105] of hospitals had no patients with PICCs), we restricted this model to the 343 hospitals that had at least 5 patients with a PICC to obtain stable estimates. For each hospital, we estimated a predicted rate of PICC use (pPICC) as the sum of predicted probabilities of PICC receipt from patient factors and the random intercept for hospital in which they were admitted. We then calculated an expected rate of PICC use (ePICC) per hospital as the sum of expected probabilities of PICC receipt from patient factors only. RSPICC for each hospital was then computed as the product of the overall unadjusted mean PICC rate (PICC) from all patients and the ratio of the predicted to expected PICC rate (uPICC*[pPICC/ePICC]).[19] Kruskal‐Wallis tests were used to evaluate the association between hospital characteristics with RSPICC rates. To evaluate the impact of the hospital in variation in PICC use, we assessed the change in likelihood ratio of a hierarchical model with hospital random effects compared to a logistic regression model with patient factors only. In addition, we estimated the intraclass correlation (ICC) to assess the proportion of variation in PICC use associated with the hospital, and the median odds ratio (MOR) from the hierarchical model. The MOR is the median of a set of odds ratios comparing 2 patients with the same set of characteristics treated at 2 randomly selected hospitals.[20, 21, 22] All analyses were performed using the Statistical Analysis System version 9.3 (SAS Institute, Inc., Cary, NC) and Stata 13 (StataCorp Inc., College Station, TX).

Ethical and Regulatory Oversight

Permission to conduct this study was obtained from the institutional review board at Baystate Medical Center, Springfield, Massachusetts. The study did not qualify as human subjects research and made use of fully deidentified data.

RESULTS

Between July 2007 and November 2011, 634,285 admissions representing 545,250 unique patients from 495 hospitals met eligibility criteria and were included in the study (Figure 1). Included patients had a median age of 71 years (interquartile range [IQR]: 5782), and 53.0% were female. Most patients were Caucasian (69.2%), unmarried (51.6%), and insured by Medicare (67.9%). Patients were admitted to the hospital by internal medicine providers (43.4%), hospitalists (21.4%), and family practice providers (14.7%); notably, critical care and pulmonary medicine providers admitted 6.5% of patients. The median Gagne comorbidity score was 2 (IQR: 15). Hypertension, chronic obstructive pulmonary disease, diabetes, and congestive heart failure were among the most common comorbidities observed (Table 1).

Figure 1
Study flow diagram. Abbreviations: CT, computed tomography; DRG, diagnosis‐related group; MS, missing; PICC, peripherally inserted central catheter; PN, pneumonia; POA, present on admission.

Among eligible patients, 41,849 (7.7%) received a PICC during hospitalization. Approximately a quarter of all patients who received PICCs did so by hospital day 2; 90% underwent insertion by hospital day 11 (mean = 5.4 days, median = 4 days). Patients who received PICCs were younger (median IQR: 69 years, 5780 years) but otherwise demographically similar to those that did not receive PICCs (median IQR: 72 years, 5782 years). Compared to other specialties, patients admitted by critical care/pulmonary providers were twice as likely to receive PICCs (12.3% vs 6.1%, P < .001). Patients who received PICCs had higher comorbidity scores than those who did not (median Gagne comorbidity score 4 vs 2, P < 0.001) and were more likely to be diagnosed with HCAP (43.1% vs 29.9%, P < 0.001) than CAP (56.9% vs 70.1%, P < 0.001).

PICC recipients were also more likely to receive intensive care unit (ICU) level of care (41.4% vs 16%, P < 0.001) and both noninvasive (17.5% vs 8.1%, P < 0.001) and invasive ventilation (28.6% vs 8.8%, P < 0.001) upon admission. Vasopressor use was also significantly more frequent in patients who received PICCs (24.7% vs 7.6%, P < 0.001) compared to those who did not receive these devices. Patients with PICCs were more often discharged to skilled nursing facilities (34.5% vs 19.3%) than those without PICCs.

Characteristics Associated With PICC Use Following Multivariable Modeling

Using HGLM with a random hospital effect, multiple patient characteristics were associated with PICC use (Table 2). Patients 65 years of age were less likely to receive a PICC compared to younger patients (odds ratio [OR]: 0.81, 95% confidence interval [CI]: 0.79‐0.84). Weight loss (OR: 2.03, 95% CI: 1.97‐2.10), sepsis on admission (OR: 1.80, 95% CI: 1.75‐1.85), and ICU status on hospital day 1 or 2 (OR: 1.70, 95% CI: 1.64‐1.75) represented 3 factors most strongly associated with PICC use.

Patient Factors Associated With PICC Use
Patient Characteristic Odds Ratio 95% Confidence Intervals
  • NOTE: Abbreviations: CAP, community‐associated pneumonia; DVT, deep vein thrombosis; FP, family practice; HCAP, healthcare‐associated pneumonia; IM, internal medicine; LMWH, low‐molecular‐weight heparin; MRSA, methicillin‐resistant Staphylococcus aureus; PICC, peripherally inserted central catheter; POA, present on admission; VTE, venous thromboembolism.

Age group (>66 vs 65 years) 0.82 0.790.84
Race/ethnicity
Other 1.02 0.971.06
Black 0.99 0.951.03
Hispanic 0.82 0.760.88
White Referent
Marital status
Other/missing 1.07 1.011.14
Single 1.02 1.001.05
Married Referent
Insurance payor
Other 0.85 0.800.89
Medicaid 1.13 1.081.18
Managed care 0.95 0.910.99
Commercialindemnity 0.93 0.871.00
Medicare Referent
Admitting physician specialty
Pulmonary/critical care medicine 1.18 1.131.24
Family practice 1.01 0.971.05
Geriatric medicine (FP and IM) 1.85 1.662.05
Hospitalist 0.94 0.910.98
Other specialties 1.02 0.971.06
Internal medicine Referent
Comorbidities
Congestive heart failure 1.27 1.241.31
Valvular disease 1.11 1.071.15
Pulmonary circulation disorders 1.37 1.321.42
Peripheral vascular disease 1.09 1.051.13
Hypertension 0.94 0.920.97
Paralysis 1.59 1.511.67
Other neurological disorders 1.20 1.161.23
Chronic lung disease 1.10 1.071.12
Diabetes 1.13 1.101.16
Hypothyroidism 1.03 1.001.06
Liver disease 1.16 1.101.23
Ulcer 1.86 1.153.02
Lymphoma 0.88 0.810.96
Metastatic cancer 0.75 0.710.80
Solid tumor without metastasis 0.93 0.880.98
Arthritis 1.22 1.161.28
Obesity 1.47 1.421.52
Weight loss 2.03 1.972.10
Blood loss 1.69 1.551.85
Deficiency anemias 1.40 1.371.44
Alcohol abuse 1.19 1.131.26
Drug abuse 1.31 1.231.39
Psychoses 1.16 1.111.21
Depression 1.10 1.061.13
Renal failure 0.96 0.930.98
Type of pneumonia
HCAP 1.03 1.011.06
CAP Referent
Sepsis (POA) 1.80 1.751.85
Antibiotic exposure
Anti‐MRSA agent 1.72 1.671.76
Anti‐Pseudomonal carbapenem 1.37 1.311.44
Non‐Pseudomonal carbapenem 1.48 1.331.66
Third‐generation cephalosporin 1.04 1.011.07
Anti‐Pseudomonal cephalosporin 1.25 1.201.30
Anti‐Pseudomonal ‐lactam 1.27 1.231.31
Aztreonam 1.31 1.231.40
Non‐Pseudomonal ‐lactam 1.36 1.231.50
‐lactam 1.55 1.261.90
Respiratory quinolone 0.90 0.870.92
Macrolide 0.85 0.820.88
Doxycycline 0.94 0.871.01
Aminoglycoside 1.21 1.141.27
Vasopressors 1.06 1.031.10
Noninvasive ventilation 1.29 1.251.34
Invasive ventilation 1.66 1.611.72
Intensive care unit on admission 1.70 1.641.75
Atrial fibrillation 1.26 1.221.29
Upper extremity chronic DVT 1.61 1.132.28
Nicotine replacement therapy/tobacco abuse 0.91 0.880.94
Aspirin 0.94 0.920.97
Warfarin 0.90 0.860.94
LMWH, prophylactic dose 1.10 1.081.13
LMWH, treatment dose 1.22 1.161.29
Intravenous steroids 1.05 1.021.08
Bacteremia (prior year) 1.14 1.021.27
VTE (prior year) 1.11 1.061.18
Pneumatic compression device 1.25 1.081.45
Invasive ventilation (prior year) 1.17 1.111.24
Irritable bowel disease 1.19 1.051.36

Therapy with potent parenteral antimicrobials including antimethicillin‐resistant Staphylococcus aureus agents (OR: 1.72, 95% CI: 1.67‐1.76), antipseudomonal ‐lactamases (OR: 1.27, 95% CI: 1.23‐1.31), and carbapenems (OR: 1.37, 95% CI: 1.31‐1.44) were significantly associated with PICC use. Conversely, use of macrolides (OR: 0.85, 95% CI: 0.82‐0.88) or respiratory fluoroquinolones (OR: 0.90, 95% CI: 0.87‐0.92) were associated with lower likelihood of PICC use. After adjusting for antimicrobial therapy, HCAP was only slightly more likely to result in PICC use than CAP (OR: 1.03, 95% CI: 1.01‐1.06). Compared to internal medicine providers, admission by geriatricians and critical care/pulmonary specialists was associated with greater likelihood of PICC use (OR: 1.85, 95% CI: 1.66‐2.05 and OR: 1.18, 95% CI: =1.13‐1.24, respectively). Admission by hospitalists was associated with a modestly lower likelihood of PICC placement (OR: 0.94, 95% CI: 0.91‐0.98).

Hospital Level Variation in PICC Use

To ensure stable estimates of hospital PICC use, we excluded 152 facilities (31%): 10% had no patients with PICCs and 21% had <5 patients who received a PICC. Therefore, RSPICC was estimated for 343 of 495 facilities (69%) (Figure 2). In these facilities, RSPICC varied from 0.3% to 41.7%. Hospital RSPICC was significantly associated with hospital location (median 11.9% vs 7.8% for urban vs rural hospitals respectively, P = 0.05). RSPICCs were also greater among hospitals in Southern (11.3%), Western (12.7%), and Midwest (12.0%) regions of the nation compared to those in the Northeast (8.4%) (P = 0.02) (Table 3).

Association Between Hospital Characteristics and Risk‐Standardized Rate of PICC Use*
Hospital Characteristic (No.) Median (IQR), % P Value
  • NOTE: Abbreviations: IQR, interquartile range; PICC, peripherally inserted central catheter.*Numbers indicate the percentage of patients with a PICC in each category, accounting for risk associated with PICC receipt. To ensure stable estimates, 152 facilities (31%) were excluded, as 10% had no patients with PICCs and 21% had <5 patients who received a PICC. Kruskal‐Wallis test.

Bed size 0.12
200 beds (106) 9.1 (4.816.3)
201 beds (237) 11.6 (5.817.6)
Rural/urban 0.05
Urban (275) 11.9 (5.517.4)
Rural (68) 7.8 (5.014.0)
Region 0.02
Northeast (50) 8.4 (3.913.0)
Midwest (69) 12.0 (5.817.4)
West (57) 12.7 (7.617.0)
South (167) 11.3 (4.817.8)
Teaching status 0.77
Nonteaching (246) 10.9 (5.017.4)
Teaching (97) 12.0 (5.816.9)
Figure 2
Observed vs risk‐standardized rate of peripherally inserted central catheter (PICC) use across 343 US hospitals (restricted to sites where >5 patients received PICCs). Horizontal axis represents rate of PICC use, whereas vertical axis represents number of hospitals. The dark shaded bars represents the observed rate of PICC use, whereas the nonshaded bars reflect risk‐standardized rate of PICC use.

A likelihood ratio test comparing the hierarchical model to a logistic model with patient factors only was highly significant (P < 0.001), indicating that the hospital where the patient was treated had a major impact on receipt of PICC after accounting for patient factors. The MOR was 2.71, which is a larger effect than we found for any of the individual patient characteristics. The proportion of variance explained by hospitals was 25% (95% CI: 22%‐28%), as measured by the ICC.

DISCUSSION

In this study of 545,250 adults hospitalized with pneumonia, we found that approximately 8% of patients received a PICC. Patients who received PICCs had more comorbidities, were more frequently diagnosed with HCAP, and were more often admitted to the ICU, where they experienced greater rates of mechanical ventilation, noninvasive ventilation, and vasopressor use compared to those who did not receive a PICC. Additionally, risk‐adjusted rates of PICC use varied as much as 10‐fold across institutions. In fact, almost 70% of the total variation in rates of PICC use remained unexplained by hospital or patient characteristics. Although use of PICCs is often clinically nuanced in ways that are difficult to capture in large datasets (eg, difficult venous access or inability to tolerate oral medications), the substantial variation of PICC use observed suggests that physician and institutional practice styles are the major determinants of PICC placement during a hospitalization for pneumonia. Because PICCs are associated with serious complications, and evidence regarding discretionary use is accumulating, a research agenda examining reasons for such use and related outcomes appears necessary.

The placement of PICCs has grown substantially in hospitalized patients all over the world.[23, 24] Although originally developed for total parenteral nutrition in surgical patients,[25] contemporary reports of PICC use in critical illness,[26] diseases such as cystic fibrosis,[27] and even pregnancy[28] are now common. Although PICCs are clinically invaluable in many of these conditions, growing use of these devices has led to the realization that benefits may be offset by complications.[9, 10, 29, 30] Additionally, recent data suggest that not all PICCs may be used for appropriate reasons. For instance, in a decade‐long study at a tertiary care center, changes in patterns of PICC use including shortened dwell times, multiple insertions in a single patient, and unclear indications for use were reported.[11] In another study at an academic medical center, a substantial proportion of PICCs were found to be idle or unjustified.[12] It comes as little surprise, then, that a recent multicenter study found that 1 out of every 5 clinicians did not even know that their patient had a PICC.[29] Although calls to improve PICC use in the hospital setting have emerged, strategies to do so are limited by data that emanate from single‐center reports or retrospective designs. No other studies reporting use of PICCs across US hospitals for any clinical condition currently exist.[31]

We found that patients with weight loss, those with greater combined comorbidity scores, and those who were critically ill or diagnosed with sepsis were more likely to receive PICCs than others. These observations suggest that PICC use may reflect underlying severity of illness, as advanced care such as ventilator support was often associated with PICC use. Additionally, discharge to a skilled nursing facility was frequently associated with PICC placement, a finding consistent with a recent study evaluating the use of PICCs in these settings.[32] However, a substantial proportion of PICC use remained unexplained by available patient or hospital factors. Although our study was not specifically designed to examine this question, a possible reason may relate to unmeasured institutional factors that influence the propensity to use a PICC, recently termed as PICC culture.[33] For example, it is plausible that hospitals with nursing‐led PICC teams or interventional radiology (such as teaching hospitals) are more likely to use PICCs than those without such operators. This hypothesis may explain why urban, larger, and teaching hospitals exhibited higher rates of PICC use. Conversely, providers may have an affinity toward PICC use that is predicated not just by operator availability, but also local hospital norms. Understanding why some facilities use PICCs at higher rates than others and implications of such variation with respect to patient safety, cost, and outcomes is important. Study designs that use mixed‐methods approaches or seek to qualitatively understand reasons behind PICC use are likely to be valuable in this enquiry.

Our study has limitations. First, we used an administrative dataset and ICD‐9‐CM codes rather than clinical data from medical records to identify cases of pneumonia or comorbidities. Our estimates of PICC use across hospitals thus may not fully account for differences in severity of illness, and it is possible that patients needed a PICC for reasons that we could not observe. However, the substantial variation observed in rates of PICC use across hospitals is unlikely to be explained by differences in patient severity of illness, documentation, or coding practices. Second, as PICC removal codes were not available, we are unable to comment on how often hospitalized pneumonia patients were discharged with PICCs or received antimicrobial therapy beyond their inpatient stay. Third, although we observed that a number of patient and hospital factors were associated with PICC receipt, our study was not designed to determine the reasons underlying these patterns.

These limitations aside, our study has important strengths. To our knowledge, this is the first study to report utilization and outcomes associated with PICC use among those hospitalized with pneumonia across the United States. The inclusion of a large number of patients receiving care in diverse facilities lends a high degree of external validity to our findings. Second, we used advanced modeling to identify factors associated with PICC use in hospitalized patients with pneumonia, producing innovative and novel findings. Third, our study is the first to show the existence of substantial variation in rates of PICC use across US hospitals within the single disease state of pneumonia. Understanding the drivers of this variability is important as it may inform future studies, policies, and practices to improve PICC use in hospitalized patients.

In conclusion, we found that PICC use in patients hospitalized with pneumonia is common and highly variable. Future studies examining the contextual factors behind PICC use and their association with outcomes are needed to facilitate efforts to standardize PICC use across hospitals.

Disclosures

Dr. Chopra is supported by a career development award (1‐K08‐HS022835‐01) from the Agency of Healthcare Research and Quality. The authors report no conflicts of interest.

Pneumonia is the most common cause of unplanned hospitalization in the United States.[1] Despite its clinical toll, the management of this disease has evolved markedly. Expanding vaccination programs, efforts to improve timeliness of antibiotic therapy, and improved processes of care are but a few developments that have improved outcomes for patients afflicted with this illness.[2, 3]

Use of peripherally inserted central catheters (PICCs) is an example of a modern development in the management of patients with pneumonia.[4, 5, 6, 7] PICCs provide many of the benefits associated with central venous catheters (CVCs) including reliable venous access for delivery of antibiotics, phlebotomy, and invasive hemodynamic monitoring. However, as they are placed in veins of the upper extremity, PICCs bypass insertion risks (eg, injury to the carotid vessels or pneumothorax) associated with placement of traditional CVCs.[8] Because they offer durable venous access, PICCs also facilitate care transitions while continuing intravenous antimicrobial therapy in patients with pneumonia.

However, accumulating evidence also suggests that PICCs are associated with important complications, including central lineassociated bloodstream infectionand venous thromboembolism.[9, 10] Furthermore, knowledge gaps in clinicians regarding indications for appropriate use and management of complications associated with PICCs have been recognized.[10, 11] These elements are problematic because reports of unjustified and inappropriate PICC use are growing in the literature.[12, 13] Such concerns have prompted a number of policy calls to improve PICC use, including Choosing Wisely recommendations by various professional societies.[14, 15]

As little is known about the prevalence or patterns of PICC use in adults hospitalized with pneumonia, we conducted a retrospective cohort study using data from a large network of US hospitals.

METHODS

Setting and Participants

We included patients from hospitals that participated in Premier's inpatient dataset, a large, fee‐supported, multipayer administrative database that has been used extensively in health services research to measure quality of care and comparative effectiveness of interventions.[16] Participating hospitals represent all regions of the United States and include teaching and nonteaching facilities in rural and urban locations. In addition to variables found in the uniform billing form, the Premier inpatient database also includes a date‐stamped list of charges for procedures conducted during hospitalization such as PICC placement. As PICC‐specific data are not available in most nationally representative datasets, Premier offers unique insights into utilization, timing, and factors associated with use of PICCs in hospitalized settings.

We included adult patients aged 18 years who were (1) admitted with a principal diagnosis of pneumonia present on admission, or secondary diagnosis of pneumonia if paired with a principal diagnosis of sepsis, respiratory failure, or influenza; (2) received at least 1 day of antibiotics between July 1, 2007 and November 30, 2011, and (3) underwent chest x‐ray or computed tomography (CT) at the time of admission. International Classification of Disease, 9th Revision, Clinical Modification (ICD‐9‐CM) codes were used for patient selection. Patients who were not admitted (eg, observation cases), had cystic fibrosis, or marked as pneumonia not present on admission were excluded. For patients who had more than 1 hospitalization during the study period, a single admission was randomly selected for inclusion.

Patient, Physician, and Hospital Data

For all patients, age, gender, marital status, insurance, race, and ethnicity were captured. Using software provided by the Healthcare Costs and Utilization Project, we categorized information on 29 comorbid conditions and computed a combined comorbidity score as described by Gagne et al.[17] Patients were considered to have healthcare‐associated pneumonia (HCAP) if they were: (1) admitted from a skilled nursing or a long‐term care facility, (2) hospitalized in the previous 90 days, (3) on dialysis, or (4) receiving immunosuppressing medications (eg, chemotherapy or steroids equivalent to at least 20 mg of prednisone per day) at the time of admission. Information on specialty of the admitting physician and hospital characteristics (eg, size, location, teaching status) were sourced through Premier data.

Receipt of PICCs and Related Therapies

Among eligible adult patients hospitalized with pneumonia, we identified patients who received a PICC at any time during hospitalization via PICC‐specific billing codes. Non‐PICC devices (eg, midlines, Hickman catheters) were not included. For all insertions, we assessed day of PICC placement relative to admission date. Data on type of PICC (eg, power‐injection capable, antibiotic coating) or PICC characteristics (size, number of lumens) were not available. We used billing codes to assess use of invasive or noninvasive ventilation, vasopressors, and administration of pneumonia‐specific antibiotics (eg, ‐lactams, macrolides, fluoroquinolones). Early exposure was defined when a billing code appeared within 2 days of hospital admission.

Outcomes of Interest

The primary outcome of interest was receipt of a PICC. Additionally, we assessed factors associated with PICC placement and variation in risk‐standardized rates of PICC use between hospitals.

Statistical Analyses

Patient and hospital characteristics were summarized using frequencies for categorical variables and medians with interquartile ranges for continuous variables. We examined association of individual patient and hospital characteristics with use of PICCs using generalized estimating equations models with a logit link for categorical variables and identity link for continuous variables, accounting for patient clustering within hospitals.

Characteristics of the Study Population
Characteristic Total, No. (%) No PICC, No. (%) PICC, No. (%) P Value*
  • NOTE: Abbreviations: CAP, community‐acquired pneumonia; GEE, generalized estimating equations; HCAP, healthcare‐associated pneumonia; ICU, intensive care unit; LMWH, low‐molecular‐weight heparin; MRSA, methicillin‐resistant Staphylococcus aureus; PICC, peripherally inserted central catheter; VTE, venous thromboembolism. *P value from GEE models that account for clustering within the hospital. Includes: discharged/transferred to cancer center/children's hospital, discharged/transferred to federal hospital; discharged/transferred to swing bed, discharged/transferred to long‐term care facility, discharged/transferred to psychiatric hospital, discharged/transferred to assisted living, discharged/transferred to other health institution not in list.

545,250 (100) 503,401 (92.3) 41,849 (7.7)
Demographics
Age, median (Q1Q3), y 71 (5782) 72 (5782) 69 (5780) <0.001
Gender <0.001
Male 256,448 (47.0) 237,232 (47.1) 19,216 (45.9)
Female 288,802 (53.0) 266,169 (52.9) 22,633 (54.1)
Race/ethnicity <0.001
White 377,255 (69.2) 346,689 (68.9) 30,566 (73.0)
Black 63,345 (11.6) 58,407 (11.6) 4,938 (11.8)
Hispanic 22,855 (4.2) 21,716 (4.3) 1,139 (2.7)
Other 81,795 (15.0) 76,589 (15.2) 5,206 (12.4)
Admitting specialty <0.001
Internal medicine 236,859 (43.4) 218,689 (43.4) 18,170 (43.4)
Hospital medicine 116,499 (21.4) 107,671 (21.4) 8,828 (21.1)
Family practice 80,388 (14.7) 75,482 (15.0) 4,906 (11.7)
Critical care and pulmonary 35,670 (6.5) 30,529 (6.1) 41,849 (12.3)
Geriatrics 4,812 (0.9) 4,098 (0.8) 714 (1.7)
Other 71,022 (13.0) 66,932 (13.3) 4,090 (9.8)
Insurance <0.001
Medicare 370,303 (67.9) 341,379 (67.8) 28,924 (69.1)
Medicaid 45,505 (8.3) 41,100 (8.2) 4,405 (10.5)
Managed care 69,984 (12.8) 65,280 (13.0) 4,704 (11.2)
Commercialindemnity 20,672 (3.8) 19,251 (3.8) 1,421 (3.4)
Other 38,786 (7.1) 36,391 (7.2) 2,395 (5.7)
Comorbidities
Gagne combined comorbidity score, median (Q1Q3) 2 (15) 2 (14) 4 (26) <0.001
Hypertension 332,347 (60.9) 306,964 (61.0) 25,383 (60.7) 0.13
Chronic pulmonary disease 255,403 (46.8) 234,619 (46.6) 20,784 (49.7) <0.001
Diabetes 171,247 (31.4) 155,540 (30.9) 15,707 (37.5) <0.001
Congestive heart failure 146,492 (26.9) 131,041 (26.0) 15,451 (36.9) <0.001
Atrial fibrillation 108,405 (19.9) 97,124 (19.3) 11,281 (27.0) <0.001
Renal failure 104,404 (19.1) 94,277 (18.7) 10,127 (24.2) <0.001
Nicotine replacement therapy/tobacco use 89,938 (16.5) 83,247 (16.5) 6,691 (16.0) <0.001
Obesity 60,242 (11.0) 53,268 (10.6) 6,974 (16.7) <0.001
Coagulopathy 41,717 (7.6) 35,371 (7.0) 6,346 (15.2) <0.001
Prior stroke (1 year) 26,787 (4.9) 24,046 (4.78) 2,741 (6.55) <0.001
Metastatic cancer 21,868 (4.0) 20,244 (4.0) 1,624 (3.9) 0.16
Solid tumor w/out metastasis 21,083 (3.9) 19,380 (3.8) 1,703 (4.1) 0.12
Prior VTE (1 year) 19,090 (3.5) 16,906 (3.4) 2,184 (5.2) <0.001
Chronic liver disease 16,273 (3.0) 14,207 (2.8) 2,066 (4.9) <0.001
Prior bacteremia (1 year) 4,106 (0.7) 3,584 (0.7) 522 (1.2) <0.001
Nephrotic syndrome 671 (0.1) 607 (0.1) 64 (0.2) 0.03
Morbidity markers
Type of pneumonia <0.001
CAP 376,370 (69.1) 352,900 (70.1) 23,830 (56.9)
HCAP 168,520 (30.9) 150,501 (29.9) 18,019 (43.1)
Sepsis present on admission 114,578 (21.0) 96,467 (19.2) 18,111 (43.3) <0.001
Non‐invasive ventilation 47,913(8.8) 40,599 (8.1) 7,314 (17.5) <0.001
Invasive mechanical ventilation 56,179 (10.3) 44,228 (8.8) 11,951 (28.6) <0.001
ICU status 97,703 (17.9) 80,380 (16.0) 17,323 (41.4) <0.001
Vasopressor use 48,353 (8.9) 38,030 (7.6) 10,323 (24.7) <0.001
Antibiotic/medication use
Anti‐MRSA agent (vancomycin) 146,068 (26.8) 123,327 (24.5) 22,741 (54.3) <0.001
Third‐generation cephalosporin 250,782 (46.0) 235,556 (46.8) 15,226 (36.4) <0.001
Anti‐Pseudomonal cephalosporin 41,798 (7.7) 36,982 (7.3) 4,816 (11.5) <0.001
Anti‐Pseudomonal ‐lactam 122,215 (22.4) 105,741 (21.0) 16,474 (39.4) <0.001
Fluroquinolone 288,051 (52.8) 267,131 (53.1) 20,920 (50.0) <0.001
Macrolide 223,737 (41.0) 210,954 (41.9) 12,783 (30.5) <0.001
Aminoglycoside 15,415 (2.8) 12,661 (2.5) 2,754 (6.6) <0.001
Oral steroids 44,486 (8.2) 41,586 (8.3) 2,900 (6.9) <0.001
Intravenous steroids 146,308 (26.8) 133,920 (26.6) 12,388 (29.6) <0.001
VTE prophylaxis with LMWH 190,735 (35.0) 174,612 (34.7) 16,123 (38.5) 0.01
Discharge disposition
Home 282,146 (51.7) 272,604(54.1) 9,542 (22.8) <0.001
Home with home health 71,977 (13.2) 65,289 (13.0) 6,688 (16.0) <0.001
Skilled nursing facility 111,541 (20.5) 97,113 (19.3) 14,428 (34.5) <0.001
Hospice 20,428 (3.7) 17,902 (3.6) 2,526 (6.0) <0.001
Expired 47,733 (8.7) 40,768 (8.1) 6,965 (16.6) <0.001
Other 11,425 (2.1) 9,725 (1.9) 1,700 (4.1) <0.001

We then developed a multivariable hierarchical generalized linear model (HGLM) for PICC placement with a random effect for hospital. In this model, we included patient demographics, comorbidities, sepsis on admission, type of pneumonia (eg, HCAP vs community‐associated pneumonia [CAP]), admitting physician specialty, and indicators for early receipt of specific treatments such as guideline‐recommended antibiotics, vasopressors, ventilation (invasive or noninvasive), and pneumatic compression devices for prophylaxis of deep vein thrombosis.

To understand and estimate between‐hospital variation in PICC use, we calculated risk‐standardized rates of PICC use (RSPICC) across hospitals using HGLM methods. These methods are also employed by the Centers for Medicare and Medicaid Services to calculate risk‐standardized measures for public reporting.[18] Because hospital rates of PICC use were highly skewed (21.2% [n = 105] of hospitals had no patients with PICCs), we restricted this model to the 343 hospitals that had at least 5 patients with a PICC to obtain stable estimates. For each hospital, we estimated a predicted rate of PICC use (pPICC) as the sum of predicted probabilities of PICC receipt from patient factors and the random intercept for hospital in which they were admitted. We then calculated an expected rate of PICC use (ePICC) per hospital as the sum of expected probabilities of PICC receipt from patient factors only. RSPICC for each hospital was then computed as the product of the overall unadjusted mean PICC rate (PICC) from all patients and the ratio of the predicted to expected PICC rate (uPICC*[pPICC/ePICC]).[19] Kruskal‐Wallis tests were used to evaluate the association between hospital characteristics with RSPICC rates. To evaluate the impact of the hospital in variation in PICC use, we assessed the change in likelihood ratio of a hierarchical model with hospital random effects compared to a logistic regression model with patient factors only. In addition, we estimated the intraclass correlation (ICC) to assess the proportion of variation in PICC use associated with the hospital, and the median odds ratio (MOR) from the hierarchical model. The MOR is the median of a set of odds ratios comparing 2 patients with the same set of characteristics treated at 2 randomly selected hospitals.[20, 21, 22] All analyses were performed using the Statistical Analysis System version 9.3 (SAS Institute, Inc., Cary, NC) and Stata 13 (StataCorp Inc., College Station, TX).

Ethical and Regulatory Oversight

Permission to conduct this study was obtained from the institutional review board at Baystate Medical Center, Springfield, Massachusetts. The study did not qualify as human subjects research and made use of fully deidentified data.

RESULTS

Between July 2007 and November 2011, 634,285 admissions representing 545,250 unique patients from 495 hospitals met eligibility criteria and were included in the study (Figure 1). Included patients had a median age of 71 years (interquartile range [IQR]: 5782), and 53.0% were female. Most patients were Caucasian (69.2%), unmarried (51.6%), and insured by Medicare (67.9%). Patients were admitted to the hospital by internal medicine providers (43.4%), hospitalists (21.4%), and family practice providers (14.7%); notably, critical care and pulmonary medicine providers admitted 6.5% of patients. The median Gagne comorbidity score was 2 (IQR: 15). Hypertension, chronic obstructive pulmonary disease, diabetes, and congestive heart failure were among the most common comorbidities observed (Table 1).

Figure 1
Study flow diagram. Abbreviations: CT, computed tomography; DRG, diagnosis‐related group; MS, missing; PICC, peripherally inserted central catheter; PN, pneumonia; POA, present on admission.

Among eligible patients, 41,849 (7.7%) received a PICC during hospitalization. Approximately a quarter of all patients who received PICCs did so by hospital day 2; 90% underwent insertion by hospital day 11 (mean = 5.4 days, median = 4 days). Patients who received PICCs were younger (median IQR: 69 years, 5780 years) but otherwise demographically similar to those that did not receive PICCs (median IQR: 72 years, 5782 years). Compared to other specialties, patients admitted by critical care/pulmonary providers were twice as likely to receive PICCs (12.3% vs 6.1%, P < .001). Patients who received PICCs had higher comorbidity scores than those who did not (median Gagne comorbidity score 4 vs 2, P < 0.001) and were more likely to be diagnosed with HCAP (43.1% vs 29.9%, P < 0.001) than CAP (56.9% vs 70.1%, P < 0.001).

PICC recipients were also more likely to receive intensive care unit (ICU) level of care (41.4% vs 16%, P < 0.001) and both noninvasive (17.5% vs 8.1%, P < 0.001) and invasive ventilation (28.6% vs 8.8%, P < 0.001) upon admission. Vasopressor use was also significantly more frequent in patients who received PICCs (24.7% vs 7.6%, P < 0.001) compared to those who did not receive these devices. Patients with PICCs were more often discharged to skilled nursing facilities (34.5% vs 19.3%) than those without PICCs.

Characteristics Associated With PICC Use Following Multivariable Modeling

Using HGLM with a random hospital effect, multiple patient characteristics were associated with PICC use (Table 2). Patients 65 years of age were less likely to receive a PICC compared to younger patients (odds ratio [OR]: 0.81, 95% confidence interval [CI]: 0.79‐0.84). Weight loss (OR: 2.03, 95% CI: 1.97‐2.10), sepsis on admission (OR: 1.80, 95% CI: 1.75‐1.85), and ICU status on hospital day 1 or 2 (OR: 1.70, 95% CI: 1.64‐1.75) represented 3 factors most strongly associated with PICC use.

Patient Factors Associated With PICC Use
Patient Characteristic Odds Ratio 95% Confidence Intervals
  • NOTE: Abbreviations: CAP, community‐associated pneumonia; DVT, deep vein thrombosis; FP, family practice; HCAP, healthcare‐associated pneumonia; IM, internal medicine; LMWH, low‐molecular‐weight heparin; MRSA, methicillin‐resistant Staphylococcus aureus; PICC, peripherally inserted central catheter; POA, present on admission; VTE, venous thromboembolism.

Age group (>66 vs 65 years) 0.82 0.790.84
Race/ethnicity
Other 1.02 0.971.06
Black 0.99 0.951.03
Hispanic 0.82 0.760.88
White Referent
Marital status
Other/missing 1.07 1.011.14
Single 1.02 1.001.05
Married Referent
Insurance payor
Other 0.85 0.800.89
Medicaid 1.13 1.081.18
Managed care 0.95 0.910.99
Commercialindemnity 0.93 0.871.00
Medicare Referent
Admitting physician specialty
Pulmonary/critical care medicine 1.18 1.131.24
Family practice 1.01 0.971.05
Geriatric medicine (FP and IM) 1.85 1.662.05
Hospitalist 0.94 0.910.98
Other specialties 1.02 0.971.06
Internal medicine Referent
Comorbidities
Congestive heart failure 1.27 1.241.31
Valvular disease 1.11 1.071.15
Pulmonary circulation disorders 1.37 1.321.42
Peripheral vascular disease 1.09 1.051.13
Hypertension 0.94 0.920.97
Paralysis 1.59 1.511.67
Other neurological disorders 1.20 1.161.23
Chronic lung disease 1.10 1.071.12
Diabetes 1.13 1.101.16
Hypothyroidism 1.03 1.001.06
Liver disease 1.16 1.101.23
Ulcer 1.86 1.153.02
Lymphoma 0.88 0.810.96
Metastatic cancer 0.75 0.710.80
Solid tumor without metastasis 0.93 0.880.98
Arthritis 1.22 1.161.28
Obesity 1.47 1.421.52
Weight loss 2.03 1.972.10
Blood loss 1.69 1.551.85
Deficiency anemias 1.40 1.371.44
Alcohol abuse 1.19 1.131.26
Drug abuse 1.31 1.231.39
Psychoses 1.16 1.111.21
Depression 1.10 1.061.13
Renal failure 0.96 0.930.98
Type of pneumonia
HCAP 1.03 1.011.06
CAP Referent
Sepsis (POA) 1.80 1.751.85
Antibiotic exposure
Anti‐MRSA agent 1.72 1.671.76
Anti‐Pseudomonal carbapenem 1.37 1.311.44
Non‐Pseudomonal carbapenem 1.48 1.331.66
Third‐generation cephalosporin 1.04 1.011.07
Anti‐Pseudomonal cephalosporin 1.25 1.201.30
Anti‐Pseudomonal ‐lactam 1.27 1.231.31
Aztreonam 1.31 1.231.40
Non‐Pseudomonal ‐lactam 1.36 1.231.50
‐lactam 1.55 1.261.90
Respiratory quinolone 0.90 0.870.92
Macrolide 0.85 0.820.88
Doxycycline 0.94 0.871.01
Aminoglycoside 1.21 1.141.27
Vasopressors 1.06 1.031.10
Noninvasive ventilation 1.29 1.251.34
Invasive ventilation 1.66 1.611.72
Intensive care unit on admission 1.70 1.641.75
Atrial fibrillation 1.26 1.221.29
Upper extremity chronic DVT 1.61 1.132.28
Nicotine replacement therapy/tobacco abuse 0.91 0.880.94
Aspirin 0.94 0.920.97
Warfarin 0.90 0.860.94
LMWH, prophylactic dose 1.10 1.081.13
LMWH, treatment dose 1.22 1.161.29
Intravenous steroids 1.05 1.021.08
Bacteremia (prior year) 1.14 1.021.27
VTE (prior year) 1.11 1.061.18
Pneumatic compression device 1.25 1.081.45
Invasive ventilation (prior year) 1.17 1.111.24
Irritable bowel disease 1.19 1.051.36

Therapy with potent parenteral antimicrobials including antimethicillin‐resistant Staphylococcus aureus agents (OR: 1.72, 95% CI: 1.67‐1.76), antipseudomonal ‐lactamases (OR: 1.27, 95% CI: 1.23‐1.31), and carbapenems (OR: 1.37, 95% CI: 1.31‐1.44) were significantly associated with PICC use. Conversely, use of macrolides (OR: 0.85, 95% CI: 0.82‐0.88) or respiratory fluoroquinolones (OR: 0.90, 95% CI: 0.87‐0.92) were associated with lower likelihood of PICC use. After adjusting for antimicrobial therapy, HCAP was only slightly more likely to result in PICC use than CAP (OR: 1.03, 95% CI: 1.01‐1.06). Compared to internal medicine providers, admission by geriatricians and critical care/pulmonary specialists was associated with greater likelihood of PICC use (OR: 1.85, 95% CI: 1.66‐2.05 and OR: 1.18, 95% CI: =1.13‐1.24, respectively). Admission by hospitalists was associated with a modestly lower likelihood of PICC placement (OR: 0.94, 95% CI: 0.91‐0.98).

Hospital Level Variation in PICC Use

To ensure stable estimates of hospital PICC use, we excluded 152 facilities (31%): 10% had no patients with PICCs and 21% had <5 patients who received a PICC. Therefore, RSPICC was estimated for 343 of 495 facilities (69%) (Figure 2). In these facilities, RSPICC varied from 0.3% to 41.7%. Hospital RSPICC was significantly associated with hospital location (median 11.9% vs 7.8% for urban vs rural hospitals respectively, P = 0.05). RSPICCs were also greater among hospitals in Southern (11.3%), Western (12.7%), and Midwest (12.0%) regions of the nation compared to those in the Northeast (8.4%) (P = 0.02) (Table 3).

Association Between Hospital Characteristics and Risk‐Standardized Rate of PICC Use*
Hospital Characteristic (No.) Median (IQR), % P Value
  • NOTE: Abbreviations: IQR, interquartile range; PICC, peripherally inserted central catheter.*Numbers indicate the percentage of patients with a PICC in each category, accounting for risk associated with PICC receipt. To ensure stable estimates, 152 facilities (31%) were excluded, as 10% had no patients with PICCs and 21% had <5 patients who received a PICC. Kruskal‐Wallis test.

Bed size 0.12
200 beds (106) 9.1 (4.816.3)
201 beds (237) 11.6 (5.817.6)
Rural/urban 0.05
Urban (275) 11.9 (5.517.4)
Rural (68) 7.8 (5.014.0)
Region 0.02
Northeast (50) 8.4 (3.913.0)
Midwest (69) 12.0 (5.817.4)
West (57) 12.7 (7.617.0)
South (167) 11.3 (4.817.8)
Teaching status 0.77
Nonteaching (246) 10.9 (5.017.4)
Teaching (97) 12.0 (5.816.9)
Figure 2
Observed vs risk‐standardized rate of peripherally inserted central catheter (PICC) use across 343 US hospitals (restricted to sites where >5 patients received PICCs). Horizontal axis represents rate of PICC use, whereas vertical axis represents number of hospitals. The dark shaded bars represents the observed rate of PICC use, whereas the nonshaded bars reflect risk‐standardized rate of PICC use.

A likelihood ratio test comparing the hierarchical model to a logistic model with patient factors only was highly significant (P < 0.001), indicating that the hospital where the patient was treated had a major impact on receipt of PICC after accounting for patient factors. The MOR was 2.71, which is a larger effect than we found for any of the individual patient characteristics. The proportion of variance explained by hospitals was 25% (95% CI: 22%‐28%), as measured by the ICC.

DISCUSSION

In this study of 545,250 adults hospitalized with pneumonia, we found that approximately 8% of patients received a PICC. Patients who received PICCs had more comorbidities, were more frequently diagnosed with HCAP, and were more often admitted to the ICU, where they experienced greater rates of mechanical ventilation, noninvasive ventilation, and vasopressor use compared to those who did not receive a PICC. Additionally, risk‐adjusted rates of PICC use varied as much as 10‐fold across institutions. In fact, almost 70% of the total variation in rates of PICC use remained unexplained by hospital or patient characteristics. Although use of PICCs is often clinically nuanced in ways that are difficult to capture in large datasets (eg, difficult venous access or inability to tolerate oral medications), the substantial variation of PICC use observed suggests that physician and institutional practice styles are the major determinants of PICC placement during a hospitalization for pneumonia. Because PICCs are associated with serious complications, and evidence regarding discretionary use is accumulating, a research agenda examining reasons for such use and related outcomes appears necessary.

The placement of PICCs has grown substantially in hospitalized patients all over the world.[23, 24] Although originally developed for total parenteral nutrition in surgical patients,[25] contemporary reports of PICC use in critical illness,[26] diseases such as cystic fibrosis,[27] and even pregnancy[28] are now common. Although PICCs are clinically invaluable in many of these conditions, growing use of these devices has led to the realization that benefits may be offset by complications.[9, 10, 29, 30] Additionally, recent data suggest that not all PICCs may be used for appropriate reasons. For instance, in a decade‐long study at a tertiary care center, changes in patterns of PICC use including shortened dwell times, multiple insertions in a single patient, and unclear indications for use were reported.[11] In another study at an academic medical center, a substantial proportion of PICCs were found to be idle or unjustified.[12] It comes as little surprise, then, that a recent multicenter study found that 1 out of every 5 clinicians did not even know that their patient had a PICC.[29] Although calls to improve PICC use in the hospital setting have emerged, strategies to do so are limited by data that emanate from single‐center reports or retrospective designs. No other studies reporting use of PICCs across US hospitals for any clinical condition currently exist.[31]

We found that patients with weight loss, those with greater combined comorbidity scores, and those who were critically ill or diagnosed with sepsis were more likely to receive PICCs than others. These observations suggest that PICC use may reflect underlying severity of illness, as advanced care such as ventilator support was often associated with PICC use. Additionally, discharge to a skilled nursing facility was frequently associated with PICC placement, a finding consistent with a recent study evaluating the use of PICCs in these settings.[32] However, a substantial proportion of PICC use remained unexplained by available patient or hospital factors. Although our study was not specifically designed to examine this question, a possible reason may relate to unmeasured institutional factors that influence the propensity to use a PICC, recently termed as PICC culture.[33] For example, it is plausible that hospitals with nursing‐led PICC teams or interventional radiology (such as teaching hospitals) are more likely to use PICCs than those without such operators. This hypothesis may explain why urban, larger, and teaching hospitals exhibited higher rates of PICC use. Conversely, providers may have an affinity toward PICC use that is predicated not just by operator availability, but also local hospital norms. Understanding why some facilities use PICCs at higher rates than others and implications of such variation with respect to patient safety, cost, and outcomes is important. Study designs that use mixed‐methods approaches or seek to qualitatively understand reasons behind PICC use are likely to be valuable in this enquiry.

