Affiliations
Department of Medicine, Johns Hopkins University School of Medicine and Johns Hopkins Bayview Medical Center, Baltimore, Maryland
Given name(s)
Olufunmilayo
Family name
Falade
Degrees
MD

Medication Warnings for Adults

Article Type
Changed
Sun, 05/21/2017 - 13:33
Display Headline
Factors associated with medication warning acceptance for hospitalized adults

Many computerized provider order entry (CPOE) systems suffer from having too much of a good thing. Few would question the beneficial effect of CPOE on medication order clarity, completeness, and transmission.[1, 2] When mechanisms for basic decision support have been added, however, such as allergy, interaction, and duplicate warnings, reductions in medication errors and adverse events have not been consistently achieved.[3, 4, 5, 6, 7] This is likely due in part to the fact that ordering providers override medication warnings at staggeringly high rates.[8, 9] Clinicians acknowledge that they are ignoring potentially valuable warnings,[10, 11] but suffer from alert fatigue due to the sheer number of messages, many of them judged by clinicians to be of low‐value.[11, 12]

Redesign of medication alert systems to increase their signal‐to‐noise ratio is badly needed,[13, 14, 15, 16] and will need to consider the clinical significance of alerts, their presentation, and context‐specific factors that potentially contribute to warning effectiveness.[17, 18, 19] Relatively few studies, however, have objectively looked at context factors such as the characteristics of providers, patients, medications, and warnings that are associated with provider responses to warnings,[9, 20, 21, 22, 23, 24, 25] and only 2 have studied how warning acceptance is associated with medication risk.[18, 26] We wished to explore these factors further. Warning acceptance has been shown to be higher, at least in the outpatient setting, when orders are entered by low‐volume prescribers for infrequently encountered warnings,[24] and there is some evidence that patients receive higher‐quality care during the day.[27] Significant attention has been placed in recent years on inappropriate prescribing in older patients,[28] and on creating a culture of safety in healthcare.[29] We therefore hypothesized that our providers would be more cautious, and medication warning acceptance rates would be higher, when orders were entered for patients who were older or with more complex medical problems, when they were entered during the day by caregivers who entered few orders, when the medications ordered were potentially associated with greater risk, and when the warnings themselves were infrequently encountered.

METHODS

Setting and Caregivers

Johns Hopkins Bayview Medical Center (JHBMC) is a 400‐bed academic medical center serving southeastern Baltimore, Maryland. Prescribing caregivers include residents and fellows who rotate to both JHBMC and Johns Hopkins Hospital, internal medicine hospitalists, other attending physicians (including teaching attendings for all departments, and hospitalists and clinical associates for departments other than internal medicine), and nurse practitioners and physician assistants from most JHBMC departments. Nearly 100% of patients on the surgery, obstetrics/gynecology, neurology, psychiatry, and chemical dependence services are hospitalized on units dedicated to their respective specialty, and the same is true for approximately 95% of medicine patients.

Order Entry

JHBMC began using a client‐server order entry system by MEDITECH (Westwood, MA) in July 2003. Provider order entry was phased in beginning in October 2003 and completed by the end of 2004. MEDITECH version 5.64 was being used during the study period. Medications may generate duplicate, interaction, allergy, adverse reaction, and dose warnings during a patient ordering session each time they are ordered. Duplicate warnings are generated when the same medication (no matter what route) is ordered that is either on their active medication list, was on the list in the preceding 24 hours, or that is being ordered simultaneously. A drug‐interaction database licensed from First DataBank (South San Francisco, CA) is utilized, and updated monthly, which classifies potential drug‐drug interactions as contraindicated, severe, intermediate, and mild. Those classified as contraindicated by First DataBank are included in the severe category in MEDITECH 5.64. During the study period, JHBMC's version of MEDITECH was configured so that providers were warned of potential severe and intermediate drug‐drug interactions, but not mild. No other customizations had been made. Patients' histories of allergies and other adverse responses to medications can be entered by any credentialed staff member. They are maintained together in an allergies section of the electronic medical record, but are identified as either allergy or adverse reactions at the time they are entered, and each generates its own warnings.

When more than 1 duplicate, interaction, allergy, or adverse reaction warning is generated for a particular medication, all appear listed on a single screen in identical fonts. No visual distinction is made between severe and intermediate drug‐drug interactions; for these, the category of medication ordered is followed by the category of the medication for which there is a potential interaction. A details button can be selected to learn specifically which medications are involved and the severity and nature of the potential interactions identified. In response to the warnings, providers can choose to either override them, erase the order, or replace the order by clicking 1 of 3 buttons at the bottom of the screen. Warnings are not repeated unless the medication is reordered for that patient. Dose warnings appear on a subsequent screen and are not addressed in this article.

Nurses are discouraged from entering verbal orders but do have the capacity to do so, at which time they encounter and must respond to the standard medication warnings, if any. Medical students are able to enter orders, at which time they also encounter and must respond to the standard medication warnings; their orders must then be cosigned by a licensed provider before they can be processed. Warnings encountered by nurses and medical students are not repeated at the time of cosignature by a licensed provider.

Data Collection

We collected data regarding all medication orders placed in our CPOE system from October 1, 2009 to April 20, 2010 for all adult patients. Intensive care unit (ICU) patients were excluded, in anticipation of a separate analysis. Hospitalizations under observation were also excluded. We then ran a report showing all medications that generated any number of warnings of any type (duplicate, interaction, allergy, or adverse reaction) for the same population. Warnings generated during readmissions that occurred at any point during the study period (ranging from 1 to 21 times) were excluded, because these patients likely had many, if not all, of the same medications ordered during their readmissions as during their initial hospitalization, which would unduly influence the analysis if retained.