Our study has limitations. First, we used an administrative dataset and ICD‐9‐CM codes rather than clinical data from medical records to identify cases of pneumonia or comorbidities. Our estimates of PICC use across hospitals thus may not fully account for differences in severity of illness, and it is possible that patients needed a PICC for reasons that we could not observe. However, the substantial variation observed in rates of PICC use across hospitals is unlikely to be explained by differences in patient severity of illness, documentation, or coding practices. Second, as PICC removal codes were not available, we are unable to comment on how often hospitalized pneumonia patients were discharged with PICCs or received antimicrobial therapy beyond their inpatient stay. Third, although we observed that a number of patient and hospital factors were associated with PICC receipt, our study was not designed to determine the reasons underlying these patterns.

These limitations aside, our study has important strengths. To our knowledge, this is the first study to report utilization and outcomes associated with PICC use among those hospitalized with pneumonia across the United States. The inclusion of a large number of patients receiving care in diverse facilities lends a high degree of external validity to our findings. Second, we used advanced modeling to identify factors associated with PICC use in hospitalized patients with pneumonia, producing innovative and novel findings. Third, our study is the first to show the existence of substantial variation in rates of PICC use across US hospitals within the single disease state of pneumonia. Understanding the drivers of this variability is important as it may inform future studies, policies, and practices to improve PICC use in hospitalized patients.

In conclusion, we found that PICC use in patients hospitalized with pneumonia is common and highly variable. Future studies examining the contextual factors behind PICC use and their association with outcomes are needed to facilitate efforts to standardize PICC use across hospitals.

Disclosures

Dr. Chopra is supported by a career development award (1‐K08‐HS022835‐01) from the Agency of Healthcare Research and Quality. The authors report no conflicts of interest.

References
  1. Elixhauser A, Owens P. Reasons for being admitted to the hospital through the emergency department, 2003. Healthcare Cost and Utilization Project Statistical Brief 2. Rockville, MD: Agency for Healthcare Research and Quality. Available at: www.hcup‐us.ahrq.gov/reports/statbriefs/sb2.pdf. Published February 2006. Accessed June 27, 2014.
  2. Suter LG, Li SX, Grady JN, et al. National patterns of risk‐standardized mortality and readmission after hospitalization for acute myocardial infarction, heart failure, and pneumonia: update on publicly reported outcomes measures based on the 2013 release. J Gen Intern Med. 2014;29(10):13331340.
  3. Lee JS, Nsa W, Hausmann LR, et al. Quality of care for elderly patients hospitalized for pneumonia in the United States, 2006 to 2010. JAMA Intern Med. 2014;174(11):18061814.
  4. Masoorli S, Angeles T. PICC lines: the latest home care challenge. RN. 1990;53(1):4451.
  5. Lam S, Scannell R, Roessler D, Smith MA. Peripherally inserted central catheters in an acute‐care hospital. Arch Intern Med. 1994;154(16):18331837.
  6. Goodwin ML, Carlson I. The peripherally inserted central catheter: a retrospective look at three years of insertions. J Intraven Nurs. 1993;16(2):92103.
  7. Ng PK, Ault MJ, Ellrodt AG, Maldonado L. Peripherally inserted central catheters in general medicine. Mayo Clin Proc. 1997;72(3):225233.
  8. Funk D, Gray J, Plourde PJ. Two‐year trends of peripherally inserted central catheter‐line complications at a tertiary‐care hospital: role of nursing expertise. Infect Control Hosp Epidemiol. 2001;22(6):377379.
  9. Chopra V, Ratz D, Kuhn L, Lopus T, Chenoweth C, Krein S. PICC‐associated bloodstream infections: prevalence, patterns, and predictors. Am J Med. 2014;127(4):319328.
  10. Chopra V, O'Horo JC, Rogers MA, Maki DG, Safdar N. The risk of bloodstream infection associated with peripherally inserted central catheters compared with central venous catheters in adults: a systematic review and meta‐analysis. Infect Control Hosp Epidemiol. 2013;34(9):908918.
  11. Gibson C, Connolly BL, Moineddin R, Mahant S, Filipescu D, Amaral JG. Peripherally inserted central catheters: use at a tertiary care pediatric center. J Vasc Interv Radiol. 2013;24(9):13231331.
  12. Tejedor SC, Tong D, Stein J, et al. Temporary central venous catheter utilization patterns in a large tertiary care center: tracking the “idle central venous catheter”. Infect Control Hosp Epidemiol. 2012;33(1):5057.
  13. Tiwari MM, Hermsen ED, Charlton ME, Anderson JR, Rupp ME. Inappropriate intravascular device use: a prospective study. Journal Hosp Infect. 2011;78(2):128132.
  14. McMahon LF, Beyth RJ, Burger A, et al. Enhancing patient‐centered care: SGIM and choosing wisely. J Gen Intern Med. 2014;29(3):432433.
  15. Williams AW, Dwyer AC, Eddy AA, et al. Critical and honest conversations: the evidence behind the “Choosing Wisely” campaign recommendations by the American Society of Nephrology. Clin J Am Soc Nephrol. 2012;7(10):16641672.
  16. Rothberg MB, Pekow PS, Priya A, et al. Using highly detailed administrative data to predict pneumonia mortality. PLoS One. 2014;9(1):e87382.
  17. Gagne JJ, Glynn RJ, Avorn J, Levin R, Schneeweiss S. A combined comorbidity score predicted mortality in elderly patients better than existing scores. J Clin Epidemiol. 2011;64(7):749759.
  18. Sjoding MW, Prescott HC, Wunsch H, Iwashyna TJ, Cooke CR. Hospitals with the highest intensive care utilization provide lower quality pneumonia care to the elderly. Crit Care Med. 2015;43(6):11781186.
  19. Normand SL, Shahian DM. Statistical and clinical aspects of hospital outcomes profiling. Stat Sci. 2007;22(2):206226.
  20. Larsen K, Merlo J. Appropriate assessment of neighborhood effects on individual health: integrating random and fixed effects in multilevel logistic regression. Am J Epidemiol. 2005;161(1):8188.
  21. Larsen K, Petersen JH, Budtz‐Jorgensen E, Endahl L. Interpreting parameters in the logistic regression model with random effects. Biometrics. 2000;56(3):909914.
  22. Sanagou M, Wolfe R, Forbes A, Reid CM. Hospital‐level associations with 30‐day patient mortality after cardiac surgery: a tutorial on the application and interpretation of marginal and multilevel logistic regression. BMC Med Res Methodol. 2012;12:28.
  23. Lisova K, Paulinova V, Zemanova K, Hromadkova J. Experiences of the first PICC team in the Czech Republic. Br J Nurs. 2015;24(suppl 2):S4S10.
  24. Konstantinou EA, Stafylarakis E, Kapritsou M, et al. Greece reports prototype intervention with first peripherally inserted central catheter: case report and literature review. J Vasc Nurs. 2012;30(3):8893.
  25. Hoshal VL Total intravenous nutrition with peripherally inserted silicone elastomer central venous catheters. Arch Surg. 1975;110(5):644646.
  26. Cotogni P, Pittiruti M. Focus on peripherally inserted central catheters in critically ill patients. World J Crit Care Med. 2014;3(4):8094.
  27. Mermis JD, Strom JC, Greenwood JP, et al. Quality improvement initiative to reduce deep vein thrombosis associated with peripherally inserted central catheters in adults with cystic fibrosis. Ann Am Thorac Soc. 2014;11(9):14041410.
  28. Cape AV, Mogensen KM, Robinson MK, Carusi DA. Peripherally Inserted central catheter (PICC) complications during pregnancy. JPEN J Parenter Enteral Nutr. 2013;38(5):595601.
  29. Chopra V, Govindan S, Kuhn L, et al. Do clinicians know which of their patients have central venous catheters?: a multicenter observational study. Ann Intern Med. 2014;161(8):562567.
  30. Chopra V, Anand S, Hickner A, et al. Risk of venous thromboembolism associated with peripherally inserted central catheters: a systematic review and meta‐analysis. Lancet. 2013;382(9889):311325.
  31. Chopra V, Flanders SA, Saint S. The problem with peripherally inserted central catheters. JAMA. 2012;308(15):15271528.
  32. Chopra V, Montoya A, Joshi D, et al. Peripherally inserted central catheter use in skilled nursing facilities: a pilot study. J Am Geriatr Soc. 2015;63(9):18941899.
  33. McGill RL, Tsukahara T, Bhardwaj R, Kapetanos AT, Marcus RJ. Inpatient venous access practices: PICC culture and the kidney patient. J Vasc Access. 2015;16(3):206210.
References
  1. Elixhauser A, Owens P. Reasons for being admitted to the hospital through the emergency department, 2003. Healthcare Cost and Utilization Project Statistical Brief 2. Rockville, MD: Agency for Healthcare Research and Quality. Available at: www.hcup‐us.ahrq.gov/reports/statbriefs/sb2.pdf. Published February 2006. Accessed June 27, 2014.
  2. Suter LG, Li SX, Grady JN, et al. National patterns of risk‐standardized mortality and readmission after hospitalization for acute myocardial infarction, heart failure, and pneumonia: update on publicly reported outcomes measures based on the 2013 release. J Gen Intern Med. 2014;29(10):13331340.
  3. Lee JS, Nsa W, Hausmann LR, et al. Quality of care for elderly patients hospitalized for pneumonia in the United States, 2006 to 2010. JAMA Intern Med. 2014;174(11):18061814.
  4. Masoorli S, Angeles T. PICC lines: the latest home care challenge. RN. 1990;53(1):4451.
  5. Lam S, Scannell R, Roessler D, Smith MA. Peripherally inserted central catheters in an acute‐care hospital. Arch Intern Med. 1994;154(16):18331837.
  6. Goodwin ML, Carlson I. The peripherally inserted central catheter: a retrospective look at three years of insertions. J Intraven Nurs. 1993;16(2):92103.
  7. Ng PK, Ault MJ, Ellrodt AG, Maldonado L. Peripherally inserted central catheters in general medicine. Mayo Clin Proc. 1997;72(3):225233.
  8. Funk D, Gray J, Plourde PJ. Two‐year trends of peripherally inserted central catheter‐line complications at a tertiary‐care hospital: role of nursing expertise. Infect Control Hosp Epidemiol. 2001;22(6):377379.
  9. Chopra V, Ratz D, Kuhn L, Lopus T, Chenoweth C, Krein S. PICC‐associated bloodstream infections: prevalence, patterns, and predictors. Am J Med. 2014;127(4):319328.
  10. Chopra V, O'Horo JC, Rogers MA, Maki DG, Safdar N. The risk of bloodstream infection associated with peripherally inserted central catheters compared with central venous catheters in adults: a systematic review and meta‐analysis. Infect Control Hosp Epidemiol. 2013;34(9):908918.
  11. Gibson C, Connolly BL, Moineddin R, Mahant S, Filipescu D, Amaral JG. Peripherally inserted central catheters: use at a tertiary care pediatric center. J Vasc Interv Radiol. 2013;24(9):13231331.
  12. Tejedor SC, Tong D, Stein J, et al. Temporary central venous catheter utilization patterns in a large tertiary care center: tracking the “idle central venous catheter”. Infect Control Hosp Epidemiol. 2012;33(1):5057.
  13. Tiwari MM, Hermsen ED, Charlton ME, Anderson JR, Rupp ME. Inappropriate intravascular device use: a prospective study. Journal Hosp Infect. 2011;78(2):128132.
  14. McMahon LF, Beyth RJ, Burger A, et al. Enhancing patient‐centered care: SGIM and choosing wisely. J Gen Intern Med. 2014;29(3):432433.
  15. Williams AW, Dwyer AC, Eddy AA, et al. Critical and honest conversations: the evidence behind the “Choosing Wisely” campaign recommendations by the American Society of Nephrology. Clin J Am Soc Nephrol. 2012;7(10):16641672.
  16. Rothberg MB, Pekow PS, Priya A, et al. Using highly detailed administrative data to predict pneumonia mortality. PLoS One. 2014;9(1):e87382.
  17. Gagne JJ, Glynn RJ, Avorn J, Levin R, Schneeweiss S. A combined comorbidity score predicted mortality in elderly patients better than existing scores. J Clin Epidemiol. 2011;64(7):749759.
  18. Sjoding MW, Prescott HC, Wunsch H, Iwashyna TJ, Cooke CR. Hospitals with the highest intensive care utilization provide lower quality pneumonia care to the elderly. Crit Care Med. 2015;43(6):11781186.
  19. Normand SL, Shahian DM. Statistical and clinical aspects of hospital outcomes profiling. Stat Sci. 2007;22(2):206226.
  20. Larsen K, Merlo J. Appropriate assessment of neighborhood effects on individual health: integrating random and fixed effects in multilevel logistic regression. Am J Epidemiol. 2005;161(1):8188.
  21. Larsen K, Petersen JH, Budtz‐Jorgensen E, Endahl L. Interpreting parameters in the logistic regression model with random effects. Biometrics. 2000;56(3):909914.
  22. Sanagou M, Wolfe R, Forbes A, Reid CM. Hospital‐level associations with 30‐day patient mortality after cardiac surgery: a tutorial on the application and interpretation of marginal and multilevel logistic regression. BMC Med Res Methodol. 2012;12:28.
  23. Lisova K, Paulinova V, Zemanova K, Hromadkova J. Experiences of the first PICC team in the Czech Republic. Br J Nurs. 2015;24(suppl 2):S4S10.
  24. Konstantinou EA, Stafylarakis E, Kapritsou M, et al. Greece reports prototype intervention with first peripherally inserted central catheter: case report and literature review. J Vasc Nurs. 2012;30(3):8893.
  25. Hoshal VL Total intravenous nutrition with peripherally inserted silicone elastomer central venous catheters. Arch Surg. 1975;110(5):644646.
  26. Cotogni P, Pittiruti M. Focus on peripherally inserted central catheters in critically ill patients. World J Crit Care Med. 2014;3(4):8094.
  27. Mermis JD, Strom JC, Greenwood JP, et al. Quality improvement initiative to reduce deep vein thrombosis associated with peripherally inserted central catheters in adults with cystic fibrosis. Ann Am Thorac Soc. 2014;11(9):14041410.
  28. Cape AV, Mogensen KM, Robinson MK, Carusi DA. Peripherally Inserted central catheter (PICC) complications during pregnancy. JPEN J Parenter Enteral Nutr. 2013;38(5):595601.
  29. Chopra V, Govindan S, Kuhn L, et al. Do clinicians know which of their patients have central venous catheters?: a multicenter observational study. Ann Intern Med. 2014;161(8):562567.
  30. Chopra V, Anand S, Hickner A, et al. Risk of venous thromboembolism associated with peripherally inserted central catheters: a systematic review and meta‐analysis. Lancet. 2013;382(9889):311325.
  31. Chopra V, Flanders SA, Saint S. The problem with peripherally inserted central catheters. JAMA. 2012;308(15):15271528.
  32. Chopra V, Montoya A, Joshi D, et al. Peripherally inserted central catheter use in skilled nursing facilities: a pilot study. J Am Geriatr Soc. 2015;63(9):18941899.
  33. McGill RL, Tsukahara T, Bhardwaj R, Kapetanos AT, Marcus RJ. Inpatient venous access practices: PICC culture and the kidney patient. J Vasc Access. 2015;16(3):206210.
Issue
Journal of Hospital Medicine - 11(8)
Issue
Journal of Hospital Medicine - 11(8)
Page Number
568-575
Page Number
568-575
Publications
Publications
Article Type
Display Headline
Variation in prevalence and patterns of peripherally inserted central catheter use in adults hospitalized with pneumonia
Display Headline
Variation in prevalence and patterns of peripherally inserted central catheter use in adults hospitalized with pneumonia
Sections
Article Source
© 2016 Society of Hospital Medicine
Disallow All Ads
Correspondence Location
Address for correspondence and reprint requests: Vineet Chopra, MD, 2800 Plymouth Road, Building 16, Room 432W, Ann Arbor, MI 48109; Telephone: 585‐922‐4331; Fax: 585‐922‐5168; E‐mail: vineetc@umich.edu
Content Gating
Gated (full article locked unless allowed per User)
Gating Strategy
First Peek Free
Article PDF Media
Media Files

Long‐term Antipsychotics in Elders

Article Type
Changed
Mon, 01/02/2017 - 19:34
Display Headline
Long‐term outcomes of elders discharged on antipsychotics

Delirium, a clinical syndrome characterized by inattention and acute cognitive dysfunction, is very common in older hospitalized patients, with a reported incidence of 18% to 35% at time of admission and overall occurrence rates of 29% to 64%.[1] Previous studies have reported that a diagnosis of delirium is not benign and is associated with other adverse outcomes including prolonged hospitalization, institutionalization, increased cost, and mortality. These outcomes occurred independent of age, prior cognitive functioning, and comorbidities.[2] Guidelines recommend that management of inpatient delirium should be focused on addressing the underlying etiology and managed with nonpharmacological interventions whenever possible.[3, 4, 5] However, implementing these recommendations can prove to be very challenging in hospital settings. Providers frequently have to resort to medical therapies, including antipsychotics (APs). Although these medications are commonly used to treat delirium in elderly patients, there is limited evidence to support their efficacy, and there are currently no proven pharmacological alternatives to these medications.[6] Furthermore, previous studies have demonstrated an increased risk of stroke, infection, cognitive impairment, and mortality in elders with dementia who receive long‐term AP therapy.[7, 8, 9] Yet as many as 48% of hospitalized elders who were newly started on APs had these drugs continued at time of discharge.[10]

There have been few studies describing the long‐term outcomes of elderly patient who are started on APs in the hospital. Most information on outcomes comes from patients with dementia. Therefore, we studied the 1‐year outcomes of a cohort of patients with and without dementia who were started on APs in the hospital and then discharged on these medications. In this cohort, we aimed to describe the number of readmissions, reasons for readmissions, duration of AP therapy, use of other sedating medications such as anxiolytics, hypnotics, and antihistamines as well as the incidence of readmission and death 1 year after the index hospital discharge.

METHODS

We previously described a retrospective cohort of 300 elders (65 years old) admitted to a tertiary care hospital between October 1, 2012 and September 31, 2013 who were newly prescribed APs while hospitalized.[10] Of patients alive at the time of discharge (260), 56% (146 patients) were discharged on APs. Two investigators extracted these 148 patient charts independently to identify and quantify the number of readmissions to the index hospital. We then limited the sample to only the first readmission per patient following the index admission and extracted this readmission for each patient. We first determined if APs were present on the admission medication reconciliation. If APs were not present on admission, we examined whether they were resumed during the hospitalization using the electronic medication administration summary. If they were present on admission, we looked to see if they were discontinued during the readmission and if additional new APs were started during the hospitalizations. We documented the circumstances around APs use and identified patients who died during their hospitalizations. We identified delirium using the same terms that were described in our prior study on the same cohort of patients.[10] We determined if patients were delirious using a predetermined algorithm (Figure 1). Briefly, we first determined delirium was documented. We then examined whether there was a Confusion Assessment Method (CAM) instrument included in the record. If a CAM instrument was not documented, we then looked for documentation using specific terms (eg, disorientation, confusions). We identified patients with dementia by determining whether dementia was documented along with other admission medical comorbidities. If it was not, we determined whether dementia was newly diagnosed during the hospital stay using progress notes or consultation notes. We did not objectively define criteria for diagnosis of dementia. We used the National Death Index (NDI) to determine mortality for all patients 1 year after discharge from the index hospitalization. The NDI is a national database of death records maintained by the National Center for Health Statistics. It has shown consistently high sensitivity and specificity for detection of death.[11]

Figure 1
Methodology for defining delirium during chart review. Abbreviations: CAM, Confusion Assessment Method.

We used descriptive statistics (means, standard deviations, range, and percents as appropriate to the scale of measurement) to describe the patient sample. We then used multiple logistic regression to identify significant predictors of death within 1 year of discharge.[12] Univariate analysis was used to select candidates for the logistic model (t tests for continuous factors and 2 for discrete factors). All factors with a significance level <0.2 on univariate analysis were included in the logistic regression, in addition to age and sex (regardless of significance). A maximum likelihood procedure was used to calculate the regression coefficients for the logistic model. The likelihood ratio criterion was used to determine the significance of individual factors in the regression model.[13] Factors with a significance level of 0.15 or less were retained in the final model, in addition to age and sex.

RESULTS

The 260 patients discharged alive from their index admissions had a 1‐year mortality rate of 29% (75/260). Of the 146/260 patients discharged on APs, 60 (41%) patients experienced at least 1 readmission (mean = 2 readmissions per patient; range, 18, with 111 total readmissions for 60 patients) within 1 year from discharge (Figure 2). Most common diagnoses at the time of readmissions were related neurological and psychiatric disorders (14%), cardiovascular and circulation disorders (13%), renal injury and electrolyte disorders (11%), and infections (6%). Among patients with at least 1 readmission, the mean age was 81.3 (range, 65.599.7), 60% were male, and 45% were admitted from a skilled nursing facility or rehabilitation facility (Table 1). Median time to readmission was 43.5 days (range, 1343 days), and 79% were readmitted to a medical service. The remaining 20% were admitted to a surgical service. Inpatient mortality during first readmissions was 8% (5/60). At the time of first readmission, 39/60 (65%) of patients were still on the same APs on which they had been discharged, and the APs were continued during the hospitalization in 79% of the patients (61% quetiapine, 19% olanzapine, and 13% risperidone). About half of patients whose APs were discontinued prior to readmission received a new AP during their hospital stays (9/20; 45%). One patient had been started on quetiapine in the outpatient setting. No patients were found to have new benzodiazepines, nonbenzodiazepine hypnotic, or antihistamines on their admission medication list.

Demographic Data of the Patients Who Were Discharged on APs and Were Readmitted the First Time Within One Year From Discharge and Circumstances Surrounding APs Initiation During Readmission
Variables Value*
  • NOTE: Abbreviations: APs, antipsychotics; ECG, electrocardiogram; QTc, QT interval; SNF, skilled nursing facility. *N = 60; patients who were discharged on APs during index admission and were readmitted within 1 year from discharge (only first readmission was included). Denominator = 31; number of patients whom APs from index admission were continued during readmission. Denominator = 18; number of patients who were started on APs during readmission. Denominator = 17; number of patients ECG was performed prior to APs administration; ∥Denominator = 4; number of patients ECG was performed after APs administration. Denominator = 55; number of patients who were alive during readmission

Age, mean (range), yr 81.3 (65.599.7)
Gender, no. (%)
Male 36 (60)
Female 24 (40)
Admitted from, no. (%)
Home 33 (55)
Rehabilitation facilities 5 (8)
SNF 22 (37)
Services, no. (%)
Medicine 48 (80)
Surgery 12 (20)
Types of APs continued on readmission (from index admission), no. (%)
Quetiapine 19 (61)
Olanzapine 6 (19)
Risperidone 4 (13)
Haloperidol 2 (7)
Types of APs started during readmission, no. (%)
Quetiapine 7 (39)
Risperidone 2 (11)
Haloperidol 16 (89)
Indications for AP use, no. (%)
Delirium 14 (77)
Undocumented 3 (17)
Other 1 (6)
ECG, no. (%)
Prior to APs administration 17 (94)
After APs administration 4 (22)
QTc prolongation >500 ms, no. (%)
Prior to APs administration 3 (18)
After APs administration∥ 2 (50)
Discharge destination, no. (%)
Home 23 (38)
Rehabilitation facilities 4 (7)
SNF 28 (47)
Death 5 (8)
Figure 2
Flowchart showing the outcomes of patients who were discharged on antipsychotics during index hospitalization and readmitted within 1 year from discharge. Abbreviations: APs, antipsychotics.

Eighteen patients received 1 or more new APs during the readmission hospitalizations. These included haloperidol (89%) and quetiapine (39%). Delirium was the main reported indication for starting APs (78%), but in 17% of cases no indication was documented. An electrocardiogram (ECG) was performed in 94% prior to APs administration and for 22% after APs administration. Corrected QT interval (QTc) of >500 ms was present in 18% of patients in pretreatment ECG and 50% of patients in post‐AP ECG. Of patients who survived readmission, 58% (32/55) were discharged to postacute facilities. Of the 39 patients who were on the same APs from index admission, 27 (69%) patients were eventually discharged on the same APs or new APs started during the readmission.

In the multivariable model (Table 2), predictors of death at 1 year included discharge to postacute facilities after index admission (odds ratio [OR]: 2.28; 95% confidence interval [CI]: 1.10‐4.73, P = 0.03) and QTc prolongation >500 ms during index admission (OR: 3.41; 95% CI: 1.34‐8.67, P = 0.01). Age and gender were not associated with 1‐year mortality.

Multivariable Analysis of One‐Year Mortality From Index Admission in Patients Who Were Started on Antipsychotics.
Odds Ratio 95% Confidence Interval P Value
  • NOTE: Abbreviations: AP, antipsychotic; QTc, QT interval. *For subset of patients for whom electrocardiogram was done.

Age 1.03 0.991.06 0.13
Male sex 0.87 0.501.52 0.63
Risperdal 3.53 0.6419.40 0.15
QTc prolongation after AP administration* 3.41 1.348.67 0.01
Presence of geriatric psychiatry consult 0.30 0.091.04 0.06
Discharged to postacute facilities vs home 2.28 1.104.73 0.03

DISCUSSION

In a cohort of elderly patients who were discharged on APs, nearly one‐third (29%) died within 1 year of the hospitalization in which APs were initiated. Nearly half of the survivors from the index admission (41%) experienced at least 1 admission within 1 year from discharge. Of readmitted patients, two‐thirds were taking the same APs that had been started during the index hospitalization. Half of the patients not on APs on readmission were started on an AP during the hospitalization, most often because they became delirious on return to the acute care setting. Compared to patients discharged home after an index admission, patients who were discharged to postacute facilities were almost 4 times as likely to die during the year subsequent to the admission. These data suggest that once patients are started on APs, most are continued on them until the next admission or are restarted during that readmission. Moreover, hospitalized elders who require an AP are at high risk for mortality in the coming year.

Prior studies have reported that patients with delirium have elevated 1‐year mortality rates.[14, 15, 16, 17, 18, 19] A secondary analysis of the Delirium Prevention Trial, which included 437 hospitalized older patients, revealed a 1‐year mortality rate of 20% in those who were never delirious during hospitalization, compared to 26% to 38% in patients with delirium.[19] Additionally, 1‐year mortality in hospitalized older patients with delirium (36%) was shown to be higher than patients with dementia (29%) or depression (26%).[17] Unlike these studies, not all of the patients in our study had documented delirium, but all received an AP. Still, it is notable that the 1‐year mortality rate for delirium in general is similar to what we found in this study.

The literature has also reported that long‐term AP use is associated with excess mortality in elder patients, especially those with dementia.[20, 21, 22] In a retrospective cohort study, older patients with dementia who were taking antipsychotics had significantly higher 1‐year mortality rates (23%29%) than patients not taking antipsychotic medications (15%). In a large Canadian propensity score‐matched cohort study that included over 13,000 demented older adults, the mortality was higher in the community‐dwelling elders who received atypical APs compared to no APs, with a difference of 1.1% in 180‐day mortality rate after initiation of APs.[21] The absolute mortality rate was 2.6% higher in patients who received typical compared to atypical APs. Unlike these studies, not every patient in our cohort had a diagnosis of dementia, but again, mortality rates in these studies appear similar to our cohort.

In contrast, other observational studies have not found an increased risk associated with receipt of APs. For example, a prospective study that enrolled approximately 950 patients with probable dementia showed that AP use was not associated with time to death after adjustment for comorbidities, demographic and cognitive variables.[23] These conflicting results highlight the difficulties of attributing outcomes in high‐risk populations. Although the excess mortality observed in patients taking APs may be related to the risks of APs, it is quite possible that patients who require APs (most often for delirium or agitated dementia) are at higher risk of death. This confounding by indication may be nearly impossible to adjust for retrospectively, even using techniques such as propensity matching.

Our report adds to the literature; we know of no studies to date describing a cohort of patients, most with delirium, who were started on APs in the hospital. We also attempted to identify the reasons that patients were started on APs, which have been infrequently reported. As noted above, our 1‐year mortality rate of 29% among older patients prescribed APs in the hospital was quite similar to mortality rates both for patients with delirium who were not necessarily treated with APs and patients with dementia who were treated with APs. This finding further supports the argument that risk factors for mortality, including dementia, delirium, and AP use are very difficult to tease apart. It is possible that the reasons that APs are prescribed (agitated delirium or dementia) have as much to do with the excess mortality reported in observational studies of APs as the use of APs themselves.

The high rate of continued AP use we observed (two‐thirds of readmitted patients) may reflect limited pharmacological alternatives to these medications with little evidence to support treating the symptoms of delirium with other drug classes, along with suboptimal environmental and behavioral modifications in postacute facilities and hospitals. This is unfortunate given that delirium is often preventable. Systematic implementation of well‐documented strategies to decrease delirium in hospitals and postacute facilities would likely reduce the prescription of APs and has the potential to slow the decline in this vulnerable population. A meta‐analysis incorporating both randomized and nonrandomized trials of medical and surgical patients showed that multicomponent nonpharmacologic interventions decreased delirium by 50%.[24] Thus, simple interventions such as reorientation, early mobilization, optimizing vision and hearing, sleepwake cycle preservation, and hydration might avoid roughly 1 million cases of delirium in hospitalized older adults annually.[24] The Hospital Elder Life Program and Acute Care for Elders units are examples of programs that have been shown to decrease the incidence of delirium.[25, 26]

Despite vigorous efforts to prevent delirium, a subgroup of patients still will become delirious. These patients are at high risk for death. Our mortality prediction model revealed that patients who were discharged to postacute facilities were 4 times more likely to die during the subsequent year compared to patients who were discharged home. Patients discharged to postacute facilities are likely to have a higher burden of disease, greater functional and cognitive impairment, and more frailty than those who are able to return to the community. Very ill and/or frail patients receiving APs in the hospital and requiring APs on discharge to postacute care facilities have limited survival and may benefit from expedited palliative care interventions to clarify prognosis and goals, and relieve suffering. At a minimum, our study identifies a need for further study to identify this very high‐risk group of elders. It is notable that 50% of patients were found to have a post‐treatment ECG with a QTc of >500 ms, a finding that has not been previously described. This would put these patients at higher risk of mortality, and as such we suggest that current guidelines should continue to emphasize the importance of post‐treatment ECGs and set clear criteria for discontinuation in elderly patients.

Our study is limited by its retrospective, single‐center design and small sample size, therefore limiting the interpretation and generalizability of the results to other hospitals. Quetiapine was the most common AP medication used in our hospital; therefore, our findings cannot be generalized to hospitals that utilize other AP agents. Future studies should examine antipsychotic use across hospitals to determine variation in prescribing patterns and outcomes. Nevertheless, the care of these patients were transitioned to a large number of geriatricians and primary care and nursing home physicians after discharge, and the reflected practice patterns extended beyond our hospital. Additionally, we were unable to determine when and why APs were discontinued or started in the outpatient setting. We were only able to detect readmissions to the 3 hospitals within our health system and therefore may have missed some readmissions to other institutions, although the majority of patients in our region tend to return to the same hospital. For patients who were not readmitted, we were also unable to identify whether they remained on the APs initiated during their index hospitalizations. Any retrospective study is limited by the difficulty of distinguishing delirium from the behavioral and psychiatric symptoms of dementia, but we identified delirium using standard terms described in previous literature.[10] We were unable to determine the types of delirium (hyperactive vs hypoactive) given that the documentations on behavioral symptoms were largely missing from the charts. The number of patients with preexisting diagnosis of dementia was likely underestimated, as we were only able to verify the diagnosis from the medical history. Additionally, the retrospective design based on chart review limited the factors that we could detect and grade accurately for inclusion in our mortality prediction model. Of note, our model did not contain objective measures of cognition, agitation, function, and markers for frailty such as walking speed, weak grip strength, weight loss, and low physical activity.

CONCLUSION

Initiating an AP (eg, haloperidol, quetiapine, olanzapine, and risperidone) in the hospital is likely to result in long‐term use of these medications despite the fact that AP use has been associated with multiple risks including falls, fractures, stroke, cardiovascular disease, and increased mortality in those with underlying dementia.[27] When possible, behavioral interventions to prevent delirium and slow the trajectory of decline should be implemented to reduce AP use. If patients with delirium are started on antipsychotics, it is important to monitor for prolonged QTc given the associated risk of mortality. In a subgroup of patients at high risk for death in the upcoming year, occurrence of delirium or use of APs during a hospitalization should both be considered triggers for early advance care planning and possibly palliative care and end‐of‐life discussions, with an emphasis on quality of life.

Disclosures: The research was supported by the Department of Medicine, Baystate Medical Center/Tufts University School of Medicine. Dr. Lagu is supported by the National Heart, Lung, and Blood Institute of the National Institutes of Health under award number K01HL114745. Drs. Lagu and Loh had full access to all of the data in the study. They take responsibility for the integrity of the data and the accuracy of the analysis. Drs. Loh, Brennan, Lindenauer, and Lagu conceived of the study. Drs. Loh, Ramdass, and Ms. Garb acquired the data. Ms Garb analyzed and interpreted the data. Drs. Loh, Ramdass, and Thim drafted the manuscript. Drs. Brennan, Lindenauer, and Lagu and Ms. Garb critically reviewed the manuscript for important intellectual content. Dr. Lagu has received consulting fees from the Institute for Healthcare Improvement, under contract to the Centers for Medicare and Medicaid Services, for her work on a project to help health systems achieve disability competence. Dr. Brennan is supported by a Geriatric Work Force Enhancement Grant from the US Department of Health and Human Services award number 1 U1QHP287020100. The authors report no conflicts of interest.

Files
References
  1. Inouye SK, Westendorp RGJ, Saczynski JS. Delirium in elderly people. Lancet. 2014;383:911922.
  2. Fong TG, Jones RN, Marcantonio ER, et al. Adverse outcomes after hospitalization and delirium in persons with Alzheimer disease. Ann Intern Med. 2012;156:848856, W296.
  3. American Geriatrics Society Expert Panel on Postoperative Delirium in Older Adults. American Geriatrics Society abstracted clinical practice guideline for postoperative delirium in older adults. J Am Geriatr Soc. 2015;63:142150.
  4. Practice guideline for the treatment of patients with delirium. American Psychiatric Association. Am J Psychiatry. 1999;156:120.
  5. Potter J, George J; Guideline Development Group. The prevention, diagnosis and management of delirium in older people: concise guidelines. Clin Med (Lond). 2006;6:303308.
  6. Seitz DP, Gill SS, Zyl LT. Antipsychotics in the treatment of delirium: a systematic review. J Clin Psychiatry. 2007;68:1121.
  7. Maust DT, Kim HM, Seyfried LS, et al. Antipsychotics, other psychotropics, and the risk of death in patients with dementia: number needed to harm. JAMA Psychiatry. 2015;72:438445.
  8. Andrade C, Radhakrishnan R. Safety and efficacy of antipsychotic drugs for the behavioral and psychological symptoms of dementia. Indian J Psychiatry. 2009;51(suppl 1):S87S92.
  9. Gareri P, Fazio P, Manfredi VGL, Sarro G. Use and safety of antipsychotics in behavioral disorders in elderly people with dementia. J Clin Psychopharmacol. 2014;34:109123.
  10. Loh KP, Ramdass S, Garb JL, Brennan MJ, Lindenauer PK, Lagu T. From hospital to community: use of antipsychotics in hospitalized elders. J Hosp Med. 2014;9:802804.
  11. Sesso HD, Paffenbarger RS, Lee I‐M. Comparison of National Death Index and World Wide Web Death Searches. Am J Epidemiol. 2000;152:107111.
  12. Cox DR. Analysis of Binary Data. London, United Kingdom: Methuen; 1970:7699.
  13. Lee ET. Statistical Methods for Survival Data Analysis. New York, NY: John Wiley 1992:233236.
  14. Evans SJ, Sayers M, Mitnitski A, Rockwood K. The risk of adverse outcomes in hospitalized older patients in relation to a frailty index based on a comprehensive geriatric assessment. Age Ageing. 2014;43:127132.
  15. Grover S, Ghormode D, Ghosh A, et al. Risk factors for delirium and inpatient mortality with delirium. J Postgrad Med. 2013;59:263270.
  16. Avelino‐Silva TJ, Farfel JM, Curiati JAE, Amaral JRG, Campora F, Jacob‐Filho W. Comprehensive geriatric assessment predicts mortality and adverse outcomes in hospitalized older adults. BMC Geriatr. 2014;14:129.
  17. Tsai M‐C, Weng H‐H, Chou S‐Y, Tsai C‐S, Hung T‐H, Su J‐A. One‐year mortality of elderly inpatients with delirium, dementia, or depression seen by a consultation‐liaison service. Psychosomatics. 2012;53:433438.
  18. Hemert AM, Mast RC, Hengeveld MW, Vorstenbosch M. Excess mortality in general hospital patients with delirium: a 5‐year follow‐up of 519 patients seen in psychiatric consultation. J Psychosom Res. 1994;38:339346.
  19. McAvay GJ, Ness PH, Bogardus ST, et al. Older adults discharged from the hospital with delirium: 1‐year outcomes. J Am Geriatr Soc. 2006;54:12451250.
  20. Ballard C, Hanney ML, Theodoulou M, et al. The dementia antipsychotic withdrawal trial (DART‐AD): long‐term follow‐up of a randomised placebo‐controlled trial. Lancet Neurol. 2009;8:151157.
  21. Gill SS, Bronskill SE, Normand S‐LT, et al. Antipsychotic drug use and mortality in older adults with dementia. Ann Intern Med. 2007;146:775786.
  22. Wang PS, Schneeweiss S, Avorn J, et al. Risk of death in elderly users of conventional vs. atypical antipsychotic medications. N Engl J Med. 2005;353:23352341.
  23. Lopez OL, Becker JT, Chang Y‐F, et al. The long‐term effects of conventional and atypical antipsychotics in patients with probable Alzheimer's disease. Am J Psychiatry. 2013;170:10511058.
  24. Hshieh TT, Yue J, Oh E, et al. Effectiveness of multicomponent nonpharmacological delirium interventions: a meta‐analysis. JAMA Intern Med. 2015;175:512520.
  25. Rubin FH, Neal K, Fenlon K, Hassan S, Inouye SK. Sustainability and scalability of the hospital elder life program at a community hospital. J Am Geriatr Soc. 2011;59:359365.
  26. Fox MT, Persaud M, Maimets I, et al. Effectiveness of acute geriatric unit care using acute care for elders components: a systematic review and meta‐analysis. J Am Geriatr Soc. 2012;60:22372245.
  27. Muench J, Hamer AM. Adverse effects of antipsychotic medications. Am Fam Physician. 2010;81:617622.
Article PDF
Issue
Journal of Hospital Medicine - 11(8)
Publications
Page Number
550-555
Sections
Files
Files
Article PDF
Article PDF

Delirium, a clinical syndrome characterized by inattention and acute cognitive dysfunction, is very common in older hospitalized patients, with a reported incidence of 18% to 35% at time of admission and overall occurrence rates of 29% to 64%.[1] Previous studies have reported that a diagnosis of delirium is not benign and is associated with other adverse outcomes including prolonged hospitalization, institutionalization, increased cost, and mortality. These outcomes occurred independent of age, prior cognitive functioning, and comorbidities.[2] Guidelines recommend that management of inpatient delirium should be focused on addressing the underlying etiology and managed with nonpharmacological interventions whenever possible.[3, 4, 5] However, implementing these recommendations can prove to be very challenging in hospital settings. Providers frequently have to resort to medical therapies, including antipsychotics (APs). Although these medications are commonly used to treat delirium in elderly patients, there is limited evidence to support their efficacy, and there are currently no proven pharmacological alternatives to these medications.[6] Furthermore, previous studies have demonstrated an increased risk of stroke, infection, cognitive impairment, and mortality in elders with dementia who receive long‐term AP therapy.[7, 8, 9] Yet as many as 48% of hospitalized elders who were newly started on APs had these drugs continued at time of discharge.[10]

There have been few studies describing the long‐term outcomes of elderly patient who are started on APs in the hospital. Most information on outcomes comes from patients with dementia. Therefore, we studied the 1‐year outcomes of a cohort of patients with and without dementia who were started on APs in the hospital and then discharged on these medications. In this cohort, we aimed to describe the number of readmissions, reasons for readmissions, duration of AP therapy, use of other sedating medications such as anxiolytics, hypnotics, and antihistamines as well as the incidence of readmission and death 1 year after the index hospital discharge.

METHODS

We previously described a retrospective cohort of 300 elders (65 years old) admitted to a tertiary care hospital between October 1, 2012 and September 31, 2013 who were newly prescribed APs while hospitalized.[10] Of patients alive at the time of discharge (260), 56% (146 patients) were discharged on APs. Two investigators extracted these 148 patient charts independently to identify and quantify the number of readmissions to the index hospital. We then limited the sample to only the first readmission per patient following the index admission and extracted this readmission for each patient. We first determined if APs were present on the admission medication reconciliation. If APs were not present on admission, we examined whether they were resumed during the hospitalization using the electronic medication administration summary. If they were present on admission, we looked to see if they were discontinued during the readmission and if additional new APs were started during the hospitalizations. We documented the circumstances around APs use and identified patients who died during their hospitalizations. We identified delirium using the same terms that were described in our prior study on the same cohort of patients.[10] We determined if patients were delirious using a predetermined algorithm (Figure 1). Briefly, we first determined delirium was documented. We then examined whether there was a Confusion Assessment Method (CAM) instrument included in the record. If a CAM instrument was not documented, we then looked for documentation using specific terms (eg, disorientation, confusions). We identified patients with dementia by determining whether dementia was documented along with other admission medical comorbidities. If it was not, we determined whether dementia was newly diagnosed during the hospital stay using progress notes or consultation notes. We did not objectively define criteria for diagnosis of dementia. We used the National Death Index (NDI) to determine mortality for all patients 1 year after discharge from the index hospitalization. The NDI is a national database of death records maintained by the National Center for Health Statistics. It has shown consistently high sensitivity and specificity for detection of death.[11]

Figure 1
Methodology for defining delirium during chart review. Abbreviations: CAM, Confusion Assessment Method.

We used descriptive statistics (means, standard deviations, range, and percents as appropriate to the scale of measurement) to describe the patient sample. We then used multiple logistic regression to identify significant predictors of death within 1 year of discharge.[12] Univariate analysis was used to select candidates for the logistic model (t tests for continuous factors and 2 for discrete factors). All factors with a significance level <0.2 on univariate analysis were included in the logistic regression, in addition to age and sex (regardless of significance). A maximum likelihood procedure was used to calculate the regression coefficients for the logistic model. The likelihood ratio criterion was used to determine the significance of individual factors in the regression model.[13] Factors with a significance level of 0.15 or less were retained in the final model, in addition to age and sex.

RESULTS

The 260 patients discharged alive from their index admissions had a 1‐year mortality rate of 29% (75/260). Of the 146/260 patients discharged on APs, 60 (41%) patients experienced at least 1 readmission (mean = 2 readmissions per patient; range, 18, with 111 total readmissions for 60 patients) within 1 year from discharge (Figure 2). Most common diagnoses at the time of readmissions were related neurological and psychiatric disorders (14%), cardiovascular and circulation disorders (13%), renal injury and electrolyte disorders (11%), and infections (6%). Among patients with at least 1 readmission, the mean age was 81.3 (range, 65.599.7), 60% were male, and 45% were admitted from a skilled nursing facility or rehabilitation facility (Table 1). Median time to readmission was 43.5 days (range, 1343 days), and 79% were readmitted to a medical service. The remaining 20% were admitted to a surgical service. Inpatient mortality during first readmissions was 8% (5/60). At the time of first readmission, 39/60 (65%) of patients were still on the same APs on which they had been discharged, and the APs were continued during the hospitalization in 79% of the patients (61% quetiapine, 19% olanzapine, and 13% risperidone). About half of patients whose APs were discontinued prior to readmission received a new AP during their hospital stays (9/20; 45%). One patient had been started on quetiapine in the outpatient setting. No patients were found to have new benzodiazepines, nonbenzodiazepine hypnotic, or antihistamines on their admission medication list.

Demographic Data of the Patients Who Were Discharged on APs and Were Readmitted the First Time Within One Year From Discharge and Circumstances Surrounding APs Initiation During Readmission
Variables Value*
  • NOTE: Abbreviations: APs, antipsychotics; ECG, electrocardiogram; QTc, QT interval; SNF, skilled nursing facility. *N = 60; patients who were discharged on APs during index admission and were readmitted within 1 year from discharge (only first readmission was included). Denominator = 31; number of patients whom APs from index admission were continued during readmission. Denominator = 18; number of patients who were started on APs during readmission. Denominator = 17; number of patients ECG was performed prior to APs administration; ∥Denominator = 4; number of patients ECG was performed after APs administration. Denominator = 55; number of patients who were alive during readmission

Age, mean (range), yr 81.3 (65.599.7)
Gender, no. (%)
Male 36 (60)
Female 24 (40)
Admitted from, no. (%)
Home 33 (55)
Rehabilitation facilities 5 (8)
SNF 22 (37)
Services, no. (%)
Medicine 48 (80)
Surgery 12 (20)
Types of APs continued on readmission (from index admission), no. (%)
Quetiapine 19 (61)
Olanzapine 6 (19)
Risperidone 4 (13)
Haloperidol 2 (7)
Types of APs started during readmission, no. (%)
Quetiapine 7 (39)
Risperidone 2 (11)
Haloperidol 16 (89)
Indications for AP use, no. (%)
Delirium 14 (77)
Undocumented 3 (17)
Other 1 (6)
ECG, no. (%)
Prior to APs administration 17 (94)
After APs administration 4 (22)
QTc prolongation >500 ms, no. (%)
Prior to APs administration 3 (18)
After APs administration∥ 2 (50)
Discharge destination, no. (%)
Home 23 (38)
Rehabilitation facilities 4 (7)
SNF 28 (47)
Death 5 (8)
Figure 2
Flowchart showing the outcomes of patients who were discharged on antipsychotics during index hospitalization and readmitted within 1 year from discharge. Abbreviations: APs, antipsychotics.

Eighteen patients received 1 or more new APs during the readmission hospitalizations. These included haloperidol (89%) and quetiapine (39%). Delirium was the main reported indication for starting APs (78%), but in 17% of cases no indication was documented. An electrocardiogram (ECG) was performed in 94% prior to APs administration and for 22% after APs administration. Corrected QT interval (QTc) of >500 ms was present in 18% of patients in pretreatment ECG and 50% of patients in post‐AP ECG. Of patients who survived readmission, 58% (32/55) were discharged to postacute facilities. Of the 39 patients who were on the same APs from index admission, 27 (69%) patients were eventually discharged on the same APs or new APs started during the readmission.

In the multivariable model (Table 2), predictors of death at 1 year included discharge to postacute facilities after index admission (odds ratio [OR]: 2.28; 95% confidence interval [CI]: 1.10‐4.73, P = 0.03) and QTc prolongation >500 ms during index admission (OR: 3.41; 95% CI: 1.34‐8.67, P = 0.01). Age and gender were not associated with 1‐year mortality.

Multivariable Analysis of One‐Year Mortality From Index Admission in Patients Who Were Started on Antipsychotics.
Odds Ratio 95% Confidence Interval P Value
  • NOTE: Abbreviations: AP, antipsychotic; QTc, QT interval. *For subset of patients for whom electrocardiogram was done.

Age 1.03 0.991.06 0.13
Male sex 0.87 0.501.52 0.63
Risperdal 3.53 0.6419.40 0.15
QTc prolongation after AP administration* 3.41 1.348.67 0.01
Presence of geriatric psychiatry consult 0.30 0.091.04 0.06
Discharged to postacute facilities vs home 2.28 1.104.73 0.03

DISCUSSION

In a cohort of elderly patients who were discharged on APs, nearly one‐third (29%) died within 1 year of the hospitalization in which APs were initiated. Nearly half of the survivors from the index admission (41%) experienced at least 1 admission within 1 year from discharge. Of readmitted patients, two‐thirds were taking the same APs that had been started during the index hospitalization. Half of the patients not on APs on readmission were started on an AP during the hospitalization, most often because they became delirious on return to the acute care setting. Compared to patients discharged home after an index admission, patients who were discharged to postacute facilities were almost 4 times as likely to die during the year subsequent to the admission. These data suggest that once patients are started on APs, most are continued on them until the next admission or are restarted during that readmission. Moreover, hospitalized elders who require an AP are at high risk for mortality in the coming year.

Prior studies have reported that patients with delirium have elevated 1‐year mortality rates.[14, 15, 16, 17, 18, 19] A secondary analysis of the Delirium Prevention Trial, which included 437 hospitalized older patients, revealed a 1‐year mortality rate of 20% in those who were never delirious during hospitalization, compared to 26% to 38% in patients with delirium.[19] Additionally, 1‐year mortality in hospitalized older patients with delirium (36%) was shown to be higher than patients with dementia (29%) or depression (26%).[17] Unlike these studies, not all of the patients in our study had documented delirium, but all received an AP. Still, it is notable that the 1‐year mortality rate for delirium in general is similar to what we found in this study.

The literature has also reported that long‐term AP use is associated with excess mortality in elder patients, especially those with dementia.[20, 21, 22] In a retrospective cohort study, older patients with dementia who were taking antipsychotics had significantly higher 1‐year mortality rates (23%29%) than patients not taking antipsychotic medications (15%). In a large Canadian propensity score‐matched cohort study that included over 13,000 demented older adults, the mortality was higher in the community‐dwelling elders who received atypical APs compared to no APs, with a difference of 1.1% in 180‐day mortality rate after initiation of APs.[21] The absolute mortality rate was 2.6% higher in patients who received typical compared to atypical APs. Unlike these studies, not every patient in our cohort had a diagnosis of dementia, but again, mortality rates in these studies appear similar to our cohort.

In contrast, other observational studies have not found an increased risk associated with receipt of APs. For example, a prospective study that enrolled approximately 950 patients with probable dementia showed that AP use was not associated with time to death after adjustment for comorbidities, demographic and cognitive variables.[23] These conflicting results highlight the difficulties of attributing outcomes in high‐risk populations. Although the excess mortality observed in patients taking APs may be related to the risks of APs, it is quite possible that patients who require APs (most often for delirium or agitated dementia) are at higher risk of death. This confounding by indication may be nearly impossible to adjust for retrospectively, even using techniques such as propensity matching.

Our report adds to the literature; we know of no studies to date describing a cohort of patients, most with delirium, who were started on APs in the hospital. We also attempted to identify the reasons that patients were started on APs, which have been infrequently reported. As noted above, our 1‐year mortality rate of 29% among older patients prescribed APs in the hospital was quite similar to mortality rates both for patients with delirium who were not necessarily treated with APs and patients with dementia who were treated with APs. This finding further supports the argument that risk factors for mortality, including dementia, delirium, and AP use are very difficult to tease apart. It is possible that the reasons that APs are prescribed (agitated delirium or dementia) have as much to do with the excess mortality reported in observational studies of APs as the use of APs themselves.

The high rate of continued AP use we observed (two‐thirds of readmitted patients) may reflect limited pharmacological alternatives to these medications with little evidence to support treating the symptoms of delirium with other drug classes, along with suboptimal environmental and behavioral modifications in postacute facilities and hospitals. This is unfortunate given that delirium is often preventable. Systematic implementation of well‐documented strategies to decrease delirium in hospitals and postacute facilities would likely reduce the prescription of APs and has the potential to slow the decline in this vulnerable population. A meta‐analysis incorporating both randomized and nonrandomized trials of medical and surgical patients showed that multicomponent nonpharmacologic interventions decreased delirium by 50%.[24] Thus, simple interventions such as reorientation, early mobilization, optimizing vision and hearing, sleepwake cycle preservation, and hydration might avoid roughly 1 million cases of delirium in hospitalized older adults annually.[24] The Hospital Elder Life Program and Acute Care for Elders units are examples of programs that have been shown to decrease the incidence of delirium.[25, 26]

Despite vigorous efforts to prevent delirium, a subgroup of patients still will become delirious. These patients are at high risk for death. Our mortality prediction model revealed that patients who were discharged to postacute facilities were 4 times more likely to die during the subsequent year compared to patients who were discharged home. Patients discharged to postacute facilities are likely to have a higher burden of disease, greater functional and cognitive impairment, and more frailty than those who are able to return to the community. Very ill and/or frail patients receiving APs in the hospital and requiring APs on discharge to postacute care facilities have limited survival and may benefit from expedited palliative care interventions to clarify prognosis and goals, and relieve suffering. At a minimum, our study identifies a need for further study to identify this very high‐risk group of elders. It is notable that 50% of patients were found to have a post‐treatment ECG with a QTc of >500 ms, a finding that has not been previously described. This would put these patients at higher risk of mortality, and as such we suggest that current guidelines should continue to emphasize the importance of post‐treatment ECGs and set clear criteria for discontinuation in elderly patients.

Our study is limited by its retrospective, single‐center design and small sample size, therefore limiting the interpretation and generalizability of the results to other hospitals. Quetiapine was the most common AP medication used in our hospital; therefore, our findings cannot be generalized to hospitals that utilize other AP agents. Future studies should examine antipsychotic use across hospitals to determine variation in prescribing patterns and outcomes. Nevertheless, the care of these patients were transitioned to a large number of geriatricians and primary care and nursing home physicians after discharge, and the reflected practice patterns extended beyond our hospital. Additionally, we were unable to determine when and why APs were discontinued or started in the outpatient setting. We were only able to detect readmissions to the 3 hospitals within our health system and therefore may have missed some readmissions to other institutions, although the majority of patients in our region tend to return to the same hospital. For patients who were not readmitted, we were also unable to identify whether they remained on the APs initiated during their index hospitalizations. Any retrospective study is limited by the difficulty of distinguishing delirium from the behavioral and psychiatric symptoms of dementia, but we identified delirium using standard terms described in previous literature.[10] We were unable to determine the types of delirium (hyperactive vs hypoactive) given that the documentations on behavioral symptoms were largely missing from the charts. The number of patients with preexisting diagnosis of dementia was likely underestimated, as we were only able to verify the diagnosis from the medical history. Additionally, the retrospective design based on chart review limited the factors that we could detect and grade accurately for inclusion in our mortality prediction model. Of note, our model did not contain objective measures of cognition, agitation, function, and markers for frailty such as walking speed, weak grip strength, weight loss, and low physical activity.

CONCLUSION

Initiating an AP (eg, haloperidol, quetiapine, olanzapine, and risperidone) in the hospital is likely to result in long‐term use of these medications despite the fact that AP use has been associated with multiple risks including falls, fractures, stroke, cardiovascular disease, and increased mortality in those with underlying dementia.[27] When possible, behavioral interventions to prevent delirium and slow the trajectory of decline should be implemented to reduce AP use. If patients with delirium are started on antipsychotics, it is important to monitor for prolonged QTc given the associated risk of mortality. In a subgroup of patients at high risk for death in the upcoming year, occurrence of delirium or use of APs during a hospitalization should both be considered triggers for early advance care planning and possibly palliative care and end‐of‐life discussions, with an emphasis on quality of life.

Disclosures: The research was supported by the Department of Medicine, Baystate Medical Center/Tufts University School of Medicine. Dr. Lagu is supported by the National Heart, Lung, and Blood Institute of the National Institutes of Health under award number K01HL114745. Drs. Lagu and Loh had full access to all of the data in the study. They take responsibility for the integrity of the data and the accuracy of the analysis. Drs. Loh, Brennan, Lindenauer, and Lagu conceived of the study. Drs. Loh, Ramdass, and Ms. Garb acquired the data. Ms Garb analyzed and interpreted the data. Drs. Loh, Ramdass, and Thim drafted the manuscript. Drs. Brennan, Lindenauer, and Lagu and Ms. Garb critically reviewed the manuscript for important intellectual content. Dr. Lagu has received consulting fees from the Institute for Healthcare Improvement, under contract to the Centers for Medicare and Medicaid Services, for her work on a project to help health systems achieve disability competence. Dr. Brennan is supported by a Geriatric Work Force Enhancement Grant from the US Department of Health and Human Services award number 1 U1QHP287020100. The authors report no conflicts of interest.

Delirium, a clinical syndrome characterized by inattention and acute cognitive dysfunction, is very common in older hospitalized patients, with a reported incidence of 18% to 35% at time of admission and overall occurrence rates of 29% to 64%.[1] Previous studies have reported that a diagnosis of delirium is not benign and is associated with other adverse outcomes including prolonged hospitalization, institutionalization, increased cost, and mortality. These outcomes occurred independent of age, prior cognitive functioning, and comorbidities.[2] Guidelines recommend that management of inpatient delirium should be focused on addressing the underlying etiology and managed with nonpharmacological interventions whenever possible.[3, 4, 5] However, implementing these recommendations can prove to be very challenging in hospital settings. Providers frequently have to resort to medical therapies, including antipsychotics (APs). Although these medications are commonly used to treat delirium in elderly patients, there is limited evidence to support their efficacy, and there are currently no proven pharmacological alternatives to these medications.[6] Furthermore, previous studies have demonstrated an increased risk of stroke, infection, cognitive impairment, and mortality in elders with dementia who receive long‐term AP therapy.[7, 8, 9] Yet as many as 48% of hospitalized elders who were newly started on APs had these drugs continued at time of discharge.[10]

There have been few studies describing the long‐term outcomes of elderly patient who are started on APs in the hospital. Most information on outcomes comes from patients with dementia. Therefore, we studied the 1‐year outcomes of a cohort of patients with and without dementia who were started on APs in the hospital and then discharged on these medications. In this cohort, we aimed to describe the number of readmissions, reasons for readmissions, duration of AP therapy, use of other sedating medications such as anxiolytics, hypnotics, and antihistamines as well as the incidence of readmission and death 1 year after the index hospital discharge.

METHODS

We previously described a retrospective cohort of 300 elders (65 years old) admitted to a tertiary care hospital between October 1, 2012 and September 31, 2013 who were newly prescribed APs while hospitalized.[10] Of patients alive at the time of discharge (260), 56% (146 patients) were discharged on APs. Two investigators extracted these 148 patient charts independently to identify and quantify the number of readmissions to the index hospital. We then limited the sample to only the first readmission per patient following the index admission and extracted this readmission for each patient. We first determined if APs were present on the admission medication reconciliation. If APs were not present on admission, we examined whether they were resumed during the hospitalization using the electronic medication administration summary. If they were present on admission, we looked to see if they were discontinued during the readmission and if additional new APs were started during the hospitalizations. We documented the circumstances around APs use and identified patients who died during their hospitalizations. We identified delirium using the same terms that were described in our prior study on the same cohort of patients.[10] We determined if patients were delirious using a predetermined algorithm (Figure 1). Briefly, we first determined delirium was documented. We then examined whether there was a Confusion Assessment Method (CAM) instrument included in the record. If a CAM instrument was not documented, we then looked for documentation using specific terms (eg, disorientation, confusions). We identified patients with dementia by determining whether dementia was documented along with other admission medical comorbidities. If it was not, we determined whether dementia was newly diagnosed during the hospital stay using progress notes or consultation notes. We did not objectively define criteria for diagnosis of dementia. We used the National Death Index (NDI) to determine mortality for all patients 1 year after discharge from the index hospitalization. The NDI is a national database of death records maintained by the National Center for Health Statistics. It has shown consistently high sensitivity and specificity for detection of death.[11]

Figure 1
Methodology for defining delirium during chart review. Abbreviations: CAM, Confusion Assessment Method.

We used descriptive statistics (means, standard deviations, range, and percents as appropriate to the scale of measurement) to describe the patient sample. We then used multiple logistic regression to identify significant predictors of death within 1 year of discharge.[12] Univariate analysis was used to select candidates for the logistic model (t tests for continuous factors and 2 for discrete factors). All factors with a significance level <0.2 on univariate analysis were included in the logistic regression, in addition to age and sex (regardless of significance). A maximum likelihood procedure was used to calculate the regression coefficients for the logistic model. The likelihood ratio criterion was used to determine the significance of individual factors in the regression model.[13] Factors with a significance level of 0.15 or less were retained in the final model, in addition to age and sex.

RESULTS

The 260 patients discharged alive from their index admissions had a 1‐year mortality rate of 29% (75/260). Of the 146/260 patients discharged on APs, 60 (41%) patients experienced at least 1 readmission (mean = 2 readmissions per patient; range, 18, with 111 total readmissions for 60 patients) within 1 year from discharge (Figure 2). Most common diagnoses at the time of readmissions were related neurological and psychiatric disorders (14%), cardiovascular and circulation disorders (13%), renal injury and electrolyte disorders (11%), and infections (6%). Among patients with at least 1 readmission, the mean age was 81.3 (range, 65.599.7), 60% were male, and 45% were admitted from a skilled nursing facility or rehabilitation facility (Table 1). Median time to readmission was 43.5 days (range, 1343 days), and 79% were readmitted to a medical service. The remaining 20% were admitted to a surgical service. Inpatient mortality during first readmissions was 8% (5/60). At the time of first readmission, 39/60 (65%) of patients were still on the same APs on which they had been discharged, and the APs were continued during the hospitalization in 79% of the patients (61% quetiapine, 19% olanzapine, and 13% risperidone). About half of patients whose APs were discontinued prior to readmission received a new AP during their hospital stays (9/20; 45%). One patient had been started on quetiapine in the outpatient setting. No patients were found to have new benzodiazepines, nonbenzodiazepine hypnotic, or antihistamines on their admission medication list.

Demographic Data of the Patients Who Were Discharged on APs and Were Readmitted the First Time Within One Year From Discharge and Circumstances Surrounding APs Initiation During Readmission
Variables Value*
  • NOTE: Abbreviations: APs, antipsychotics; ECG, electrocardiogram; QTc, QT interval; SNF, skilled nursing facility. *N = 60; patients who were discharged on APs during index admission and were readmitted within 1 year from discharge (only first readmission was included). Denominator = 31; number of patients whom APs from index admission were continued during readmission. Denominator = 18; number of patients who were started on APs during readmission. Denominator = 17; number of patients ECG was performed prior to APs administration; ∥Denominator = 4; number of patients ECG was performed after APs administration. Denominator = 55; number of patients who were alive during readmission

Age, mean (range), yr 81.3 (65.599.7)
Gender, no. (%)
Male 36 (60)
Female 24 (40)
Admitted from, no. (%)
Home 33 (55)
Rehabilitation facilities 5 (8)
SNF 22 (37)
Services, no. (%)
Medicine 48 (80)
Surgery 12 (20)
Types of APs continued on readmission (from index admission), no. (%)
Quetiapine 19 (61)
Olanzapine 6 (19)
Risperidone 4 (13)
Haloperidol 2 (7)
Types of APs started during readmission, no. (%)
Quetiapine 7 (39)
Risperidone 2 (11)
Haloperidol 16 (89)
Indications for AP use, no. (%)
Delirium 14 (77)
Undocumented 3 (17)
Other 1 (6)
ECG, no. (%)
Prior to APs administration 17 (94)
After APs administration 4 (22)
QTc prolongation >500 ms, no. (%)
Prior to APs administration 3 (18)
After APs administration∥ 2 (50)
Discharge destination, no. (%)
Home 23 (38)
Rehabilitation facilities 4 (7)
SNF 28 (47)
Death 5 (8)
Figure 2
Flowchart showing the outcomes of patients who were discharged on antipsychotics during index hospitalization and readmitted within 1 year from discharge. Abbreviations: APs, antipsychotics.

Eighteen patients received 1 or more new APs during the readmission hospitalizations. These included haloperidol (89%) and quetiapine (39%). Delirium was the main reported indication for starting APs (78%), but in 17% of cases no indication was documented. An electrocardiogram (ECG) was performed in 94% prior to APs administration and for 22% after APs administration. Corrected QT interval (QTc) of >500 ms was present in 18% of patients in pretreatment ECG and 50% of patients in post‐AP ECG. Of patients who survived readmission, 58% (32/55) were discharged to postacute facilities. Of the 39 patients who were on the same APs from index admission, 27 (69%) patients were eventually discharged on the same APs or new APs started during the readmission.

In the multivariable model (Table 2), predictors of death at 1 year included discharge to postacute facilities after index admission (odds ratio [OR]: 2.28; 95% confidence interval [CI]: 1.10‐4.73, P = 0.03) and QTc prolongation >500 ms during index admission (OR: 3.41; 95% CI: 1.34‐8.67, P = 0.01). Age and gender were not associated with 1‐year mortality.

Multivariable Analysis of One‐Year Mortality From Index Admission in Patients Who Were Started on Antipsychotics.
Odds Ratio 95% Confidence Interval P Value
  • NOTE: Abbreviations: AP, antipsychotic; QTc, QT interval. *For subset of patients for whom electrocardiogram was done.

Age 1.03 0.991.06 0.13
Male sex 0.87 0.501.52 0.63
Risperdal 3.53 0.6419.40 0.15
QTc prolongation after AP administration* 3.41 1.348.67 0.01
Presence of geriatric psychiatry consult 0.30 0.091.04 0.06
Discharged to postacute facilities vs home 2.28 1.104.73 0.03

DISCUSSION

In a cohort of elderly patients who were discharged on APs, nearly one‐third (29%) died within 1 year of the hospitalization in which APs were initiated. Nearly half of the survivors from the index admission (41%) experienced at least 1 admission within 1 year from discharge. Of readmitted patients, two‐thirds were taking the same APs that had been started during the index hospitalization. Half of the patients not on APs on readmission were started on an AP during the hospitalization, most often because they became delirious on return to the acute care setting. Compared to patients discharged home after an index admission, patients who were discharged to postacute facilities were almost 4 times as likely to die during the year subsequent to the admission. These data suggest that once patients are started on APs, most are continued on them until the next admission or are restarted during that readmission. Moreover, hospitalized elders who require an AP are at high risk for mortality in the coming year.

Prior studies have reported that patients with delirium have elevated 1‐year mortality rates.[14, 15, 16, 17, 18, 19] A secondary analysis of the Delirium Prevention Trial, which included 437 hospitalized older patients, revealed a 1‐year mortality rate of 20% in those who were never delirious during hospitalization, compared to 26% to 38% in patients with delirium.[19] Additionally, 1‐year mortality in hospitalized older patients with delirium (36%) was shown to be higher than patients with dementia (29%) or depression (26%).[17] Unlike these studies, not all of the patients in our study had documented delirium, but all received an AP. Still, it is notable that the 1‐year mortality rate for delirium in general is similar to what we found in this study.

The literature has also reported that long‐term AP use is associated with excess mortality in elder patients, especially those with dementia.[20, 21, 22] In a retrospective cohort study, older patients with dementia who were taking antipsychotics had significantly higher 1‐year mortality rates (23%29%) than patients not taking antipsychotic medications (15%). In a large Canadian propensity score‐matched cohort study that included over 13,000 demented older adults, the mortality was higher in the community‐dwelling elders who received atypical APs compared to no APs, with a difference of 1.1% in 180‐day mortality rate after initiation of APs.[21] The absolute mortality rate was 2.6% higher in patients who received typical compared to atypical APs. Unlike these studies, not every patient in our cohort had a diagnosis of dementia, but again, mortality rates in these studies appear similar to our cohort.

In contrast, other observational studies have not found an increased risk associated with receipt of APs. For example, a prospective study that enrolled approximately 950 patients with probable dementia showed that AP use was not associated with time to death after adjustment for comorbidities, demographic and cognitive variables.[23] These conflicting results highlight the difficulties of attributing outcomes in high‐risk populations. Although the excess mortality observed in patients taking APs may be related to the risks of APs, it is quite possible that patients who require APs (most often for delirium or agitated dementia) are at higher risk of death. This confounding by indication may be nearly impossible to adjust for retrospectively, even using techniques such as propensity matching.

Our report adds to the literature; we know of no studies to date describing a cohort of patients, most with delirium, who were started on APs in the hospital. We also attempted to identify the reasons that patients were started on APs, which have been infrequently reported. As noted above, our 1‐year mortality rate of 29% among older patients prescribed APs in the hospital was quite similar to mortality rates both for patients with delirium who were not necessarily treated with APs and patients with dementia who were treated with APs. This finding further supports the argument that risk factors for mortality, including dementia, delirium, and AP use are very difficult to tease apart. It is possible that the reasons that APs are prescribed (agitated delirium or dementia) have as much to do with the excess mortality reported in observational studies of APs as the use of APs themselves.

The high rate of continued AP use we observed (two‐thirds of readmitted patients) may reflect limited pharmacological alternatives to these medications with little evidence to support treating the symptoms of delirium with other drug classes, along with suboptimal environmental and behavioral modifications in postacute facilities and hospitals. This is unfortunate given that delirium is often preventable. Systematic implementation of well‐documented strategies to decrease delirium in hospitals and postacute facilities would likely reduce the prescription of APs and has the potential to slow the decline in this vulnerable population. A meta‐analysis incorporating both randomized and nonrandomized trials of medical and surgical patients showed that multicomponent nonpharmacologic interventions decreased delirium by 50%.[24] Thus, simple interventions such as reorientation, early mobilization, optimizing vision and hearing, sleepwake cycle preservation, and hydration might avoid roughly 1 million cases of delirium in hospitalized older adults annually.[24] The Hospital Elder Life Program and Acute Care for Elders units are examples of programs that have been shown to decrease the incidence of delirium.[25, 26]

Despite vigorous efforts to prevent delirium, a subgroup of patients still will become delirious. These patients are at high risk for death. Our mortality prediction model revealed that patients who were discharged to postacute facilities were 4 times more likely to die during the subsequent year compared to patients who were discharged home. Patients discharged to postacute facilities are likely to have a higher burden of disease, greater functional and cognitive impairment, and more frailty than those who are able to return to the community. Very ill and/or frail patients receiving APs in the hospital and requiring APs on discharge to postacute care facilities have limited survival and may benefit from expedited palliative care interventions to clarify prognosis and goals, and relieve suffering. At a minimum, our study identifies a need for further study to identify this very high‐risk group of elders. It is notable that 50% of patients were found to have a post‐treatment ECG with a QTc of >500 ms, a finding that has not been previously described. This would put these patients at higher risk of mortality, and as such we suggest that current guidelines should continue to emphasize the importance of post‐treatment ECGs and set clear criteria for discontinuation in elderly patients.

Our study is limited by its retrospective, single‐center design and small sample size, therefore limiting the interpretation and generalizability of the results to other hospitals. Quetiapine was the most common AP medication used in our hospital; therefore, our findings cannot be generalized to hospitals that utilize other AP agents. Future studies should examine antipsychotic use across hospitals to determine variation in prescribing patterns and outcomes. Nevertheless, the care of these patients were transitioned to a large number of geriatricians and primary care and nursing home physicians after discharge, and the reflected practice patterns extended beyond our hospital. Additionally, we were unable to determine when and why APs were discontinued or started in the outpatient setting. We were only able to detect readmissions to the 3 hospitals within our health system and therefore may have missed some readmissions to other institutions, although the majority of patients in our region tend to return to the same hospital. For patients who were not readmitted, we were also unable to identify whether they remained on the APs initiated during their index hospitalizations. Any retrospective study is limited by the difficulty of distinguishing delirium from the behavioral and psychiatric symptoms of dementia, but we identified delirium using standard terms described in previous literature.[10] We were unable to determine the types of delirium (hyperactive vs hypoactive) given that the documentations on behavioral symptoms were largely missing from the charts. The number of patients with preexisting diagnosis of dementia was likely underestimated, as we were only able to verify the diagnosis from the medical history. Additionally, the retrospective design based on chart review limited the factors that we could detect and grade accurately for inclusion in our mortality prediction model. Of note, our model did not contain objective measures of cognition, agitation, function, and markers for frailty such as walking speed, weak grip strength, weight loss, and low physical activity.

CONCLUSION

Initiating an AP (eg, haloperidol, quetiapine, olanzapine, and risperidone) in the hospital is likely to result in long‐term use of these medications despite the fact that AP use has been associated with multiple risks including falls, fractures, stroke, cardiovascular disease, and increased mortality in those with underlying dementia.[27] When possible, behavioral interventions to prevent delirium and slow the trajectory of decline should be implemented to reduce AP use. If patients with delirium are started on antipsychotics, it is important to monitor for prolonged QTc given the associated risk of mortality. In a subgroup of patients at high risk for death in the upcoming year, occurrence of delirium or use of APs during a hospitalization should both be considered triggers for early advance care planning and possibly palliative care and end‐of‐life discussions, with an emphasis on quality of life.

Disclosures: The research was supported by the Department of Medicine, Baystate Medical Center/Tufts University School of Medicine. Dr. Lagu is supported by the National Heart, Lung, and Blood Institute of the National Institutes of Health under award number K01HL114745. Drs. Lagu and Loh had full access to all of the data in the study. They take responsibility for the integrity of the data and the accuracy of the analysis. Drs. Loh, Brennan, Lindenauer, and Lagu conceived of the study. Drs. Loh, Ramdass, and Ms. Garb acquired the data. Ms Garb analyzed and interpreted the data. Drs. Loh, Ramdass, and Thim drafted the manuscript. Drs. Brennan, Lindenauer, and Lagu and Ms. Garb critically reviewed the manuscript for important intellectual content. Dr. Lagu has received consulting fees from the Institute for Healthcare Improvement, under contract to the Centers for Medicare and Medicaid Services, for her work on a project to help health systems achieve disability competence. Dr. Brennan is supported by a Geriatric Work Force Enhancement Grant from the US Department of Health and Human Services award number 1 U1QHP287020100. The authors report no conflicts of interest.

References
  1. Inouye SK, Westendorp RGJ, Saczynski JS. Delirium in elderly people. Lancet. 2014;383:911922.
  2. Fong TG, Jones RN, Marcantonio ER, et al. Adverse outcomes after hospitalization and delirium in persons with Alzheimer disease. Ann Intern Med. 2012;156:848856, W296.
  3. American Geriatrics Society Expert Panel on Postoperative Delirium in Older Adults. American Geriatrics Society abstracted clinical practice guideline for postoperative delirium in older adults. J Am Geriatr Soc. 2015;63:142150.
  4. Practice guideline for the treatment of patients with delirium. American Psychiatric Association. Am J Psychiatry. 1999;156:120.
  5. Potter J, George J; Guideline Development Group. The prevention, diagnosis and management of delirium in older people: concise guidelines. Clin Med (Lond). 2006;6:303308.
  6. Seitz DP, Gill SS, Zyl LT. Antipsychotics in the treatment of delirium: a systematic review. J Clin Psychiatry. 2007;68:1121.
  7. Maust DT, Kim HM, Seyfried LS, et al. Antipsychotics, other psychotropics, and the risk of death in patients with dementia: number needed to harm. JAMA Psychiatry. 2015;72:438445.
  8. Andrade C, Radhakrishnan R. Safety and efficacy of antipsychotic drugs for the behavioral and psychological symptoms of dementia. Indian J Psychiatry. 2009;51(suppl 1):S87S92.
  9. Gareri P, Fazio P, Manfredi VGL, Sarro G. Use and safety of antipsychotics in behavioral disorders in elderly people with dementia. J Clin Psychopharmacol. 2014;34:109123.
  10. Loh KP, Ramdass S, Garb JL, Brennan MJ, Lindenauer PK, Lagu T. From hospital to community: use of antipsychotics in hospitalized elders. J Hosp Med. 2014;9:802804.
  11. Sesso HD, Paffenbarger RS, Lee I‐M. Comparison of National Death Index and World Wide Web Death Searches. Am J Epidemiol. 2000;152:107111.
  12. Cox DR. Analysis of Binary Data. London, United Kingdom: Methuen; 1970:7699.
  13. Lee ET. Statistical Methods for Survival Data Analysis. New York, NY: John Wiley 1992:233236.
  14. Evans SJ, Sayers M, Mitnitski A, Rockwood K. The risk of adverse outcomes in hospitalized older patients in relation to a frailty index based on a comprehensive geriatric assessment. Age Ageing. 2014;43:127132.
  15. Grover S, Ghormode D, Ghosh A, et al. Risk factors for delirium and inpatient mortality with delirium. J Postgrad Med. 2013;59:263270.
  16. Avelino‐Silva TJ, Farfel JM, Curiati JAE, Amaral JRG, Campora F, Jacob‐Filho W. Comprehensive geriatric assessment predicts mortality and adverse outcomes in hospitalized older adults. BMC Geriatr. 2014;14:129.
  17. Tsai M‐C, Weng H‐H, Chou S‐Y, Tsai C‐S, Hung T‐H, Su J‐A. One‐year mortality of elderly inpatients with delirium, dementia, or depression seen by a consultation‐liaison service. Psychosomatics. 2012;53:433438.
  18. Hemert AM, Mast RC, Hengeveld MW, Vorstenbosch M. Excess mortality in general hospital patients with delirium: a 5‐year follow‐up of 519 patients seen in psychiatric consultation. J Psychosom Res. 1994;38:339346.
  19. McAvay GJ, Ness PH, Bogardus ST, et al. Older adults discharged from the hospital with delirium: 1‐year outcomes. J Am Geriatr Soc. 2006;54:12451250.
  20. Ballard C, Hanney ML, Theodoulou M, et al. The dementia antipsychotic withdrawal trial (DART‐AD): long‐term follow‐up of a randomised placebo‐controlled trial. Lancet Neurol. 2009;8:151157.
  21. Gill SS, Bronskill SE, Normand S‐LT, et al. Antipsychotic drug use and mortality in older adults with dementia. Ann Intern Med. 2007;146:775786.
  22. Wang PS, Schneeweiss S, Avorn J, et al. Risk of death in elderly users of conventional vs. atypical antipsychotic medications. N Engl J Med. 2005;353:23352341.
  23. Lopez OL, Becker JT, Chang Y‐F, et al. The long‐term effects of conventional and atypical antipsychotics in patients with probable Alzheimer's disease. Am J Psychiatry. 2013;170:10511058.
  24. Hshieh TT, Yue J, Oh E, et al. Effectiveness of multicomponent nonpharmacological delirium interventions: a meta‐analysis. JAMA Intern Med. 2015;175:512520.
  25. Rubin FH, Neal K, Fenlon K, Hassan S, Inouye SK. Sustainability and scalability of the hospital elder life program at a community hospital. J Am Geriatr Soc. 2011;59:359365.
  26. Fox MT, Persaud M, Maimets I, et al. Effectiveness of acute geriatric unit care using acute care for elders components: a systematic review and meta‐analysis. J Am Geriatr Soc. 2012;60:22372245.
  27. Muench J, Hamer AM. Adverse effects of antipsychotic medications. Am Fam Physician. 2010;81:617622.
References
  1. Inouye SK, Westendorp RGJ, Saczynski JS. Delirium in elderly people. Lancet. 2014;383:911922.
  2. Fong TG, Jones RN, Marcantonio ER, et al. Adverse outcomes after hospitalization and delirium in persons with Alzheimer disease. Ann Intern Med. 2012;156:848856, W296.
  3. American Geriatrics Society Expert Panel on Postoperative Delirium in Older Adults. American Geriatrics Society abstracted clinical practice guideline for postoperative delirium in older adults. J Am Geriatr Soc. 2015;63:142150.
  4. Practice guideline for the treatment of patients with delirium. American Psychiatric Association. Am J Psychiatry. 1999;156:120.
  5. Potter J, George J; Guideline Development Group. The prevention, diagnosis and management of delirium in older people: concise guidelines. Clin Med (Lond). 2006;6:303308.
  6. Seitz DP, Gill SS, Zyl LT. Antipsychotics in the treatment of delirium: a systematic review. J Clin Psychiatry. 2007;68:1121.
  7. Maust DT, Kim HM, Seyfried LS, et al. Antipsychotics, other psychotropics, and the risk of death in patients with dementia: number needed to harm. JAMA Psychiatry. 2015;72:438445.
  8. Andrade C, Radhakrishnan R. Safety and efficacy of antipsychotic drugs for the behavioral and psychological symptoms of dementia. Indian J Psychiatry. 2009;51(suppl 1):S87S92.
  9. Gareri P, Fazio P, Manfredi VGL, Sarro G. Use and safety of antipsychotics in behavioral disorders in elderly people with dementia. J Clin Psychopharmacol. 2014;34:109123.
  10. Loh KP, Ramdass S, Garb JL, Brennan MJ, Lindenauer PK, Lagu T. From hospital to community: use of antipsychotics in hospitalized elders. J Hosp Med. 2014;9:802804.
  11. Sesso HD, Paffenbarger RS, Lee I‐M. Comparison of National Death Index and World Wide Web Death Searches. Am J Epidemiol. 2000;152:107111.
  12. Cox DR. Analysis of Binary Data. London, United Kingdom: Methuen; 1970:7699.
  13. Lee ET. Statistical Methods for Survival Data Analysis. New York, NY: John Wiley 1992:233236.
  14. Evans SJ, Sayers M, Mitnitski A, Rockwood K. The risk of adverse outcomes in hospitalized older patients in relation to a frailty index based on a comprehensive geriatric assessment. Age Ageing. 2014;43:127132.
  15. Grover S, Ghormode D, Ghosh A, et al. Risk factors for delirium and inpatient mortality with delirium. J Postgrad Med. 2013;59:263270.
  16. Avelino‐Silva TJ, Farfel JM, Curiati JAE, Amaral JRG, Campora F, Jacob‐Filho W. Comprehensive geriatric assessment predicts mortality and adverse outcomes in hospitalized older adults. BMC Geriatr. 2014;14:129.
  17. Tsai M‐C, Weng H‐H, Chou S‐Y, Tsai C‐S, Hung T‐H, Su J‐A. One‐year mortality of elderly inpatients with delirium, dementia, or depression seen by a consultation‐liaison service. Psychosomatics. 2012;53:433438.
  18. Hemert AM, Mast RC, Hengeveld MW, Vorstenbosch M. Excess mortality in general hospital patients with delirium: a 5‐year follow‐up of 519 patients seen in psychiatric consultation. J Psychosom Res. 1994;38:339346.
  19. McAvay GJ, Ness PH, Bogardus ST, et al. Older adults discharged from the hospital with delirium: 1‐year outcomes. J Am Geriatr Soc. 2006;54:12451250.
  20. Ballard C, Hanney ML, Theodoulou M, et al. The dementia antipsychotic withdrawal trial (DART‐AD): long‐term follow‐up of a randomised placebo‐controlled trial. Lancet Neurol. 2009;8:151157.
  21. Gill SS, Bronskill SE, Normand S‐LT, et al. Antipsychotic drug use and mortality in older adults with dementia. Ann Intern Med. 2007;146:775786.
  22. Wang PS, Schneeweiss S, Avorn J, et al. Risk of death in elderly users of conventional vs. atypical antipsychotic medications. N Engl J Med. 2005;353:23352341.
  23. Lopez OL, Becker JT, Chang Y‐F, et al. The long‐term effects of conventional and atypical antipsychotics in patients with probable Alzheimer's disease. Am J Psychiatry. 2013;170:10511058.
  24. Hshieh TT, Yue J, Oh E, et al. Effectiveness of multicomponent nonpharmacological delirium interventions: a meta‐analysis. JAMA Intern Med. 2015;175:512520.
  25. Rubin FH, Neal K, Fenlon K, Hassan S, Inouye SK. Sustainability and scalability of the hospital elder life program at a community hospital. J Am Geriatr Soc. 2011;59:359365.
  26. Fox MT, Persaud M, Maimets I, et al. Effectiveness of acute geriatric unit care using acute care for elders components: a systematic review and meta‐analysis. J Am Geriatr Soc. 2012;60:22372245.
  27. Muench J, Hamer AM. Adverse effects of antipsychotic medications. Am Fam Physician. 2010;81:617622.
Issue
Journal of Hospital Medicine - 11(8)
Issue
Journal of Hospital Medicine - 11(8)
Page Number
550-555
Page Number
550-555
Publications
Publications
Article Type
Display Headline
Long‐term outcomes of elders discharged on antipsychotics
Display Headline
Long‐term outcomes of elders discharged on antipsychotics
Sections
Article Source
© 2016 Society of Hospital Medicine
Disallow All Ads
Correspondence Location
Address for correspondence and reprint requests: Kah Poh Loh, BMedSci, James P. Wilmot Cancer Institute, University of Rochester Medical Center, 601 Elmwood Avenue, Box 704, Rochester, NY 14642; Telephone: 413‐306‐9767; Fax: +1585‐273‐1042; E‐mail: melissalkp@gmail.com
Content Gating
Gated (full article locked unless allowed per User)
Gating Strategy
First Peek Free
Article PDF Media
Media Files

Direct Admissions to the Hospital

Article Type
Changed
Mon, 01/02/2017 - 19:34
Display Headline
Direct admission to the hospital: An alternative approach to hospitalization

Increasing use of emergency departments (EDs) throughout the United States has become a focus of national healthcare policy and reform efforts. ED growth continues to outpace population growth, with the Institute of Medicine describing our ED systems as fragmented, overburdened, and at the breaking point.[1] Associations between ED crowding and patient dissatisfaction, delays in treatment, medical errors, and patient mortality speak to the urgency of systems improvements.[2] One major factor contributing to ED volumes is the growing number of hospital admissions that begin in EDs. From 1993 to 2006, the proportion of hospitalizations originating in EDs increased from 33.5% to 43.8%, with more than 17 million hospital admissions originating in EDs annually.[3, 4] Despite these challenges, discussions about alternative approaches to hospital admission remain at the periphery of healthcare policy conversations.

Direct admission to the hospital, defined as hospitalization without first receiving care in the hospital's ED, is an alternative approach to hospital admission, and may be a vehicle to both observation and inpatient hospital stays. Direct admissions account for 25% of all nonelective pediatric hospitalizations and 15% of nonelective adult hospitalizations in the United States.[5, 6] This admission approach was considerably more common in the past, facilitated by primary care providers (PCPs) or specialists who provided both outpatient and hospital‐based care for their patients.[4] However, as the number of hospitalists in the United States has grown over the last 30 years, the number of direct admissions has decreased concurrently. In fact, from 2003 to 2009, the number of direct admissions from clinics and physicians' offices decreased by a total of 1.6 million.[4] Although this decline is undoubtedly multifactorial, hospitalists may have contributed, both deliberately and inadvertently, to the shifting epidemiology of hospital admissions. Although many factors influence the source of hospital admissions and admission processes, direct admission has 2 important prerequisites: patients require timely access to outpatient providers for acute care, and hospitals, in partnership with outpatient‐based clinics and practices, require systems to safely and efficiently facilitate admissions without ED involvement. However, we know little about hospital admission systems, developed in the era of hospital medicine, to facilitate admissions independent of the ED.

Direct admission offers a number of potential benefits for both patients and healthcare delivery systems including reductions in the number of sites and providers of care, improved communication and coordination between outpatient and hospital‐based healthcare providers, greater patient and referring physician satisfaction, and reduced ED volumes and subsequent costs.[7] However, there are also risks and potential harms associated with direct admission, including potential delays in initial evaluation and management, inconsistent admission processes, and difficulties determining direct admission appropriateness, all of which could adversely impact patient safety and quality of care.[7, 8, 9] One study of adults with sepsis found that direct admission was associated with increased mortality compared to ED admission, which the authors speculated to be related to less timely care.[9] Similarly, a study of unscheduled adult hospitalizations found that patients admitted directly had higher mortality for time‐sensitive conditions such as acute myocardial infarction and sepsis than patients admitted through EDs, differences not observed among adults admitted with pneumonia, asthma, cellulitis, and several other common, yet frequently less emergent, reasons for hospitalization.[8] Among children with pneumonia, the most common reason for pediatric hospitalization, direct admission has been associated with significantly lower costs than admissions originating in the ED, with no significant differences in rates of transfer to the intensive care unit or hospital readmission.[10]

There is significant variation across both diagnoses and hospitals in rates of direct admission, raising questions about the contextual factors unique to hospital medicine programs that perform a substantial proportion of direct admissions.[5] This variation also highlights opportunities to identify the populations, conditions, and systems that facilitate safe and effective direct admissions. Certainly, direct admission is unlikely to be appropriate for all populations or conditions. Patients requiring emergent care or rapid diagnostic imaging are likely to receive more timely care in the ED; sepsis, acute myocardial infarction, and trauma are but a few examples of conditions for which rapid ED care decreases morbidity and mortality. Similarly, patients for whom the need for hospitalization is uncertainfor example, dehydration, asthmamay be more appropriate for initial ED management followed by re‐evaluation to inform the need for hospitalization. Finally, patients for whom the admitting diagnosis is uncertain and who require consultation for several subspecialists may be more efficiently evaluated in EDs. In our national survey of pediatric direct admission guidelines, less than one‐third of hospitals reported having formal criteria to assess the appropriateness of direct admissions, and respondents' perspectives regarding populations and diagnoses appropriate for this admission approach varied considerably.[7] These results point to the need for further research and quality‐improvement initiatives to inform the development of direct admission guidelines and protocols.