There was wide variation in the number of warnings generated per medication and in the number of each warning type per medication that generated multiple warnings. Therefore, for ease of analysis and to ensure that we could accurately determine varying response to each individual warning type, we thereafter focused on the medications that generated single warnings during the study period. For each single warning we obtained patient name, account number, event date and time, hospital unit at the time of the event, ordered medication, ordering staff member, warning type, and staff member response to the warning (eg, override warning or erase order [accept the warning]). The response replace was used very infrequently, and therefore warnings that resulted in this response were excluded. Medications available in more than 1 form included the route of administration in their name, and from this they were categorized as parenteral or nonparenteral. All nonparenteral or parenteral forms of a given medication were grouped together as 1 medication (eg, morphine sustained release and morphine elixir were classified as a single‐medication, nonparenteral morphine). Medications were further categorized according to whether or not they were on the Institute for Safe Medication Practice (ISMP) List of High‐Alert Medications.[30]

The study was approved by the Johns Hopkins Institutional Review Board.

Analysis

We collected descriptive data about patients and providers. Age and length of stay (LOS) at the time of the event were determined based on the patients' admit date and date of birth, and grouped into quartiles. Hospital units were grouped according to which service or services they primarily served. Medications were grouped into quartiles according to the total number of warnings they generated during the study period. Warnings were dichotomously categorized according to whether they were overridden or accepted. Unpaired t tests were used to compare continuous variables for the 2 groups, and [2] tests were used to compare categorical variables. A multivariate logistic regression was then performed, using variables with a P value of <0.10 in the univariate analysis, to control for confounders and identify independent predictors of medication warning acceptance. All analyses were performed using Intercooled Stata 12 (StataCorp, College Station, TX).

RESULTS

A total of 259,656 medication orders were placed for adult non‐ICU patients during the 7‐month study period. Of those orders, 45,835 generated some number of medication warnings.[1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20] The median number of warnings per patient was 4 (interquartile range [IQR]=28; mean=5.9, standard deviation [SD]=6.2), with a range from 1 to 84. The median number of warnings generated per provider during the study period was 36 (IQR=6106, mean=87.4, SD=133.7), with a range of 1 to 1096.

There were 40,391 orders placed for 454 medications for adult non‐ICU patients, which generated a single‐medication warning (excluding those with the response replace, which was used 20 times) during the 7‐month study period. Data regarding the patients and providers associated with the orders generating single warnings are shown in Table 1. Most patients were on medicine units, and most orders were entered by residents. Patients' LOS at the time the orders were placed ranged from 0 to 118 days (median=1, IQR=04; mean=4.0, SD=7.2). The median number of single warnings per patient was 4 (IQR=28; mean=6.1, SD=6.5), with a range from 1 to 84. The median number of single warnings generated per provider during the study period was 15 (IQR=373; mean=61.7, SD=109.6), with a range of 1 to 1057.

Patient and Provider Features
 No. (%)
  • NOTE: Abbreviations: GYN, gynecology; IM, internal medicine; Neuro/psych/chem dep, neurology/psychiatry/chemical dependence; NP, nurse practitioner; OB, obstetrics; PA, physician assistant.

  • Hospital unit at the time of order entry.

  • Total is >100% due to rounding.

Patients (N=6,646) 
Age 
1545 years2,048 (31%)
4657 years1,610 (24%)
5872 years1,520 (23%)
73104 years1,468 (22%)
Gender 
Male2,934 (44%)
Hospital unita 
Medicine2,992 (45%)
Surgery1,836 (28%)
Neuro/psych/chem dep1,337 (20%)
OB/GYN481 (7%)
Caregivers (N=655) 
Resident248 (38%)b
Nurse154 (24%)
Attending or other97 (15%)
NP/PA69 (11%)
IM hospitalist31 (5%)
Fellow27 (4%)
Medical student23 (4%)
Pharmacist6 (1%)

Patient and caregiver characteristics for the medication orders that generated single warnings are shown in Table 2. The majority of medications were nonparenteral and not on the ISMP list (Table 3). Most warnings generated were either duplicate (47%) or interaction warnings (47%). Warnings of a particular type were repeated 14.5% of the time for a particular medication and patient (from 2 to 24 times, median=2, IQR=22, mean=2.7, SD=1.4), and 9.8% of the time for a particular caregiver, medication, and patient (from 2 to 18 times, median=2, IQR=22, mean=2.4, SD=1.1).

Characteristics of Patients, Caregivers, Orders, Medications, and Warnings for Medication Orders Generating Single Warnings, and Association With Warning Acceptance
VariableNo. of Warnings (%)aNo. of Warnings Accepted (%)aP
  • NOTE: Abbreviations: GYN, gynecology; IM, internal medicine; ISMP, Institute for Safe Medication Practices; Neuro/psych/chem dep, neurology/psychiatry/chemical dependence; NP, nurse practitioner; OB, obstetrics; PA, physician assistant.

  • Totals may not equal 100% due to rounding.

  • Total number of medications is >454 because many medications generated more than 1 warning type.

Patient age   
1545 years10,881 (27)602 (5.5%)<0.001
4657 years9,733 (24)382 (3.9%) 
5872 years10,000 (25)308 (3.1%) 
73104 years9,777 (24)262 (2.7%) 
Patient gender   
Female23,395 (58)866 (3.7%)0.074
Male16,996 (42)688 (4.1%) 
Patient length of stay   
<1 day10,721 (27)660 (6.2%)<0.001
1 day10,854 (27)385 (3.5%) 
24 days10,424 (26)277 (2.7%) 
5118 days8,392 (21)232 (2.8%) 
Patient hospital unit   
Medicine20,057 (50)519 (2.6%)<0.001
Surgery10,274 (25)477 (4.6%) 
Neuro/psych/chem dep8,279 (21)417 (5.0%) 
OB/GYN1,781 (4)141 (7.9%) 
Ordering caregiver   
Resident22,523 (56)700 (3.1%)<0.001
NP/PA7,534 (19)369 (4.9%) 
IM hospitalist5,048 (13)155 (3.1%) 
Attending3225 (8)219 (6.8%) 
Fellow910 (2)34 (3.7%) 
Nurse865 (2)58 (6.7%) 
Medical student265 (<1)17 (6.4%) 
Pharmacist21 (<1)2 (9.5%) 
Day ordered   
Weekday31,499 (78%)1276 (4.1%)<0.001
Weekend8,892 (22%)278 (3.1%) 
Time ordered   
000005594,231 (11%)117 (2.8%)<0.001
0600115911,696 (29%)348 (3.0%) 
1200175915,879 (39%)722 (4.6%) 
180023598,585 (21%)367 (4.3%) 
Administration route (no. of meds)  
Nonparenteral (339)27,086 (67%)956 (3.5%)<0.001
Parenteral (115)13,305 (33%)598 (4.5%) 
ISMP List of High‐Alert Medications status (no. of meds)[30]  
Not on ISMP list (394)27,503 (68%)1251 (4.5%)<0.001
On ISMP list (60)12,888 (32%)303 (2.4%) 
No. of warnings per med (no. of meds)  
11062133 (7)9,869 (24%)191 (1.9%)<0.001
4681034 (13)10,014 (25%)331 (3.3%) 
170444 (40)10,182 (25%)314 (3.1%) 
1169 (394)10,326 (26%)718 (7.0%) 
Warning type (no. of meds)b  
Duplicate (369)19,083 (47%)1041 (5.5%)<0.001
Interaction (315)18,894 (47%)254 (1.3%) 
Allergy (138)2,371 (6%)243 (10.0%) 
Adverse reaction (14)43 (0.1%)16 (37%) 
Multivariate Analysis of Factors Associated With Acceptance of Medication Warnings
VariableAdjusted OR95% CI
  • NOTE: Abbreviations: CI, confidence interval; GYN, gynecology; IM, internal medicine; ISMP, Institute for Safe Medication Practices; Neuro/psych/chem dep, neurology/psychiatry/chemical dependence; NP, nurse practitioner; OB, obstetrics; OR, odds ratio; PA, physician assistant.

  • Day ordered and time of order entry were included but were not significant in the multivariate model.

Patient age  
1545 years1.00Reference
4657 years0.890.771.02
5872 years0.850.730.99
73104 years0.910.771.08
Patient gender  
Female1.00Reference
Male1.261.131.41
Patient length of stay 
<1 day1.00Reference
1 day0.650.550.76
24 days0.490.420.58
5118 days0.490.410.58
Patient hospital unit  
Medicine1.00Reference
Surgery1.451.251.68
Neuro/psych/chem dep1.351.151.58
OB/GYN2.431.923.08
Ordering caregiver  
Resident1.00Reference
NP/PA1.631.421.88
IM hospitalist1.241.021.50
Attending1.831.542.18
Fellow1.410.982.03
Nurse1.921.442.57
Medical student1.170.701.95
Pharmacist3.080.6714.03
Medication factors  
Nonparenteral1.00Reference
Parenteral1.791.592.03
HighAlert Medication status (no. of meds)[30]
Not on ISMP list1.00Reference
On ISMP list0.370.320.43
No. of warnings per medication 
110621331.00Reference
46810342.301.902.79
1704442.251.852.73
11694.103.424.92
Warning type  
Duplicate1.00Reference
Interaction0.240.210.28
Allergy2.281.942.68
Adverse reaction9.244.5218.90

One thousand five hundred fifty‐four warnings were erased (ie, accepted by clinicians [4%]). In univariate analysis, only patient gender was not associated with warning acceptance. Patient age, LOS, hospital unit at the time of order entry, ordering caregiver type, day and time the medication was ordered, administration route, presence on the ISMP list, warning frequency, and warning type were all significantly associated with warning acceptance (Table 2).

Older patient age, longer LOS, presence of the medication on the ISMP list, and interaction warning type were all negatively associated with warning acceptance in multivariable analysis. Warning acceptance was positively associated with male patient gender, being on a service other than medicine, being a caregiver other than a resident, parenteral medications, lower warning frequency, and allergy or adverse reaction warning types (Table 3).

The 20 medications that generated the most single warnings are shown in Table 4. Medications on the ISMP list accounted for 8 of these top 20 medications. For most of them, duplicate and interaction warnings accounted for most of the warnings generated, except for parenteral hydromorphone, oral oxycodone, parenteral morphine, and oral hydromorphone, which each had more allergy than interaction warnings.

Top 20 Medications Generating Single Warnings and Warning Type Distribution for Each
MedicationISMP ListbNo. of WarningsDuplicate, No. (%)cInteraction, No. (%)cAllergy, No. (%)cAdverse Reaction, No. (%)c
  • NOTE: Abbreviations: ISMP, Institute for Safe Medication Practices.

  • Medications not noted as injectable should be presumed not parenteral.

  • SMP List of High‐Alert Medications.[30]

  • Total may not add up to 100% due to rounding.

Hydromorphone injectableYes2,1331,584 (74.3)127 (6.0)422 (19.8) 
Metoprolol 1,432550 (38.4)870 (60.8)12 (0.8) 
Aspirin 1,375212 (15.4)1,096 (79.7)67 (4.9) 
OxycodoneYes1,360987 (72.6) 364 (26.8)9 (0.7)
Potassium chloride 1,296379 (29.2)917 (70.8)  
Ondansetron injectable 1,1671,013 (86.8)153 (13.1)1 (0.1) 
Aspart insulin injectableYes1,106643 (58.1)463 (41.9)  
WarfarinYes1,034298 (28.8)736 (71.2)  
Heparin injectableYes1,030205 (19.9)816 (79.2)9 (0.3) 
Furosemide injectable 980438 (45.0)542 (55.3)  
Lisinopril 926225 (24.3)698 (75.4)3 (0.3) 
Acetaminophen 860686 (79.8)118 (13.7)54 (6.3)2 (0.2)
Morphine injectableYes804467 (58.1)100 (12.4)233 (29.0)4 (0.5)
Diazepam 786731 (93.0)41 (5.2)14 (1.8) 
Glargine insulin injectableYes746268 (35.9)478 (64.1)  
Ibuprofen 713125 (17.5)529 (74.2)54 (7.6)5 (0.7)
HydromorphoneYes594372 (62.6)31 (5.2)187 (31.5)4 (0.7)
Furosemide 586273 (46.6)312 (53.2)1 (0.2) 
Ketorolac injectable 48739 (8.0)423 (86.9)23 (4.7)2 (0.4)
Prednisone 468166 (35.5)297 (63.5)5 (1.1) 