During the last decade, hospitals' discharge processes have been the focus of tremendous research, policy, and quality improvement efforts. The phrase transition of care is now widely understood to describe the changes in patient care that begin with discharge planning and conclude when patients' have established care at home or another healthcare facility. Transitions of care have been a focus of the Journal of Hospital Medicine since its inception, including publication of the Transitions of Care Consensus Policy Statement in 2009, as well as numerous other studies highlighting both risks associated with transitions of care as well as methods to address these.[11, 12, 13, 14, 15, 16] Similar to hospital discharge, hospital admission is an inherent feature of every hospitalization, and admission and discharge processes share many commonalities. Both involve transitions in sites of care, and handoffs between healthcare providers. Most involve changes in medical therapies, including both the addition of new medications and changes to existing treatments. Moreover, both are associated with significant stress to patients and their families. As a result, hospital admissions expose patients to many of same risks that have been the focus of hospital discharge reform: unstructured patient handoffs, poor communication between healthcare providers, and costly, inefficient care. The Society of Hospital Medicine has been a leader in articulating the importance of patient‐centered, clinically relevant medication reconciliation across the healthcare continuum.[17] However, with the exception of this important work, research and policy focused on understanding and improving transitions of care into the hospital have received disproportionately little attention.

To facilitate research and quality improvement efforts focused on hospital admission, we suggest that the transitions of care framework, typically discussed in the context of hospital discharge, be expanded to reflect the different origins of hospitalizations and multiple transitions that can be experienced by patients as they enter the hospital. A broadening of the transitions of care framework to incorporate hospital admissions brings numerous questions previously addressed in hospital‐to‐home transitions to the forefront. How do transitions into the hospital impact patients and healthcare systems? When is direct admission safe and effective, and how does this vary across conditions and hospital settings? What protocols and tools might optimize the associated transitions and reduce the risks of error and harm? There are numerous stakeholders who will undoubtedly bring diverse perspectives to these questionspatients and their families, hospital‐based healthcare providers, PCPs and specialists, ED physicians, and payors.

Increasing ED volumes, long wait times, and rising ED costs speak to the importance of better understanding hospital admission alternatives and the associated risks and benefits. Encouraging more direct admissions may be a solution, but evidence to guide best practices must precede this. The growing presence of round‐the‐clock pediatric and adult hospitalists across the country creates unique opportunities to transform hospital admission systems for the vast number of patients who do not require emergent care. The Affordable Care Act's expansion of insurance coverage and incentivized coordinated care within patient‐centered medical homes creates a unique opportunity for this broadened view of transitions of care. This suggests that the time is ripe for pursuing strategies that will both improve patients' transitions from outpatient to inpatient care and reduce stress on our overburdened emergency departments.

Disclosure: Dr. Lagu was supported by the National Heart, Lung, and Blood Institute of the National Institutes of Health under award number K01HL114745. She has received consulting fees from the Institute for Healthcare Improvement, under contract to the Centers for Medicare and Medicaid Services (CMS), for her work on a project to help health systems achieve disability competence, and from the Island Peer Review Organization, under contract to CMS, for her work on development of episodes of care for CMS payment purposes (both unrelated to the current work). Dr. Leyenaar was supported by grant number K08HS024133 from the Agency for Healthcare Research and Quality. The content is solely the responsibility of the authors and does not necessarily represent the official views of the Agency for Healthcare Research and Quality. The authors report no conflicts of interest.

Files
References
  1. Institute of Medicine. Hospital‐based emergency care: At the breaking point. Washington, DC: National Academies Press; 2006. Available at: http://www.nap.edu/openbook.php?record_id=11621. Accessed September 13, 2015.
  2. Pitts SR, Pines JM, Handrigan MT, Kellermann AL. National trends in emergency department occupancy, 2001 to 2008: effect of inpatient admissions versus emergency department practice intensity. Ann Emerg Med. 2012;60(6):679686.e3.
  3. Schuur J, Venkatesh A. The growing role of emergency departments in hospital admissions. N Engl J Med. 2012;367(5):391393.
  4. Morganti KG, Bauhoff S, Blanchard J, et al. The Evolving Role of Emergency Departments in the United States. Santa Monica, CA: RAND Corp.; 2013:179.
  5. Leyenaar J, Shieh M‐S, Lagu T, Pekow PS, Lindenauer PK. Direct admission to hospitals among children in the United States. JAMA Pediatr. 2015;169(5):500502.
  6. Healthcare Cost and Utilization Project. National Inpatient Sample. 2012. Agency for Healthcare Research and Quality, Rockville, MD. Available at: www.hcup‐us.ahrq.gov/nisoverview.jsp. Accessed October 11, 2014.
  7. Leyenaar JK, O'Brien ER, Malkani N, Lagu T, Lindenauer PK. Direct admission to hospital: a mixed methods survey of pediatric practices, benefits, and challenges [published online August 17, 2015]. Acad Pediatr.
  8. Kocher KE, Dimick JB, Nallamothu BK. Changes in the source of unscheduled hospitalizations in the United States. Med Care. 2013;51(8):689698.
  9. Powell ES, Khare RK, Courtney DM, Feinglass J. Lower mortality in sepsis patients admitted through the ED vs direct admission. Am J Emerg Med. 2012;30(3):432439.
  10. Leyenaar JK, Shieh M, Lagu T, Pekow PS, Lindenauer PK. Variation and outcomes associated with direct admission among children with pneumonia in the United States. JAMA Pediatr. 2014;168(9):829836.
  11. Snow V, Beck D, Budnitz T, et al. Transitions of Care Consensus policy statement: American College of Physicians, Society of General Internal Medicine, Society of Hospital Medicine, American Geriatrics Society, American College Of Emergency Physicians, and Society for Academic Emergency Medicine. J Hosp Med. 2009;4(6):364370.
  12. Kripalani S, Jackson AT, Schnipper JL, Coleman EA. Promoting effective transitions of care at hospital discharge: a review of key issues for hospitalists. J Hosp Med. 2007;2(5):314323.
  13. Coleman EA. Safety in numbers: physicians joining forces to seal the cracks during transitions. J Hosp Med. 2009;4(6):329330.
  14. Soong C, Daub S, Lee J, et al. Development of a checklist of safe discharge practices for hospital patients. J Hosp Med. 2013;8(8):444449.
  15. Hansen LO, Greenwald JL, Budnitz T, et al. Project BOOST: effectiveness of a multihospital effort to reduce rehospitalization. J Hosp Med. 2013;8(8):421427.
  16. Solan LG, Ranji SR, Shah SS. The successes and challenges of hospital to home transitions. J Hosp Med. 2014;9(4):271273.
  17. Greenwald JL, Halasyamani L, Greene J, et al. Making inpatient medication reconciliation patient centered, clinically relevant and implementable: a consensus statement on key principles and necessary first steps. J Hosp Med. 2010;5(8):477485.
Article PDF
Issue
Journal of Hospital Medicine - 11(4)
Publications
Page Number
303-305
Sections
Files
Files
Article PDF
Article PDF

Increasing use of emergency departments (EDs) throughout the United States has become a focus of national healthcare policy and reform efforts. ED growth continues to outpace population growth, with the Institute of Medicine describing our ED systems as fragmented, overburdened, and at the breaking point.[1] Associations between ED crowding and patient dissatisfaction, delays in treatment, medical errors, and patient mortality speak to the urgency of systems improvements.[2] One major factor contributing to ED volumes is the growing number of hospital admissions that begin in EDs. From 1993 to 2006, the proportion of hospitalizations originating in EDs increased from 33.5% to 43.8%, with more than 17 million hospital admissions originating in EDs annually.[3, 4] Despite these challenges, discussions about alternative approaches to hospital admission remain at the periphery of healthcare policy conversations.

Direct admission to the hospital, defined as hospitalization without first receiving care in the hospital's ED, is an alternative approach to hospital admission, and may be a vehicle to both observation and inpatient hospital stays. Direct admissions account for 25% of all nonelective pediatric hospitalizations and 15% of nonelective adult hospitalizations in the United States.[5, 6] This admission approach was considerably more common in the past, facilitated by primary care providers (PCPs) or specialists who provided both outpatient and hospital‐based care for their patients.[4] However, as the number of hospitalists in the United States has grown over the last 30 years, the number of direct admissions has decreased concurrently. In fact, from 2003 to 2009, the number of direct admissions from clinics and physicians' offices decreased by a total of 1.6 million.[4] Although this decline is undoubtedly multifactorial, hospitalists may have contributed, both deliberately and inadvertently, to the shifting epidemiology of hospital admissions. Although many factors influence the source of hospital admissions and admission processes, direct admission has 2 important prerequisites: patients require timely access to outpatient providers for acute care, and hospitals, in partnership with outpatient‐based clinics and practices, require systems to safely and efficiently facilitate admissions without ED involvement. However, we know little about hospital admission systems, developed in the era of hospital medicine, to facilitate admissions independent of the ED.

Direct admission offers a number of potential benefits for both patients and healthcare delivery systems including reductions in the number of sites and providers of care, improved communication and coordination between outpatient and hospital‐based healthcare providers, greater patient and referring physician satisfaction, and reduced ED volumes and subsequent costs.[7] However, there are also risks and potential harms associated with direct admission, including potential delays in initial evaluation and management, inconsistent admission processes, and difficulties determining direct admission appropriateness, all of which could adversely impact patient safety and quality of care.[7, 8, 9] One study of adults with sepsis found that direct admission was associated with increased mortality compared to ED admission, which the authors speculated to be related to less timely care.[9] Similarly, a study of unscheduled adult hospitalizations found that patients admitted directly had higher mortality for time‐sensitive conditions such as acute myocardial infarction and sepsis than patients admitted through EDs, differences not observed among adults admitted with pneumonia, asthma, cellulitis, and several other common, yet frequently less emergent, reasons for hospitalization.[8] Among children with pneumonia, the most common reason for pediatric hospitalization, direct admission has been associated with significantly lower costs than admissions originating in the ED, with no significant differences in rates of transfer to the intensive care unit or hospital readmission.[10]

There is significant variation across both diagnoses and hospitals in rates of direct admission, raising questions about the contextual factors unique to hospital medicine programs that perform a substantial proportion of direct admissions.[5] This variation also highlights opportunities to identify the populations, conditions, and systems that facilitate safe and effective direct admissions. Certainly, direct admission is unlikely to be appropriate for all populations or conditions. Patients requiring emergent care or rapid diagnostic imaging are likely to receive more timely care in the ED; sepsis, acute myocardial infarction, and trauma are but a few examples of conditions for which rapid ED care decreases morbidity and mortality. Similarly, patients for whom the need for hospitalization is uncertainfor example, dehydration, asthmamay be more appropriate for initial ED management followed by re‐evaluation to inform the need for hospitalization. Finally, patients for whom the admitting diagnosis is uncertain and who require consultation for several subspecialists may be more efficiently evaluated in EDs. In our national survey of pediatric direct admission guidelines, less than one‐third of hospitals reported having formal criteria to assess the appropriateness of direct admissions, and respondents' perspectives regarding populations and diagnoses appropriate for this admission approach varied considerably.[7] These results point to the need for further research and quality‐improvement initiatives to inform the development of direct admission guidelines and protocols.

During the last decade, hospitals' discharge processes have been the focus of tremendous research, policy, and quality improvement efforts. The phrase transition of care is now widely understood to describe the changes in patient care that begin with discharge planning and conclude when patients' have established care at home or another healthcare facility. Transitions of care have been a focus of the Journal of Hospital Medicine since its inception, including publication of the Transitions of Care Consensus Policy Statement in 2009, as well as numerous other studies highlighting both risks associated with transitions of care as well as methods to address these.[11, 12, 13, 14, 15, 16] Similar to hospital discharge, hospital admission is an inherent feature of every hospitalization, and admission and discharge processes share many commonalities. Both involve transitions in sites of care, and handoffs between healthcare providers. Most involve changes in medical therapies, including both the addition of new medications and changes to existing treatments. Moreover, both are associated with significant stress to patients and their families. As a result, hospital admissions expose patients to many of same risks that have been the focus of hospital discharge reform: unstructured patient handoffs, poor communication between healthcare providers, and costly, inefficient care. The Society of Hospital Medicine has been a leader in articulating the importance of patient‐centered, clinically relevant medication reconciliation across the healthcare continuum.[17] However, with the exception of this important work, research and policy focused on understanding and improving transitions of care into the hospital have received disproportionately little attention.

To facilitate research and quality improvement efforts focused on hospital admission, we suggest that the transitions of care framework, typically discussed in the context of hospital discharge, be expanded to reflect the different origins of hospitalizations and multiple transitions that can be experienced by patients as they enter the hospital. A broadening of the transitions of care framework to incorporate hospital admissions brings numerous questions previously addressed in hospital‐to‐home transitions to the forefront. How do transitions into the hospital impact patients and healthcare systems? When is direct admission safe and effective, and how does this vary across conditions and hospital settings? What protocols and tools might optimize the associated transitions and reduce the risks of error and harm? There are numerous stakeholders who will undoubtedly bring diverse perspectives to these questionspatients and their families, hospital‐based healthcare providers, PCPs and specialists, ED physicians, and payors.

Increasing ED volumes, long wait times, and rising ED costs speak to the importance of better understanding hospital admission alternatives and the associated risks and benefits. Encouraging more direct admissions may be a solution, but evidence to guide best practices must precede this. The growing presence of round‐the‐clock pediatric and adult hospitalists across the country creates unique opportunities to transform hospital admission systems for the vast number of patients who do not require emergent care. The Affordable Care Act's expansion of insurance coverage and incentivized coordinated care within patient‐centered medical homes creates a unique opportunity for this broadened view of transitions of care. This suggests that the time is ripe for pursuing strategies that will both improve patients' transitions from outpatient to inpatient care and reduce stress on our overburdened emergency departments.

Disclosure: Dr. Lagu was supported by the National Heart, Lung, and Blood Institute of the National Institutes of Health under award number K01HL114745. She has received consulting fees from the Institute for Healthcare Improvement, under contract to the Centers for Medicare and Medicaid Services (CMS), for her work on a project to help health systems achieve disability competence, and from the Island Peer Review Organization, under contract to CMS, for her work on development of episodes of care for CMS payment purposes (both unrelated to the current work). Dr. Leyenaar was supported by grant number K08HS024133 from the Agency for Healthcare Research and Quality. The content is solely the responsibility of the authors and does not necessarily represent the official views of the Agency for Healthcare Research and Quality. The authors report no conflicts of interest.

Increasing use of emergency departments (EDs) throughout the United States has become a focus of national healthcare policy and reform efforts. ED growth continues to outpace population growth, with the Institute of Medicine describing our ED systems as fragmented, overburdened, and at the breaking point.[1] Associations between ED crowding and patient dissatisfaction, delays in treatment, medical errors, and patient mortality speak to the urgency of systems improvements.[2] One major factor contributing to ED volumes is the growing number of hospital admissions that begin in EDs. From 1993 to 2006, the proportion of hospitalizations originating in EDs increased from 33.5% to 43.8%, with more than 17 million hospital admissions originating in EDs annually.[3, 4] Despite these challenges, discussions about alternative approaches to hospital admission remain at the periphery of healthcare policy conversations.

Direct admission to the hospital, defined as hospitalization without first receiving care in the hospital's ED, is an alternative approach to hospital admission, and may be a vehicle to both observation and inpatient hospital stays. Direct admissions account for 25% of all nonelective pediatric hospitalizations and 15% of nonelective adult hospitalizations in the United States.[5, 6] This admission approach was considerably more common in the past, facilitated by primary care providers (PCPs) or specialists who provided both outpatient and hospital‐based care for their patients.[4] However, as the number of hospitalists in the United States has grown over the last 30 years, the number of direct admissions has decreased concurrently. In fact, from 2003 to 2009, the number of direct admissions from clinics and physicians' offices decreased by a total of 1.6 million.[4] Although this decline is undoubtedly multifactorial, hospitalists may have contributed, both deliberately and inadvertently, to the shifting epidemiology of hospital admissions. Although many factors influence the source of hospital admissions and admission processes, direct admission has 2 important prerequisites: patients require timely access to outpatient providers for acute care, and hospitals, in partnership with outpatient‐based clinics and practices, require systems to safely and efficiently facilitate admissions without ED involvement. However, we know little about hospital admission systems, developed in the era of hospital medicine, to facilitate admissions independent of the ED.

Direct admission offers a number of potential benefits for both patients and healthcare delivery systems including reductions in the number of sites and providers of care, improved communication and coordination between outpatient and hospital‐based healthcare providers, greater patient and referring physician satisfaction, and reduced ED volumes and subsequent costs.[7] However, there are also risks and potential harms associated with direct admission, including potential delays in initial evaluation and management, inconsistent admission processes, and difficulties determining direct admission appropriateness, all of which could adversely impact patient safety and quality of care.[7, 8, 9] One study of adults with sepsis found that direct admission was associated with increased mortality compared to ED admission, which the authors speculated to be related to less timely care.[9] Similarly, a study of unscheduled adult hospitalizations found that patients admitted directly had higher mortality for time‐sensitive conditions such as acute myocardial infarction and sepsis than patients admitted through EDs, differences not observed among adults admitted with pneumonia, asthma, cellulitis, and several other common, yet frequently less emergent, reasons for hospitalization.[8] Among children with pneumonia, the most common reason for pediatric hospitalization, direct admission has been associated with significantly lower costs than admissions originating in the ED, with no significant differences in rates of transfer to the intensive care unit or hospital readmission.[10]

There is significant variation across both diagnoses and hospitals in rates of direct admission, raising questions about the contextual factors unique to hospital medicine programs that perform a substantial proportion of direct admissions.[5] This variation also highlights opportunities to identify the populations, conditions, and systems that facilitate safe and effective direct admissions. Certainly, direct admission is unlikely to be appropriate for all populations or conditions. Patients requiring emergent care or rapid diagnostic imaging are likely to receive more timely care in the ED; sepsis, acute myocardial infarction, and trauma are but a few examples of conditions for which rapid ED care decreases morbidity and mortality. Similarly, patients for whom the need for hospitalization is uncertainfor example, dehydration, asthmamay be more appropriate for initial ED management followed by re‐evaluation to inform the need for hospitalization. Finally, patients for whom the admitting diagnosis is uncertain and who require consultation for several subspecialists may be more efficiently evaluated in EDs. In our national survey of pediatric direct admission guidelines, less than one‐third of hospitals reported having formal criteria to assess the appropriateness of direct admissions, and respondents' perspectives regarding populations and diagnoses appropriate for this admission approach varied considerably.[7] These results point to the need for further research and quality‐improvement initiatives to inform the development of direct admission guidelines and protocols.

During the last decade, hospitals' discharge processes have been the focus of tremendous research, policy, and quality improvement efforts. The phrase transition of care is now widely understood to describe the changes in patient care that begin with discharge planning and conclude when patients' have established care at home or another healthcare facility. Transitions of care have been a focus of the Journal of Hospital Medicine since its inception, including publication of the Transitions of Care Consensus Policy Statement in 2009, as well as numerous other studies highlighting both risks associated with transitions of care as well as methods to address these.[11, 12, 13, 14, 15, 16] Similar to hospital discharge, hospital admission is an inherent feature of every hospitalization, and admission and discharge processes share many commonalities. Both involve transitions in sites of care, and handoffs between healthcare providers. Most involve changes in medical therapies, including both the addition of new medications and changes to existing treatments. Moreover, both are associated with significant stress to patients and their families. As a result, hospital admissions expose patients to many of same risks that have been the focus of hospital discharge reform: unstructured patient handoffs, poor communication between healthcare providers, and costly, inefficient care. The Society of Hospital Medicine has been a leader in articulating the importance of patient‐centered, clinically relevant medication reconciliation across the healthcare continuum.[17] However, with the exception of this important work, research and policy focused on understanding and improving transitions of care into the hospital have received disproportionately little attention.

To facilitate research and quality improvement efforts focused on hospital admission, we suggest that the transitions of care framework, typically discussed in the context of hospital discharge, be expanded to reflect the different origins of hospitalizations and multiple transitions that can be experienced by patients as they enter the hospital. A broadening of the transitions of care framework to incorporate hospital admissions brings numerous questions previously addressed in hospital‐to‐home transitions to the forefront. How do transitions into the hospital impact patients and healthcare systems? When is direct admission safe and effective, and how does this vary across conditions and hospital settings? What protocols and tools might optimize the associated transitions and reduce the risks of error and harm? There are numerous stakeholders who will undoubtedly bring diverse perspectives to these questionspatients and their families, hospital‐based healthcare providers, PCPs and specialists, ED physicians, and payors.

Increasing ED volumes, long wait times, and rising ED costs speak to the importance of better understanding hospital admission alternatives and the associated risks and benefits. Encouraging more direct admissions may be a solution, but evidence to guide best practices must precede this. The growing presence of round‐the‐clock pediatric and adult hospitalists across the country creates unique opportunities to transform hospital admission systems for the vast number of patients who do not require emergent care. The Affordable Care Act's expansion of insurance coverage and incentivized coordinated care within patient‐centered medical homes creates a unique opportunity for this broadened view of transitions of care. This suggests that the time is ripe for pursuing strategies that will both improve patients' transitions from outpatient to inpatient care and reduce stress on our overburdened emergency departments.

Disclosure: Dr. Lagu was supported by the National Heart, Lung, and Blood Institute of the National Institutes of Health under award number K01HL114745. She has received consulting fees from the Institute for Healthcare Improvement, under contract to the Centers for Medicare and Medicaid Services (CMS), for her work on a project to help health systems achieve disability competence, and from the Island Peer Review Organization, under contract to CMS, for her work on development of episodes of care for CMS payment purposes (both unrelated to the current work). Dr. Leyenaar was supported by grant number K08HS024133 from the Agency for Healthcare Research and Quality. The content is solely the responsibility of the authors and does not necessarily represent the official views of the Agency for Healthcare Research and Quality. The authors report no conflicts of interest.

References
  1. Institute of Medicine. Hospital‐based emergency care: At the breaking point. Washington, DC: National Academies Press; 2006. Available at: http://www.nap.edu/openbook.php?record_id=11621. Accessed September 13, 2015.
  2. Pitts SR, Pines JM, Handrigan MT, Kellermann AL. National trends in emergency department occupancy, 2001 to 2008: effect of inpatient admissions versus emergency department practice intensity. Ann Emerg Med. 2012;60(6):679686.e3.
  3. Schuur J, Venkatesh A. The growing role of emergency departments in hospital admissions. N Engl J Med. 2012;367(5):391393.
  4. Morganti KG, Bauhoff S, Blanchard J, et al. The Evolving Role of Emergency Departments in the United States. Santa Monica, CA: RAND Corp.; 2013:179.
  5. Leyenaar J, Shieh M‐S, Lagu T, Pekow PS, Lindenauer PK. Direct admission to hospitals among children in the United States. JAMA Pediatr. 2015;169(5):500502.
  6. Healthcare Cost and Utilization Project. National Inpatient Sample. 2012. Agency for Healthcare Research and Quality, Rockville, MD. Available at: www.hcup‐us.ahrq.gov/nisoverview.jsp. Accessed October 11, 2014.
  7. Leyenaar JK, O'Brien ER, Malkani N, Lagu T, Lindenauer PK. Direct admission to hospital: a mixed methods survey of pediatric practices, benefits, and challenges [published online August 17, 2015]. Acad Pediatr.
  8. Kocher KE, Dimick JB, Nallamothu BK. Changes in the source of unscheduled hospitalizations in the United States. Med Care. 2013;51(8):689698.
  9. Powell ES, Khare RK, Courtney DM, Feinglass J. Lower mortality in sepsis patients admitted through the ED vs direct admission. Am J Emerg Med. 2012;30(3):432439.
  10. Leyenaar JK, Shieh M, Lagu T, Pekow PS, Lindenauer PK. Variation and outcomes associated with direct admission among children with pneumonia in the United States. JAMA Pediatr. 2014;168(9):829836.
  11. Snow V, Beck D, Budnitz T, et al. Transitions of Care Consensus policy statement: American College of Physicians, Society of General Internal Medicine, Society of Hospital Medicine, American Geriatrics Society, American College Of Emergency Physicians, and Society for Academic Emergency Medicine. J Hosp Med. 2009;4(6):364370.
  12. Kripalani S, Jackson AT, Schnipper JL, Coleman EA. Promoting effective transitions of care at hospital discharge: a review of key issues for hospitalists. J Hosp Med. 2007;2(5):314323.
  13. Coleman EA. Safety in numbers: physicians joining forces to seal the cracks during transitions. J Hosp Med. 2009;4(6):329330.
  14. Soong C, Daub S, Lee J, et al. Development of a checklist of safe discharge practices for hospital patients. J Hosp Med. 2013;8(8):444449.
  15. Hansen LO, Greenwald JL, Budnitz T, et al. Project BOOST: effectiveness of a multihospital effort to reduce rehospitalization. J Hosp Med. 2013;8(8):421427.
  16. Solan LG, Ranji SR, Shah SS. The successes and challenges of hospital to home transitions. J Hosp Med. 2014;9(4):271273.
  17. Greenwald JL, Halasyamani L, Greene J, et al. Making inpatient medication reconciliation patient centered, clinically relevant and implementable: a consensus statement on key principles and necessary first steps. J Hosp Med. 2010;5(8):477485.
References
  1. Institute of Medicine. Hospital‐based emergency care: At the breaking point. Washington, DC: National Academies Press; 2006. Available at: http://www.nap.edu/openbook.php?record_id=11621. Accessed September 13, 2015.
  2. Pitts SR, Pines JM, Handrigan MT, Kellermann AL. National trends in emergency department occupancy, 2001 to 2008: effect of inpatient admissions versus emergency department practice intensity. Ann Emerg Med. 2012;60(6):679686.e3.
  3. Schuur J, Venkatesh A. The growing role of emergency departments in hospital admissions. N Engl J Med. 2012;367(5):391393.
  4. Morganti KG, Bauhoff S, Blanchard J, et al. The Evolving Role of Emergency Departments in the United States. Santa Monica, CA: RAND Corp.; 2013:179.
  5. Leyenaar J, Shieh M‐S, Lagu T, Pekow PS, Lindenauer PK. Direct admission to hospitals among children in the United States. JAMA Pediatr. 2015;169(5):500502.
  6. Healthcare Cost and Utilization Project. National Inpatient Sample. 2012. Agency for Healthcare Research and Quality, Rockville, MD. Available at: www.hcup‐us.ahrq.gov/nisoverview.jsp. Accessed October 11, 2014.
  7. Leyenaar JK, O'Brien ER, Malkani N, Lagu T, Lindenauer PK. Direct admission to hospital: a mixed methods survey of pediatric practices, benefits, and challenges [published online August 17, 2015]. Acad Pediatr.
  8. Kocher KE, Dimick JB, Nallamothu BK. Changes in the source of unscheduled hospitalizations in the United States. Med Care. 2013;51(8):689698.
  9. Powell ES, Khare RK, Courtney DM, Feinglass J. Lower mortality in sepsis patients admitted through the ED vs direct admission. Am J Emerg Med. 2012;30(3):432439.
  10. Leyenaar JK, Shieh M, Lagu T, Pekow PS, Lindenauer PK. Variation and outcomes associated with direct admission among children with pneumonia in the United States. JAMA Pediatr. 2014;168(9):829836.
  11. Snow V, Beck D, Budnitz T, et al. Transitions of Care Consensus policy statement: American College of Physicians, Society of General Internal Medicine, Society of Hospital Medicine, American Geriatrics Society, American College Of Emergency Physicians, and Society for Academic Emergency Medicine. J Hosp Med. 2009;4(6):364370.
  12. Kripalani S, Jackson AT, Schnipper JL, Coleman EA. Promoting effective transitions of care at hospital discharge: a review of key issues for hospitalists. J Hosp Med. 2007;2(5):314323.
  13. Coleman EA. Safety in numbers: physicians joining forces to seal the cracks during transitions. J Hosp Med. 2009;4(6):329330.
  14. Soong C, Daub S, Lee J, et al. Development of a checklist of safe discharge practices for hospital patients. J Hosp Med. 2013;8(8):444449.
  15. Hansen LO, Greenwald JL, Budnitz T, et al. Project BOOST: effectiveness of a multihospital effort to reduce rehospitalization. J Hosp Med. 2013;8(8):421427.
  16. Solan LG, Ranji SR, Shah SS. The successes and challenges of hospital to home transitions. J Hosp Med. 2014;9(4):271273.
  17. Greenwald JL, Halasyamani L, Greene J, et al. Making inpatient medication reconciliation patient centered, clinically relevant and implementable: a consensus statement on key principles and necessary first steps. J Hosp Med. 2010;5(8):477485.
Issue
Journal of Hospital Medicine - 11(4)
Issue
Journal of Hospital Medicine - 11(4)
Page Number
303-305
Page Number
303-305
Publications
Publications
Article Type
Display Headline
Direct admission to the hospital: An alternative approach to hospitalization
Display Headline
Direct admission to the hospital: An alternative approach to hospitalization
Sections
Article Source
© 2015 Society of Hospital Medicine
Disallow All Ads
Correspondence Location
Address for correspondence and reprint requests: JoAnna K. Leyenaar, MD, Pediatric Hospitalist, Assistant Professor of Pediatrics, Tufts Medical Center, Boston, MA 02111; Telephone: 857‐244‐1381; Fax: 413‐794‐8866; E‐mail: jleyenaar@post.harvard.edu
Content Gating
Gated (full article locked unless allowed per User)
Gating Strategy
First Peek Free
Article PDF Media
Media Files

Using Social Media as a Hospital QI Tool

Article Type
Changed
Mon, 01/02/2017 - 19:34
Display Headline
Can social media be used as a hospital quality improvement tool?

Patient experience has become a major component of the Center for Medicare and Medicaid Services Value‐Based Purchasing initiative.[1] Hospitals have therefore focused quality improvement (QI) efforts on this area.[2] Hospital performance in the realm of patient experience is generally determined using systematic surveys with closed‐ended questions, but patient‐generated narrative feedback can help hospitals identify the components of care that contribute to patient satisfaction and or are in need of improvement.[3] Online narrative responses posted by patients on rating websites or social media have been criticized because they may not be representative of the population,[4] but they also have some advantages.[5] Any patient may leave a comment, not just those who are selected for a survey. Patients may also experience benefits through the act of sharing their story with others. Moreover, most US hospitals use some form of social media,[6] which they can theoretically use to self‐collect narrative data online. To realize the full potential of patient‐generated online narratives, we need a clearer understanding of the best practices for collecting and using these narratives. We therefore solicited patient feedback on the Facebook page of a large tertiary academic medical center to determine whether it is feasible to use social media platforms for learning about and improving hospital quality.

METHODS

Baystate Medical Center (BMC) is a tertiary care medical center in western Massachusetts. We identified key BMC stakeholders in the areas of QI and public affairs. Noting that patients have expressed interest in leaving comments via social media,[7] the group opted to perform a pilot study to obtain patient narratives via a Facebook prompt (Facebook is a social media site used by an estimated 58% of US adults[8]). The BMC public affairs department delivered a press release to the local media describing a 3‐week period during which patients were invited to leave narrative feedback on the BMC Facebook wall. The BMC Institutional Review Board deemed that this study did not constitute human subjects research.

During March 2014 (March 10, 2014March 24, 2014), we posted (once a week) an open‐ended prompt on BMC's Facebook wall. The prompt was designed to elicit novel descriptions of patient experience that could help to drive QI. It read: We want to hear about your experiences. In the comment section below, please tell us what we do well and how we can improve your care. Because of concerns about the potential reputational risks of allowing open feedback on a public social media page, the prompt also reminded patients of the social media ground rules: there should be no mention of specific physicians, nurses, or other caregivers by name (for liability reasons); and patients should not include details about their medical history (for privacy reasons).

We collected all posts to preserve comments and used directed qualitative content analysis to examine them.[9] Two research team members[3, 10, 11] independently coded the responses. Starting with an a priori codebook that was developed during a previous study,[3] they amended the codebook through an iterative process to incorporate new concepts. After independently coding all blocks of text, the coders reviewed their coding selections and resolved discrepancies through discussion. We then performed second‐level coding, in which codes were organized into major pertinent themes. We reviewed the coded text after applying secondary codes in order to check for accuracy of coding and theme assignment as well as completeness of second‐level coding. We calculated percent agreement, defined as both raters scoring a block of text with the same code divided by total number of codes. We also calculated the Spearman correlation between the 2 reviewers. We used descriptive statistics to assess the frequency of select codes and themes (see Supporting Information, Appendix 1 and Appendix 2, in the online version of this article).[9, 12, 13]

RESULTS

Over a 3‐week study period, 47 comments were submitted by 37 respondents. This yielded 148 codable statements (Table 1). Despite limited information on respondents, we ascertained from Facebook that 32 (86%) were women and 5 (14%) were men.

Number of Total, Positive, and Negative Comments and Representative Quotations for Themes
Theme Total Respondents, N (%) % Positive Positive Quotation % Negative Negative Quotation
  • NOTE: Abbreviations: ER, emergency room; IV, intravenous; NICU, neonatal intensive care unit.

Staff 17 (46) 45% The nurses in the pediatric unit, as well as the doctors in radiology and x‐ray department were AMAZING! 55% My 24‐year‐old daughter had to go for 5 days of IV treatmentwhile getting her infusion there was a fire alarm. She has a video showing the flashing of the light and the sound of the alarm and the closing of doors and NOT A SINGLE staff member to be found. Her infusions take about 2 hours. They set it and forget it. Luckily there wasn't a fire and someone did finally come to disconnect her.
Had a fabulous experience with Wesson women's this week! Had a C section and 3‐day admission. All staff from preoperative to inpatient were so helpful and really anticipated my needs before I could even ask for things. My mother was hospitalized for at least 3 weeks right after the cardiovascular center openedwhen she went into cardiac arrest and in acute care and the step unit the care was great, very attentive nurses and doctors. When she was starting to recover and moved upstairs, downhill it went. She'd ring for assistance because she wanted to walk to the bathrooms and more times she was left to her own devices because no one would respond.
Facility 9 (24) 25% New buildings are beautiful and the new signs are way better. 75% The parking situation was disappointing and the waiting room was also very dirty.
I really like the individual pods in the ER. I could have used a single room as my roommate was very annoying and demanding.
Departments 22 (60) 44% The NICU was great when my son was in there. The children's unit was great with my daughter and respected my needs. 56% Revamp maternity; it needs it desperately.
Labor and delivery was a great place. Love Baystate but hate the ER.
Technical aspects of care (eg, errors) 9 (24) 0 100% Day 2 of my 24 year old getting her 2‐hour IV infusion....she was set up with her IV. When checked 2 hours later, the staff member was very upset to find that only the saline had run. She never opened the medication clamp. So now they gave her the medication in 1 hour instead of 2.
If I had 1 suggestion it would be to re‐evaluate patient comfort when patients are waiting to be admitted.

From coded text, several broad themes were identified (see Table 1 for representative quotes): (1) comments about staff (17/37 respondents, 45.9%). These included positive descriptions of efficiency, caring behavior, good training, and good communication, whereas negative comments included descriptions of unfriendliness, apparent lack of caring, inattentiveness, poor training, unprofessional behavior, and poor communication; (2) comments about specific departments (22/37, 59.5%); (3) comments on technical aspects of care, including perceived errors, incorrect diagnoses, and inattention to pain control (9/37, 24.3%); and (4) comments describing the hospital physical plant, parking, and amenities (9/37, 24.3%). There were a few miscellaneous comments that did not fit into these broad themes, such as expressions of gratitude for our solicitation of narratives. Percent agreement between coders was 80% and Spearman's Rho was 0.82 (p<0.001).

A small number (n=3) of respondents repeatedly made comments over the 3‐week period, accounting for 30% (45/148) of codes. These repetitive commenters tended to dominate the Facebook conversation, at times describing the same experience more than once.

DISCUSSION

In this study evaluating the potential utility of social media as a hospital QI tool, several broad themes emerged. From these themes, we identified several areas that could be deemed as QI targets, including: training staff to be more responsive and sensitive to patients needs and concerns, improving patient and visitor parking, and reducing emergency department waiting times. However, the insight gained from solicited Facebook comments was similar to feedback gained from more traditional approaches of soliciting patient perspectives on care, such as patient experience surveys.[14]

Our findings should be viewed in the context of prior work focused on patient narratives in healthcare. Greaves et al. used sentiment analysis to describe the content of nearly 200,000 tweets (comments posted on the social networking website Twitter) sent to National Health Service (NHS) hospitals.[15] Themes were similar to those found in our study: (1) interaction with staff, (2) environment and facilities, and (3) issues of access and timeliness of service. Notably, these themes mirrored prior work examining narratives at NHS hospitals[3] and were similar to domains of commonly used surveys of patient experience.[14] The authors noted that there were issues with the signal to noise ratio (only about 10% of tweets were about quality) and the enforced brevity of Twitter (tweets must be 140 characters or less). These limitations suggest that using Twitter to identify QI targets would be difficult.

In contrast to Greaves et al., we chose to solicit feedback on our hospital's Facebook page. Facebook does not have Twitter's enforced brevity, allowing for more detailed narratives. In addition, we did not encounter the signal‐to‐noise problem, because our prompt was designed to request feedback that was relevant to recent experiences of care. However, a few respondents dominated the conversation, supporting the hypothesis that those most likely to comment may be the patients or families who have had the best or worst experiences. In the future, we will attempt to address this limitation and reduce the influence of repeat commenters by changing our prompt (eg, Please tell us about your experience, but please do not post descriptions of the same experience more than once.).

This pilot demonstrated some of the previously described benefits of online narratives.[5] First, there appears to be value in allowing patients to share their experiences and to read the experiences of others (as indicated in a few grateful patients comments). Second, soliciting online narratives offers a way for hospitals to demonstrate a commitment to transparency. Third, in contrast to closed‐ended survey questions, narrative comments help to identify why patients were satisfied or unsatisfied with their care. Although some surveys with closed‐ended questions also allow for narratives, these comments may or may not be carefully reviewed by the hospital. Using social media to solicit and respond to comments enhances existing methods for evaluating patient experience by engaging patients in a public space, which increases the likelihood that hospitals will attempt to improve care in response.

Notably, none of the identified areas for improvement could be considered novel QI targets for BMC. For example, our hospital has been very focused on training staff around patient experience, and emergency department wait times are the focus of a system‐wide improvement effort called Patient Progress.

This study has other limitations. We conducted this study over a 3‐week time period in a single center and on a single social media site whose members may not be representative of the overall patient population at BMC. Although we do not know how generalizable our findings are (in terms of identifying QI targets), we feel that we have demonstrated how using social media to collect data on patient experience is feasible and could be informative for other hospitals in other locations. It is possible that we did not allow the experiment to run long enough; a longer time or broader outreach (eg, a handout given to every discharged patient over a longer period) may be needed to allow patients adequate opportunity to comment. Of note, we did not specifically examine responses by time period, but it does seem, in post hoc analysis, that after 2 weeks of data collection we reached theoretical saturation with no new themes emerging in the third week (eg, third‐week comments included I heart your nurses. and Love Baystate but hate the ER.). More work is also needed that includes a broader range of social media platforms and more participating hospitals.

In conclusion, the opportunity to provide feedback on Facebook has the potential to engage and empower patients, and hospitals can use these online narratives to help to drive improvement efforts. Yet potential benefits must be weighed against reputational risks, a lack of representative respondents, and the paucity of novel QI targets obtained in this study.

Disclosures: Dr. Lagu is supported by the National Heart, Lung, and Blood Institute of the National Institutes of Health under award number K01HL114745. The authors report no conflicts of interest.

Files
References
  1. Centers for Medicare 47(2):193219.
  2. Lagu T, Goff SL, Hannon NS, Shatz A, Lindenauer PK. A mixed‐methods analysis of patient reviews of hospital care in England: implications for public reporting of health care quality data in the United States. Jt Comm J Qual Patient Saf. 2013;39(1):715.
  3. Schlesinger M, Grob R, Shaller D, Martino SC, Parker AM, Finucane ML, Cerully JL, Rybowski L. Taking Patients' Narratives about Clinicians from Anecdote to Science. NEJM. 2015;373(7):675679.
  4. Lagu T, Lindenauer PK. Putting the public back in public reporting of health care quality. JAMA. 2010;304(15):17111712.
  5. Griffis HM, Kilaru AS, Werner RM, et al. Use of social media across US hospitals: descriptive analysis of adoption and utilization. J Med Internet Res. 2014;16(11):e264.
  6. Lee JL, Choudhry NK, Wu AW, Matlin OS, Brennan TA, Shrank WH. Patient use of email, Facebook, and physician websites to communicate with physicians: a national online survey of retail pharmacy users [published online June 24, 2015]. J Gen Intern Med. doi:10.1007/s11606-015-3374-7.
  7. Pew Research Center. Social networking fact sheet. Available at: http://www.pewinternet.org/fact‐sheets/social‐networking‐fact‐sheet. Accessed March 4, 2015.
  8. Hsieh H‐F, Shannon SE. Three approaches to qualitative content analysis. Qual Health Res. 2005;15(9):12771288.
  9. Lagu T, Hannon NS, Rothberg MB, Lindenauer PK. Patients’ evaluations of health care providers in the era of social networking: an analysis of physician‐rating websites. J Gen Intern Med. 2010;25(9):942946.
  10. Goff SL, Mazor KM, Gagne SJ, Corey KC, Blake DR. Vaccine counseling: a content analysis of patient‐physician discussions regarding human papilloma virus vaccine. Vaccine. 2011;29(43):73437349.
  11. Sofaer S. Qualitative research methods. Int J Qual Health Care. 2002;14(4):329336.
  12. Crabtree BF, Miller WL. Doing Qualitative Research. Vol 2. Thousand Oaks, CA: Sage Publications; 1999.
  13. Jha AK, Orav EJ, Zheng J, Epstein AM. Patients’ perception of hospital care in the United States. N Engl J Med. 2008;359(18):19211931.
  14. Greaves F, Laverty AA, Cano DR, et al. Tweets about hospital quality: a mixed methods study. BMJ Qual Saf. 2014;23(10):838846.
Article PDF
Issue
Journal of Hospital Medicine - 11(1)
Publications
Page Number
52-55
Sections
Files
Files
Article PDF
Article PDF

Patient experience has become a major component of the Center for Medicare and Medicaid Services Value‐Based Purchasing initiative.[1] Hospitals have therefore focused quality improvement (QI) efforts on this area.[2] Hospital performance in the realm of patient experience is generally determined using systematic surveys with closed‐ended questions, but patient‐generated narrative feedback can help hospitals identify the components of care that contribute to patient satisfaction and or are in need of improvement.[3] Online narrative responses posted by patients on rating websites or social media have been criticized because they may not be representative of the population,[4] but they also have some advantages.[5] Any patient may leave a comment, not just those who are selected for a survey. Patients may also experience benefits through the act of sharing their story with others. Moreover, most US hospitals use some form of social media,[6] which they can theoretically use to self‐collect narrative data online. To realize the full potential of patient‐generated online narratives, we need a clearer understanding of the best practices for collecting and using these narratives. We therefore solicited patient feedback on the Facebook page of a large tertiary academic medical center to determine whether it is feasible to use social media platforms for learning about and improving hospital quality.

METHODS

Baystate Medical Center (BMC) is a tertiary care medical center in western Massachusetts. We identified key BMC stakeholders in the areas of QI and public affairs. Noting that patients have expressed interest in leaving comments via social media,[7] the group opted to perform a pilot study to obtain patient narratives via a Facebook prompt (Facebook is a social media site used by an estimated 58% of US adults[8]). The BMC public affairs department delivered a press release to the local media describing a 3‐week period during which patients were invited to leave narrative feedback on the BMC Facebook wall. The BMC Institutional Review Board deemed that this study did not constitute human subjects research.