DISCUSSION

Medication warnings in our study were frequently overridden, particularly when encountered by residents, for patients with a long LOS and on the internal medicine service, and for medications generating the most warnings and on the ISMP list. Disturbingly, this means that potentially important warnings for medications with the highest potential for causing harm, for possibly the sickest and most complex patients, were those that were most often ignored by young physicians in training who should have had the most to gain from them. Of course, this is not entirely surprising. Despite our hope that a culture of safety would influence young physicians' actions when caring for these patients and prescribing these medications, these patients and medications are those for whom the most warnings are generated, and these physicians are the ones entering the most orders. Only 13% of the medications studied were on the ISMP list, but they generated 32% of the warnings. We controlled for number of warnings and ISMP list status, but not for warning validity. Most likely, high‐risk medications have been set up with more warnings, many of them of lower quality, in an errant but well‐intentioned effort to make them safer. If developers of CPOE systems want to gain serious traction in using decision support to promote prescribing safe medications, they must take substantial action to increase attention to important warnings and decrease the number of clinically insignificant, low‐value warnings encountered by active caregivers on a daily basis.

Only 2 prior studies, both by Seidling et al., have specifically looked at provider response to warnings for high risk medications. Interaction warnings were rarely accepted in 1,[18] as in our study; however, in contrast to our findings, warning acceptance in both studies was higher for drugs with dose‐dependent toxicity.[18, 26] The effect of physician experience on warning acceptance has been addressed in 2 prior studies. In Weingart et al., residents were more likely than staff physicians to erase medication orders when presented with allergy and interaction warnings in a primary care setting.[20] Long et al. found that physicians younger than 40 years were less likely than older physicians to accept duplicate warnings, but those who had been at the study hospital for a longer period of time were more likely to accept them.[23] The influence of patient LOS and service on warning acceptance has not previously been described. Further study is needed looking at each of these factors.

Individual hospitals tend to avoid making modifications to order entry warning systems, because monitoring and maintaining these changes is labor intensive. Some institutions may make the decision to turn off certain categories of alerts, such as intermediate interaction warnings, to minimize the noise their providers encounter. There are even tools for disabling individual alerts or groups of alerts, such as that available for purchase from our interaction database vendor.[31] However, institutions may fear litigation should an adverse event be attributed to a disabled warning.[15, 16] Clearly, a comprehensive, health system‐wide approach is warranted.[13, 15] To date, published efforts describing ways to improve the effectiveness of medication warning systems have focused on either heightening the clinical significance of alerts[14, 21, 22, 32, 33, 34, 35, 36] or altering their presentation and how providers experience them.[21, 36, 37, 38, 39, 40, 41, 42, 43] The single medication warnings our providers receive are all presented in an identical font, and presumably response to each would be different if they were better distinguished from each other. We also found that a small but significant number of warnings were repeated for a given patient and even a given provider. If the providers knew they would only be presented with warnings the first time they occurred for a given patient and medication, they might be more attuned to the remaining warnings. Previous studies describe context‐specific decision support for medication ordering[44, 45, 46]; however, only 1 has described the use of patient context factors to modify when or how warnings are presented to providers.[47] None have described tailoring allergy, duplicate, and interaction warnings according to medication or provider types. If further study confirms our findings, modulating basic warning systems according to severity of illness, provider experience, and medication risk could powerfully increase their effectiveness. Of course, this would be extremely challenging to achieve, and is likely outside the capabilities of most, if not all, CPOE systems, at least for now.

Our study has some limitations. First, it was limited to medications that generated a single warning. We did this for ease of analysis and so that we could ensure understanding of provider response to each warning type without bias from simultaneously occurring warnings; however, caregiver response to multiple warnings appearing simultaneously for a particular medication order might be quite different. Second, we did not include any assessment of the number of medications ordered by each provider type or for each patient, either of which could significantly affect provider response to warnings. Third, as previously noted, we did not include any assessment of the validity of the warnings, beyond the 4 main categories described, which could also significantly affect provider response. However, it should be noted that although the validity of interaction warnings varies significantly from 1 medication to another, the validity of duplicate, allergy, and adverse reaction warnings in the described system are essentially the same for all medications. Fourth, it is possible that providers did modify or even erase their orders even after selecting override in response to the warning; it is also possible that providers reentered the same order after choosing erase. Unfortunately auditing for actions such as these would be extremely laborious. Finally, the study was conducted at a single medical center using a single order‐entry system. The system in use at our medical center is in use at one‐third of the 6000 hospitals in the United States, though certainly not all are using our version. Even if a hospital was using the same CPOE version and interaction database as our institution, variations in patient population and local decisions modifying how the database interacts with the warning presentation system might affect reproducibility at that institution.

Commonly encountered medication warnings are overridden at extremely high rates, and in our study this was particularly so for medications on the ISMP list, when ordered by physicians in training. Warnings of little clinical significance must be identified and eliminated, the most important warnings need to be visually distinct to increase user attention, and further research should be done into the patient, provider, setting, and medication factors that affect user responses to warnings, so that they may be customized accordingly and their significance increased. Doing so will enable us to reap the maximum possible potential from our CPOE systems, and increase the CPOE's power to protect our most vulnerable patients from our most dangerous medications, particularly when cared for by our most inexperienced physicians.

Acknowledgements

The authors thank, in particular, Scott Carey, Research Informatics Manager, for assistance with data collection. Additional thanks go to Olga Sherman and Kathleen Ancinich for assistance with data collection and management.

Disclosures: This research was supported in part by the Johns Hopkins Institute for Clinical and Translational Research. All listed authors contributed substantially to the study conception and design, analysis and interpretation of data, drafting the article or revising it critically for important intellectual content, and final approval of the version to be published. No one who fulfills these criteria has been excluded from authorship. This research received no specific grant from any funding agency in the public, commercial, or not‐for‐profit sectors. The authors have no competing interests to declare.

Files
References
  1. Bates DW, Leape L, Cullen DJ, et al., Effect of computerized physician order entry and a team intervention on prevention of serious medication errors. JAMA. 1998;280:13111316.
  2. Teich JM, Merchia PR, Schmiz JL, Kuperman GJ, Spurr CD, Bates DW. Effects of computerized provider order entry on prescribing practices. Arch Intern Med. 2000;160:27412747.
  3. Garg AX, Adhikari NKJ, McDonald H, et al. Effects of computerized clinician decision support systems on practitioner performance and patient outcomes: a systematic review. JAMA. 2005;293:12231238.
  4. Wolfstadt JI, Gurwitz JH, Field TS, et al. The effect of computerized physician order entry with clinical decision support on the rates of adverse drug events: a systematic review. J Gen Intern Med. 2008;23:451458.
  5. Eslami S, Keizer NF, Abu‐Hanna A. The impact of computerized physician medication order entry in hospitalized patients—a systematic review. Int J Med Inform. 2008;77:365376.
  6. Schedlbauer A, Prasad V, Mulvaney C, et al. What evidence supports the use of computerized alerts and prompts to improve clinicians' prescribing behavior? J Am Med Inform Assoc. 2009;16:531538.
  7. Reckmann MH, Westbrook JI, Koh Y, Lo C, Day RO. Does computerized provider order entry reduce prescribing errors for hospital inpatients? A systematic review. J Am Med Inform Assoc. 2009;16:613623.
  8. Sijs H, Aarts J, Vulto A, Berg M. Overriding of drug safety alerts in computerized physician order entry. J Am Med Inform Assoc. 2006;13:138147.
  9. Lin CP, Payne TH, Nichol WP, Hoey PJ, Anderson CL, Gennari JH. Evaluating clinical decision support systems: monitoring CPOE order check override rates in the Department of Veterans Affairs' Computerized Patient Record System. J Am Med Inform Assoc. 2008;15:620626.
  10. Magnus D, Rodger S, Avery AJ. GPs' views on computerized drug interaction alerts: questionnaire survey. J Clin Pharm Ther. 2002;27:377382.
  11. Weingart SN, Simchowitz B, Shiman L, et al. Clinicians' assessments of electronic medication safety alerts in ambulatory care. Arch Intern Med. 2009;169:16271632.
  12. Lapane KL, Waring ME, Schneider KL, Dube C, Quilliam BJ. A mixed method study of the merits of e‐prescribing drug alerts in primary care. J Gen Intern Med. 2008;23:442446.
  13. Bates DW. CPOE and clinical decision support in hospitals: getting the benefits: comment on “Unintended effects of a computerized physician order entry nearly hard‐stop alert to prevent a drug interaction.” Arch Intern Med. 2010;170:15831584.
  14. Classen DC, Phansalkar S, Bates DW. Critical drug‐drug interactions for use in electronic health records systems with computerized physician order entry: review of leading approaches. J Patient Saf. 2011;7:6165.
  15. Kesselheim AS, Cresswell K, Phansalkar S, Bates DW, Sheikh A. Clinical decision support systems could be modified to reduce 'alert fatigue' while still minimizing the risk of litigation. Health Aff (Millwood). 2011;30:23102317.
  16. Hines LE, Murphy JE, Grizzle AJ, Malone DC. Critical issues associated with drug‐drug interactions: highlights of a multistakeholder conference. Am J Health Syst Pharm. 2011;68:941946.
  17. Riedmann D, Jung M, Hackl WO, Stuhlinger W, der Sijs H, Ammenwerth E. Development of a context model to prioritize drug safety alerts in CPOE systems. BMC Med Inform Decis Mak. 2011;11:35.
  18. Seidling HM, Phansalkar S, Seger DL, et al. Factors influencing alert acceptance: a novel approach for predicting the success of clinical decision support. J Am Med Inform Assoc. 2011;18:479484.
  19. Riedmann D, Jung M, Hackl WO, Ammenwerth E. How to improve the delivery of medication alerts within computerized physician order entry systems: an international Delphi study. J Am Med Inform Assoc. 2011;18:760766.
  20. Weingart SN, Toth M, Sands DZ, Aronson MD, Davis RB, Phillips RS. Physicians' decisions to override computerized drug alerts in primary care. Arch Intern Med. 2003;163:26252631.
  21. Shah NR, Seger AC, Seger DL, et al. Improving acceptance of computerized prescribing alerts in ambulatory care. J Am Med Inform Assoc. 2006;13:511.
  22. Stutman HR, Fineman R, Meyer K, Jones D. Optimizing the acceptance of medication‐based alerts by physicians during CPOE implementation in a community hospital environment. AMIA Annu Symp Proc. 2007:701705.
  23. Long AJ, Chang P, Li YC, Chiu WT. The use of a CPOE log for the analysis of physicians' behavior when responding to drug‐duplication reminders. Int J Med Inform. 2008;77:499506.
  24. Isaac T, Weissman JS, Davis RB, et al. Overrides of medication alerts in ambulatory care. Arch Intern Med. 2009;169:305311.
  25. der Sijs H, Mulder A, Gelder T, Aarts J, Berg M, Vulto A. Drug safety alert generation and overriding in a large Dutch university medical centre. Pharmacoepidemiol Drug Saf. 2009;18:941947.
  26. Seidling HM, Schmitt SP, Bruckner T, et al. Patient‐specific electronic decision support reduces prescription of excessive doses. Qual Saf Health Care. 2010;19:e15.
  27. Peberdy MA, Ornato JP, Larkin GL, et al. Survival from in‐hospital cardiac arrest during nights and weekends. JAMA. 2008;299:785792.
  28. Steinman MA, Hanlon JT. Managing medications in clinically complex elders: “There's got to be a happy medium.” JAMA. 2010;304:15921601.
  29. Agency for Healthcare Research and Quality. Safety culture. Available at: http://psnet.ahrq.gov/primer.aspx?primerID=5. Accessed October 29, 2013.
  30. Institute for Safe Medication Practice. List of High‐Alert Medications. Available at: http://www.ismp.org/Tools/highalertmedications.pdf. Accessed June 18, 2013.
  31. First Databank. FDB AlertSpace. Available at: http://www.fdbhealth.com/solutions/fdb‐alertspace. Accessed July 3, 2014.
  32. Abookire SA, Teich JM, Sandige H, et al. Improving allergy alerting in a computerized physician order entry system. Proc AMIA Symp. 2000:26.
  33. Boussadi A, Caruba T, Zapletal E, Sabatier B, Durieux P, Degoulet P. A clinical data warehouse‐based process for refining medication orders alerts. J Am Med Inform Assoc. 2012;19:782785.
  34. Phansalkar S, der Sijs H, Tucker AD, et al. Drug‐drug interactions that should be non‐interruptive in order to reduce alert fatigue in electronic health records. J Am Med Inform Assoc. 2013;20:489493.
  35. Phansalkar S, Desai AA, Bell D, et al. High‐priority drug‐drug interactions for use in electronic health records. J Am Med Inform Assoc. 2012;19:735743.
  36. Horsky J, Phansalkar S, Desai A, Bell D, Middleton B. Design of decision support interventions for medication prescribing. Int J Med Inform. 2013;82:492503.
  37. Tamblyn R, Huang A, Taylor L, et al. A randomized trial of the effectiveness of on‐demand versus computer‐triggered drug decision support in primary care. J Am Med Inform Assoc. 2008;15:430438.
  38. Paterno MD, Maviglia SM, Gorman PN, et al. Tiering drug‐drug interaction alerts by severity increases compliance rates. J Am Med Inform Assoc. 2009;16:4046.
  39. Phansalkar S, Edworthy J, Hellier E, et al. A review of human factors principles for the design and implementation of medication safety alerts in clinical information systems. J Am Med Inform Assoc. 2010;17:493501.
  40. Strom BL, Schinnar R, Aberra F, et al. Unintended effects of a computerized physician order entry nearly hard‐stop alert to prevent a drug interaction: a randomized controlled trial. Arch Intern Med. 2010;170:15781583.
  41. Strom BL, Schinnar R, Bilker W, Hennessy S, Leonard CE, Pifer E. Randomized clinical trial of a customized electronic alert requiring an affirmative response compared to a control group receiving a commercial passive CPOE alert: NSAID—warfarin co‐prescribing as a test case. J Am Med Inform Assoc. 2010;17:411415.
  42. Scott GP, Shah P, Wyatt JC, Makubate B, Cross FW. Making electronic prescribing alerts more effective: scenario‐based experimental study in junior doctors. J Am Med Inform Assoc. 2011;18:789798.
  43. Zachariah M, Phansalkar S, Seidling HM, et al. Development and preliminary evidence for the validity of an instrument assessing implementation of human‐factors principles in medication‐related decision‐support systems—I‐MeDeSA. J Am Med Inform Assoc. 2011;18(suppl 1):i62i72.
  44. Kuperman GJ, Bobb A, Payne TH, et al. Medication‐related clinical decision support in computerized provider order entry systems: a review. J Am Med Inform Assoc. 2007;14:2940.
  45. Jung M, Riedmann D, Hackl WO, et al. Physicians' perceptions on the usefulness of contextual information for prioritizing and presenting alerts in Computerized Physician Order Entry systems. BMC Med Inform Decis Mak. 2012;12:111.
  46. Hemens BJ, Holbrook A, Tonkin M, et al. Computerized clinical decision support systems for drug prescribing and management: a decision‐maker‐researcher partnership systematic review. Implement Sci. 2011;6:89.
  47. Duke JD, Bolchini D. A successful model and visual design for creating context‐aware drug‐drug interaction alerts. AMIA Annu Symp Proc. 2011;2011:339348.
Article PDF
Issue
Journal of Hospital Medicine - 10(1)
Publications
Page Number
19-25
Sections
Files
Files
Article PDF
Article PDF