During March 2014 (March 10, 2014March 24, 2014), we posted (once a week) an open‐ended prompt on BMC's Facebook wall. The prompt was designed to elicit novel descriptions of patient experience that could help to drive QI. It read: We want to hear about your experiences. In the comment section below, please tell us what we do well and how we can improve your care. Because of concerns about the potential reputational risks of allowing open feedback on a public social media page, the prompt also reminded patients of the social media ground rules: there should be no mention of specific physicians, nurses, or other caregivers by name (for liability reasons); and patients should not include details about their medical history (for privacy reasons).

We collected all posts to preserve comments and used directed qualitative content analysis to examine them.[9] Two research team members[3, 10, 11] independently coded the responses. Starting with an a priori codebook that was developed during a previous study,[3] they amended the codebook through an iterative process to incorporate new concepts. After independently coding all blocks of text, the coders reviewed their coding selections and resolved discrepancies through discussion. We then performed second‐level coding, in which codes were organized into major pertinent themes. We reviewed the coded text after applying secondary codes in order to check for accuracy of coding and theme assignment as well as completeness of second‐level coding. We calculated percent agreement, defined as both raters scoring a block of text with the same code divided by total number of codes. We also calculated the Spearman correlation between the 2 reviewers. We used descriptive statistics to assess the frequency of select codes and themes (see Supporting Information, Appendix 1 and Appendix 2, in the online version of this article).[9, 12, 13]

RESULTS

Over a 3‐week study period, 47 comments were submitted by 37 respondents. This yielded 148 codable statements (Table 1). Despite limited information on respondents, we ascertained from Facebook that 32 (86%) were women and 5 (14%) were men.

Number of Total, Positive, and Negative Comments and Representative Quotations for Themes
Theme Total Respondents, N (%) % Positive Positive Quotation % Negative Negative Quotation
  • NOTE: Abbreviations: ER, emergency room; IV, intravenous; NICU, neonatal intensive care unit.

Staff 17 (46) 45% The nurses in the pediatric unit, as well as the doctors in radiology and x‐ray department were AMAZING! 55% My 24‐year‐old daughter had to go for 5 days of IV treatmentwhile getting her infusion there was a fire alarm. She has a video showing the flashing of the light and the sound of the alarm and the closing of doors and NOT A SINGLE staff member to be found. Her infusions take about 2 hours. They set it and forget it. Luckily there wasn't a fire and someone did finally come to disconnect her.
Had a fabulous experience with Wesson women's this week! Had a C section and 3‐day admission. All staff from preoperative to inpatient were so helpful and really anticipated my needs before I could even ask for things. My mother was hospitalized for at least 3 weeks right after the cardiovascular center openedwhen she went into cardiac arrest and in acute care and the step unit the care was great, very attentive nurses and doctors. When she was starting to recover and moved upstairs, downhill it went. She'd ring for assistance because she wanted to walk to the bathrooms and more times she was left to her own devices because no one would respond.
Facility 9 (24) 25% New buildings are beautiful and the new signs are way better. 75% The parking situation was disappointing and the waiting room was also very dirty.
I really like the individual pods in the ER. I could have used a single room as my roommate was very annoying and demanding.
Departments 22 (60) 44% The NICU was great when my son was in there. The children's unit was great with my daughter and respected my needs. 56% Revamp maternity; it needs it desperately.
Labor and delivery was a great place. Love Baystate but hate the ER.
Technical aspects of care (eg, errors) 9 (24) 0 100% Day 2 of my 24 year old getting her 2‐hour IV infusion....she was set up with her IV. When checked 2 hours later, the staff member was very upset to find that only the saline had run. She never opened the medication clamp. So now they gave her the medication in 1 hour instead of 2.
If I had 1 suggestion it would be to re‐evaluate patient comfort when patients are waiting to be admitted.

From coded text, several broad themes were identified (see Table 1 for representative quotes): (1) comments about staff (17/37 respondents, 45.9%). These included positive descriptions of efficiency, caring behavior, good training, and good communication, whereas negative comments included descriptions of unfriendliness, apparent lack of caring, inattentiveness, poor training, unprofessional behavior, and poor communication; (2) comments about specific departments (22/37, 59.5%); (3) comments on technical aspects of care, including perceived errors, incorrect diagnoses, and inattention to pain control (9/37, 24.3%); and (4) comments describing the hospital physical plant, parking, and amenities (9/37, 24.3%). There were a few miscellaneous comments that did not fit into these broad themes, such as expressions of gratitude for our solicitation of narratives. Percent agreement between coders was 80% and Spearman's Rho was 0.82 (p<0.001).

A small number (n=3) of respondents repeatedly made comments over the 3‐week period, accounting for 30% (45/148) of codes. These repetitive commenters tended to dominate the Facebook conversation, at times describing the same experience more than once.

DISCUSSION

In this study evaluating the potential utility of social media as a hospital QI tool, several broad themes emerged. From these themes, we identified several areas that could be deemed as QI targets, including: training staff to be more responsive and sensitive to patients needs and concerns, improving patient and visitor parking, and reducing emergency department waiting times. However, the insight gained from solicited Facebook comments was similar to feedback gained from more traditional approaches of soliciting patient perspectives on care, such as patient experience surveys.[14]

Our findings should be viewed in the context of prior work focused on patient narratives in healthcare. Greaves et al. used sentiment analysis to describe the content of nearly 200,000 tweets (comments posted on the social networking website Twitter) sent to National Health Service (NHS) hospitals.[15] Themes were similar to those found in our study: (1) interaction with staff, (2) environment and facilities, and (3) issues of access and timeliness of service. Notably, these themes mirrored prior work examining narratives at NHS hospitals[3] and were similar to domains of commonly used surveys of patient experience.[14] The authors noted that there were issues with the signal to noise ratio (only about 10% of tweets were about quality) and the enforced brevity of Twitter (tweets must be 140 characters or less). These limitations suggest that using Twitter to identify QI targets would be difficult.

In contrast to Greaves et al., we chose to solicit feedback on our hospital's Facebook page. Facebook does not have Twitter's enforced brevity, allowing for more detailed narratives. In addition, we did not encounter the signal‐to‐noise problem, because our prompt was designed to request feedback that was relevant to recent experiences of care. However, a few respondents dominated the conversation, supporting the hypothesis that those most likely to comment may be the patients or families who have had the best or worst experiences. In the future, we will attempt to address this limitation and reduce the influence of repeat commenters by changing our prompt (eg, Please tell us about your experience, but please do not post descriptions of the same experience more than once.).

This pilot demonstrated some of the previously described benefits of online narratives.[5] First, there appears to be value in allowing patients to share their experiences and to read the experiences of others (as indicated in a few grateful patients comments). Second, soliciting online narratives offers a way for hospitals to demonstrate a commitment to transparency. Third, in contrast to closed‐ended survey questions, narrative comments help to identify why patients were satisfied or unsatisfied with their care. Although some surveys with closed‐ended questions also allow for narratives, these comments may or may not be carefully reviewed by the hospital. Using social media to solicit and respond to comments enhances existing methods for evaluating patient experience by engaging patients in a public space, which increases the likelihood that hospitals will attempt to improve care in response.

Notably, none of the identified areas for improvement could be considered novel QI targets for BMC. For example, our hospital has been very focused on training staff around patient experience, and emergency department wait times are the focus of a system‐wide improvement effort called Patient Progress.

This study has other limitations. We conducted this study over a 3‐week time period in a single center and on a single social media site whose members may not be representative of the overall patient population at BMC. Although we do not know how generalizable our findings are (in terms of identifying QI targets), we feel that we have demonstrated how using social media to collect data on patient experience is feasible and could be informative for other hospitals in other locations. It is possible that we did not allow the experiment to run long enough; a longer time or broader outreach (eg, a handout given to every discharged patient over a longer period) may be needed to allow patients adequate opportunity to comment. Of note, we did not specifically examine responses by time period, but it does seem, in post hoc analysis, that after 2 weeks of data collection we reached theoretical saturation with no new themes emerging in the third week (eg, third‐week comments included I heart your nurses. and Love Baystate but hate the ER.). More work is also needed that includes a broader range of social media platforms and more participating hospitals.

In conclusion, the opportunity to provide feedback on Facebook has the potential to engage and empower patients, and hospitals can use these online narratives to help to drive improvement efforts. Yet potential benefits must be weighed against reputational risks, a lack of representative respondents, and the paucity of novel QI targets obtained in this study.

Disclosures: Dr. Lagu is supported by the National Heart, Lung, and Blood Institute of the National Institutes of Health under award number K01HL114745. The authors report no conflicts of interest.

Patient experience has become a major component of the Center for Medicare and Medicaid Services Value‐Based Purchasing initiative.[1] Hospitals have therefore focused quality improvement (QI) efforts on this area.[2] Hospital performance in the realm of patient experience is generally determined using systematic surveys with closed‐ended questions, but patient‐generated narrative feedback can help hospitals identify the components of care that contribute to patient satisfaction and or are in need of improvement.[3] Online narrative responses posted by patients on rating websites or social media have been criticized because they may not be representative of the population,[4] but they also have some advantages.[5] Any patient may leave a comment, not just those who are selected for a survey. Patients may also experience benefits through the act of sharing their story with others. Moreover, most US hospitals use some form of social media,[6] which they can theoretically use to self‐collect narrative data online. To realize the full potential of patient‐generated online narratives, we need a clearer understanding of the best practices for collecting and using these narratives. We therefore solicited patient feedback on the Facebook page of a large tertiary academic medical center to determine whether it is feasible to use social media platforms for learning about and improving hospital quality.

METHODS

Baystate Medical Center (BMC) is a tertiary care medical center in western Massachusetts. We identified key BMC stakeholders in the areas of QI and public affairs. Noting that patients have expressed interest in leaving comments via social media,[7] the group opted to perform a pilot study to obtain patient narratives via a Facebook prompt (Facebook is a social media site used by an estimated 58% of US adults[8]). The BMC public affairs department delivered a press release to the local media describing a 3‐week period during which patients were invited to leave narrative feedback on the BMC Facebook wall. The BMC Institutional Review Board deemed that this study did not constitute human subjects research.

During March 2014 (March 10, 2014March 24, 2014), we posted (once a week) an open‐ended prompt on BMC's Facebook wall. The prompt was designed to elicit novel descriptions of patient experience that could help to drive QI. It read: We want to hear about your experiences. In the comment section below, please tell us what we do well and how we can improve your care. Because of concerns about the potential reputational risks of allowing open feedback on a public social media page, the prompt also reminded patients of the social media ground rules: there should be no mention of specific physicians, nurses, or other caregivers by name (for liability reasons); and patients should not include details about their medical history (for privacy reasons).

We collected all posts to preserve comments and used directed qualitative content analysis to examine them.[9] Two research team members[3, 10, 11] independently coded the responses. Starting with an a priori codebook that was developed during a previous study,[3] they amended the codebook through an iterative process to incorporate new concepts. After independently coding all blocks of text, the coders reviewed their coding selections and resolved discrepancies through discussion. We then performed second‐level coding, in which codes were organized into major pertinent themes. We reviewed the coded text after applying secondary codes in order to check for accuracy of coding and theme assignment as well as completeness of second‐level coding. We calculated percent agreement, defined as both raters scoring a block of text with the same code divided by total number of codes. We also calculated the Spearman correlation between the 2 reviewers. We used descriptive statistics to assess the frequency of select codes and themes (see Supporting Information, Appendix 1 and Appendix 2, in the online version of this article).[9, 12, 13]

RESULTS

Over a 3‐week study period, 47 comments were submitted by 37 respondents. This yielded 148 codable statements (Table 1). Despite limited information on respondents, we ascertained from Facebook that 32 (86%) were women and 5 (14%) were men.

Number of Total, Positive, and Negative Comments and Representative Quotations for Themes
Theme Total Respondents, N (%) % Positive Positive Quotation % Negative Negative Quotation
  • NOTE: Abbreviations: ER, emergency room; IV, intravenous; NICU, neonatal intensive care unit.

Staff 17 (46) 45% The nurses in the pediatric unit, as well as the doctors in radiology and x‐ray department were AMAZING! 55% My 24‐year‐old daughter had to go for 5 days of IV treatmentwhile getting her infusion there was a fire alarm. She has a video showing the flashing of the light and the sound of the alarm and the closing of doors and NOT A SINGLE staff member to be found. Her infusions take about 2 hours. They set it and forget it. Luckily there wasn't a fire and someone did finally come to disconnect her.
Had a fabulous experience with Wesson women's this week! Had a C section and 3‐day admission. All staff from preoperative to inpatient were so helpful and really anticipated my needs before I could even ask for things. My mother was hospitalized for at least 3 weeks right after the cardiovascular center openedwhen she went into cardiac arrest and in acute care and the step unit the care was great, very attentive nurses and doctors. When she was starting to recover and moved upstairs, downhill it went. She'd ring for assistance because she wanted to walk to the bathrooms and more times she was left to her own devices because no one would respond.
Facility 9 (24) 25% New buildings are beautiful and the new signs are way better. 75% The parking situation was disappointing and the waiting room was also very dirty.
I really like the individual pods in the ER. I could have used a single room as my roommate was very annoying and demanding.
Departments 22 (60) 44% The NICU was great when my son was in there. The children's unit was great with my daughter and respected my needs. 56% Revamp maternity; it needs it desperately.
Labor and delivery was a great place. Love Baystate but hate the ER.
Technical aspects of care (eg, errors) 9 (24) 0 100% Day 2 of my 24 year old getting her 2‐hour IV infusion....she was set up with her IV. When checked 2 hours later, the staff member was very upset to find that only the saline had run. She never opened the medication clamp. So now they gave her the medication in 1 hour instead of 2.
If I had 1 suggestion it would be to re‐evaluate patient comfort when patients are waiting to be admitted.

From coded text, several broad themes were identified (see Table 1 for representative quotes): (1) comments about staff (17/37 respondents, 45.9%). These included positive descriptions of efficiency, caring behavior, good training, and good communication, whereas negative comments included descriptions of unfriendliness, apparent lack of caring, inattentiveness, poor training, unprofessional behavior, and poor communication; (2) comments about specific departments (22/37, 59.5%); (3) comments on technical aspects of care, including perceived errors, incorrect diagnoses, and inattention to pain control (9/37, 24.3%); and (4) comments describing the hospital physical plant, parking, and amenities (9/37, 24.3%). There were a few miscellaneous comments that did not fit into these broad themes, such as expressions of gratitude for our solicitation of narratives. Percent agreement between coders was 80% and Spearman's Rho was 0.82 (p<0.001).

A small number (n=3) of respondents repeatedly made comments over the 3‐week period, accounting for 30% (45/148) of codes. These repetitive commenters tended to dominate the Facebook conversation, at times describing the same experience more than once.

DISCUSSION

In this study evaluating the potential utility of social media as a hospital QI tool, several broad themes emerged. From these themes, we identified several areas that could be deemed as QI targets, including: training staff to be more responsive and sensitive to patients needs and concerns, improving patient and visitor parking, and reducing emergency department waiting times. However, the insight gained from solicited Facebook comments was similar to feedback gained from more traditional approaches of soliciting patient perspectives on care, such as patient experience surveys.[14]

Our findings should be viewed in the context of prior work focused on patient narratives in healthcare. Greaves et al. used sentiment analysis to describe the content of nearly 200,000 tweets (comments posted on the social networking website Twitter) sent to National Health Service (NHS) hospitals.[15] Themes were similar to those found in our study: (1) interaction with staff, (2) environment and facilities, and (3) issues of access and timeliness of service. Notably, these themes mirrored prior work examining narratives at NHS hospitals[3] and were similar to domains of commonly used surveys of patient experience.[14] The authors noted that there were issues with the signal to noise ratio (only about 10% of tweets were about quality) and the enforced brevity of Twitter (tweets must be 140 characters or less). These limitations suggest that using Twitter to identify QI targets would be difficult.

In contrast to Greaves et al., we chose to solicit feedback on our hospital's Facebook page. Facebook does not have Twitter's enforced brevity, allowing for more detailed narratives. In addition, we did not encounter the signal‐to‐noise problem, because our prompt was designed to request feedback that was relevant to recent experiences of care. However, a few respondents dominated the conversation, supporting the hypothesis that those most likely to comment may be the patients or families who have had the best or worst experiences. In the future, we will attempt to address this limitation and reduce the influence of repeat commenters by changing our prompt (eg, Please tell us about your experience, but please do not post descriptions of the same experience more than once.).

This pilot demonstrated some of the previously described benefits of online narratives.[5] First, there appears to be value in allowing patients to share their experiences and to read the experiences of others (as indicated in a few grateful patients comments). Second, soliciting online narratives offers a way for hospitals to demonstrate a commitment to transparency. Third, in contrast to closed‐ended survey questions, narrative comments help to identify why patients were satisfied or unsatisfied with their care. Although some surveys with closed‐ended questions also allow for narratives, these comments may or may not be carefully reviewed by the hospital. Using social media to solicit and respond to comments enhances existing methods for evaluating patient experience by engaging patients in a public space, which increases the likelihood that hospitals will attempt to improve care in response.

Notably, none of the identified areas for improvement could be considered novel QI targets for BMC. For example, our hospital has been very focused on training staff around patient experience, and emergency department wait times are the focus of a system‐wide improvement effort called Patient Progress.

This study has other limitations. We conducted this study over a 3‐week time period in a single center and on a single social media site whose members may not be representative of the overall patient population at BMC. Although we do not know how generalizable our findings are (in terms of identifying QI targets), we feel that we have demonstrated how using social media to collect data on patient experience is feasible and could be informative for other hospitals in other locations. It is possible that we did not allow the experiment to run long enough; a longer time or broader outreach (eg, a handout given to every discharged patient over a longer period) may be needed to allow patients adequate opportunity to comment. Of note, we did not specifically examine responses by time period, but it does seem, in post hoc analysis, that after 2 weeks of data collection we reached theoretical saturation with no new themes emerging in the third week (eg, third‐week comments included I heart your nurses. and Love Baystate but hate the ER.). More work is also needed that includes a broader range of social media platforms and more participating hospitals.

In conclusion, the opportunity to provide feedback on Facebook has the potential to engage and empower patients, and hospitals can use these online narratives to help to drive improvement efforts. Yet potential benefits must be weighed against reputational risks, a lack of representative respondents, and the paucity of novel QI targets obtained in this study.

Disclosures: Dr. Lagu is supported by the National Heart, Lung, and Blood Institute of the National Institutes of Health under award number K01HL114745. The authors report no conflicts of interest.

References
  1. Centers for Medicare 47(2):193219.
  2. Lagu T, Goff SL, Hannon NS, Shatz A, Lindenauer PK. A mixed‐methods analysis of patient reviews of hospital care in England: implications for public reporting of health care quality data in the United States. Jt Comm J Qual Patient Saf. 2013;39(1):715.
  3. Schlesinger M, Grob R, Shaller D, Martino SC, Parker AM, Finucane ML, Cerully JL, Rybowski L. Taking Patients' Narratives about Clinicians from Anecdote to Science. NEJM. 2015;373(7):675679.
  4. Lagu T, Lindenauer PK. Putting the public back in public reporting of health care quality. JAMA. 2010;304(15):17111712.
  5. Griffis HM, Kilaru AS, Werner RM, et al. Use of social media across US hospitals: descriptive analysis of adoption and utilization. J Med Internet Res. 2014;16(11):e264.
  6. Lee JL, Choudhry NK, Wu AW, Matlin OS, Brennan TA, Shrank WH. Patient use of email, Facebook, and physician websites to communicate with physicians: a national online survey of retail pharmacy users [published online June 24, 2015]. J Gen Intern Med. doi:10.1007/s11606-015-3374-7.
  7. Pew Research Center. Social networking fact sheet. Available at: http://www.pewinternet.org/fact‐sheets/social‐networking‐fact‐sheet. Accessed March 4, 2015.
  8. Hsieh H‐F, Shannon SE. Three approaches to qualitative content analysis. Qual Health Res. 2005;15(9):12771288.
  9. Lagu T, Hannon NS, Rothberg MB, Lindenauer PK. Patients’ evaluations of health care providers in the era of social networking: an analysis of physician‐rating websites. J Gen Intern Med. 2010;25(9):942946.
  10. Goff SL, Mazor KM, Gagne SJ, Corey KC, Blake DR. Vaccine counseling: a content analysis of patient‐physician discussions regarding human papilloma virus vaccine. Vaccine. 2011;29(43):73437349.
  11. Sofaer S. Qualitative research methods. Int J Qual Health Care. 2002;14(4):329336.
  12. Crabtree BF, Miller WL. Doing Qualitative Research. Vol 2. Thousand Oaks, CA: Sage Publications; 1999.
  13. Jha AK, Orav EJ, Zheng J, Epstein AM. Patients’ perception of hospital care in the United States. N Engl J Med. 2008;359(18):19211931.
  14. Greaves F, Laverty AA, Cano DR, et al. Tweets about hospital quality: a mixed methods study. BMJ Qual Saf. 2014;23(10):838846.
References
  1. Centers for Medicare 47(2):193219.
  2. Lagu T, Goff SL, Hannon NS, Shatz A, Lindenauer PK. A mixed‐methods analysis of patient reviews of hospital care in England: implications for public reporting of health care quality data in the United States. Jt Comm J Qual Patient Saf. 2013;39(1):715.
  3. Schlesinger M, Grob R, Shaller D, Martino SC, Parker AM, Finucane ML, Cerully JL, Rybowski L. Taking Patients' Narratives about Clinicians from Anecdote to Science. NEJM. 2015;373(7):675679.
  4. Lagu T, Lindenauer PK. Putting the public back in public reporting of health care quality. JAMA. 2010;304(15):17111712.
  5. Griffis HM, Kilaru AS, Werner RM, et al. Use of social media across US hospitals: descriptive analysis of adoption and utilization. J Med Internet Res. 2014;16(11):e264.
  6. Lee JL, Choudhry NK, Wu AW, Matlin OS, Brennan TA, Shrank WH. Patient use of email, Facebook, and physician websites to communicate with physicians: a national online survey of retail pharmacy users [published online June 24, 2015]. J Gen Intern Med. doi:10.1007/s11606-015-3374-7.
  7. Pew Research Center. Social networking fact sheet. Available at: http://www.pewinternet.org/fact‐sheets/social‐networking‐fact‐sheet. Accessed March 4, 2015.
  8. Hsieh H‐F, Shannon SE. Three approaches to qualitative content analysis. Qual Health Res. 2005;15(9):12771288.
  9. Lagu T, Hannon NS, Rothberg MB, Lindenauer PK. Patients’ evaluations of health care providers in the era of social networking: an analysis of physician‐rating websites. J Gen Intern Med. 2010;25(9):942946.
  10. Goff SL, Mazor KM, Gagne SJ, Corey KC, Blake DR. Vaccine counseling: a content analysis of patient‐physician discussions regarding human papilloma virus vaccine. Vaccine. 2011;29(43):73437349.
  11. Sofaer S. Qualitative research methods. Int J Qual Health Care. 2002;14(4):329336.
  12. Crabtree BF, Miller WL. Doing Qualitative Research. Vol 2. Thousand Oaks, CA: Sage Publications; 1999.
  13. Jha AK, Orav EJ, Zheng J, Epstein AM. Patients’ perception of hospital care in the United States. N Engl J Med. 2008;359(18):19211931.
  14. Greaves F, Laverty AA, Cano DR, et al. Tweets about hospital quality: a mixed methods study. BMJ Qual Saf. 2014;23(10):838846.
Issue
Journal of Hospital Medicine - 11(1)
Issue
Journal of Hospital Medicine - 11(1)
Page Number
52-55
Page Number
52-55
Publications
Publications
Article Type
Display Headline
Can social media be used as a hospital quality improvement tool?
Display Headline
Can social media be used as a hospital quality improvement tool?
Sections
Article Source
© 2015 Society of Hospital Medicine
Disallow All Ads
Correspondence Location
Address for correspondence and reprint requests: Tara Lagu, MD, MPH, Center for Quality of Care Research, Baystate Medical Center, 280 Chestnut St., Springfield, MA 01199; Telephone: 413‐794‐7688; Fax: 413‐794‐8866; E‐mail: lagutc@gmail.com
Content Gating
Gated (full article locked unless allowed per User)
Gating Strategy
First Peek Free
Article PDF Media
Media Files

Dyspnea Assessment and Management Survey

Article Type
Changed
Tue, 05/16/2017 - 22:57
Display Headline
Hospitalist attitudes toward the assessment and management of dyspnea in patients with acute cardiopulmonary diseases

Dyspnea, defined as a subjective experience of breathing discomfort,[1] is the seventh most frequent reason adult patients present to the emergency room and the most frequent cause for emergency room visits in patients 65 years or older.[2] Moreover, dyspnea is experienced by 49% of patients hospitalized with a medical condition[3, 4, 5] and by 70% of patients who are seriously ill.[6]

Based on evidence that patients are not treated consistently and effectively for relief of their shortness of breath, the American College of Chest Physicians (ACCP) statement on dyspnea management in patients with advanced lung or heart disease recommended that patients should be asked to rate their dyspnea, and the rating should be routinely documented in the medical record to guide management.[7] Although clinicians may question the utility of routine assessment of dyspnea using a standardized scale, studies have found that the prevalence of dyspnea reported from chart review is much lower than when patients are directly interviewed.[8] This may be the result of underrecognition of dyspnea or poor documentation by physicians, or that patients may not communicate their symptoms unless the physician specifically asks. As is the case with pain, routine assessment of dyspnea severity could lead to improved clinical management and greater patient‐centered care. However, unlike in the case of pain, regulatory bodies, such as the Joint Commission for Accreditation of Healthcare Organization, do not require routine dyspnea assessment.[9]

Currently, there are more than 40,000 hospitalists in the United States, and the vast majority of hospitals with >200 beds have a hospitalist group.[10] Hospitalists care for over 60% of inpatients[11] and play a major role in the management of patients with acute cardiopulmonary diseases. If standardized approaches for the assessment and documentation of dyspnea are to be implemented, hospitalists would be a key stakeholder group for utilizing enhanced clinical information about dyspnea. Therefore, we evaluated attitudes and practices of hospitalists in regard to the assessment and management of dyspnea, including the potential benefits and challenges related to the implementation of standardized assessment. We hypothesized that hospitalists would believe that a dyspnea scale for assessment of severity could improve their management of patients with cardiovascular diseases. Further, we hypothesized that physicians who agreed with the general statement that dyspnea is an important clinical problem would be more likely to believe that routine dyspnea assessment would be valuable.

METHODS

Study Sample

We invited 255 attending hospitalists from 9 geographically and structurally diverse hospitals to complete a survey about the assessment and management of dyspnea. The 9 hospitals represent range of practice environments including 4 academic medical centers, 2 community teaching and 3 nonteaching hospitals, 1 Veterans Administration hospital, and 2 staff‐model HMOs (see Supporting Table 1 in the online version of this article). The survey was distributed online using REDCap (Research Electronic Data Capture), a secure web‐based interface application.[12] A coinvestigator who was a pulmonary critical‐care physician at each site sent an initial email to their hospitalist groups that alerted them to expect a survey from the principal investigator. This notification was subsequently followed by an email invitation containing an informational cover letter and a link to the online survey. The cover letter stated that the completed surveys would not be stored at the local sites and that all the analyzed data would be deidentified. Nonrespondents were sent reminders at 2 and 4 weeks after the initial mailing. A $25 electronic gift card was provided as a gesture of appreciation for their time. The survey was conducted between September 2013 and December 2013.

The study was approved by the Baystate Health Institutional Review Board, Springfield, Massachusetts, with a waiver for written informed consent.

Questionnaire

We developed a 17‐item instrument based on a review of the dyspnea literature and a prior ACCP survey.[12] Questions were piloted with 4 hospitalists at a single institution and modified to improve face validity and clarity (see Supporting Information in the online version of this article for the full survey).

Hospitalists were asked to consider the care of patients admitted for acute cardiopulmonary disease, including heart failure, chronic obstructive pulmonary disease, and pneumonia. A series of 5‐point Likert scales were used to assess the respondents level of agreement with statements related to the following domains: the importance of dyspnea in clinical care, the potential benefits and challenges of routine dyspnea assessment (statements such as: Having a standardized assessment of dyspnea severity would be helpful in management of patients with cardiopulmonary diseases. Dyspnea assessment by a scale should be part of the vital signs for patients with cardiopulmonary diseases.), and management of dyspnea (questions regarding the use of opioids and other nonpharmacological therapies). Additional questions were asked about current assessment practices (questions such as: How often do you assess severity of dyspnea? What is your approach in assessing dyspnea? with options of choosing a categorical or numerical scale), if dyspnea is assessed in their institution by nurses and how often, and the influence of dyspnea severity assessment on their management. The survey had 1 question that solicited comments from the participants: If you don't think that it would be useful to have a standardized dyspnea assessment, please tell us why.

Data Analysis

Responses to survey questions were summarized via counts and percentages in each response category. Adopting the methodology used in the ACCP consensus statement, strongly agree and somewhat agree were combined into a single category of agreement. We also presented percentage of responses in the 2 levels of agreement (strongly agree and somewhat agree) for each question in a bar graph.

Associations between tertiles of physicians' time in practice and attitude toward dyspnea were evaluated via 2 or Fisher exact test.

To examine how answers to the first 2 questions, which assessed attitude toward importance of dyspnea in clinical care, affect answers to the remaining questions, we grouped respondents in 3 categories (strongly agree, agree to these questions, do not agree) and tested the associations using 2 or Fisher exact test.

All analyses were performed using SAS version 9.3 (SAS institute, Inc., Cary, NC) and Stata release 13.1 (StataCorp, College Station, TX).

RESULTS

Overall, 178 (69.8%) of 255 identified hospitalists completed the survey, and all 9 participating hospitals had a response rate greater than 50%. The median number of years in practice was 6 (range, 038 years). A majority (77.5%) of respondents agreed with the statement that dyspnea is 1 of the major symptoms of patients with cardiopulmonary disease, and that its treatment is central to the management of these patients (77.0%) (Figure 1).

Figure 1
Attitude and practices regarding dyspnea assessment and management.

Attitude and Practices Surrounding Dyspnea Assessment

When asked about their current assessment of dyspnea, a majority (84.3%) of the hospitalists stated that they assess dyspnea on a daily basis; two‐thirds indicated that they use a categorical scale (ie, no shortness of breath, improved or worsened compared with a prior date), and one‐third indicated that they ask whether the patient is dyspneic or not. Fifty‐six percent of hospitalists stated that dyspnea is regularly assessed by nurses in their hospital.

The majority of respondents agreed (78.6%, 23.0% strongly and 55.6% somewhat agree) that standardized assessment of dyspnea severity, using a numeric scale and serial measurements as part of the vital signs, would benefit the management of patients with cardiopulmonary diseases. Furthermore, 79.6% (33.0% strongly and 46.6% somewhat agree) reported that using a dyspnea scale that included information to further characterize the patient‐reported experience, such as the level of distress associated with dyspnea, would be helpful in management.

Approximately 90% of the hospitalists indicated that awareness of dyspnea severity has an influence on clinical decision making, including whether to intensify treatment of underlying conditions, to pursue additional diagnostic testing, or to modify discharge timing. Additionally, two‐thirds of hospitalists agreed that awareness of dyspnea severity influences their decision to add opioids, whereas only one‐third prescribed nonpharmacologic symptom‐oriented treatment (Table 1).

Responses of Hospitalists to Questions Regarding Current Assessment and Management of Dyspnea
 Frequency (%)
  • NOTE: Abbreviations: SOB, shortness of breath. *Survey respondents selected all that apply, so these numbers are not mutually exclusive.

When caring for patients with acute cardiopulmonary diseases, how often do you assess severity of dyspnea?*
At admission66 (37.1)
At discharge59 (33.2)
Daily until discharge150 (84.3)
More often than daily58 (32.6)
Which description best characterizes your approach to assessing dyspnea severity?
I don't regularly ask about dyspnea severity3 (1.7)
I ask the patient whether or not they are having shortness of breath50 (28.3)
I ask the patient to rate the severity of shortness of breath using a numeric scale4 (2.3)
I ask the patient to rate the severity of shortness of breath using a categorical scale (eg, somewhat SOB, no SOB, improved or worsened compared with a prior date)120 (67.8)
When is dyspnea severity assessed and documented by nursing at your hospital?*
Dyspnea is not routinely assessed60 (33.7)
At admission30 (16.9)
Daily43 (24.2)
Each shift64 (36.0)
Awareness of dyspnea severity affects my management by:*
Influencing my decision to intensify treatment of the patient's underlying condition170 (95.5)
Influencing my decision to pursue additional diagnostic testing160 (89.9)
Influencing my decision to add pharmacologic‐based, symptom‐oriented treatment for dyspnea, such as opioids115 (64.6)
Influencing my decision to add nonpharmacologic‐based, symptom‐oriented treatment for dyspnea, such as fans or pursed lip breathing technique58 (32.6)
Influencing my decision regarding timing of discharge162 (91.0)
Which of the following nonpharmacologic therapies are effective for the relief of dyspnea?*
Pursed lip breathing113 (63.5)
Relaxation techniques137 (77.0)
Noninvasive ventilation143 (80.3)
O2 for nonhypoxemic patients89 (50.0)
Cool air/fan125 (70.2)
Cognitive behavioral strategies101 (56.7)

Forty‐two percent of the respondents agreed that patients are able to rate their dyspnea on a scale (2.3% strongly agree and 40.0% agree), and 73.0% indicated that patient experience of dyspnea should guide management independent of physiologic measures such as respiratory rate and oxygen saturation (Figure 1).

Several potential barriers were identified among the 18 participants who did not think that a standardized assessment of dyspnea would be beneficial, including concerns that (1) a dyspnea severity scale is too subjective and numerical scales are not useful for a subjective symptom (19.0%), (2) patients may overrate their symptom or will not be able to rate their dyspnea using a scale (31.0%), or (3) categorical description is sufficient (31.0%).

Practices in Dyspnea Management

Seventy‐nine percent of respondents agreed with the statement that judicious use of opioids can provide relief of dyspnea (26.1% strongly and 52.8% agreed), and 88.7% hospitalists identified the risk of respiratory depression as 1 of the barriers for the limited use of opioids. The majority of physicians (60%80%) considered nonpharmacologic therapies effective for symptomatic treatment of dyspnea, including in the order of agreement: noninvasive ventilation, relaxation techniques, cool air/fan, use of pursed lip breathing, and oxygen for nonhypoxemic patients (Table 1).

Physician Experience and Attitudes Toward Dyspnea Management

When we stratified hospitalists in tertiles of median years of time in practice (median [range]: 2 [04], 6 [58] and 15 [938]), we did not find an association with any of the responses to the questions.

Attitude Regarding the Importance of Dyspnea in Clinical Care and Responses to Subsequent Questions

Respondents who strongly agree or agree that dyspnea is the primary presenting symptom in patients with cardiovascular condition and that dyspnea relief is central to the management of these patients were more likely to believe that patients would like to be asked about their dyspnea (61.2% vs 30.2% vs 29.7%). They also had a more positive attitude about the usefulness of a standardized assessment of dyspnea and the inclusion of the assessment of dyspnea by a scale in the vital signs (Table 2).

Attitude Regarding the Importance of Dyspnea in Clinical Care and Responses to Subsequent Questions
DescriptionDo Not Agree, n (%)Somewhat Agree, n (%)Strongly Agree, n (%)P Value*
  • Chi‐square test

  • Fisher's exact test

 37 (20.9)43 (24.3)97 (54.8) 
Which description best characterizes your approach to assessing dyspnea severity?   0.552
I don't regularly ask about dyspnea severity0 (0)0 (0)3 (3.1) 
I ask the patient whether or not they are having shortness of breath11 (29.7)14 (32.6)25 (25.8) 
I ask the patient to rate the severity of shortness of breath using a numeric scale2 (5.4)1 (2.3)1 (1.0) 
I ask the patient to rate the severity of shortness of breath using a categorical scale (e.g., somewhat shortness of breath, no shortness of breath, improved or worsened compared with a prior date)24 (64.9)28 (65.1)68 (70.1) 
Patients would like me to ask them about their dyspnea.   <0.0001
Somewhat agree9 (24.3)21 (48.8)32 (32.7) 
Strongly agree11 (29.7)13 (30.2)60 (61.2) 
Patients are able to rate their own dyspnea intensity on a scale of 0‐10.   0.432
Somewhat agree12 (32.4)16 (37.2)42 (43.3) 
Strongly agree2 (5.4)0 (0)2 (2.1) 
Having a standardized assessment of dyspnea severity would be helpful to me in management of patients with cardiopulmonary diseases.   0.026
Somewhat agree17 (46.0)25 (58.1)57 (58.2) 
Strongly agree7 (18.9)6 (14.0)28 (28.6) 
Serial measurements of dyspnea would be useful for assessing response to therapy.   0.042
Somewhat agree14 (37.8)28 (65.1)48 (49.5) 
Strongly agree16 (43.2)12 (27.9)43 (44.3) 
Dyspnea assessment by a scale should be part of the vital signs for patients with cardiopulmonary diseases.   0.042
Somewhat agree13 (35.1)17 (39.5)51 (52.0) 
Strongly agree4 (10.8)5 (11.6)19 (19.4) 
Using an enhanced dyspnea scale that includes information about the following 4 features 1) Current dyspnea severity, 2) Worst dyspnea ever, 3) Improvement of dyspnea since admission, 4) Acceptability of current level of dyspnea, would be more helpful for my management than a single question focused on dyspnea severity.   0.03
Somewhat agree14 (40.0)24 (55.8)44 (44.9) 
Strongly agree9 (25.7)9 (20.9)40 (40.8) 
The patients experience of dyspnea should be used to guide treatment decisions independent of objective measures such as respiratory rate and oxygen saturation.   0.10
Somewhat agree20 (54.0)21 (48.8)51 (52.0) 
Strongly agree5 (13.5)6 (14.0)27 (27.6) 
Judicious use of oral and/or parenteral opioids can provide relief of dyspnea.   0.21
Somewhat agree20 (54.0)23 (54.8)50 (51.6) 
Strongly agree10 (27.0)6 (14.3)30 (30.9) 
Limited use of opioids for relief of dyspnea in patients with advanced cardiopulmonary disorders is often due to concerns of respiratory depression.   0.71
Somewhat agree17 (46.0)23 (54.8)43 (43.9) 
Strongly agree15 (40.5)14 (33.3)45 (45.9) 

DISCUSSION

In this survey of 178 most hospitalists from a diverse group of 9 US hospitals, we found that most indicate that severity of dyspnea has a profound influence on their clinical practice (including their decision whether to intensify treatments such as diuretics or bronchodilators, to pursue additional diagnostic testing, add opioids or other nonpharmacological treatments) and ultimately their decision regarding the timing of hospital discharge. More importantly, whereas less than half reported experience with standardized assessment of dyspnea severity, most stated that such data would be very useful in their practice.

Despite being a highly prevalent symptom in diverse patient populations, several studies have shown that documentation of dyspnea is sporadic and evaluation of dyspnea quality of care is not routinely performed.[13, 14, 15] Statements from a number of professional societies, including the ACCP, the American Thoracic Society and the Canadian Respiratory Society, recommend that dyspnea management should rely on patient reporting, and that dyspnea severity should be recorded.[1, 4, 7] Assessment is an essential step to guide interventions; however, simply asking about the presence or absence of dyspnea is insufficient.

Several rating scales have been validated and might be implementable in the acute care setting, including the Numerical Rating Scale and the Visual Assessment Scale.[16, 17, 18, 19, 20] Our survey shows that standardized documentation of dyspnea severity in clinical practice is uncommon. However, most hospitalists in our study believed that assessment of dyspnea, using a standardized scale, would positively impact their management of patients with cardiopulmonary disease.

There are a number of potential benefits of routine assessment of dyspnea in hospitalized patients. Implementation of a standardized approach to dyspnea measurement would result in more uniform assessment and documentation practices, and in turn greater awareness among members of the patient‐care team. Though not sufficient to improve care, measurement is necessary because physicians do not always recognize the severity of patients' dyspnea or may not recognize its presence. A retrospective study that assessed the prevalence of symptoms in 410 ambulatory patients showed that one‐quarter of patients had dyspnea, but only half of them told their doctor about it.[21] Two other studies of patients with cancer diagnoses found that 30%70% of patients had dyspnea, but the symptom was recognized in only half of them; even when recognized, dyspnea severity was frequently underrated by physicians.[21, 22] Importantly, underestimation appears to correlate with underutilization of symptomatic management of dyspnea.[8]

Although the results of our survey are encouraging, they highlight a number of potential barriers and misconceptions among hospitalists. For example, although dyspnea can be characterized only by the person experiencing it, only 42% of our survey respondents believed that patients are able to rate their dyspnea intensity on a scale. Some of these responses may be influenced by the fact that dyspnea scales are not currently available to patients under their care. Another explanation is that similar to the case for pain, some hospitalists may believe that patients will exaggerate dyspnea severity. Almost one‐third of the respondents stated that objective measures, such as respiratory rate or oxygen saturation, are more important than a patient's experience of dyspnea in guiding the treatment, and that dyspnea is a subjective symptom and not a vital sign itself. Hospitalists who appreciated the importance of dyspnea in clinical practice were more likely to support the implementation of a standardized dyspnea scale for dyspnea assessment.

Although the potential benefits of including routine measurement of dyspnea in standard hospital practice may seem obvious, evidence that implementing routine assessment improves patient care or outcomes is lacking. Even if hospitalists see the value of dyspnea assessment, asking nurses to collect and document additional information would represent a substantial change in hospital workflow. Finally, without specific protocols to guide care, it is unclear whether physicians will be able to use new information about dyspnea severity effectively. Future studies need to evaluate the impact of implementing routine dyspnea assessment on the management of patients with cardiopulmonary diseases including the use of evidence‐based interventions and reducing the use of less valuable care.

Most hospitalists agreed with the basic principles of dyspnea treatment in patients with advanced cardiopulmonary disease after the primary disease had been stabilized. Effective measures are available, and several guidelines endorse opioids in dyspnea management.[1, 4, 7] However, many clinicians are uncomfortable with this approach for dyspnea, and opioids remain underused. In our study, almost 90% of physicians recognized that concerns about respiratory depression limits opioids use as a treatment. A qualitative study that explored the physicians' perspective toward opioids showed that most physicians were reluctant to prescribe opioids for refractory dyspnea, describing a lack of related knowledge and experience, and fears related to the potential adverse effects. The findings of our study also outline the need to better educate residents and hospitalists on the assessment and management of dyspnea, including prescribing opioids for refractory dyspnea.[23]

Study Strengths and Limitations

This study has several strengths. To our knowledge, it is the first to explore hospitalists' perspectives on incorporating dyspnea assessment in their clinical practice. Hospitalists are the attending physicians for a large majority of inpatients and would be the main users of a dyspnea severity scale. Our questionnaire survey included a large number of hospitalists, from 9 geographically and structurally diverse hospitals, which increased the generalizability of the findings to other hospitals around the country.

The study also has several limitations that need be kept in mind in interpreting the study results. First, desirability bias may have exaggerated some of the positive views expressed by hospitalists toward implementation of routine assessment of dyspnea. Second, because this was a survey, the estimates of dyspnea assessment and documentation practices of both physicians and nurses were based on the respondents' perception and not an objective review of medical records, and the results may be different from actual practice. Third, this was not a population‐based random sample of hospitalists, and it may not be entirely representative; however, those surveyed were from a diverse set of sites with different geographical location, size, academic affiliation, and practice environment, and their time in practice varied widely. Last, we do not have information on nonrespondents, and there is a possibility of nonresponse bias, although the high response rate lessens the risk.

CONCLUSIONS

The results of this survey suggest that most hospitalists believe that routine assessment of dyspnea severity would enhance their clinical decision making and improve patient care. Standardized assessment of dyspnea might result in better awareness of this symptom among providers, reduce undertreatment and mistreatment, and ultimately result in better outcomes for patients. However, implementation of the routine assessment of dyspnea would change current clinical practices and may have a significant effect on existing nursing and physician workflows. Additional research is needed to determine the feasibility and impact on outcomes of routine dyspnea assessment.

Acknowledgements

The authors wish to acknowledge Ms. Anu Joshi for her help with editing the manuscript and assisting with table preparations.

Disclosures

Dr. Stefan is supported by grant K01HL114631‐01A1 from the National Heart, Lung, and Blood Institute of the National Institutes of Health, and by the National Center for Research Resources and the National Center for Advancing Translational Sciences, National Institutes of Health, through grant UL1RR025752. The funder had no role in the design and conduct of the study; collection, management, analysis, and interpretation of the data; preparation, review, or approval of the manuscript; and decision to submit the manuscript for publication. M.S.S. and P.K.L. conceived of the study. M.S.S. acquired the data with the help of all collaborators. M.S.S., P.K.L., P.S.P., and A.P. analyzed and interpreted the data. M.S.S. drafted the manuscript. All authors critically reviewed the manuscript for intellectual content. M.S.S., P.K.L., and A.P. had full access to all of the data in the study and take responsibility for the integrity of the data and the accuracy of the data analysis. M.S.S. is the guarantor for this article, and is responsible for the content of the article, including data and analysis. The authors report no conflicts of interest.

Files
References
  1. Parshall MB, Schwartzstein RM, Adams L, et al. An Official American Thoracic Society Statement: Update on the Mechanisms, Assessment, and Management of Dyspnea. Am J Respir Crit Care Med. 2012;185(4):435452.
  2. CDC/ National Center for Health Statistics. National Hospital Amulatory Medical Care Survey: 2011 Emergency Department Summary Tables. http://www.cdc.gov/nchs/data/ahcd/nhamcs_emergency/2011_ed_web_tables.pdf. Accessed May 15, 2015.
  3. Albert N, Trochelman K, Li J, Lin S. Signs and symptoms of heart failure: are you asking the right questions? Am J Crit Care. 2010;19(5):443452.
  4. Marciniuk DD, Goodridge D, Hernandez P, et al. Managing dyspnea in patients with advanced chronic obstructive pulmonary disease: a Canadian Thoracic Society clinical practice guideline. Can Respir J. 2011;18(2):6978.
  5. Sigurdardottir KR, Haugen DF. Prevalence of distressing symptoms in hospitalised patients on medical wards: A cross‐sectional study. BMC Palliat Care. 2008;7:16.
  6. Reuben DB, Mor V. Dyspnea in terminally ill cancer patients. Chest. 1986;89(2):234236.
  7. Mahler DA, Selecky PA, Harrod CG, et al. American College of Chest Physicians consensus statement on the management of dyspnea in patients with advanced lung or heart disease. Chest. 2010;137(3):674691.
  8. Kroenke K, Mangelsdorff AD. Common symptoms in ambulatory care: incidence, evaluation, therapy, and outcome. Am J Med. 1989;86(3):262266.
  9. The Joint Commission. Facts about Pain Management. http://www.jointcommission.org/pain_management/. Accessed May, 15, 2015.
  10. Buser M. Hospitalist programs in the age of healthcare reform. J Healthc Manag. 2010;55(6):378380.
  11. Casey MM, Hung P, Moscovice I, Prasad S. The Use of Hospitalists by Small Rural Hospitals: Results of a National Survey. Med Care Res Rev. 2014;71(4):356366.
  12. Tufts CTSI. REDCap [Internet]. Tufts Clinical and Translational Science Institute. http://www.tuftsctsi.org/Services-and-Consultation/REDCap.aspx. Accessed May, 15, 2015.
  13. Carrieri‐Kohlman V, Dudgeon DJ. Multi‐dimensional Assessment of Dyspnea. Dyspnoea in Advanced Disease: A guide to clinical management; 2005.
  14. Lorenz K, Lynn J, Dy S, et al. Cancer care quality measures: symptoms and end‐of‐life care. Evid Rep Technol Assess (Full Rep). 2006(137):177.
  15. Mularski RA. Defining and measuring quality palliative and end‐of‐life care in the intensive care unit. Crit Care Med. 2006;34(11 Suppl):S309316.
  16. Gift AG. Validation of a vertical visual analogue scale as a measure of clinical dyspnea. Rehabil Nurs. 1989;14(6):323325.
  17. Kendrick KR. Can a self‐rating 0‐10 scale for dyspnea yield a common language that is understood by ED nurses, patients, and their families? J Emerg Nurs. 2000;26(3):233234.
  18. Lansing RW, Moosavi SH, Banzett RB. Measurement of dyspnea: word labeled visual analog scale vs. verbal ordinal scale. Respir Physiol Neurobiol. 2003;134(2):7783.
  19. Morris NR, Sabapathy S, Adams L, Kingsley RA, Schneider DA, Stulbarg MS. Verbal numerical scales are as reliable and sensitive as visual analog scales for rating dyspnea in young and older subjects. Respir Physiol Neurobiol. 2007;157(2‐3):360365.
  20. Parshall MB, Carle AC, Ice U, Taylor R, Powers J. Validation of a three‐factor measurement model of dyspnea in hospitalized adults with heart failure. Heart Lung. 2011;41(1):4456.
  21. Hayes AW, Philip J, Spruyt OW. Patient reporting and doctor recognition of dyspnoea in a comprehensive cancer centre. Intern Med J. 2006;36(6):381384.
  22. Brown ML, Carrieri V, Janson B, Dodd MJ. Lung cancer and dyspnea: the patient's perception. Oncol Nurs Forum. 1986;13(5):1924.
  23. LeGrand SB, Khawam EA, Walsh D, Rivera NI. Opioids, respiratory function, and dyspnea. Am J Hosp Palliat Care. 2003;20(1):5761.
Article PDF
Issue
Journal of Hospital Medicine - 10(11)
Publications
Page Number
724-730
Sections
Files
Files
Article PDF
Article PDF

Dyspnea, defined as a subjective experience of breathing discomfort,[1] is the seventh most frequent reason adult patients present to the emergency room and the most frequent cause for emergency room visits in patients 65 years or older.[2] Moreover, dyspnea is experienced by 49% of patients hospitalized with a medical condition[3, 4, 5] and by 70% of patients who are seriously ill.[6]

Based on evidence that patients are not treated consistently and effectively for relief of their shortness of breath, the American College of Chest Physicians (ACCP) statement on dyspnea management in patients with advanced lung or heart disease recommended that patients should be asked to rate their dyspnea, and the rating should be routinely documented in the medical record to guide management.[7] Although clinicians may question the utility of routine assessment of dyspnea using a standardized scale, studies have found that the prevalence of dyspnea reported from chart review is much lower than when patients are directly interviewed.[8] This may be the result of underrecognition of dyspnea or poor documentation by physicians, or that patients may not communicate their symptoms unless the physician specifically asks. As is the case with pain, routine assessment of dyspnea severity could lead to improved clinical management and greater patient‐centered care. However, unlike in the case of pain, regulatory bodies, such as the Joint Commission for Accreditation of Healthcare Organization, do not require routine dyspnea assessment.[9]

Currently, there are more than 40,000 hospitalists in the United States, and the vast majority of hospitals with >200 beds have a hospitalist group.[10] Hospitalists care for over 60% of inpatients[11] and play a major role in the management of patients with acute cardiopulmonary diseases. If standardized approaches for the assessment and documentation of dyspnea are to be implemented, hospitalists would be a key stakeholder group for utilizing enhanced clinical information about dyspnea. Therefore, we evaluated attitudes and practices of hospitalists in regard to the assessment and management of dyspnea, including the potential benefits and challenges related to the implementation of standardized assessment. We hypothesized that hospitalists would believe that a dyspnea scale for assessment of severity could improve their management of patients with cardiovascular diseases. Further, we hypothesized that physicians who agreed with the general statement that dyspnea is an important clinical problem would be more likely to believe that routine dyspnea assessment would be valuable.

METHODS

Study Sample

We invited 255 attending hospitalists from 9 geographically and structurally diverse hospitals to complete a survey about the assessment and management of dyspnea. The 9 hospitals represent range of practice environments including 4 academic medical centers, 2 community teaching and 3 nonteaching hospitals, 1 Veterans Administration hospital, and 2 staff‐model HMOs (see Supporting Table 1 in the online version of this article). The survey was distributed online using REDCap (Research Electronic Data Capture), a secure web‐based interface application.[12] A coinvestigator who was a pulmonary critical‐care physician at each site sent an initial email to their hospitalist groups that alerted them to expect a survey from the principal investigator. This notification was subsequently followed by an email invitation containing an informational cover letter and a link to the online survey. The cover letter stated that the completed surveys would not be stored at the local sites and that all the analyzed data would be deidentified. Nonrespondents were sent reminders at 2 and 4 weeks after the initial mailing. A $25 electronic gift card was provided as a gesture of appreciation for their time. The survey was conducted between September 2013 and December 2013.

The study was approved by the Baystate Health Institutional Review Board, Springfield, Massachusetts, with a waiver for written informed consent.

Questionnaire

We developed a 17‐item instrument based on a review of the dyspnea literature and a prior ACCP survey.[12] Questions were piloted with 4 hospitalists at a single institution and modified to improve face validity and clarity (see Supporting Information in the online version of this article for the full survey).

Hospitalists were asked to consider the care of patients admitted for acute cardiopulmonary disease, including heart failure, chronic obstructive pulmonary disease, and pneumonia. A series of 5‐point Likert scales were used to assess the respondents level of agreement with statements related to the following domains: the importance of dyspnea in clinical care, the potential benefits and challenges of routine dyspnea assessment (statements such as: Having a standardized assessment of dyspnea severity would be helpful in management of patients with cardiopulmonary diseases. Dyspnea assessment by a scale should be part of the vital signs for patients with cardiopulmonary diseases.), and management of dyspnea (questions regarding the use of opioids and other nonpharmacological therapies). Additional questions were asked about current assessment practices (questions such as: How often do you assess severity of dyspnea? What is your approach in assessing dyspnea? with options of choosing a categorical or numerical scale), if dyspnea is assessed in their institution by nurses and how often, and the influence of dyspnea severity assessment on their management. The survey had 1 question that solicited comments from the participants: If you don't think that it would be useful to have a standardized dyspnea assessment, please tell us why.

Data Analysis

Responses to survey questions were summarized via counts and percentages in each response category. Adopting the methodology used in the ACCP consensus statement, strongly agree and somewhat agree were combined into a single category of agreement. We also presented percentage of responses in the 2 levels of agreement (strongly agree and somewhat agree) for each question in a bar graph.

Associations between tertiles of physicians' time in practice and attitude toward dyspnea were evaluated via 2 or Fisher exact test.

To examine how answers to the first 2 questions, which assessed attitude toward importance of dyspnea in clinical care, affect answers to the remaining questions, we grouped respondents in 3 categories (strongly agree, agree to these questions, do not agree) and tested the associations using 2 or Fisher exact test.

All analyses were performed using SAS version 9.3 (SAS institute, Inc., Cary, NC) and Stata release 13.1 (StataCorp, College Station, TX).

RESULTS

Overall, 178 (69.8%) of 255 identified hospitalists completed the survey, and all 9 participating hospitals had a response rate greater than 50%. The median number of years in practice was 6 (range, 038 years). A majority (77.5%) of respondents agreed with the statement that dyspnea is 1 of the major symptoms of patients with cardiopulmonary disease, and that its treatment is central to the management of these patients (77.0%) (Figure 1).

Figure 1
Attitude and practices regarding dyspnea assessment and management.

Attitude and Practices Surrounding Dyspnea Assessment

When asked about their current assessment of dyspnea, a majority (84.3%) of the hospitalists stated that they assess dyspnea on a daily basis; two‐thirds indicated that they use a categorical scale (ie, no shortness of breath, improved or worsened compared with a prior date), and one‐third indicated that they ask whether the patient is dyspneic or not. Fifty‐six percent of hospitalists stated that dyspnea is regularly assessed by nurses in their hospital.

The majority of respondents agreed (78.6%, 23.0% strongly and 55.6% somewhat agree) that standardized assessment of dyspnea severity, using a numeric scale and serial measurements as part of the vital signs, would benefit the management of patients with cardiopulmonary diseases. Furthermore, 79.6% (33.0% strongly and 46.6% somewhat agree) reported that using a dyspnea scale that included information to further characterize the patient‐reported experience, such as the level of distress associated with dyspnea, would be helpful in management.

Approximately 90% of the hospitalists indicated that awareness of dyspnea severity has an influence on clinical decision making, including whether to intensify treatment of underlying conditions, to pursue additional diagnostic testing, or to modify discharge timing. Additionally, two‐thirds of hospitalists agreed that awareness of dyspnea severity influences their decision to add opioids, whereas only one‐third prescribed nonpharmacologic symptom‐oriented treatment (Table 1).

Responses of Hospitalists to Questions Regarding Current Assessment and Management of Dyspnea
 Frequency (%)
  • NOTE: Abbreviations: SOB, shortness of breath. *Survey respondents selected all that apply, so these numbers are not mutually exclusive.

When caring for patients with acute cardiopulmonary diseases, how often do you assess severity of dyspnea?*
At admission66 (37.1)
At discharge59 (33.2)
Daily until discharge150 (84.3)
More often than daily58 (32.6)
Which description best characterizes your approach to assessing dyspnea severity?
I don't regularly ask about dyspnea severity3 (1.7)
I ask the patient whether or not they are having shortness of breath50 (28.3)
I ask the patient to rate the severity of shortness of breath using a numeric scale4 (2.3)
I ask the patient to rate the severity of shortness of breath using a categorical scale (eg, somewhat SOB, no SOB, improved or worsened compared with a prior date)120 (67.8)
When is dyspnea severity assessed and documented by nursing at your hospital?*
Dyspnea is not routinely assessed60 (33.7)
At admission30 (16.9)
Daily43 (24.2)
Each shift64 (36.0)
Awareness of dyspnea severity affects my management by:*
Influencing my decision to intensify treatment of the patient's underlying condition170 (95.5)
Influencing my decision to pursue additional diagnostic testing160 (89.9)
Influencing my decision to add pharmacologic‐based, symptom‐oriented treatment for dyspnea, such as opioids115 (64.6)
Influencing my decision to add nonpharmacologic‐based, symptom‐oriented treatment for dyspnea, such as fans or pursed lip breathing technique58 (32.6)
Influencing my decision regarding timing of discharge162 (91.0)
Which of the following nonpharmacologic therapies are effective for the relief of dyspnea?*
Pursed lip breathing113 (63.5)
Relaxation techniques137 (77.0)
Noninvasive ventilation143 (80.3)
O2 for nonhypoxemic patients89 (50.0)
Cool air/fan125 (70.2)
Cognitive behavioral strategies101 (56.7)

Forty‐two percent of the respondents agreed that patients are able to rate their dyspnea on a scale (2.3% strongly agree and 40.0% agree), and 73.0% indicated that patient experience of dyspnea should guide management independent of physiologic measures such as respiratory rate and oxygen saturation (Figure 1).

Several potential barriers were identified among the 18 participants who did not think that a standardized assessment of dyspnea would be beneficial, including concerns that (1) a dyspnea severity scale is too subjective and numerical scales are not useful for a subjective symptom (19.0%), (2) patients may overrate their symptom or will not be able to rate their dyspnea using a scale (31.0%), or (3) categorical description is sufficient (31.0%).

Practices in Dyspnea Management

Seventy‐nine percent of respondents agreed with the statement that judicious use of opioids can provide relief of dyspnea (26.1% strongly and 52.8% agreed), and 88.7% hospitalists identified the risk of respiratory depression as 1 of the barriers for the limited use of opioids. The majority of physicians (60%80%) considered nonpharmacologic therapies effective for symptomatic treatment of dyspnea, including in the order of agreement: noninvasive ventilation, relaxation techniques, cool air/fan, use of pursed lip breathing, and oxygen for nonhypoxemic patients (Table 1).

Physician Experience and Attitudes Toward Dyspnea Management

When we stratified hospitalists in tertiles of median years of time in practice (median [range]: 2 [04], 6 [58] and 15 [938]), we did not find an association with any of the responses to the questions.

Attitude Regarding the Importance of Dyspnea in Clinical Care and Responses to Subsequent Questions

Respondents who strongly agree or agree that dyspnea is the primary presenting symptom in patients with cardiovascular condition and that dyspnea relief is central to the management of these patients were more likely to believe that patients would like to be asked about their dyspnea (61.2% vs 30.2% vs 29.7%). They also had a more positive attitude about the usefulness of a standardized assessment of dyspnea and the inclusion of the assessment of dyspnea by a scale in the vital signs (Table 2).

Attitude Regarding the Importance of Dyspnea in Clinical Care and Responses to Subsequent Questions
DescriptionDo Not Agree, n (%)Somewhat Agree, n (%)Strongly Agree, n (%)P Value*
  • Chi‐square test

  • Fisher's exact test

 37 (20.9)43 (24.3)97 (54.8) 
Which description best characterizes your approach to assessing dyspnea severity?   0.552
I don't regularly ask about dyspnea severity0 (0)0 (0)3 (3.1) 
I ask the patient whether or not they are having shortness of breath11 (29.7)14 (32.6)25 (25.8) 
I ask the patient to rate the severity of shortness of breath using a numeric scale2 (5.4)1 (2.3)1 (1.0) 
I ask the patient to rate the severity of shortness of breath using a categorical scale (e.g., somewhat shortness of breath, no shortness of breath, improved or worsened compared with a prior date)24 (64.9)28 (65.1)68 (70.1) 
Patients would like me to ask them about their dyspnea.   <0.0001
Somewhat agree9 (24.3)21 (48.8)32 (32.7) 
Strongly agree11 (29.7)13 (30.2)60 (61.2) 
Patients are able to rate their own dyspnea intensity on a scale of 0‐10.   0.432
Somewhat agree12 (32.4)16 (37.2)42 (43.3) 
Strongly agree2 (5.4)0 (0)2 (2.1) 
Having a standardized assessment of dyspnea severity would be helpful to me in management of patients with cardiopulmonary diseases.   0.026
Somewhat agree17 (46.0)25 (58.1)57 (58.2) 
Strongly agree7 (18.9)6 (14.0)28 (28.6) 
Serial measurements of dyspnea would be useful for assessing response to therapy.   0.042
Somewhat agree14 (37.8)28 (65.1)48 (49.5) 
Strongly agree16 (43.2)12 (27.9)43 (44.3) 
Dyspnea assessment by a scale should be part of the vital signs for patients with cardiopulmonary diseases.   0.042
Somewhat agree13 (35.1)17 (39.5)51 (52.0) 
Strongly agree4 (10.8)5 (11.6)19 (19.4) 
Using an enhanced dyspnea scale that includes information about the following 4 features 1) Current dyspnea severity, 2) Worst dyspnea ever, 3) Improvement of dyspnea since admission, 4) Acceptability of current level of dyspnea, would be more helpful for my management than a single question focused on dyspnea severity.   0.03
Somewhat agree14 (40.0)24 (55.8)44 (44.9) 
Strongly agree9 (25.7)9 (20.9)40 (40.8) 
The patients experience of dyspnea should be used to guide treatment decisions independent of objective measures such as respiratory rate and oxygen saturation.   0.10
Somewhat agree20 (54.0)21 (48.8)51 (52.0) 
Strongly agree5 (13.5)6 (14.0)27 (27.6) 
Judicious use of oral and/or parenteral opioids can provide relief of dyspnea.   0.21
Somewhat agree20 (54.0)23 (54.8)50 (51.6) 
Strongly agree10 (27.0)6 (14.3)30 (30.9) 
Limited use of opioids for relief of dyspnea in patients with advanced cardiopulmonary disorders is often due to concerns of respiratory depression.   0.71
Somewhat agree17 (46.0)23 (54.8)43 (43.9) 
Strongly agree15 (40.5)14 (33.3)45 (45.9) 

DISCUSSION

In this survey of 178 most hospitalists from a diverse group of 9 US hospitals, we found that most indicate that severity of dyspnea has a profound influence on their clinical practice (including their decision whether to intensify treatments such as diuretics or bronchodilators, to pursue additional diagnostic testing, add opioids or other nonpharmacological treatments) and ultimately their decision regarding the timing of hospital discharge. More importantly, whereas less than half reported experience with standardized assessment of dyspnea severity, most stated that such data would be very useful in their practice.

Despite being a highly prevalent symptom in diverse patient populations, several studies have shown that documentation of dyspnea is sporadic and evaluation of dyspnea quality of care is not routinely performed.[13, 14, 15] Statements from a number of professional societies, including the ACCP, the American Thoracic Society and the Canadian Respiratory Society, recommend that dyspnea management should rely on patient reporting, and that dyspnea severity should be recorded.[1, 4, 7] Assessment is an essential step to guide interventions; however, simply asking about the presence or absence of dyspnea is insufficient.

Several rating scales have been validated and might be implementable in the acute care setting, including the Numerical Rating Scale and the Visual Assessment Scale.[16, 17, 18, 19, 20] Our survey shows that standardized documentation of dyspnea severity in clinical practice is uncommon. However, most hospitalists in our study believed that assessment of dyspnea, using a standardized scale, would positively impact their management of patients with cardiopulmonary disease.

There are a number of potential benefits of routine assessment of dyspnea in hospitalized patients. Implementation of a standardized approach to dyspnea measurement would result in more uniform assessment and documentation practices, and in turn greater awareness among members of the patient‐care team. Though not sufficient to improve care, measurement is necessary because physicians do not always recognize the severity of patients' dyspnea or may not recognize its presence. A retrospective study that assessed the prevalence of symptoms in 410 ambulatory patients showed that one‐quarter of patients had dyspnea, but only half of them told their doctor about it.[21] Two other studies of patients with cancer diagnoses found that 30%70% of patients had dyspnea, but the symptom was recognized in only half of them; even when recognized, dyspnea severity was frequently underrated by physicians.[21, 22] Importantly, underestimation appears to correlate with underutilization of symptomatic management of dyspnea.[8]

Although the results of our survey are encouraging, they highlight a number of potential barriers and misconceptions among hospitalists. For example, although dyspnea can be characterized only by the person experiencing it, only 42% of our survey respondents believed that patients are able to rate their dyspnea intensity on a scale. Some of these responses may be influenced by the fact that dyspnea scales are not currently available to patients under their care. Another explanation is that similar to the case for pain, some hospitalists may believe that patients will exaggerate dyspnea severity. Almost one‐third of the respondents stated that objective measures, such as respiratory rate or oxygen saturation, are more important than a patient's experience of dyspnea in guiding the treatment, and that dyspnea is a subjective symptom and not a vital sign itself. Hospitalists who appreciated the importance of dyspnea in clinical practice were more likely to support the implementation of a standardized dyspnea scale for dyspnea assessment.

Although the potential benefits of including routine measurement of dyspnea in standard hospital practice may seem obvious, evidence that implementing routine assessment improves patient care or outcomes is lacking. Even if hospitalists see the value of dyspnea assessment, asking nurses to collect and document additional information would represent a substantial change in hospital workflow. Finally, without specific protocols to guide care, it is unclear whether physicians will be able to use new information about dyspnea severity effectively. Future studies need to evaluate the impact of implementing routine dyspnea assessment on the management of patients with cardiopulmonary diseases including the use of evidence‐based interventions and reducing the use of less valuable care.

Most hospitalists agreed with the basic principles of dyspnea treatment in patients with advanced cardiopulmonary disease after the primary disease had been stabilized. Effective measures are available, and several guidelines endorse opioids in dyspnea management.[1, 4, 7] However, many clinicians are uncomfortable with this approach for dyspnea, and opioids remain underused. In our study, almost 90% of physicians recognized that concerns about respiratory depression limits opioids use as a treatment. A qualitative study that explored the physicians' perspective toward opioids showed that most physicians were reluctant to prescribe opioids for refractory dyspnea, describing a lack of related knowledge and experience, and fears related to the potential adverse effects. The findings of our study also outline the need to better educate residents and hospitalists on the assessment and management of dyspnea, including prescribing opioids for refractory dyspnea.[23]

Study Strengths and Limitations

This study has several strengths. To our knowledge, it is the first to explore hospitalists' perspectives on incorporating dyspnea assessment in their clinical practice. Hospitalists are the attending physicians for a large majority of inpatients and would be the main users of a dyspnea severity scale. Our questionnaire survey included a large number of hospitalists, from 9 geographically and structurally diverse hospitals, which increased the generalizability of the findings to other hospitals around the country.

The study also has several limitations that need be kept in mind in interpreting the study results. First, desirability bias may have exaggerated some of the positive views expressed by hospitalists toward implementation of routine assessment of dyspnea. Second, because this was a survey, the estimates of dyspnea assessment and documentation practices of both physicians and nurses were based on the respondents' perception and not an objective review of medical records, and the results may be different from actual practice. Third, this was not a population‐based random sample of hospitalists, and it may not be entirely representative; however, those surveyed were from a diverse set of sites with different geographical location, size, academic affiliation, and practice environment, and their time in practice varied widely. Last, we do not have information on nonrespondents, and there is a possibility of nonresponse bias, although the high response rate lessens the risk.

CONCLUSIONS

The results of this survey suggest that most hospitalists believe that routine assessment of dyspnea severity would enhance their clinical decision making and improve patient care. Standardized assessment of dyspnea might result in better awareness of this symptom among providers, reduce undertreatment and mistreatment, and ultimately result in better outcomes for patients. However, implementation of the routine assessment of dyspnea would change current clinical practices and may have a significant effect on existing nursing and physician workflows. Additional research is needed to determine the feasibility and impact on outcomes of routine dyspnea assessment.

Acknowledgements

The authors wish to acknowledge Ms. Anu Joshi for her help with editing the manuscript and assisting with table preparations.

Disclosures

Dr. Stefan is supported by grant K01HL114631‐01A1 from the National Heart, Lung, and Blood Institute of the National Institutes of Health, and by the National Center for Research Resources and the National Center for Advancing Translational Sciences, National Institutes of Health, through grant UL1RR025752. The funder had no role in the design and conduct of the study; collection, management, analysis, and interpretation of the data; preparation, review, or approval of the manuscript; and decision to submit the manuscript for publication. M.S.S. and P.K.L. conceived of the study. M.S.S. acquired the data with the help of all collaborators. M.S.S., P.K.L., P.S.P., and A.P. analyzed and interpreted the data. M.S.S. drafted the manuscript. All authors critically reviewed the manuscript for intellectual content. M.S.S., P.K.L., and A.P. had full access to all of the data in the study and take responsibility for the integrity of the data and the accuracy of the data analysis. M.S.S. is the guarantor for this article, and is responsible for the content of the article, including data and analysis. The authors report no conflicts of interest.

Dyspnea, defined as a subjective experience of breathing discomfort,[1] is the seventh most frequent reason adult patients present to the emergency room and the most frequent cause for emergency room visits in patients 65 years or older.[2] Moreover, dyspnea is experienced by 49% of patients hospitalized with a medical condition[3, 4, 5] and by 70% of patients who are seriously ill.[6]

Based on evidence that patients are not treated consistently and effectively for relief of their shortness of breath, the American College of Chest Physicians (ACCP) statement on dyspnea management in patients with advanced lung or heart disease recommended that patients should be asked to rate their dyspnea, and the rating should be routinely documented in the medical record to guide management.[7] Although clinicians may question the utility of routine assessment of dyspnea using a standardized scale, studies have found that the prevalence of dyspnea reported from chart review is much lower than when patients are directly interviewed.[8] This may be the result of underrecognition of dyspnea or poor documentation by physicians, or that patients may not communicate their symptoms unless the physician specifically asks. As is the case with pain, routine assessment of dyspnea severity could lead to improved clinical management and greater patient‐centered care. However, unlike in the case of pain, regulatory bodies, such as the Joint Commission for Accreditation of Healthcare Organization, do not require routine dyspnea assessment.[9]

Currently, there are more than 40,000 hospitalists in the United States, and the vast majority of hospitals with >200 beds have a hospitalist group.[10] Hospitalists care for over 60% of inpatients[11] and play a major role in the management of patients with acute cardiopulmonary diseases. If standardized approaches for the assessment and documentation of dyspnea are to be implemented, hospitalists would be a key stakeholder group for utilizing enhanced clinical information about dyspnea. Therefore, we evaluated attitudes and practices of hospitalists in regard to the assessment and management of dyspnea, including the potential benefits and challenges related to the implementation of standardized assessment. We hypothesized that hospitalists would believe that a dyspnea scale for assessment of severity could improve their management of patients with cardiovascular diseases. Further, we hypothesized that physicians who agreed with the general statement that dyspnea is an important clinical problem would be more likely to believe that routine dyspnea assessment would be valuable.

METHODS

Study Sample

We invited 255 attending hospitalists from 9 geographically and structurally diverse hospitals to complete a survey about the assessment and management of dyspnea. The 9 hospitals represent range of practice environments including 4 academic medical centers, 2 community teaching and 3 nonteaching hospitals, 1 Veterans Administration hospital, and 2 staff‐model HMOs (see Supporting Table 1 in the online version of this article). The survey was distributed online using REDCap (Research Electronic Data Capture), a secure web‐based interface application.[12] A coinvestigator who was a pulmonary critical‐care physician at each site sent an initial email to their hospitalist groups that alerted them to expect a survey from the principal investigator. This notification was subsequently followed by an email invitation containing an informational cover letter and a link to the online survey. The cover letter stated that the completed surveys would not be stored at the local sites and that all the analyzed data would be deidentified. Nonrespondents were sent reminders at 2 and 4 weeks after the initial mailing. A $25 electronic gift card was provided as a gesture of appreciation for their time. The survey was conducted between September 2013 and December 2013.

The study was approved by the Baystate Health Institutional Review Board, Springfield, Massachusetts, with a waiver for written informed consent.

Questionnaire

We developed a 17‐item instrument based on a review of the dyspnea literature and a prior ACCP survey.[12] Questions were piloted with 4 hospitalists at a single institution and modified to improve face validity and clarity (see Supporting Information in the online version of this article for the full survey).

Hospitalists were asked to consider the care of patients admitted for acute cardiopulmonary disease, including heart failure, chronic obstructive pulmonary disease, and pneumonia. A series of 5‐point Likert scales were used to assess the respondents level of agreement with statements related to the following domains: the importance of dyspnea in clinical care, the potential benefits and challenges of routine dyspnea assessment (statements such as: Having a standardized assessment of dyspnea severity would be helpful in management of patients with cardiopulmonary diseases. Dyspnea assessment by a scale should be part of the vital signs for patients with cardiopulmonary diseases.), and management of dyspnea (questions regarding the use of opioids and other nonpharmacological therapies). Additional questions were asked about current assessment practices (questions such as: How often do you assess severity of dyspnea? What is your approach in assessing dyspnea? with options of choosing a categorical or numerical scale), if dyspnea is assessed in their institution by nurses and how often, and the influence of dyspnea severity assessment on their management. The survey had 1 question that solicited comments from the participants: If you don't think that it would be useful to have a standardized dyspnea assessment, please tell us why.

Data Analysis

Responses to survey questions were summarized via counts and percentages in each response category. Adopting the methodology used in the ACCP consensus statement, strongly agree and somewhat agree were combined into a single category of agreement. We also presented percentage of responses in the 2 levels of agreement (strongly agree and somewhat agree) for each question in a bar graph.

Associations between tertiles of physicians' time in practice and attitude toward dyspnea were evaluated via 2 or Fisher exact test.

To examine how answers to the first 2 questions, which assessed attitude toward importance of dyspnea in clinical care, affect answers to the remaining questions, we grouped respondents in 3 categories (strongly agree, agree to these questions, do not agree) and tested the associations using 2 or Fisher exact test.

All analyses were performed using SAS version 9.3 (SAS institute, Inc., Cary, NC) and Stata release 13.1 (StataCorp, College Station, TX).

RESULTS

Overall, 178 (69.8%) of 255 identified hospitalists completed the survey, and all 9 participating hospitals had a response rate greater than 50%. The median number of years in practice was 6 (range, 038 years). A majority (77.5%) of respondents agreed with the statement that dyspnea is 1 of the major symptoms of patients with cardiopulmonary disease, and that its treatment is central to the management of these patients (77.0%) (Figure 1).

Figure 1
Attitude and practices regarding dyspnea assessment and management.

Attitude and Practices Surrounding Dyspnea Assessment

When asked about their current assessment of dyspnea, a majority (84.3%) of the hospitalists stated that they assess dyspnea on a daily basis; two‐thirds indicated that they use a categorical scale (ie, no shortness of breath, improved or worsened compared with a prior date), and one‐third indicated that they ask whether the patient is dyspneic or not. Fifty‐six percent of hospitalists stated that dyspnea is regularly assessed by nurses in their hospital.

The majority of respondents agreed (78.6%, 23.0% strongly and 55.6% somewhat agree) that standardized assessment of dyspnea severity, using a numeric scale and serial measurements as part of the vital signs, would benefit the management of patients with cardiopulmonary diseases. Furthermore, 79.6% (33.0% strongly and 46.6% somewhat agree) reported that using a dyspnea scale that included information to further characterize the patient‐reported experience, such as the level of distress associated with dyspnea, would be helpful in management.

Approximately 90% of the hospitalists indicated that awareness of dyspnea severity has an influence on clinical decision making, including whether to intensify treatment of underlying conditions, to pursue additional diagnostic testing, or to modify discharge timing. Additionally, two‐thirds of hospitalists agreed that awareness of dyspnea severity influences their decision to add opioids, whereas only one‐third prescribed nonpharmacologic symptom‐oriented treatment (Table 1).

Responses of Hospitalists to Questions Regarding Current Assessment and Management of Dyspnea
 Frequency (%)
  • NOTE: Abbreviations: SOB, shortness of breath. *Survey respondents selected all that apply, so these numbers are not mutually exclusive.

When caring for patients with acute cardiopulmonary diseases, how often do you assess severity of dyspnea?*
At admission66 (37.1)
At discharge59 (33.2)
Daily until discharge150 (84.3)
More often than daily58 (32.6)
Which description best characterizes your approach to assessing dyspnea severity?
I don't regularly ask about dyspnea severity3 (1.7)
I ask the patient whether or not they are having shortness of breath50 (28.3)
I ask the patient to rate the severity of shortness of breath using a numeric scale4 (2.3)
I ask the patient to rate the severity of shortness of breath using a categorical scale (eg, somewhat SOB, no SOB, improved or worsened compared with a prior date)120 (67.8)
When is dyspnea severity assessed and documented by nursing at your hospital?*
Dyspnea is not routinely assessed60 (33.7)
At admission30 (16.9)
Daily43 (24.2)
Each shift64 (36.0)
Awareness of dyspnea severity affects my management by:*
Influencing my decision to intensify treatment of the patient's underlying condition170 (95.5)
Influencing my decision to pursue additional diagnostic testing160 (89.9)
Influencing my decision to add pharmacologic‐based, symptom‐oriented treatment for dyspnea, such as opioids115 (64.6)
Influencing my decision to add nonpharmacologic‐based, symptom‐oriented treatment for dyspnea, such as fans or pursed lip breathing technique58 (32.6)
Influencing my decision regarding timing of discharge162 (91.0)
Which of the following nonpharmacologic therapies are effective for the relief of dyspnea?*
Pursed lip breathing113 (63.5)
Relaxation techniques137 (77.0)
Noninvasive ventilation143 (80.3)
O2 for nonhypoxemic patients89 (50.0)
Cool air/fan125 (70.2)
Cognitive behavioral strategies101 (56.7)

Forty‐two percent of the respondents agreed that patients are able to rate their dyspnea on a scale (2.3% strongly agree and 40.0% agree), and 73.0% indicated that patient experience of dyspnea should guide management independent of physiologic measures such as respiratory rate and oxygen saturation (Figure 1).

Several potential barriers were identified among the 18 participants who did not think that a standardized assessment of dyspnea would be beneficial, including concerns that (1) a dyspnea severity scale is too subjective and numerical scales are not useful for a subjective symptom (19.0%), (2) patients may overrate their symptom or will not be able to rate their dyspnea using a scale (31.0%), or (3) categorical description is sufficient (31.0%).

Practices in Dyspnea Management

Seventy‐nine percent of respondents agreed with the statement that judicious use of opioids can provide relief of dyspnea (26.1% strongly and 52.8% agreed), and 88.7% hospitalists identified the risk of respiratory depression as 1 of the barriers for the limited use of opioids. The majority of physicians (60%80%) considered nonpharmacologic therapies effective for symptomatic treatment of dyspnea, including in the order of agreement: noninvasive ventilation, relaxation techniques, cool air/fan, use of pursed lip breathing, and oxygen for nonhypoxemic patients (Table 1).

Physician Experience and Attitudes Toward Dyspnea Management

When we stratified hospitalists in tertiles of median years of time in practice (median [range]: 2 [04], 6 [58] and 15 [938]), we did not find an association with any of the responses to the questions.

Attitude Regarding the Importance of Dyspnea in Clinical Care and Responses to Subsequent Questions

Respondents who strongly agree or agree that dyspnea is the primary presenting symptom in patients with cardiovascular condition and that dyspnea relief is central to the management of these patients were more likely to believe that patients would like to be asked about their dyspnea (61.2% vs 30.2% vs 29.7%). They also had a more positive attitude about the usefulness of a standardized assessment of dyspnea and the inclusion of the assessment of dyspnea by a scale in the vital signs (Table 2).

Attitude Regarding the Importance of Dyspnea in Clinical Care and Responses to Subsequent Questions
DescriptionDo Not Agree, n (%)Somewhat Agree, n (%)Strongly Agree, n (%)P Value*
  • Chi‐square test

  • Fisher's exact test

 37 (20.9)43 (24.3)97 (54.8) 
Which description best characterizes your approach to assessing dyspnea severity?   0.552
I don't regularly ask about dyspnea severity0 (0)0 (0)3 (3.1) 
I ask the patient whether or not they are having shortness of breath11 (29.7)14 (32.6)25 (25.8) 
I ask the patient to rate the severity of shortness of breath using a numeric scale2 (5.4)1 (2.3)1 (1.0) 
I ask the patient to rate the severity of shortness of breath using a categorical scale (e.g., somewhat shortness of breath, no shortness of breath, improved or worsened compared with a prior date)24 (64.9)28 (65.1)68 (70.1) 
Patients would like me to ask them about their dyspnea.   <0.0001
Somewhat agree9 (24.3)21 (48.8)32 (32.7) 
Strongly agree11 (29.7)13 (30.2)60 (61.2) 
Patients are able to rate their own dyspnea intensity on a scale of 0‐10.   0.432
Somewhat agree12 (32.4)16 (37.2)42 (43.3) 
Strongly agree2 (5.4)0 (0)2 (2.1) 
Having a standardized assessment of dyspnea severity would be helpful to me in management of patients with cardiopulmonary diseases.   0.026
Somewhat agree17 (46.0)25 (58.1)57 (58.2) 
Strongly agree7 (18.9)6 (14.0)28 (28.6) 
Serial measurements of dyspnea would be useful for assessing response to therapy.   0.042
Somewhat agree14 (37.8)28 (65.1)48 (49.5) 
Strongly agree16 (43.2)12 (27.9)43 (44.3) 
Dyspnea assessment by a scale should be part of the vital signs for patients with cardiopulmonary diseases.   0.042
Somewhat agree13 (35.1)17 (39.5)51 (52.0) 
Strongly agree4 (10.8)5 (11.6)19 (19.4) 
Using an enhanced dyspnea scale that includes information about the following 4 features 1) Current dyspnea severity, 2) Worst dyspnea ever, 3) Improvement of dyspnea since admission, 4) Acceptability of current level of dyspnea, would be more helpful for my management than a single question focused on dyspnea severity.   0.03
Somewhat agree14 (40.0)24 (55.8)44 (44.9) 
Strongly agree9 (25.7)9 (20.9)40 (40.8) 
The patients experience of dyspnea should be used to guide treatment decisions independent of objective measures such as respiratory rate and oxygen saturation.   0.10
Somewhat agree20 (54.0)21 (48.8)51 (52.0) 
Strongly agree5 (13.5)6 (14.0)27 (27.6) 
Judicious use of oral and/or parenteral opioids can provide relief of dyspnea.   0.21
Somewhat agree20 (54.0)23 (54.8)50 (51.6) 
Strongly agree10 (27.0)6 (14.3)30 (30.9) 
Limited use of opioids for relief of dyspnea in patients with advanced cardiopulmonary disorders is often due to concerns of respiratory depression.   0.71
Somewhat agree17 (46.0)23 (54.8)43 (43.9) 
Strongly agree15 (40.5)14 (33.3)45 (45.9) 

DISCUSSION

In this survey of 178 most hospitalists from a diverse group of 9 US hospitals, we found that most indicate that severity of dyspnea has a profound influence on their clinical practice (including their decision whether to intensify treatments such as diuretics or bronchodilators, to pursue additional diagnostic testing, add opioids or other nonpharmacological treatments) and ultimately their decision regarding the timing of hospital discharge. More importantly, whereas less than half reported experience with standardized assessment of dyspnea severity, most stated that such data would be very useful in their practice.

Despite being a highly prevalent symptom in diverse patient populations, several studies have shown that documentation of dyspnea is sporadic and evaluation of dyspnea quality of care is not routinely performed.[13, 14, 15] Statements from a number of professional societies, including the ACCP, the American Thoracic Society and the Canadian Respiratory Society, recommend that dyspnea management should rely on patient reporting, and that dyspnea severity should be recorded.[1, 4, 7] Assessment is an essential step to guide interventions; however, simply asking about the presence or absence of dyspnea is insufficient.

Several rating scales have been validated and might be implementable in the acute care setting, including the Numerical Rating Scale and the Visual Assessment Scale.[16, 17, 18, 19, 20] Our survey shows that standardized documentation of dyspnea severity in clinical practice is uncommon. However, most hospitalists in our study believed that assessment of dyspnea, using a standardized scale, would positively impact their management of patients with cardiopulmonary disease.

There are a number of potential benefits of routine assessment of dyspnea in hospitalized patients. Implementation of a standardized approach to dyspnea measurement would result in more uniform assessment and documentation practices, and in turn greater awareness among members of the patient‐care team. Though not sufficient to improve care, measurement is necessary because physicians do not always recognize the severity of patients' dyspnea or may not recognize its presence. A retrospective study that assessed the prevalence of symptoms in 410 ambulatory patients showed that one‐quarter of patients had dyspnea, but only half of them told their doctor about it.[21] Two other studies of patients with cancer diagnoses found that 30%70% of patients had dyspnea, but the symptom was recognized in only half of them; even when recognized, dyspnea severity was frequently underrated by physicians.[21, 22] Importantly, underestimation appears to correlate with underutilization of symptomatic management of dyspnea.[8]

Although the results of our survey are encouraging, they highlight a number of potential barriers and misconceptions among hospitalists. For example, although dyspnea can be characterized only by the person experiencing it, only 42% of our survey respondents believed that patients are able to rate their dyspnea intensity on a scale. Some of these responses may be influenced by the fact that dyspnea scales are not currently available to patients under their care. Another explanation is that similar to the case for pain, some hospitalists may believe that patients will exaggerate dyspnea severity. Almost one‐third of the respondents stated that objective measures, such as respiratory rate or oxygen saturation, are more important than a patient's experience of dyspnea in guiding the treatment, and that dyspnea is a subjective symptom and not a vital sign itself. Hospitalists who appreciated the importance of dyspnea in clinical practice were more likely to support the implementation of a standardized dyspnea scale for dyspnea assessment.

Although the potential benefits of including routine measurement of dyspnea in standard hospital practice may seem obvious, evidence that implementing routine assessment improves patient care or outcomes is lacking. Even if hospitalists see the value of dyspnea assessment, asking nurses to collect and document additional information would represent a substantial change in hospital workflow. Finally, without specific protocols to guide care, it is unclear whether physicians will be able to use new information about dyspnea severity effectively. Future studies need to evaluate the impact of implementing routine dyspnea assessment on the management of patients with cardiopulmonary diseases including the use of evidence‐based interventions and reducing the use of less valuable care.

Most hospitalists agreed with the basic principles of dyspnea treatment in patients with advanced cardiopulmonary disease after the primary disease had been stabilized. Effective measures are available, and several guidelines endorse opioids in dyspnea management.[1, 4, 7] However, many clinicians are uncomfortable with this approach for dyspnea, and opioids remain underused. In our study, almost 90% of physicians recognized that concerns about respiratory depression limits opioids use as a treatment. A qualitative study that explored the physicians' perspective toward opioids showed that most physicians were reluctant to prescribe opioids for refractory dyspnea, describing a lack of related knowledge and experience, and fears related to the potential adverse effects. The findings of our study also outline the need to better educate residents and hospitalists on the assessment and management of dyspnea, including prescribing opioids for refractory dyspnea.[23]

Study Strengths and Limitations

This study has several strengths. To our knowledge, it is the first to explore hospitalists' perspectives on incorporating dyspnea assessment in their clinical practice. Hospitalists are the attending physicians for a large majority of inpatients and would be the main users of a dyspnea severity scale. Our questionnaire survey included a large number of hospitalists, from 9 geographically and structurally diverse hospitals, which increased the generalizability of the findings to other hospitals around the country.