Many computerized provider order entry (CPOE) systems suffer from having too much of a good thing. Few would question the beneficial effect of CPOE on medication order clarity, completeness, and transmission.[1, 2] When mechanisms for basic decision support have been added, however, such as allergy, interaction, and duplicate warnings, reductions in medication errors and adverse events have not been consistently achieved.[3, 4, 5, 6, 7] This is likely due in part to the fact that ordering providers override medication warnings at staggeringly high rates.[8, 9] Clinicians acknowledge that they are ignoring potentially valuable warnings,[10, 11] but suffer from alert fatigue due to the sheer number of messages, many of them judged by clinicians to be of low‐value.[11, 12]

Redesign of medication alert systems to increase their signal‐to‐noise ratio is badly needed,[13, 14, 15, 16] and will need to consider the clinical significance of alerts, their presentation, and context‐specific factors that potentially contribute to warning effectiveness.[17, 18, 19] Relatively few studies, however, have objectively looked at context factors such as the characteristics of providers, patients, medications, and warnings that are associated with provider responses to warnings,[9, 20, 21, 22, 23, 24, 25] and only 2 have studied how warning acceptance is associated with medication risk.[18, 26] We wished to explore these factors further. Warning acceptance has been shown to be higher, at least in the outpatient setting, when orders are entered by low‐volume prescribers for infrequently encountered warnings,[24] and there is some evidence that patients receive higher‐quality care during the day.[27] Significant attention has been placed in recent years on inappropriate prescribing in older patients,[28] and on creating a culture of safety in healthcare.[29] We therefore hypothesized that our providers would be more cautious, and medication warning acceptance rates would be higher, when orders were entered for patients who were older or with more complex medical problems, when they were entered during the day by caregivers who entered few orders, when the medications ordered were potentially associated with greater risk, and when the warnings themselves were infrequently encountered.