The study also has several limitations that need be kept in mind in interpreting the study results. First, desirability bias may have exaggerated some of the positive views expressed by hospitalists toward implementation of routine assessment of dyspnea. Second, because this was a survey, the estimates of dyspnea assessment and documentation practices of both physicians and nurses were based on the respondents' perception and not an objective review of medical records, and the results may be different from actual practice. Third, this was not a population‐based random sample of hospitalists, and it may not be entirely representative; however, those surveyed were from a diverse set of sites with different geographical location, size, academic affiliation, and practice environment, and their time in practice varied widely. Last, we do not have information on nonrespondents, and there is a possibility of nonresponse bias, although the high response rate lessens the risk.

CONCLUSIONS

The results of this survey suggest that most hospitalists believe that routine assessment of dyspnea severity would enhance their clinical decision making and improve patient care. Standardized assessment of dyspnea might result in better awareness of this symptom among providers, reduce undertreatment and mistreatment, and ultimately result in better outcomes for patients. However, implementation of the routine assessment of dyspnea would change current clinical practices and may have a significant effect on existing nursing and physician workflows. Additional research is needed to determine the feasibility and impact on outcomes of routine dyspnea assessment.

Acknowledgements

The authors wish to acknowledge Ms. Anu Joshi for her help with editing the manuscript and assisting with table preparations.

Disclosures

Dr. Stefan is supported by grant K01HL114631‐01A1 from the National Heart, Lung, and Blood Institute of the National Institutes of Health, and by the National Center for Research Resources and the National Center for Advancing Translational Sciences, National Institutes of Health, through grant UL1RR025752. The funder had no role in the design and conduct of the study; collection, management, analysis, and interpretation of the data; preparation, review, or approval of the manuscript; and decision to submit the manuscript for publication. M.S.S. and P.K.L. conceived of the study. M.S.S. acquired the data with the help of all collaborators. M.S.S., P.K.L., P.S.P., and A.P. analyzed and interpreted the data. M.S.S. drafted the manuscript. All authors critically reviewed the manuscript for intellectual content. M.S.S., P.K.L., and A.P. had full access to all of the data in the study and take responsibility for the integrity of the data and the accuracy of the data analysis. M.S.S. is the guarantor for this article, and is responsible for the content of the article, including data and analysis. The authors report no conflicts of interest.

References
  1. Parshall MB, Schwartzstein RM, Adams L, et al. An Official American Thoracic Society Statement: Update on the Mechanisms, Assessment, and Management of Dyspnea. Am J Respir Crit Care Med. 2012;185(4):435452.
  2. CDC/ National Center for Health Statistics. National Hospital Amulatory Medical Care Survey: 2011 Emergency Department Summary Tables. http://www.cdc.gov/nchs/data/ahcd/nhamcs_emergency/2011_ed_web_tables.pdf. Accessed May 15, 2015.
  3. Albert N, Trochelman K, Li J, Lin S. Signs and symptoms of heart failure: are you asking the right questions? Am J Crit Care. 2010;19(5):443452.
  4. Marciniuk DD, Goodridge D, Hernandez P, et al. Managing dyspnea in patients with advanced chronic obstructive pulmonary disease: a Canadian Thoracic Society clinical practice guideline. Can Respir J. 2011;18(2):6978.
  5. Sigurdardottir KR, Haugen DF. Prevalence of distressing symptoms in hospitalised patients on medical wards: A cross‐sectional study. BMC Palliat Care. 2008;7:16.
  6. Reuben DB, Mor V. Dyspnea in terminally ill cancer patients. Chest. 1986;89(2):234236.
  7. Mahler DA, Selecky PA, Harrod CG, et al. American College of Chest Physicians consensus statement on the management of dyspnea in patients with advanced lung or heart disease. Chest. 2010;137(3):674691.
  8. Kroenke K, Mangelsdorff AD. Common symptoms in ambulatory care: incidence, evaluation, therapy, and outcome. Am J Med. 1989;86(3):262266.
  9. The Joint Commission. Facts about Pain Management. http://www.jointcommission.org/pain_management/. Accessed May, 15, 2015.
  10. Buser M. Hospitalist programs in the age of healthcare reform. J Healthc Manag. 2010;55(6):378380.
  11. Casey MM, Hung P, Moscovice I, Prasad S. The Use of Hospitalists by Small Rural Hospitals: Results of a National Survey. Med Care Res Rev. 2014;71(4):356366.
  12. Tufts CTSI. REDCap [Internet]. Tufts Clinical and Translational Science Institute. http://www.tuftsctsi.org/Services-and-Consultation/REDCap.aspx. Accessed May, 15, 2015.
  13. Carrieri‐Kohlman V, Dudgeon DJ. Multi‐dimensional Assessment of Dyspnea. Dyspnoea in Advanced Disease: A guide to clinical management; 2005.
  14. Lorenz K, Lynn J, Dy S, et al. Cancer care quality measures: symptoms and end‐of‐life care. Evid Rep Technol Assess (Full Rep). 2006(137):177.
  15. Mularski RA. Defining and measuring quality palliative and end‐of‐life care in the intensive care unit. Crit Care Med. 2006;34(11 Suppl):S309316.
  16. Gift AG. Validation of a vertical visual analogue scale as a measure of clinical dyspnea. Rehabil Nurs. 1989;14(6):323325.
  17. Kendrick KR. Can a self‐rating 0‐10 scale for dyspnea yield a common language that is understood by ED nurses, patients, and their families? J Emerg Nurs. 2000;26(3):233234.
  18. Lansing RW, Moosavi SH, Banzett RB. Measurement of dyspnea: word labeled visual analog scale vs. verbal ordinal scale. Respir Physiol Neurobiol. 2003;134(2):7783.
  19. Morris NR, Sabapathy S, Adams L, Kingsley RA, Schneider DA, Stulbarg MS. Verbal numerical scales are as reliable and sensitive as visual analog scales for rating dyspnea in young and older subjects. Respir Physiol Neurobiol. 2007;157(2‐3):360365.
  20. Parshall MB, Carle AC, Ice U, Taylor R, Powers J. Validation of a three‐factor measurement model of dyspnea in hospitalized adults with heart failure. Heart Lung. 2011;41(1):4456.
  21. Hayes AW, Philip J, Spruyt OW. Patient reporting and doctor recognition of dyspnoea in a comprehensive cancer centre. Intern Med J. 2006;36(6):381384.
  22. Brown ML, Carrieri V, Janson B, Dodd MJ. Lung cancer and dyspnea: the patient's perception. Oncol Nurs Forum. 1986;13(5):1924.
  23. LeGrand SB, Khawam EA, Walsh D, Rivera NI. Opioids, respiratory function, and dyspnea. Am J Hosp Palliat Care. 2003;20(1):5761.
References
  1. Parshall MB, Schwartzstein RM, Adams L, et al. An Official American Thoracic Society Statement: Update on the Mechanisms, Assessment, and Management of Dyspnea. Am J Respir Crit Care Med. 2012;185(4):435452.
  2. CDC/ National Center for Health Statistics. National Hospital Amulatory Medical Care Survey: 2011 Emergency Department Summary Tables. http://www.cdc.gov/nchs/data/ahcd/nhamcs_emergency/2011_ed_web_tables.pdf. Accessed May 15, 2015.
  3. Albert N, Trochelman K, Li J, Lin S. Signs and symptoms of heart failure: are you asking the right questions? Am J Crit Care. 2010;19(5):443452.
  4. Marciniuk DD, Goodridge D, Hernandez P, et al. Managing dyspnea in patients with advanced chronic obstructive pulmonary disease: a Canadian Thoracic Society clinical practice guideline. Can Respir J. 2011;18(2):6978.
  5. Sigurdardottir KR, Haugen DF. Prevalence of distressing symptoms in hospitalised patients on medical wards: A cross‐sectional study. BMC Palliat Care. 2008;7:16.
  6. Reuben DB, Mor V. Dyspnea in terminally ill cancer patients. Chest. 1986;89(2):234236.
  7. Mahler DA, Selecky PA, Harrod CG, et al. American College of Chest Physicians consensus statement on the management of dyspnea in patients with advanced lung or heart disease. Chest. 2010;137(3):674691.
  8. Kroenke K, Mangelsdorff AD. Common symptoms in ambulatory care: incidence, evaluation, therapy, and outcome. Am J Med. 1989;86(3):262266.
  9. The Joint Commission. Facts about Pain Management. http://www.jointcommission.org/pain_management/. Accessed May, 15, 2015.
  10. Buser M. Hospitalist programs in the age of healthcare reform. J Healthc Manag. 2010;55(6):378380.
  11. Casey MM, Hung P, Moscovice I, Prasad S. The Use of Hospitalists by Small Rural Hospitals: Results of a National Survey. Med Care Res Rev. 2014;71(4):356366.
  12. Tufts CTSI. REDCap [Internet]. Tufts Clinical and Translational Science Institute. http://www.tuftsctsi.org/Services-and-Consultation/REDCap.aspx. Accessed May, 15, 2015.
  13. Carrieri‐Kohlman V, Dudgeon DJ. Multi‐dimensional Assessment of Dyspnea. Dyspnoea in Advanced Disease: A guide to clinical management; 2005.
  14. Lorenz K, Lynn J, Dy S, et al. Cancer care quality measures: symptoms and end‐of‐life care. Evid Rep Technol Assess (Full Rep). 2006(137):177.
  15. Mularski RA. Defining and measuring quality palliative and end‐of‐life care in the intensive care unit. Crit Care Med. 2006;34(11 Suppl):S309316.
  16. Gift AG. Validation of a vertical visual analogue scale as a measure of clinical dyspnea. Rehabil Nurs. 1989;14(6):323325.
  17. Kendrick KR. Can a self‐rating 0‐10 scale for dyspnea yield a common language that is understood by ED nurses, patients, and their families? J Emerg Nurs. 2000;26(3):233234.
  18. Lansing RW, Moosavi SH, Banzett RB. Measurement of dyspnea: word labeled visual analog scale vs. verbal ordinal scale. Respir Physiol Neurobiol. 2003;134(2):7783.
  19. Morris NR, Sabapathy S, Adams L, Kingsley RA, Schneider DA, Stulbarg MS. Verbal numerical scales are as reliable and sensitive as visual analog scales for rating dyspnea in young and older subjects. Respir Physiol Neurobiol. 2007;157(2‐3):360365.
  20. Parshall MB, Carle AC, Ice U, Taylor R, Powers J. Validation of a three‐factor measurement model of dyspnea in hospitalized adults with heart failure. Heart Lung. 2011;41(1):4456.
  21. Hayes AW, Philip J, Spruyt OW. Patient reporting and doctor recognition of dyspnoea in a comprehensive cancer centre. Intern Med J. 2006;36(6):381384.
  22. Brown ML, Carrieri V, Janson B, Dodd MJ. Lung cancer and dyspnea: the patient's perception. Oncol Nurs Forum. 1986;13(5):1924.
  23. LeGrand SB, Khawam EA, Walsh D, Rivera NI. Opioids, respiratory function, and dyspnea. Am J Hosp Palliat Care. 2003;20(1):5761.
Issue
Journal of Hospital Medicine - 10(11)
Issue
Journal of Hospital Medicine - 10(11)
Page Number
724-730
Page Number
724-730
Publications
Publications
Article Type
Display Headline
Hospitalist attitudes toward the assessment and management of dyspnea in patients with acute cardiopulmonary diseases
Display Headline
Hospitalist attitudes toward the assessment and management of dyspnea in patients with acute cardiopulmonary diseases
Sections
Article Source

© 2015 Society of Hospital Medicine

Disallow All Ads
Correspondence Location
Address for correspondence and reprint requests: Mihaela S. Stefan, MD, Department of Medicine, Baystate Medical Center, 759 Chestnut Street, 2nd Floor, Springfield, MA 01199; Telephone: 413‐704‐8121; Fax: 413‐794‐8054; E‐mail: Mihaela.Stefan@baystatehealth.org
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Article PDF Media
Media Files

Impact of MC Intervention on QIs

Article Type
Changed
Sun, 05/21/2017 - 13:17
Display Headline
Outcomes associated with a mandatory gastroenterology consultation to improve the quality of care of patients hospitalized with decompensated cirrhosis

Decompensated cirrhosis (DC) is defined as cirrhosis with at least 1 of the following complications: ascites, hepatocellular carcinoma, bleeding from portal hypertension, or hepatic encephalopathy. Patients with DC have a median survival estimated at 2 years compared to the 12‐year median survival of compensated cirrhotics.[1] In an era where quality of hospital care is being measured, and where progress is being made in the management of several conditions including congestive heart failure and nosocomial infections, little attention has been paid to DC. The burden of chronic liver failure is clear in the United States, where DC leads to more than 150,000 annual admissions to the hospital and accounts for 40,000 deaths annually.[2]

This burden of disease spurred quality improvement efforts in 2010, when a team of experts identified a set of literature‐based parameters or quality indicators (QI) for patients with cirrhosis.[3] We have demonstrated that adherence to these indicators fell far short of desired targets.[4] A year before their publication, an overall compliance of <50% with these metrics was measured at a single medical center.

We sought to improve the quality of care for patients with DC through implementation of mandatory consultation (MC) with a gastroenterologist for all patients admitted with DC. We assessed whether MC was associated with better care and improved outcomes (hospitalization length of stay [LOS], 30‐day readmission, and inpatient mortality) when compared to usual care (UC).[4]

MATERIALS AND METHODS

Design, Setting, and Patients

We conducted a cohort study comparing adherence to QI and outcomes of patients admitted with DC after the institution of an MC to a historical cohort of patients managed with UC (ie, before MC, adherence to QI for this group has been reported elsewhere).[4] Both cohorts included all patients aged >18 years with DC admitted to Baystate Medical Center, a tertiary care medical center in western Massachusetts. The UC cohort was collected between January 1, 2009 and December 31, 2009, and the MC cohort was assembled between June 1, 2011 and June 30, 2012.

As previously reported,[4] patients were considered for inclusion in the historical cohort if their International Classification of DiseasesNinth Revision discharge code pertained to chronic liver disease (see Supporting Information, Appendix 1, in the online version of this article). This list was broad by design to identify all patients with decompensated cirrhosis. A gastroenterologist (R.G.) then manually extracted charts from electronic medical records (EMRs) using a set of predefined clinical criteria, the same in both cohorts, to identify the patients with DC: cirrhosis with concomitant ascites, hepatic encephalopathy, or gastrointestinal (GI) bleeding secondary to portal hypertension. Other types of decompensated states, such as hepatocellular carcinoma, were not included as their management was not detailed in the QI.[3]

We included patients with suspected or established cirrhosis who had ascites confirmed radiographically or by exam, noting shifting dullness or fluid wave. However, patients were excluded if they lacked sufficient peritoneal fluid for bedside or image‐guided paracentesis. Cirrhotic patients were defined as having hepatic encephalopathy if the patient had altered mental status not secondary to seizures, cerebrovascular accident, or alcohol withdrawal. Finally, gastrointestinal bleeding in cirrhotic patients was defined as any upper or lower bleeding prompting hospital admission, or identified in the medical record as clinically significant by the attending physician.

The same QIs were measured in both cohorts. From the QI set,[3] we selected the 16 QIs that would apply to the management of inpatients (see Supporting Information, Appendix 2, in the online version of this article). Indicators developed for outpatient settings were not included. A quality score was calculated for each admission, defined as the proportion of QIs met divided by the number of QIs for which the patient was eligible. For example, a patient with hepatic encephalopathy but without GI bleeding or ascites would have a score calculated as the number of QIs met for hepatic encephalopathy and documentation of transplant evaluation divided by 3 (2 QIs for hepatic encephalopathy and 1 QI for transplant evaluation). If the patient met both QIs for hepatic encephalopathy, but the consultant failed to address liver transplant eligibility, the score would be 2/3=0.666.

After the institution of the MC, all inpatients with DC were identified within 24 hours of admission by a gastroenterologist (R.G., D.D.), who manually reviewed on a daily basis all admissions from EMRs. An author (R.G.) would then contact the admitting team (hospitalist or resident) to make sure that a gastroenterology consult was called and would then obtain the QI by manual extraction from the EMRs.

Of the 16 gastroenterologists who work at the hospital, 12 of them belong to several private practice groups, whereas 4 are employed by the hospital. As part of the intervention, all gastroenterologists were made aware of the intervention 1 month before the starting date, were provided with a checklist of the QIs of interest, and were encouraged to work with the hospitalist attendings to achieve compliance with the QIs. We reminded the gastroenterologists of the ongoing study during routine division meetings and regularly sought feedback from the hospitalists

The MC consisted of a systematic consultation by a gastroenterologist: any identified patient with DC would generate a mandatory GI consultation and would be assigned to a specialist depending on the roster coverage for that day. A close monitoring of the process allowed us to confirm that all patients admitted with DC were seen by a gastroenterologist. Patients were followed until their discharge, death, or readmission to our institution during the study period.

Outcomes

The primary outcome was defined as the rate of adherence to the QIs and overall QI score expressed as a proportion as noted above. Secondary outcomes included in‐hospital mortality, LOS, and 30‐day readmission rate. These parameters were abstracted from the medical record.

Covariates

The hospital EMR (Cerner Corporation, North Kansas City, MO) was used to extract patient demographic parameters such as gender, race, language, and age at time of admission. Other admission‐level details were extracted from the EMR including Model for End‐Stage Liver Disease (MELD) scores, documented comorbidities (including substance abuse, psychiatric diagnosis, diabetes mellitus, renal failure, congestive heart failure, coronary artery disease, and cancer), underlying etiology for cirrhosis, and reason for admission.

The study was approved by Baystate Medical Center's institutional review board.

Statistical Analysis

Summary statistics for outcomes and covariates were calculated as means/standard deviations (SDs), medians/emnterquartile range, and proportions. Univariable statistics (unpaired t tests, 1‐way analysis of variance, Fisher exact test, Spearman correlation) were used to identify possible demographic (eg, age, race) and clinical (eg, admission complaint) predictors of quality score and with 30‐day outcomes. For each admission, a composite quality score, also known as an opportunity model score,[5, 6] was calculated as a fraction (ie, the number of QIs met divided by the total number of possible QIs indicated by the patient's presentation). This fraction was then multiplied by 100 so as to express the QI score as a percent. Possible scores, therefore, ranged from 0 to 100%.

Calculation of the 30‐day incidence proportion of readmission after the first admission was restricted to patients whose readmission occurred in this hospital, and occurring up to 30‐days before study closure (June 1, 2012). In‐hospital death was examined as a function of QI score during that admission. To derive an unbiased, risk‐adjusted estimate of the association between quality score and outcomes, multiple linear regression (opportunity model score [OMS], LOS) or multiple Poisson regression models (30‐day readmission, in‐hospital death) were built. These included a dummy variable for the study period, as well as any potential confounder that was associated at P0.10, with both study period and the outcome in univariable analyses. Robust standard errors were specified to account for multiple admissions within patients. Marginal means or proportions were then estimated with 95% confidence intervals derived using the delta method. All analyses were performed using Stata 12.1 for Windows (StataCorp, College Station, TX).

RESULTS

A total of 303 patients were observed in 695 hospitalizations;149 patients in 379 admissions were observed in the UC cohort, and 154 patients in 316 admissions were observed in the MC cohort. Baseline demographics of all study admissions appear in Table 1. Patients seen in the MC cohort were younger, more likely to speak English, and less likely to be male or have comorbid diabetes mellitus. Most admissions (n=217, 57.2%; 95% confidence interval: 52.3%‐62.3%) were not evaluated by a gastroenterologist in the UC cohort but all were in the MC cohort.

Patient Characteristics
 UC, N=379, N (%) or Mean/SDMC, N=316, N (%) or Mean/SDP Value*
  • NOTE: Abbreviations: CHF, congestive heart failure; CAD, coronary artery disease; GI, gastrointestinal; MELD, Model for End‐Stage Liver Disease; SD, standard deviation; UC, usual care. *Independent samples t test (continuous), Fisher exact (categorical).

Age, y55.3/12.153.3/13.60.05
English speaking261 (68.9%)261 (82.6%)<0.001
Male251 (66.2%)163 (53.5%)0.001
Race  <0.001
White301 (79.4%)262 (82.9%) 
Black31 (8.2%)40 (12.7%) 
Asian16 (4.2%)0 (0.0%) 
Other31 (8.2%)14 (4.4%) 
Comorbidities   
Substance75 (19.8%)58 (18.4%)0.70
abuse
Psychiatric123 (32.5%)103 (32.9%)0.94
Diabetes mellitus175 (45.4%)115 (36.5%)0.02
Renal failure74 (19.3%)55 (17.4%)0.50
CHF38 (10.0%)24 (7.6%)0.35
CAD26 (6.9%)17 (5.4%)0.43
Cancer48 (12.7%)40 (12.7%)1.00
Admission MELD15.6/6.917.0/7.00.006
Serum creatinine1.43/1.941.42/1.300.91
Reason for admission   
Hepatology/GI318 (83.9%)257 (81.3%)0.42
Renal failure85 (22.4%)90 (28.5%)0.08
Encephalopathy151 (39.3%)113 (34.9%)0.24
GI bleed78 (20.5%)57 (18.0%)1.00
Abdominal pain116 (30.7%)114 (36.2%)0.15
Ascites246 (64.9%)185 (58.5%)0.10

Admission Characteristics

The baseline clinical measures of all study admissions appear in Table 1. The UC and MC cohorts had similar characteristics, with the majority of patients with DC admitted for a gastrointestinal/hepatology‐related reason specifically for the management of ascites and hepatic encephalopathy. The patients in the MC cohort had a statistically higher MELD score on admission, which was not clinically relevant.

Quality Measures

Adherence to individual quality indices is shown in Table 2.

Percent Quality Indicators Met per Admission by Indication
Condition (Denominator)Quality Indicator (Numerator)UC (n=379), Met/IndicatedMC (n=316), Met/IndicatedP Value
  • NOTE: Abbreviations: GI, gastrointestinal; INR, International Normalized Ratio; MELD, Model for End‐Stage Liver Disease; SD, standard deviation; UC, usual care.

Admissions with ascites    
1Admissions to the hospital because of ascites or encephalopathy.Diagnostic paracentesis during admission.77/193, 39.9%, (32.9%, 46.9%)111/135, 82.2% (75.7%, 88.8%)<0.001
2No fibrinolysis or disseminated intravascular coagulation before paracentesis INR <2.5, >100,000 platelets.No fresh frozen plasma or platelet replacement given.36/37, 97.3% (91.8%, 103.0%)41/42, 97.6% (92.8%, 102.4%)1.00
3All admissions with diagnostic paracentesis (not limited to admissions for ascites or hepatic encephalopathy).Cell count differential, total protein, albumin, and culture/sensitivity all performed.31/49, 63.3% (49.3%‐77.3%)46/72 63.9% (52.7%, 75.0%)1.00
4Admissions with known portal hypertension‐related ascites receiving a paracentesis.Ascitic fluid cell count and differential performed.15/104, 14.4% (7.6%‐ 21.3%)47/62, 75.8% (63.2%, 88.4%)<0.001
5Serum sodium 110 mEq/L.Fluid restriction and discontinuation of diuretics.NANANA
6Polymorphonuclear count of 250/mm3 in ascites.Empiric antibiotics, 6 hours of results.10/13, 76.9% (50.4%‐ 103.4%)16/20, 80.0% (60.8%, 99.2%)1.00
7Ascitic fluid, total protein 1.1 gm/dL, serum bilirubin 2.5 mg/dL.Prophylactic antibiotics.4/12, 33.3% (2.0%‐ 64.6%)18/30, 60.0%, (41.4%, 78.6%)0.18
8Normal renal function.Salt restriction and diuretics (spironolactone and loop diuretics).57/186, 30.6%, (24.0%‐ 37.3%)81/122, 66.4%, (57.9%, 74.9%)<0.001
Total ascites subscore, mean/SD30%/36%67%/34%<0.001
GI bleeding    
9Admissions with GI bleeding: variceal and nonvariceal, hematemesis and melena.Upper endoscopy 24 hours of presentation.60/78, 76.9% (67.4%, 86.4%)52/57, 91.2% (83.7%, 98.8%)0.04
10Esophageal varices (active, stigmata of recent bleeding, or no other causes to explain bleeding).Endoscopic variceal ligation/sublerotherapy.40/46, 87.0% (76.8%‐97.1%)30/32, 93.8% (84.9%, 100.0%)0.46
11Admissions with established/suspected upper GI bleeding.Antibiotics within 24 hours of admission.27/69, 39.1% (27.3%‐ 50.9%)26/58, 44.8% (31.6%, 58.0%)0.59
12Admissions with established/suspected variceal bleeding.Somatostatin/octreotide given within 12 hours of presentation.53/69, 76.8%, (66.6%‐ 87.0%)49/58, 84.5% (73.8%, 95.2%)0.37
13Recurrent bleeding within 72 hours of initial endoscopic hemostasis.Repeat endoscopy or transjugular intrahepatic portosystemic shunt.5/5 100%2/3, 66.7% (76.8%, 210.0%)0.38
Total GI subscore, mean/SD61%/38%74%/28%0.04
Liver transplantation    
14Admissions with MELD 15 or MELD 15 and decompensated status (ie, all admissions in our study).Documented evaluation for liver transplantation.112/379, 29.6% (24.9%‐ 34.2%)231/316, 73.6% (68.7%, 78.5%)<0.001
Hepatic encephalopathy    
15Admissions with hepatic encephalopathy.Search for reversible factors documented.81/151, 53.6% (45.6%‐ 61.7 %)97/113, 85.8% (79.4%, 92.3%)<0.001
16Admissions with hepatic encephalopathy.Oral disaccharides/ rifaximin.144/151, 95.3% (91.9 %‐ 98.7 %)107/113, 94.7% (90.7%. 98.69%)1.00
Total encephalopathy subscore, mean/SD75%/28%90%/24%<0.001

Ascites

The management of ascites yielded 3 main differences between the 2 cohorts. Following the implementation of the MC, 82.2 % (111/135) of ascites‐related admissions led to a diagnostic paracentesis as compared to 39.9% (77/193) in the UC group (P<0.001).

In the MC cohort, 75.8% (47/62) of admissions with known portal hypertensionrelated ascites who received a paracentesis had an ascites cell count checked. In contrast, only 14.4% (15/104) in the UC group receiving paracentesis had a fluid cell count (P<0.001). The management of ascites in patients with normal renal function was optimal, with sodium restriction and diuretics combination in 66.4% (81/122) of the MC cohort, whereas this parameter in the UC cohort was only 30.6% (57/186) (P<0.001). There were no significant differences between the groups for the other QIs.

Variceal Bleeding

The MC group had a higher frequency of endoscopy within 24 hours of admissions than the UC group (91.2% [52/57] vs 76.9% [60/78], respectively; P<0.04). The rest had endoscopy later in the admission. Among admissions with bleeding from varices, banding was done 93.8% of the time for patients in the MC group (30/32), which was not statistically different than 87.0% (40/46) for patients seen in the UC group. In the remaining admissions, endoscopy only revealed nonbleeding large esophageal varices, and the endoscopist opted not to proceed with therapy. There were no statistically significant differences in the rest of the management.

Hepatic Encephalopathy

For hepatic encephalopathy, an empirical treatment was given to 95.3 % (144/151) patients in the UC group and 94.7% (107/113) of the patients in the MC group. We found better documentation of a search for underlying etiologies leading to hepatic encephalopathy in the MC cohort 85.8% (97/113) versus the UC cohort, which was only 53.6% (81/151) (P<0.001).

Evaluation for Liver Transplantation

Better documentation of evaluation for liver transplantation was seen in the MC group 73.6% (231/316) in comparison to the UC group 29.4% (111/379) (P<0.001).

Opportunity Score and Clinical Outcomes

As detailed above, care provided during the MC achieved a higher compliance with the QI shown with the QI score or OMS (Table 3). These improvements were not associated with statistically significant differences in in‐hospital death, LOS, or 30‐day readmission. To explore this further we also examined the direct association between the OMS and outcomes in the MC group by dividing patients into 2 groups: patients whose OMS was 80% and those whose OMS was <80% (see Supporting Information, Appendix 4, in the online version of this article). Although there were trends toward decreased in‐hospital death (6.4% vs 8.6%, P=0.26), increased 30‐day readmission (33.8% vs 23.0%, P=0.27), and decreased LOS (6.2 days vs 6.6 days, P=0.77), none of these differences achieved statistical significance.

Outcomes
 UnadjustedAdjusted*
UCMCDifferenceUCMCDifference
  • NOTE: Abbreviations: MC, mandatory consultation; MELD, Model for End‐Stage Liver Disease; UC, usual care. *Quality indicators score adjusted for baseline MELD and age. In‐hospital death adjusted for baseline MELD score and ascites‐related admission. Thirty‐day readmission adjusted for baseline MELD score and race. Length of stay adjusted for baseline MELD ascites‐related admission.

Opportunity model score0.460.77+0.31 (0.24, 0.39)0.460.77+0.30(0.23, 0.37)
In‐hospital death7.1%8.5%+1.4 (0.3, +5.6)7.5%7.9%+0.4% (4.0%, +4.5%)
Readmission within 30 days39.6%32.6%7.0% (16.4%, +2.5%)40.0%31.8%8.2%(18.0%, +1.5%)
Length of stay6.1d6.2d+0.1d (1.0 d, +1.2 d)6.1d6.2d+0.1d (1.0 d, +1.2d)

Mandatory Consultation Subgroups: Employed Versus Private Physicians

The type of employment of the gastroenterologist on consultation (employed by the hospital vs private practice) affected the management of the patients admitted with DC (see Supporting Information, Appendix 3, in the online version of this article). Patients seen by a hospital‐employed gastroenterologist were more likely to have a better documentation in regard to evaluation for liver transplantation and better management of ascites. Except for the prescription of antibiotics in patients presenting with GI bleeding, which were more often given by the employed physician (63% vs 23%, P=0.004), the management of hepatic encephalopathy and GI bleeding was similar between employed and private‐practice physicians.

DISCUSSION

In this evaluation of an MC intervention for patients with DC cared for at a large tertiary academic medical center, we found that the implementation of a routine consultation by a gastroenterologist led to greater adherence to recommended care processes when compared to UC. Overall, the management of ascites and the documentation of evaluation for liver transplantation were statistically superior in the intervention (MC) group. UC and MC were similar with respect to treatment of variceal bleeding and hepatic encephalopathy. Although we did not demonstrate changes in mortality, readmission, or LOS as a result of the MC intervention, our study was underpowered to detect clinically meaningful effects.

The gaps in care of patients with cirrhosis were reported before and after the publication of the formal QIs.[7, 8, 9, 10] These gaps remain relevant in the face of an increasing prevalence of DC along with a recent publication suggesting an underestimation of the burden of liver disease in the United States.[11] Ours is the first study to evaluate the impact on inpatients with DC of a liver service with a systematic, mandatory, specialist consultation. A previous study[12] had shown that a GI consultation would improve the care of patients with DC, but excluded patients with variceal bleeding, did not specifically measure the compliance with QIs, and more important, the GI consult was not mandatory.

Our study has several limitations that must be considered while weighing its findings. The patients were not randomly assigned but followed a pre‐established distribution depending on the call schedule. Some of the improvement we noted might be the result of secular trends; however, this remains unlikely given the lack of national initiatives or pay for performance programs. In the UC cohort, patients who were nonEnglish‐speaking were associated with a lower QI score, which could account for part of the improvement seen in the MC group that has a more prominent English‐speaking cohort. Readmissions could have occurred at other hospitals, and patients were not monitored in an outpatient setting. We did not observe a change in the secondary outcomes (30‐day readmission, LOS, in‐hospital death); however, our study was underpowered for that purpose. Given the complexity of the billing process we did not collect the costs of the MC, which is another limitation of our work. Future studies are needed to determine the cost‐effectiveness of the intervention.

This study shows that a dedicated team of physicians focused on compliance with QIs can achieve a rapid improvement, over a year, in providing higher‐quality care. This may be relevant at other institutions. The strength of our study is that our large tertiary academic medical center serves a large catchment area, with a mix of patients from both rural and urban communities. It is located in Massachusetts, where most of the population has had access to healthcare since 2006. Therefore, although this is a single‐center study, we expect our findings to be more generalizable and less subject to selection bias than other single‐center studies.

Importantly, the compliance with QIs was often far from being perfect in the MC group and was different across type of employment of providers, reflecting the challenges in changing practice among physicians.[13] In fact the QI scores of the private practice group did not change, and mirror the compliance observed at our institution in the previous study, before the implementation of the MC.[4] The difference in performance according to the type of employment of providers stems from 2 factors. First, a better documentation of the need of formal evaluation for liver transplantation by the employed gastroenterologists resulted in better compliance with this QI. Second, and more important, among the employed physicians, there was a readiness to assist the hospitalist with diagnostic/therapeutic paracentesis without relying on, for example, an interventional radiologist. This is reflected by the higher score in the management of ascites. Although our study was not designed to answer this directly, employed physicians may have been more engaged in the project and showed a greater willingness to change practice. In the future, linking reimbursement to quality of care will lead to improved accountability of consultants.

In this study we show that a direct involvement of a gastroenterologist improves the care of inpatients as measured by QIs. We theorize that a better coordination of the transition to outpatient care involving the specialist should lead to better outcomes, specifically a reduction in the 22% observed readmission rate within 30 days of patients with DC.[14, 15] As we move forward, a broader definition of outcomes should be addressed, taking into account patient‐related outcomes and preferences.[16] Future studies should define the relationship between the gastroenterologist and the hospitalist service, the role of physician assistants and nurse practitioners in implementing and monitoring compliance with QIs, and define how physicians and patients can be made accountable in the transition to the outpatient setting.

Disclosures

R.G.: Conception, data collection and interpretation, manuscript. J.F.: Data management, data analysis, manuscript. P.V.: Conception, data analysis, manuscript. P.L.: Conception, data interpretation, manuscript. T.L.: Dr. Lagu is supported by the National Heart, Lung, and Blood Institute of the National Institutes of Health under award number K01HL114745. Conception, data interpretation, manuscript. D.D.: Conception, data collection and interpretation, manuscript. A.B.: Data collection. J.S.: Data collection. Source of funding: internal. The authors report no conflicts of interest.

Files
References
  1. D'Amico G, Garcia‐Tsao G, Pagliaro L. Natural history and prognostic indicators of survival in cirrhosis: a systematic review of 118 studies. J Hepatol. 2006;44:217231.
  2. Wigg AJ, McCormick R, Wundke R, Woodman RJ. Efficacy of a chronic disease management model for patients with chronic liver failure. Clin Gastroenterol Hepatol. 2013;11:850858.
  3. Kanwal F, Kramer J, Asch SM, et al. An Explicit Quality Indicator Set for Measurement of Quality of Care in Patients with Cirrhosis. Clin Gastroenterol Hepatol. 2010;8:709717.
  4. Ghaoui R, Friderici J, Visintainer P, Lindenauer PK, Lagu T, Desilets D. Measurement of the quality of care of patients admitted with decompensated cirrhosis. Liver Int. 2014:34:204210.
  5. Nolan T, Berwick DM. All‐or‐none measurement raises the bar on performance. JAMA. 2006;295:11681170.
  6. Joint Commission on Accreditation of Healthcare Organizations. Quality report user guide. Available at: http://www.jointcommission.org. Accessed May 30, 2011.
  7. Saab S, Nguyen S, Ibrahim A, et al. Management of patients with cirrhosis in Southern California: results of a practitioner survey. J Clin Gastroenterol. 2006;40:156161.
  8. Lucena MI, Andrade RJ, Tognoni G, et al. Spanish Collaborative Study Group on Therapeutic Management In Liver Disease. Multicenter hospital study on prescribing patterns for prophylaxis and treatment of complications of cirrhosis. Eur J Clin Pharmacol. 2002;58:435440.
  9. Kanwal F, Kramer JR, Buchanan P, et al. The quality of care provided to patients with cirrhosis and ascites in the Department of Veterans Affairs. Gastroenterology. 2012 143(1):7077.
  10. Chalasani N, Kahi C, Francois F, et al. Improved patient survival after acute variceal bleeding: a multicenter, cohort study. Am J Gastroenterol. 2003;98:653659.
  11. Asrani SK, Larson JJ, Yawn B, Therneau TM, Kim WR. Underestimation of liver‐related mortality in the United States. Gastroenterology. 2013;145:375382.
  12. Bini E, Weisnshel E, Generoso R, et al. Impact of gastroenterology consultation on the outcomes of patients admitted to the hospital with decompensated cirrhosis. Hepatology. 2001;34:10891095.
  13. Cabana MD, Rand CS, Powe NR, et al. Why don't physicians follow clinical practice guidelines? A framework for improvement. JAMA. 1999;282:14581465.
  14. Berman K, Tandra S, Forssell K, et al. Incidence and predictors of 30‐day readmission among patients hospitalized with advanced liver disease. Clin Gastroenterol Hepatol. 2011;9:254259.
  15. Volk M, Tocco R, Bazick J, et al. Hospital Readmissions among patients with decompensated cirrhosis. Am J Gastroenterol. 2012;107:247252.
  16. Kanwal F. Patient‐reported outcomes of cirrhosis. Clin Gastroenterol Hepatol. 2013;11:10431045.
Article PDF
Issue
Journal of Hospital Medicine - 10(4)
Publications
Page Number
236-241
Sections
Files
Files
Article PDF
Article PDF

Decompensated cirrhosis (DC) is defined as cirrhosis with at least 1 of the following complications: ascites, hepatocellular carcinoma, bleeding from portal hypertension, or hepatic encephalopathy. Patients with DC have a median survival estimated at 2 years compared to the 12‐year median survival of compensated cirrhotics.[1] In an era where quality of hospital care is being measured, and where progress is being made in the management of several conditions including congestive heart failure and nosocomial infections, little attention has been paid to DC. The burden of chronic liver failure is clear in the United States, where DC leads to more than 150,000 annual admissions to the hospital and accounts for 40,000 deaths annually.[2]

This burden of disease spurred quality improvement efforts in 2010, when a team of experts identified a set of literature‐based parameters or quality indicators (QI) for patients with cirrhosis.[3] We have demonstrated that adherence to these indicators fell far short of desired targets.[4] A year before their publication, an overall compliance of <50% with these metrics was measured at a single medical center.

We sought to improve the quality of care for patients with DC through implementation of mandatory consultation (MC) with a gastroenterologist for all patients admitted with DC. We assessed whether MC was associated with better care and improved outcomes (hospitalization length of stay [LOS], 30‐day readmission, and inpatient mortality) when compared to usual care (UC).[4]

MATERIALS AND METHODS

Design, Setting, and Patients

We conducted a cohort study comparing adherence to QI and outcomes of patients admitted with DC after the institution of an MC to a historical cohort of patients managed with UC (ie, before MC, adherence to QI for this group has been reported elsewhere).[4] Both cohorts included all patients aged >18 years with DC admitted to Baystate Medical Center, a tertiary care medical center in western Massachusetts. The UC cohort was collected between January 1, 2009 and December 31, 2009, and the MC cohort was assembled between June 1, 2011 and June 30, 2012.

As previously reported,[4] patients were considered for inclusion in the historical cohort if their International Classification of DiseasesNinth Revision discharge code pertained to chronic liver disease (see Supporting Information, Appendix 1, in the online version of this article). This list was broad by design to identify all patients with decompensated cirrhosis. A gastroenterologist (R.G.) then manually extracted charts from electronic medical records (EMRs) using a set of predefined clinical criteria, the same in both cohorts, to identify the patients with DC: cirrhosis with concomitant ascites, hepatic encephalopathy, or gastrointestinal (GI) bleeding secondary to portal hypertension. Other types of decompensated states, such as hepatocellular carcinoma, were not included as their management was not detailed in the QI.[3]

We included patients with suspected or established cirrhosis who had ascites confirmed radiographically or by exam, noting shifting dullness or fluid wave. However, patients were excluded if they lacked sufficient peritoneal fluid for bedside or image‐guided paracentesis. Cirrhotic patients were defined as having hepatic encephalopathy if the patient had altered mental status not secondary to seizures, cerebrovascular accident, or alcohol withdrawal. Finally, gastrointestinal bleeding in cirrhotic patients was defined as any upper or lower bleeding prompting hospital admission, or identified in the medical record as clinically significant by the attending physician.

The same QIs were measured in both cohorts. From the QI set,[3] we selected the 16 QIs that would apply to the management of inpatients (see Supporting Information, Appendix 2, in the online version of this article). Indicators developed for outpatient settings were not included. A quality score was calculated for each admission, defined as the proportion of QIs met divided by the number of QIs for which the patient was eligible. For example, a patient with hepatic encephalopathy but without GI bleeding or ascites would have a score calculated as the number of QIs met for hepatic encephalopathy and documentation of transplant evaluation divided by 3 (2 QIs for hepatic encephalopathy and 1 QI for transplant evaluation). If the patient met both QIs for hepatic encephalopathy, but the consultant failed to address liver transplant eligibility, the score would be 2/3=0.666.

After the institution of the MC, all inpatients with DC were identified within 24 hours of admission by a gastroenterologist (R.G., D.D.), who manually reviewed on a daily basis all admissions from EMRs. An author (R.G.) would then contact the admitting team (hospitalist or resident) to make sure that a gastroenterology consult was called and would then obtain the QI by manual extraction from the EMRs.

Of the 16 gastroenterologists who work at the hospital, 12 of them belong to several private practice groups, whereas 4 are employed by the hospital. As part of the intervention, all gastroenterologists were made aware of the intervention 1 month before the starting date, were provided with a checklist of the QIs of interest, and were encouraged to work with the hospitalist attendings to achieve compliance with the QIs. We reminded the gastroenterologists of the ongoing study during routine division meetings and regularly sought feedback from the hospitalists

The MC consisted of a systematic consultation by a gastroenterologist: any identified patient with DC would generate a mandatory GI consultation and would be assigned to a specialist depending on the roster coverage for that day. A close monitoring of the process allowed us to confirm that all patients admitted with DC were seen by a gastroenterologist. Patients were followed until their discharge, death, or readmission to our institution during the study period.

Outcomes

The primary outcome was defined as the rate of adherence to the QIs and overall QI score expressed as a proportion as noted above. Secondary outcomes included in‐hospital mortality, LOS, and 30‐day readmission rate. These parameters were abstracted from the medical record.

Covariates

The hospital EMR (Cerner Corporation, North Kansas City, MO) was used to extract patient demographic parameters such as gender, race, language, and age at time of admission. Other admission‐level details were extracted from the EMR including Model for End‐Stage Liver Disease (MELD) scores, documented comorbidities (including substance abuse, psychiatric diagnosis, diabetes mellitus, renal failure, congestive heart failure, coronary artery disease, and cancer), underlying etiology for cirrhosis, and reason for admission.

The study was approved by Baystate Medical Center's institutional review board.

Statistical Analysis

Summary statistics for outcomes and covariates were calculated as means/standard deviations (SDs), medians/emnterquartile range, and proportions. Univariable statistics (unpaired t tests, 1‐way analysis of variance, Fisher exact test, Spearman correlation) were used to identify possible demographic (eg, age, race) and clinical (eg, admission complaint) predictors of quality score and with 30‐day outcomes. For each admission, a composite quality score, also known as an opportunity model score,[5, 6] was calculated as a fraction (ie, the number of QIs met divided by the total number of possible QIs indicated by the patient's presentation). This fraction was then multiplied by 100 so as to express the QI score as a percent. Possible scores, therefore, ranged from 0 to 100%.

Calculation of the 30‐day incidence proportion of readmission after the first admission was restricted to patients whose readmission occurred in this hospital, and occurring up to 30‐days before study closure (June 1, 2012). In‐hospital death was examined as a function of QI score during that admission. To derive an unbiased, risk‐adjusted estimate of the association between quality score and outcomes, multiple linear regression (opportunity model score [OMS], LOS) or multiple Poisson regression models (30‐day readmission, in‐hospital death) were built. These included a dummy variable for the study period, as well as any potential confounder that was associated at P0.10, with both study period and the outcome in univariable analyses. Robust standard errors were specified to account for multiple admissions within patients. Marginal means or proportions were then estimated with 95% confidence intervals derived using the delta method. All analyses were performed using Stata 12.1 for Windows (StataCorp, College Station, TX).

RESULTS

A total of 303 patients were observed in 695 hospitalizations;149 patients in 379 admissions were observed in the UC cohort, and 154 patients in 316 admissions were observed in the MC cohort. Baseline demographics of all study admissions appear in Table 1. Patients seen in the MC cohort were younger, more likely to speak English, and less likely to be male or have comorbid diabetes mellitus. Most admissions (n=217, 57.2%; 95% confidence interval: 52.3%‐62.3%) were not evaluated by a gastroenterologist in the UC cohort but all were in the MC cohort.

Patient Characteristics
 UC, N=379, N (%) or Mean/SDMC, N=316, N (%) or Mean/SDP Value*
  • NOTE: Abbreviations: CHF, congestive heart failure; CAD, coronary artery disease; GI, gastrointestinal; MELD, Model for End‐Stage Liver Disease; SD, standard deviation; UC, usual care. *Independent samples t test (continuous), Fisher exact (categorical).

Age, y55.3/12.153.3/13.60.05
English speaking261 (68.9%)261 (82.6%)<0.001
Male251 (66.2%)163 (53.5%)0.001
Race  <0.001
White301 (79.4%)262 (82.9%) 
Black31 (8.2%)40 (12.7%) 
Asian16 (4.2%)0 (0.0%) 
Other31 (8.2%)14 (4.4%) 
Comorbidities   
Substance75 (19.8%)58 (18.4%)0.70
abuse
Psychiatric123 (32.5%)103 (32.9%)0.94
Diabetes mellitus175 (45.4%)115 (36.5%)0.02
Renal failure74 (19.3%)55 (17.4%)0.50
CHF38 (10.0%)24 (7.6%)0.35
CAD26 (6.9%)17 (5.4%)0.43
Cancer48 (12.7%)40 (12.7%)1.00
Admission MELD15.6/6.917.0/7.00.006
Serum creatinine1.43/1.941.42/1.300.91
Reason for admission   
Hepatology/GI318 (83.9%)257 (81.3%)0.42
Renal failure85 (22.4%)90 (28.5%)0.08
Encephalopathy151 (39.3%)113 (34.9%)0.24
GI bleed78 (20.5%)57 (18.0%)1.00
Abdominal pain116 (30.7%)114 (36.2%)0.15
Ascites246 (64.9%)185 (58.5%)0.10

Admission Characteristics

The baseline clinical measures of all study admissions appear in Table 1. The UC and MC cohorts had similar characteristics, with the majority of patients with DC admitted for a gastrointestinal/hepatology‐related reason specifically for the management of ascites and hepatic encephalopathy. The patients in the MC cohort had a statistically higher MELD score on admission, which was not clinically relevant.

Quality Measures

Adherence to individual quality indices is shown in Table 2.

Percent Quality Indicators Met per Admission by Indication
Condition (Denominator)Quality Indicator (Numerator)UC (n=379), Met/IndicatedMC (n=316), Met/IndicatedP Value
  • NOTE: Abbreviations: GI, gastrointestinal; INR, International Normalized Ratio; MELD, Model for End‐Stage Liver Disease; SD, standard deviation; UC, usual care.

Admissions with ascites    
1Admissions to the hospital because of ascites or encephalopathy.Diagnostic paracentesis during admission.77/193, 39.9%, (32.9%, 46.9%)111/135, 82.2% (75.7%, 88.8%)<0.001
2No fibrinolysis or disseminated intravascular coagulation before paracentesis INR <2.5, >100,000 platelets.No fresh frozen plasma or platelet replacement given.36/37, 97.3% (91.8%, 103.0%)41/42, 97.6% (92.8%, 102.4%)1.00
3All admissions with diagnostic paracentesis (not limited to admissions for ascites or hepatic encephalopathy).Cell count differential, total protein, albumin, and culture/sensitivity all performed.31/49, 63.3% (49.3%‐77.3%)46/72 63.9% (52.7%, 75.0%)1.00
4Admissions with known portal hypertension‐related ascites receiving a paracentesis.Ascitic fluid cell count and differential performed.15/104, 14.4% (7.6%‐ 21.3%)47/62, 75.8% (63.2%, 88.4%)<0.001
5Serum sodium 110 mEq/L.Fluid restriction and discontinuation of diuretics.NANANA
6Polymorphonuclear count of 250/mm3 in ascites.Empiric antibiotics, 6 hours of results.10/13, 76.9% (50.4%‐ 103.4%)16/20, 80.0% (60.8%, 99.2%)1.00
7Ascitic fluid, total protein 1.1 gm/dL, serum bilirubin 2.5 mg/dL.Prophylactic antibiotics.4/12, 33.3% (2.0%‐ 64.6%)18/30, 60.0%, (41.4%, 78.6%)0.18
8Normal renal function.Salt restriction and diuretics (spironolactone and loop diuretics).57/186, 30.6%, (24.0%‐ 37.3%)81/122, 66.4%, (57.9%, 74.9%)<0.001
Total ascites subscore, mean/SD30%/36%67%/34%<0.001
GI bleeding    
9Admissions with GI bleeding: variceal and nonvariceal, hematemesis and melena.Upper endoscopy 24 hours of presentation.60/78, 76.9% (67.4%, 86.4%)52/57, 91.2% (83.7%, 98.8%)0.04
10Esophageal varices (active, stigmata of recent bleeding, or no other causes to explain bleeding).Endoscopic variceal ligation/sublerotherapy.40/46, 87.0% (76.8%‐97.1%)30/32, 93.8% (84.9%, 100.0%)0.46
11Admissions with established/suspected upper GI bleeding.Antibiotics within 24 hours of admission.27/69, 39.1% (27.3%‐ 50.9%)26/58, 44.8% (31.6%, 58.0%)0.59
12Admissions with established/suspected variceal bleeding.Somatostatin/octreotide given within 12 hours of presentation.53/69, 76.8%, (66.6%‐ 87.0%)49/58, 84.5% (73.8%, 95.2%)0.37
13Recurrent bleeding within 72 hours of initial endoscopic hemostasis.Repeat endoscopy or transjugular intrahepatic portosystemic shunt.5/5 100%2/3, 66.7% (76.8%, 210.0%)0.38
Total GI subscore, mean/SD61%/38%74%/28%0.04
Liver transplantation    
14Admissions with MELD 15 or MELD 15 and decompensated status (ie, all admissions in our study).Documented evaluation for liver transplantation.112/379, 29.6% (24.9%‐ 34.2%)231/316, 73.6% (68.7%, 78.5%)<0.001
Hepatic encephalopathy    
15Admissions with hepatic encephalopathy.Search for reversible factors documented.81/151, 53.6% (45.6%‐ 61.7 %)97/113, 85.8% (79.4%, 92.3%)<0.001
16Admissions with hepatic encephalopathy.Oral disaccharides/ rifaximin.144/151, 95.3% (91.9 %‐ 98.7 %)107/113, 94.7% (90.7%. 98.69%)1.00
Total encephalopathy subscore, mean/SD75%/28%90%/24%<0.001

Ascites

The management of ascites yielded 3 main differences between the 2 cohorts. Following the implementation of the MC, 82.2 % (111/135) of ascites‐related admissions led to a diagnostic paracentesis as compared to 39.9% (77/193) in the UC group (P<0.001).

In the MC cohort, 75.8% (47/62) of admissions with known portal hypertensionrelated ascites who received a paracentesis had an ascites cell count checked. In contrast, only 14.4% (15/104) in the UC group receiving paracentesis had a fluid cell count (P<0.001). The management of ascites in patients with normal renal function was optimal, with sodium restriction and diuretics combination in 66.4% (81/122) of the MC cohort, whereas this parameter in the UC cohort was only 30.6% (57/186) (P<0.001). There were no significant differences between the groups for the other QIs.

Variceal Bleeding

The MC group had a higher frequency of endoscopy within 24 hours of admissions than the UC group (91.2% [52/57] vs 76.9% [60/78], respectively; P<0.04). The rest had endoscopy later in the admission. Among admissions with bleeding from varices, banding was done 93.8% of the time for patients in the MC group (30/32), which was not statistically different than 87.0% (40/46) for patients seen in the UC group. In the remaining admissions, endoscopy only revealed nonbleeding large esophageal varices, and the endoscopist opted not to proceed with therapy. There were no statistically significant differences in the rest of the management.

Hepatic Encephalopathy

For hepatic encephalopathy, an empirical treatment was given to 95.3 % (144/151) patients in the UC group and 94.7% (107/113) of the patients in the MC group. We found better documentation of a search for underlying etiologies leading to hepatic encephalopathy in the MC cohort 85.8% (97/113) versus the UC cohort, which was only 53.6% (81/151) (P<0.001).

Evaluation for Liver Transplantation

Better documentation of evaluation for liver transplantation was seen in the MC group 73.6% (231/316) in comparison to the UC group 29.4% (111/379) (P<0.001).

Opportunity Score and Clinical Outcomes

As detailed above, care provided during the MC achieved a higher compliance with the QI shown with the QI score or OMS (Table 3). These improvements were not associated with statistically significant differences in in‐hospital death, LOS, or 30‐day readmission. To explore this further we also examined the direct association between the OMS and outcomes in the MC group by dividing patients into 2 groups: patients whose OMS was 80% and those whose OMS was <80% (see Supporting Information, Appendix 4, in the online version of this article). Although there were trends toward decreased in‐hospital death (6.4% vs 8.6%, P=0.26), increased 30‐day readmission (33.8% vs 23.0%, P=0.27), and decreased LOS (6.2 days vs 6.6 days, P=0.77), none of these differences achieved statistical significance.

Outcomes
 UnadjustedAdjusted*
UCMCDifferenceUCMCDifference
  • NOTE: Abbreviations: MC, mandatory consultation; MELD, Model for End‐Stage Liver Disease; UC, usual care. *Quality indicators score adjusted for baseline MELD and age. In‐hospital death adjusted for baseline MELD score and ascites‐related admission. Thirty‐day readmission adjusted for baseline MELD score and race. Length of stay adjusted for baseline MELD ascites‐related admission.

Opportunity model score0.460.77+0.31 (0.24, 0.39)0.460.77+0.30(0.23, 0.37)
In‐hospital death7.1%8.5%+1.4 (0.3, +5.6)7.5%7.9%+0.4% (4.0%, +4.5%)
Readmission within 30 days39.6%32.6%7.0% (16.4%, +2.5%)40.0%31.8%8.2%(18.0%, +1.5%)
Length of stay6.1d6.2d+0.1d (1.0 d, +1.2 d)6.1d6.2d+0.1d (1.0 d, +1.2d)

Mandatory Consultation Subgroups: Employed Versus Private Physicians

The type of employment of the gastroenterologist on consultation (employed by the hospital vs private practice) affected the management of the patients admitted with DC (see Supporting Information, Appendix 3, in the online version of this article). Patients seen by a hospital‐employed gastroenterologist were more likely to have a better documentation in regard to evaluation for liver transplantation and better management of ascites. Except for the prescription of antibiotics in patients presenting with GI bleeding, which were more often given by the employed physician (63% vs 23%, P=0.004), the management of hepatic encephalopathy and GI bleeding was similar between employed and private‐practice physicians.

DISCUSSION

In this evaluation of an MC intervention for patients with DC cared for at a large tertiary academic medical center, we found that the implementation of a routine consultation by a gastroenterologist led to greater adherence to recommended care processes when compared to UC. Overall, the management of ascites and the documentation of evaluation for liver transplantation were statistically superior in the intervention (MC) group. UC and MC were similar with respect to treatment of variceal bleeding and hepatic encephalopathy. Although we did not demonstrate changes in mortality, readmission, or LOS as a result of the MC intervention, our study was underpowered to detect clinically meaningful effects.

The gaps in care of patients with cirrhosis were reported before and after the publication of the formal QIs.[7, 8, 9, 10] These gaps remain relevant in the face of an increasing prevalence of DC along with a recent publication suggesting an underestimation of the burden of liver disease in the United States.[11] Ours is the first study to evaluate the impact on inpatients with DC of a liver service with a systematic, mandatory, specialist consultation. A previous study[12] had shown that a GI consultation would improve the care of patients with DC, but excluded patients with variceal bleeding, did not specifically measure the compliance with QIs, and more important, the GI consult was not mandatory.

Our study has several limitations that must be considered while weighing its findings. The patients were not randomly assigned but followed a pre‐established distribution depending on the call schedule. Some of the improvement we noted might be the result of secular trends; however, this remains unlikely given the lack of national initiatives or pay for performance programs. In the UC cohort, patients who were nonEnglish‐speaking were associated with a lower QI score, which could account for part of the improvement seen in the MC group that has a more prominent English‐speaking cohort. Readmissions could have occurred at other hospitals, and patients were not monitored in an outpatient setting. We did not observe a change in the secondary outcomes (30‐day readmission, LOS, in‐hospital death); however, our study was underpowered for that purpose. Given the complexity of the billing process we did not collect the costs of the MC, which is another limitation of our work. Future studies are needed to determine the cost‐effectiveness of the intervention.

This study shows that a dedicated team of physicians focused on compliance with QIs can achieve a rapid improvement, over a year, in providing higher‐quality care. This may be relevant at other institutions. The strength of our study is that our large tertiary academic medical center serves a large catchment area, with a mix of patients from both rural and urban communities. It is located in Massachusetts, where most of the population has had access to healthcare since 2006. Therefore, although this is a single‐center study, we expect our findings to be more generalizable and less subject to selection bias than other single‐center studies.

Importantly, the compliance with QIs was often far from being perfect in the MC group and was different across type of employment of providers, reflecting the challenges in changing practice among physicians.[13] In fact the QI scores of the private practice group did not change, and mirror the compliance observed at our institution in the previous study, before the implementation of the MC.[4] The difference in performance according to the type of employment of providers stems from 2 factors. First, a better documentation of the need of formal evaluation for liver transplantation by the employed gastroenterologists resulted in better compliance with this QI. Second, and more important, among the employed physicians, there was a readiness to assist the hospitalist with diagnostic/therapeutic paracentesis without relying on, for example, an interventional radiologist. This is reflected by the higher score in the management of ascites. Although our study was not designed to answer this directly, employed physicians may have been more engaged in the project and showed a greater willingness to change practice. In the future, linking reimbursement to quality of care will lead to improved accountability of consultants.

In this study we show that a direct involvement of a gastroenterologist improves the care of inpatients as measured by QIs. We theorize that a better coordination of the transition to outpatient care involving the specialist should lead to better outcomes, specifically a reduction in the 22% observed readmission rate within 30 days of patients with DC.[14, 15] As we move forward, a broader definition of outcomes should be addressed, taking into account patient‐related outcomes and preferences.[16] Future studies should define the relationship between the gastroenterologist and the hospitalist service, the role of physician assistants and nurse practitioners in implementing and monitoring compliance with QIs, and define how physicians and patients can be made accountable in the transition to the outpatient setting.

Disclosures

R.G.: Conception, data collection and interpretation, manuscript. J.F.: Data management, data analysis, manuscript. P.V.: Conception, data analysis, manuscript. P.L.: Conception, data interpretation, manuscript. T.L.: Dr. Lagu is supported by the National Heart, Lung, and Blood Institute of the National Institutes of Health under award number K01HL114745. Conception, data interpretation, manuscript. D.D.: Conception, data collection and interpretation, manuscript. A.B.: Data collection. J.S.: Data collection. Source of funding: internal. The authors report no conflicts of interest.

Decompensated cirrhosis (DC) is defined as cirrhosis with at least 1 of the following complications: ascites, hepatocellular carcinoma, bleeding from portal hypertension, or hepatic encephalopathy. Patients with DC have a median survival estimated at 2 years compared to the 12‐year median survival of compensated cirrhotics.[1] In an era where quality of hospital care is being measured, and where progress is being made in the management of several conditions including congestive heart failure and nosocomial infections, little attention has been paid to DC. The burden of chronic liver failure is clear in the United States, where DC leads to more than 150,000 annual admissions to the hospital and accounts for 40,000 deaths annually.[2]

This burden of disease spurred quality improvement efforts in 2010, when a team of experts identified a set of literature‐based parameters or quality indicators (QI) for patients with cirrhosis.[3] We have demonstrated that adherence to these indicators fell far short of desired targets.[4] A year before their publication, an overall compliance of <50% with these metrics was measured at a single medical center.

We sought to improve the quality of care for patients with DC through implementation of mandatory consultation (MC) with a gastroenterologist for all patients admitted with DC. We assessed whether MC was associated with better care and improved outcomes (hospitalization length of stay [LOS], 30‐day readmission, and inpatient mortality) when compared to usual care (UC).[4]

MATERIALS AND METHODS

Design, Setting, and Patients

We conducted a cohort study comparing adherence to QI and outcomes of patients admitted with DC after the institution of an MC to a historical cohort of patients managed with UC (ie, before MC, adherence to QI for this group has been reported elsewhere).[4] Both cohorts included all patients aged >18 years with DC admitted to Baystate Medical Center, a tertiary care medical center in western Massachusetts. The UC cohort was collected between January 1, 2009 and December 31, 2009, and the MC cohort was assembled between June 1, 2011 and June 30, 2012.

As previously reported,[4] patients were considered for inclusion in the historical cohort if their International Classification of DiseasesNinth Revision discharge code pertained to chronic liver disease (see Supporting Information, Appendix 1, in the online version of this article). This list was broad by design to identify all patients with decompensated cirrhosis. A gastroenterologist (R.G.) then manually extracted charts from electronic medical records (EMRs) using a set of predefined clinical criteria, the same in both cohorts, to identify the patients with DC: cirrhosis with concomitant ascites, hepatic encephalopathy, or gastrointestinal (GI) bleeding secondary to portal hypertension. Other types of decompensated states, such as hepatocellular carcinoma, were not included as their management was not detailed in the QI.[3]

We included patients with suspected or established cirrhosis who had ascites confirmed radiographically or by exam, noting shifting dullness or fluid wave. However, patients were excluded if they lacked sufficient peritoneal fluid for bedside or image‐guided paracentesis. Cirrhotic patients were defined as having hepatic encephalopathy if the patient had altered mental status not secondary to seizures, cerebrovascular accident, or alcohol withdrawal. Finally, gastrointestinal bleeding in cirrhotic patients was defined as any upper or lower bleeding prompting hospital admission, or identified in the medical record as clinically significant by the attending physician.

The same QIs were measured in both cohorts. From the QI set,[3] we selected the 16 QIs that would apply to the management of inpatients (see Supporting Information, Appendix 2, in the online version of this article). Indicators developed for outpatient settings were not included. A quality score was calculated for each admission, defined as the proportion of QIs met divided by the number of QIs for which the patient was eligible. For example, a patient with hepatic encephalopathy but without GI bleeding or ascites would have a score calculated as the number of QIs met for hepatic encephalopathy and documentation of transplant evaluation divided by 3 (2 QIs for hepatic encephalopathy and 1 QI for transplant evaluation). If the patient met both QIs for hepatic encephalopathy, but the consultant failed to address liver transplant eligibility, the score would be 2/3=0.666.

After the institution of the MC, all inpatients with DC were identified within 24 hours of admission by a gastroenterologist (R.G., D.D.), who manually reviewed on a daily basis all admissions from EMRs. An author (R.G.) would then contact the admitting team (hospitalist or resident) to make sure that a gastroenterology consult was called and would then obtain the QI by manual extraction from the EMRs.

Of the 16 gastroenterologists who work at the hospital, 12 of them belong to several private practice groups, whereas 4 are employed by the hospital. As part of the intervention, all gastroenterologists were made aware of the intervention 1 month before the starting date, were provided with a checklist of the QIs of interest, and were encouraged to work with the hospitalist attendings to achieve compliance with the QIs. We reminded the gastroenterologists of the ongoing study during routine division meetings and regularly sought feedback from the hospitalists

The MC consisted of a systematic consultation by a gastroenterologist: any identified patient with DC would generate a mandatory GI consultation and would be assigned to a specialist depending on the roster coverage for that day. A close monitoring of the process allowed us to confirm that all patients admitted with DC were seen by a gastroenterologist. Patients were followed until their discharge, death, or readmission to our institution during the study period.

Outcomes

The primary outcome was defined as the rate of adherence to the QIs and overall QI score expressed as a proportion as noted above. Secondary outcomes included in‐hospital mortality, LOS, and 30‐day readmission rate. These parameters were abstracted from the medical record.

Covariates

The hospital EMR (Cerner Corporation, North Kansas City, MO) was used to extract patient demographic parameters such as gender, race, language, and age at time of admission. Other admission‐level details were extracted from the EMR including Model for End‐Stage Liver Disease (MELD) scores, documented comorbidities (including substance abuse, psychiatric diagnosis, diabetes mellitus, renal failure, congestive heart failure, coronary artery disease, and cancer), underlying etiology for cirrhosis, and reason for admission.

The study was approved by Baystate Medical Center's institutional review board.

Statistical Analysis

Summary statistics for outcomes and covariates were calculated as means/standard deviations (SDs), medians/emnterquartile range, and proportions. Univariable statistics (unpaired t tests, 1‐way analysis of variance, Fisher exact test, Spearman correlation) were used to identify possible demographic (eg, age, race) and clinical (eg, admission complaint) predictors of quality score and with 30‐day outcomes. For each admission, a composite quality score, also known as an opportunity model score,[5, 6] was calculated as a fraction (ie, the number of QIs met divided by the total number of possible QIs indicated by the patient's presentation). This fraction was then multiplied by 100 so as to express the QI score as a percent. Possible scores, therefore, ranged from 0 to 100%.

Calculation of the 30‐day incidence proportion of readmission after the first admission was restricted to patients whose readmission occurred in this hospital, and occurring up to 30‐days before study closure (June 1, 2012). In‐hospital death was examined as a function of QI score during that admission. To derive an unbiased, risk‐adjusted estimate of the association between quality score and outcomes, multiple linear regression (opportunity model score [OMS], LOS) or multiple Poisson regression models (30‐day readmission, in‐hospital death) were built. These included a dummy variable for the study period, as well as any potential confounder that was associated at P0.10, with both study period and the outcome in univariable analyses. Robust standard errors were specified to account for multiple admissions within patients. Marginal means or proportions were then estimated with 95% confidence intervals derived using the delta method. All analyses were performed using Stata 12.1 for Windows (StataCorp, College Station, TX).

RESULTS

A total of 303 patients were observed in 695 hospitalizations;149 patients in 379 admissions were observed in the UC cohort, and 154 patients in 316 admissions were observed in the MC cohort. Baseline demographics of all study admissions appear in Table 1. Patients seen in the MC cohort were younger, more likely to speak English, and less likely to be male or have comorbid diabetes mellitus. Most admissions (n=217, 57.2%; 95% confidence interval: 52.3%‐62.3%) were not evaluated by a gastroenterologist in the UC cohort but all were in the MC cohort.

Patient Characteristics
 UC, N=379, N (%) or Mean/SDMC, N=316, N (%) or Mean/SDP Value*
  • NOTE: Abbreviations: CHF, congestive heart failure; CAD, coronary artery disease; GI, gastrointestinal; MELD, Model for End‐Stage Liver Disease; SD, standard deviation; UC, usual care. *Independent samples t test (continuous), Fisher exact (categorical).

Age, y55.3/12.153.3/13.60.05
English speaking261 (68.9%)261 (82.6%)<0.001
Male251 (66.2%)163 (53.5%)0.001
Race  <0.001
White301 (79.4%)262 (82.9%) 
Black31 (8.2%)40 (12.7%) 
Asian16 (4.2%)0 (0.0%) 
Other31 (8.2%)14 (4.4%) 
Comorbidities   
Substance75 (19.8%)58 (18.4%)0.70
abuse
Psychiatric123 (32.5%)103 (32.9%)0.94
Diabetes mellitus175 (45.4%)115 (36.5%)0.02
Renal failure74 (19.3%)55 (17.4%)0.50
CHF38 (10.0%)24 (7.6%)0.35
CAD26 (6.9%)17 (5.4%)0.43
Cancer48 (12.7%)40 (12.7%)1.00
Admission MELD15.6/6.917.0/7.00.006
Serum creatinine1.43/1.941.42/1.300.91
Reason for admission   
Hepatology/GI318 (83.9%)257 (81.3%)0.42
Renal failure85 (22.4%)90 (28.5%)0.08
Encephalopathy151 (39.3%)113 (34.9%)0.24
GI bleed78 (20.5%)57 (18.0%)1.00
Abdominal pain116 (30.7%)114 (36.2%)0.15
Ascites246 (64.9%)185 (58.5%)0.10

Admission Characteristics

The baseline clinical measures of all study admissions appear in Table 1. The UC and MC cohorts had similar characteristics, with the majority of patients with DC admitted for a gastrointestinal/hepatology‐related reason specifically for the management of ascites and hepatic encephalopathy. The patients in the MC cohort had a statistically higher MELD score on admission, which was not clinically relevant.

Quality Measures

Adherence to individual quality indices is shown in Table 2.

Percent Quality Indicators Met per Admission by Indication
Condition (Denominator)Quality Indicator (Numerator)UC (n=379), Met/IndicatedMC (n=316), Met/IndicatedP Value
  • NOTE: Abbreviations: GI, gastrointestinal; INR, International Normalized Ratio; MELD, Model for End‐Stage Liver Disease; SD, standard deviation; UC, usual care.

Admissions with ascites    
1Admissions to the hospital because of ascites or encephalopathy.Diagnostic paracentesis during admission.77/193, 39.9%, (32.9%, 46.9%)111/135, 82.2% (75.7%, 88.8%)<0.001
2No fibrinolysis or disseminated intravascular coagulation before paracentesis INR <2.5, >100,000 platelets.No fresh frozen plasma or platelet replacement given.36/37, 97.3% (91.8%, 103.0%)41/42, 97.6% (92.8%, 102.4%)1.00
3All admissions with diagnostic paracentesis (not limited to admissions for ascites or hepatic encephalopathy).Cell count differential, total protein, albumin, and culture/sensitivity all performed.31/49, 63.3% (49.3%‐77.3%)46/72 63.9% (52.7%, 75.0%)1.00
4Admissions with known portal hypertension‐related ascites receiving a paracentesis.Ascitic fluid cell count and differential performed.15/104, 14.4% (7.6%‐ 21.3%)47/62, 75.8% (63.2%, 88.4%)<0.001
5Serum sodium 110 mEq/L.Fluid restriction and discontinuation of diuretics.NANANA
6Polymorphonuclear count of 250/mm3 in ascites.Empiric antibiotics, 6 hours of results.10/13, 76.9% (50.4%‐ 103.4%)16/20, 80.0% (60.8%, 99.2%)1.00
7Ascitic fluid, total protein 1.1 gm/dL, serum bilirubin 2.5 mg/dL.Prophylactic antibiotics.4/12, 33.3% (2.0%‐ 64.6%)18/30, 60.0%, (41.4%, 78.6%)0.18
8Normal renal function.Salt restriction and diuretics (spironolactone and loop diuretics).57/186, 30.6%, (24.0%‐ 37.3%)81/122, 66.4%, (57.9%, 74.9%)<0.001
Total ascites subscore, mean/SD30%/36%67%/34%<0.001
GI bleeding    
9Admissions with GI bleeding: variceal and nonvariceal, hematemesis and melena.Upper endoscopy 24 hours of presentation.60/78, 76.9% (67.4%, 86.4%)52/57, 91.2% (83.7%, 98.8%)0.04
10Esophageal varices (active, stigmata of recent bleeding, or no other causes to explain bleeding).Endoscopic variceal ligation/sublerotherapy.40/46, 87.0% (76.8%‐97.1%)30/32, 93.8% (84.9%, 100.0%)0.46
11Admissions with established/suspected upper GI bleeding.Antibiotics within 24 hours of admission.27/69, 39.1% (27.3%‐ 50.9%)26/58, 44.8% (31.6%, 58.0%)0.59
12Admissions with established/suspected variceal bleeding.Somatostatin/octreotide given within 12 hours of presentation.53/69, 76.8%, (66.6%‐ 87.0%)49/58, 84.5% (73.8%, 95.2%)0.37
13Recurrent bleeding within 72 hours of initial endoscopic hemostasis.Repeat endoscopy or transjugular intrahepatic portosystemic shunt.5/5 100%2/3, 66.7% (76.8%, 210.0%)0.38
Total GI subscore, mean/SD61%/38%74%/28%0.04
Liver transplantation    
14Admissions with MELD 15 or MELD 15 and decompensated status (ie, all admissions in our study).Documented evaluation for liver transplantation.112/379, 29.6% (24.9%‐ 34.2%)231/316, 73.6% (68.7%, 78.5%)<0.001
Hepatic encephalopathy    
15Admissions with hepatic encephalopathy.Search for reversible factors documented.81/151, 53.6% (45.6%‐ 61.7 %)97/113, 85.8% (79.4%, 92.3%)<0.001
16Admissions with hepatic encephalopathy.Oral disaccharides/ rifaximin.144/151, 95.3% (91.9 %‐ 98.7 %)107/113, 94.7% (90.7%. 98.69%)1.00
Total encephalopathy subscore, mean/SD75%/28%90%/24%<0.001

Ascites

The management of ascites yielded 3 main differences between the 2 cohorts. Following the implementation of the MC, 82.2 % (111/135) of ascites‐related admissions led to a diagnostic paracentesis as compared to 39.9% (77/193) in the UC group (P<0.001).

In the MC cohort, 75.8% (47/62) of admissions with known portal hypertensionrelated ascites who received a paracentesis had an ascites cell count checked. In contrast, only 14.4% (15/104) in the UC group receiving paracentesis had a fluid cell count (P<0.001). The management of ascites in patients with normal renal function was optimal, with sodium restriction and diuretics combination in 66.4% (81/122) of the MC cohort, whereas this parameter in the UC cohort was only 30.6% (57/186) (P<0.001). There were no significant differences between the groups for the other QIs.

Variceal Bleeding

The MC group had a higher frequency of endoscopy within 24 hours of admissions than the UC group (91.2% [52/57] vs 76.9% [60/78], respectively; P<0.04). The rest had endoscopy later in the admission. Among admissions with bleeding from varices, banding was done 93.8% of the time for patients in the MC group (30/32), which was not statistically different than 87.0% (40/46) for patients seen in the UC group. In the remaining admissions, endoscopy only revealed nonbleeding large esophageal varices, and the endoscopist opted not to proceed with therapy. There were no statistically significant differences in the rest of the management.

Hepatic Encephalopathy

For hepatic encephalopathy, an empirical treatment was given to 95.3 % (144/151) patients in the UC group and 94.7% (107/113) of the patients in the MC group. We found better documentation of a search for underlying etiologies leading to hepatic encephalopathy in the MC cohort 85.8% (97/113) versus the UC cohort, which was only 53.6% (81/151) (P<0.001).

Evaluation for Liver Transplantation

Better documentation of evaluation for liver transplantation was seen in the MC group 73.6% (231/316) in comparison to the UC group 29.4% (111/379) (P<0.001).

Opportunity Score and Clinical Outcomes

As detailed above, care provided during the MC achieved a higher compliance with the QI shown with the QI score or OMS (Table 3). These improvements were not associated with statistically significant differences in in‐hospital death, LOS, or 30‐day readmission. To explore this further we also examined the direct association between the OMS and outcomes in the MC group by dividing patients into 2 groups: patients whose OMS was 80% and those whose OMS was <80% (see Supporting Information, Appendix 4, in the online version of this article). Although there were trends toward decreased in‐hospital death (6.4% vs 8.6%, P=0.26), increased 30‐day readmission (33.8% vs 23.0%, P=0.27), and decreased LOS (6.2 days vs 6.6 days, P=0.77), none of these differences achieved statistical significance.

Outcomes
 UnadjustedAdjusted*
UCMCDifferenceUCMCDifference
  • NOTE: Abbreviations: MC, mandatory consultation; MELD, Model for End‐Stage Liver Disease; UC, usual care. *Quality indicators score adjusted for baseline MELD and age. In‐hospital death adjusted for baseline MELD score and ascites‐related admission. Thirty‐day readmission adjusted for baseline MELD score and race. Length of stay adjusted for baseline MELD ascites‐related admission.

Opportunity model score0.460.77+0.31 (0.24, 0.39)0.460.77+0.30(0.23, 0.37)
In‐hospital death7.1%8.5%+1.4 (0.3, +5.6)7.5%7.9%+0.4% (4.0%, +4.5%)
Readmission within 30 days39.6%32.6%7.0% (16.4%, +2.5%)40.0%31.8%8.2%(18.0%, +1.5%)
Length of stay6.1d6.2d+0.1d (1.0 d, +1.2 d)6.1d6.2d+0.1d (1.0 d, +1.2d)

Mandatory Consultation Subgroups: Employed Versus Private Physicians

The type of employment of the gastroenterologist on consultation (employed by the hospital vs private practice) affected the management of the patients admitted with DC (see Supporting Information, Appendix 3, in the online version of this article). Patients seen by a hospital‐employed gastroenterologist were more likely to have a better documentation in regard to evaluation for liver transplantation and better management of ascites. Except for the prescription of antibiotics in patients presenting with GI bleeding, which were more often given by the employed physician (63% vs 23%, P=0.004), the management of hepatic encephalopathy and GI bleeding was similar between employed and private‐practice physicians.

DISCUSSION

In this evaluation of an MC intervention for patients with DC cared for at a large tertiary academic medical center, we found that the implementation of a routine consultation by a gastroenterologist led to greater adherence to recommended care processes when compared to UC. Overall, the management of ascites and the documentation of evaluation for liver transplantation were statistically superior in the intervention (MC) group. UC and MC were similar with respect to treatment of variceal bleeding and hepatic encephalopathy. Although we did not demonstrate changes in mortality, readmission, or LOS as a result of the MC intervention, our study was underpowered to detect clinically meaningful effects.

The gaps in care of patients with cirrhosis were reported before and after the publication of the formal QIs.[7, 8, 9, 10] These gaps remain relevant in the face of an increasing prevalence of DC along with a recent publication suggesting an underestimation of the burden of liver disease in the United States.[11] Ours is the first study to evaluate the impact on inpatients with DC of a liver service with a systematic, mandatory, specialist consultation. A previous study[12] had shown that a GI consultation would improve the care of patients with DC, but excluded patients with variceal bleeding, did not specifically measure the compliance with QIs, and more important, the GI consult was not mandatory.

Our study has several limitations that must be considered while weighing its findings. The patients were not randomly assigned but followed a pre‐established distribution depending on the call schedule. Some of the improvement we noted might be the result of secular trends; however, this remains unlikely given the lack of national initiatives or pay for performance programs. In the UC cohort, patients who were nonEnglish‐speaking were associated with a lower QI score, which could account for part of the improvement seen in the MC group that has a more prominent English‐speaking cohort. Readmissions could have occurred at other hospitals, and patients were not monitored in an outpatient setting. We did not observe a change in the secondary outcomes (30‐day readmission, LOS, in‐hospital death); however, our study was underpowered for that purpose. Given the complexity of the billing process we did not collect the costs of the MC, which is another limitation of our work. Future studies are needed to determine the cost‐effectiveness of the intervention.

This study shows that a dedicated team of physicians focused on compliance with QIs can achieve a rapid improvement, over a year, in providing higher‐quality care. This may be relevant at other institutions. The strength of our study is that our large tertiary academic medical center serves a large catchment area, with a mix of patients from both rural and urban communities. It is located in Massachusetts, where most of the population has had access to healthcare since 2006. Therefore, although this is a single‐center study, we expect our findings to be more generalizable and less subject to selection bias than other single‐center studies.

Importantly, the compliance with QIs was often far from being perfect in the MC group and was different across type of employment of providers, reflecting the challenges in changing practice among physicians.[13] In fact the QI scores of the private practice group did not change, and mirror the compliance observed at our institution in the previous study, before the implementation of the MC.[4] The difference in performance according to the type of employment of providers stems from 2 factors. First, a better documentation of the need of formal evaluation for liver transplantation by the employed gastroenterologists resulted in better compliance with this QI. Second, and more important, among the employed physicians, there was a readiness to assist the hospitalist with diagnostic/therapeutic paracentesis without relying on, for example, an interventional radiologist. This is reflected by the higher score in the management of ascites. Although our study was not designed to answer this directly, employed physicians may have been more engaged in the project and showed a greater willingness to change practice. In the future, linking reimbursement to quality of care will lead to improved accountability of consultants.

In this study we show that a direct involvement of a gastroenterologist improves the care of inpatients as measured by QIs. We theorize that a better coordination of the transition to outpatient care involving the specialist should lead to better outcomes, specifically a reduction in the 22% observed readmission rate within 30 days of patients with DC.[14, 15] As we move forward, a broader definition of outcomes should be addressed, taking into account patient‐related outcomes and preferences.[16] Future studies should define the relationship between the gastroenterologist and the hospitalist service, the role of physician assistants and nurse practitioners in implementing and monitoring compliance with QIs, and define how physicians and patients can be made accountable in the transition to the outpatient setting.

Disclosures

R.G.: Conception, data collection and interpretation, manuscript. J.F.: Data management, data analysis, manuscript. P.V.: Conception, data analysis, manuscript. P.L.: Conception, data interpretation, manuscript. T.L.: Dr. Lagu is supported by the National Heart, Lung, and Blood Institute of the National Institutes of Health under award number K01HL114745. Conception, data interpretation, manuscript. D.D.: Conception, data collection and interpretation, manuscript. A.B.: Data collection. J.S.: Data collection. Source of funding: internal. The authors report no conflicts of interest.

References
  1. D'Amico G, Garcia‐Tsao G, Pagliaro L. Natural history and prognostic indicators of survival in cirrhosis: a systematic review of 118 studies. J Hepatol. 2006;44:217231.
  2. Wigg AJ, McCormick R, Wundke R, Woodman RJ. Efficacy of a chronic disease management model for patients with chronic liver failure. Clin Gastroenterol Hepatol. 2013;11:850858.
  3. Kanwal F, Kramer J, Asch SM, et al. An Explicit Quality Indicator Set for Measurement of Quality of Care in Patients with Cirrhosis. Clin Gastroenterol Hepatol. 2010;8:709717.
  4. Ghaoui R, Friderici J, Visintainer P, Lindenauer PK, Lagu T, Desilets D. Measurement of the quality of care of patients admitted with decompensated cirrhosis. Liver Int. 2014:34:204210.
  5. Nolan T, Berwick DM. All‐or‐none measurement raises the bar on performance. JAMA. 2006;295:11681170.
  6. Joint Commission on Accreditation of Healthcare Organizations. Quality report user guide. Available at: http://www.jointcommission.org. Accessed May 30, 2011.
  7. Saab S, Nguyen S, Ibrahim A, et al. Management of patients with cirrhosis in Southern California: results of a practitioner survey. J Clin Gastroenterol. 2006;40:156161.
  8. Lucena MI, Andrade RJ, Tognoni G, et al. Spanish Collaborative Study Group on Therapeutic Management In Liver Disease. Multicenter hospital study on prescribing patterns for prophylaxis and treatment of complications of cirrhosis. Eur J Clin Pharmacol. 2002;58:435440.
  9. Kanwal F, Kramer JR, Buchanan P, et al. The quality of care provided to patients with cirrhosis and ascites in the Department of Veterans Affairs. Gastroenterology. 2012 143(1):7077.
  10. Chalasani N, Kahi C, Francois F, et al. Improved patient survival after acute variceal bleeding: a multicenter, cohort study. Am J Gastroenterol. 2003;98:653659.
  11. Asrani SK, Larson JJ, Yawn B, Therneau TM, Kim WR. Underestimation of liver‐related mortality in the United States. Gastroenterology. 2013;145:375382.
  12. Bini E, Weisnshel E, Generoso R, et al. Impact of gastroenterology consultation on the outcomes of patients admitted to the hospital with decompensated cirrhosis. Hepatology. 2001;34:10891095.
  13. Cabana MD, Rand CS, Powe NR, et al. Why don't physicians follow clinical practice guidelines? A framework for improvement. JAMA. 1999;282:14581465.
  14. Berman K, Tandra S, Forssell K, et al. Incidence and predictors of 30‐day readmission among patients hospitalized with advanced liver disease. Clin Gastroenterol Hepatol. 2011;9:254259.
  15. Volk M, Tocco R, Bazick J, et al. Hospital Readmissions among patients with decompensated cirrhosis. Am J Gastroenterol. 2012;107:247252.
  16. Kanwal F. Patient‐reported outcomes of cirrhosis. Clin Gastroenterol Hepatol. 2013;11:10431045.
References
  1. D'Amico G, Garcia‐Tsao G, Pagliaro L. Natural history and prognostic indicators of survival in cirrhosis: a systematic review of 118 studies. J Hepatol. 2006;44:217231.
  2. Wigg AJ, McCormick R, Wundke R, Woodman RJ. Efficacy of a chronic disease management model for patients with chronic liver failure. Clin Gastroenterol Hepatol. 2013;11:850858.
  3. Kanwal F, Kramer J, Asch SM, et al. An Explicit Quality Indicator Set for Measurement of Quality of Care in Patients with Cirrhosis. Clin Gastroenterol Hepatol. 2010;8:709717.
  4. Ghaoui R, Friderici J, Visintainer P, Lindenauer PK, Lagu T, Desilets D. Measurement of the quality of care of patients admitted with decompensated cirrhosis. Liver Int. 2014:34:204210.
  5. Nolan T, Berwick DM. All‐or‐none measurement raises the bar on performance. JAMA. 2006;295:11681170.
  6. Joint Commission on Accreditation of Healthcare Organizations. Quality report user guide. Available at: http://www.jointcommission.org. Accessed May 30, 2011.
  7. Saab S, Nguyen S, Ibrahim A, et al. Management of patients with cirrhosis in Southern California: results of a practitioner survey. J Clin Gastroenterol. 2006;40:156161.
  8. Lucena MI, Andrade RJ, Tognoni G, et al. Spanish Collaborative Study Group on Therapeutic Management In Liver Disease. Multicenter hospital study on prescribing patterns for prophylaxis and treatment of complications of cirrhosis. Eur J Clin Pharmacol. 2002;58:435440.
  9. Kanwal F, Kramer JR, Buchanan P, et al. The quality of care provided to patients with cirrhosis and ascites in the Department of Veterans Affairs. Gastroenterology. 2012 143(1):7077.
  10. Chalasani N, Kahi C, Francois F, et al. Improved patient survival after acute variceal bleeding: a multicenter, cohort study. Am J Gastroenterol. 2003;98:653659.
  11. Asrani SK, Larson JJ, Yawn B, Therneau TM, Kim WR. Underestimation of liver‐related mortality in the United States. Gastroenterology. 2013;145:375382.
  12. Bini E, Weisnshel E, Generoso R, et al. Impact of gastroenterology consultation on the outcomes of patients admitted to the hospital with decompensated cirrhosis. Hepatology. 2001;34:10891095.
  13. Cabana MD, Rand CS, Powe NR, et al. Why don't physicians follow clinical practice guidelines? A framework for improvement. JAMA. 1999;282:14581465.
  14. Berman K, Tandra S, Forssell K, et al. Incidence and predictors of 30‐day readmission among patients hospitalized with advanced liver disease. Clin Gastroenterol Hepatol. 2011;9:254259.
  15. Volk M, Tocco R, Bazick J, et al. Hospital Readmissions among patients with decompensated cirrhosis. Am J Gastroenterol. 2012;107:247252.
  16. Kanwal F. Patient‐reported outcomes of cirrhosis. Clin Gastroenterol Hepatol. 2013;11:10431045.
Issue
Journal of Hospital Medicine - 10(4)
Issue
Journal of Hospital Medicine - 10(4)
Page Number
236-241
Page Number
236-241
Publications
Publications
Article Type
Display Headline
Outcomes associated with a mandatory gastroenterology consultation to improve the quality of care of patients hospitalized with decompensated cirrhosis
Display Headline
Outcomes associated with a mandatory gastroenterology consultation to improve the quality of care of patients hospitalized with decompensated cirrhosis
Sections
Article Source

© 2014 Society of Hospital Medicine

Disallow All Ads
Correspondence Location
Address for correspondence and reprint requests: Rony Ghaoui, MD, Division of Gastroenterology, Baystate Medical Center, 759 Chestnut St., S2606, Springfield, MA 01199; Telephone: 413‐794‐3570; Fax: 413‐794‐8828; E‐mail: rony.ghaoui@bhs.org
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Article PDF Media
Media Files