METHODS

Setting and Caregivers

Johns Hopkins Bayview Medical Center (JHBMC) is a 400‐bed academic medical center serving southeastern Baltimore, Maryland. Prescribing caregivers include residents and fellows who rotate to both JHBMC and Johns Hopkins Hospital, internal medicine hospitalists, other attending physicians (including teaching attendings for all departments, and hospitalists and clinical associates for departments other than internal medicine), and nurse practitioners and physician assistants from most JHBMC departments. Nearly 100% of patients on the surgery, obstetrics/gynecology, neurology, psychiatry, and chemical dependence services are hospitalized on units dedicated to their respective specialty, and the same is true for approximately 95% of medicine patients.

Order Entry

JHBMC began using a client‐server order entry system by MEDITECH (Westwood, MA) in July 2003. Provider order entry was phased in beginning in October 2003 and completed by the end of 2004. MEDITECH version 5.64 was being used during the study period. Medications may generate duplicate, interaction, allergy, adverse reaction, and dose warnings during a patient ordering session each time they are ordered. Duplicate warnings are generated when the same medication (no matter what route) is ordered that is either on their active medication list, was on the list in the preceding 24 hours, or that is being ordered simultaneously. A drug‐interaction database licensed from First DataBank (South San Francisco, CA) is utilized, and updated monthly, which classifies potential drug‐drug interactions as contraindicated, severe, intermediate, and mild. Those classified as contraindicated by First DataBank are included in the severe category in MEDITECH 5.64. During the study period, JHBMC's version of MEDITECH was configured so that providers were warned of potential severe and intermediate drug‐drug interactions, but not mild. No other customizations had been made. Patients' histories of allergies and other adverse responses to medications can be entered by any credentialed staff member. They are maintained together in an allergies section of the electronic medical record, but are identified as either allergy or adverse reactions at the time they are entered, and each generates its own warnings.

When more than 1 duplicate, interaction, allergy, or adverse reaction warning is generated for a particular medication, all appear listed on a single screen in identical fonts. No visual distinction is made between severe and intermediate drug‐drug interactions; for these, the category of medication ordered is followed by the category of the medication for which there is a potential interaction. A details button can be selected to learn specifically which medications are involved and the severity and nature of the potential interactions identified. In response to the warnings, providers can choose to either override them, erase the order, or replace the order by clicking 1 of 3 buttons at the bottom of the screen. Warnings are not repeated unless the medication is reordered for that patient. Dose warnings appear on a subsequent screen and are not addressed in this article.

Nurses are discouraged from entering verbal orders but do have the capacity to do so, at which time they encounter and must respond to the standard medication warnings, if any. Medical students are able to enter orders, at which time they also encounter and must respond to the standard medication warnings; their orders must then be cosigned by a licensed provider before they can be processed. Warnings encountered by nurses and medical students are not repeated at the time of cosignature by a licensed provider.

Data Collection

We collected data regarding all medication orders placed in our CPOE system from October 1, 2009 to April 20, 2010 for all adult patients. Intensive care unit (ICU) patients were excluded, in anticipation of a separate analysis. Hospitalizations under observation were also excluded. We then ran a report showing all medications that generated any number of warnings of any type (duplicate, interaction, allergy, or adverse reaction) for the same population. Warnings generated during readmissions that occurred at any point during the study period (ranging from 1 to 21 times) were excluded, because these patients likely had many, if not all, of the same medications ordered during their readmissions as during their initial hospitalization, which would unduly influence the analysis if retained.

There was wide variation in the number of warnings generated per medication and in the number of each warning type per medication that generated multiple warnings. Therefore, for ease of analysis and to ensure that we could accurately determine varying response to each individual warning type, we thereafter focused on the medications that generated single warnings during the study period. For each single warning we obtained patient name, account number, event date and time, hospital unit at the time of the event, ordered medication, ordering staff member, warning type, and staff member response to the warning (eg, override warning or erase order [accept the warning]). The response replace was used very infrequently, and therefore warnings that resulted in this response were excluded. Medications available in more than 1 form included the route of administration in their name, and from this they were categorized as parenteral or nonparenteral. All nonparenteral or parenteral forms of a given medication were grouped together as 1 medication (eg, morphine sustained release and morphine elixir were classified as a single‐medication, nonparenteral morphine). Medications were further categorized according to whether or not they were on the Institute for Safe Medication Practice (ISMP) List of High‐Alert Medications.[30]

The study was approved by the Johns Hopkins Institutional Review Board.

Analysis

We collected descriptive data about patients and providers. Age and length of stay (LOS) at the time of the event were determined based on the patients' admit date and date of birth, and grouped into quartiles. Hospital units were grouped according to which service or services they primarily served. Medications were grouped into quartiles according to the total number of warnings they generated during the study period. Warnings were dichotomously categorized according to whether they were overridden or accepted. Unpaired t tests were used to compare continuous variables for the 2 groups, and [2] tests were used to compare categorical variables. A multivariate logistic regression was then performed, using variables with a P value of <0.10 in the univariate analysis, to control for confounders and identify independent predictors of medication warning acceptance. All analyses were performed using Intercooled Stata 12 (StataCorp, College Station, TX).

RESULTS

A total of 259,656 medication orders were placed for adult non‐ICU patients during the 7‐month study period. Of those orders, 45,835 generated some number of medication warnings.[1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20] The median number of warnings per patient was 4 (interquartile range [IQR]=28; mean=5.9, standard deviation [SD]=6.2), with a range from 1 to 84. The median number of warnings generated per provider during the study period was 36 (IQR=6106, mean=87.4, SD=133.7), with a range of 1 to 1096.

There were 40,391 orders placed for 454 medications for adult non‐ICU patients, which generated a single‐medication warning (excluding those with the response replace, which was used 20 times) during the 7‐month study period. Data regarding the patients and providers associated with the orders generating single warnings are shown in Table 1. Most patients were on medicine units, and most orders were entered by residents. Patients' LOS at the time the orders were placed ranged from 0 to 118 days (median=1, IQR=04; mean=4.0, SD=7.2). The median number of single warnings per patient was 4 (IQR=28; mean=6.1, SD=6.5), with a range from 1 to 84. The median number of single warnings generated per provider during the study period was 15 (IQR=373; mean=61.7, SD=109.6), with a range of 1 to 1057.

Patient and Provider Features
 No. (%)
  • NOTE: Abbreviations: GYN, gynecology; IM, internal medicine; Neuro/psych/chem dep, neurology/psychiatry/chemical dependence; NP, nurse practitioner; OB, obstetrics; PA, physician assistant.

  • Hospital unit at the time of order entry.

  • Total is >100% due to rounding.

Patients (N=6,646) 
Age 
1545 years2,048 (31%)
4657 years1,610 (24%)
5872 years1,520 (23%)
73104 years1,468 (22%)
Gender 
Male2,934 (44%)
Hospital unita 
Medicine2,992 (45%)
Surgery1,836 (28%)
Neuro/psych/chem dep1,337 (20%)
OB/GYN481 (7%)
Caregivers (N=655) 
Resident248 (38%)b
Nurse154 (24%)
Attending or other97 (15%)
NP/PA69 (11%)
IM hospitalist31 (5%)
Fellow27 (4%)
Medical student23 (4%)
Pharmacist6 (1%)

Patient and caregiver characteristics for the medication orders that generated single warnings are shown in Table 2. The majority of medications were nonparenteral and not on the ISMP list (Table 3). Most warnings generated were either duplicate (47%) or interaction warnings (47%). Warnings of a particular type were repeated 14.5% of the time for a particular medication and patient (from 2 to 24 times, median=2, IQR=22, mean=2.7, SD=1.4), and 9.8% of the time for a particular caregiver, medication, and patient (from 2 to 18 times, median=2, IQR=22, mean=2.4, SD=1.1).

Characteristics of Patients, Caregivers, Orders, Medications, and Warnings for Medication Orders Generating Single Warnings, and Association With Warning Acceptance
VariableNo. of Warnings (%)aNo. of Warnings Accepted (%)aP
  • NOTE: Abbreviations: GYN, gynecology; IM, internal medicine; ISMP, Institute for Safe Medication Practices; Neuro/psych/chem dep, neurology/psychiatry/chemical dependence; NP, nurse practitioner; OB, obstetrics; PA, physician assistant.

  • Totals may not equal 100% due to rounding.

  • Total number of medications is >454 because many medications generated more than 1 warning type.

Patient age   
1545 years10,881 (27)602 (5.5%)<0.001
4657 years9,733 (24)382 (3.9%) 
5872 years10,000 (25)308 (3.1%) 
73104 years9,777 (24)262 (2.7%) 
Patient gender   
Female23,395 (58)866 (3.7%)0.074
Male16,996 (42)688 (4.1%) 
Patient length of stay   
<1 day10,721 (27)660 (6.2%)<0.001
1 day10,854 (27)385 (3.5%) 
24 days10,424 (26)277 (2.7%) 
5118 days8,392 (21)232 (2.8%) 
Patient hospital unit   
Medicine20,057 (50)519 (2.6%)<0.001
Surgery10,274 (25)477 (4.6%) 
Neuro/psych/chem dep8,279 (21)417 (5.0%) 
OB/GYN1,781 (4)141 (7.9%) 
Ordering caregiver   
Resident22,523 (56)700 (3.1%)<0.001
NP/PA7,534 (19)369 (4.9%) 
IM hospitalist5,048 (13)155 (3.1%) 
Attending3225 (8)219 (6.8%) 
Fellow910 (2)34 (3.7%) 
Nurse865 (2)58 (6.7%) 
Medical student265 (<1)17 (6.4%) 
Pharmacist21 (<1)2 (9.5%) 
Day ordered   
Weekday31,499 (78%)1276 (4.1%)<0.001
Weekend8,892 (22%)278 (3.1%) 
Time ordered   
000005594,231 (11%)117 (2.8%)<0.001
0600115911,696 (29%)348 (3.0%) 
1200175915,879 (39%)722 (4.6%) 
180023598,585 (21%)367 (4.3%) 
Administration route (no. of meds)  
Nonparenteral (339)27,086 (67%)956 (3.5%)<0.001
Parenteral (115)13,305 (33%)598 (4.5%) 
ISMP List of High‐Alert Medications status (no. of meds)[30]  
Not on ISMP list (394)27,503 (68%)1251 (4.5%)<0.001
On ISMP list (60)12,888 (32%)303 (2.4%) 
No. of warnings per med (no. of meds)  
11062133 (7)9,869 (24%)191 (1.9%)<0.001
4681034 (13)10,014 (25%)331 (3.3%) 
170444 (40)10,182 (25%)314 (3.1%) 
1169 (394)10,326 (26%)718 (7.0%) 
Warning type (no. of meds)b  
Duplicate (369)19,083 (47%)1041 (5.5%)<0.001
Interaction (315)18,894 (47%)254 (1.3%) 
Allergy (138)2,371 (6%)243 (10.0%) 
Adverse reaction (14)43 (0.1%)16 (37%) 
Multivariate Analysis of Factors Associated With Acceptance of Medication Warnings
VariableAdjusted OR95% CI
  • NOTE: Abbreviations: CI, confidence interval; GYN, gynecology; IM, internal medicine; ISMP, Institute for Safe Medication Practices; Neuro/psych/chem dep, neurology/psychiatry/chemical dependence; NP, nurse practitioner; OB, obstetrics; OR, odds ratio; PA, physician assistant.

  • Day ordered and time of order entry were included but were not significant in the multivariate model.

Patient age  
1545 years1.00Reference
4657 years0.890.771.02
5872 years0.850.730.99
73104 years0.910.771.08
Patient gender  
Female1.00Reference
Male1.261.131.41
Patient length of stay 
<1 day1.00Reference
1 day0.650.550.76
24 days0.490.420.58
5118 days0.490.410.58
Patient hospital unit  
Medicine1.00Reference
Surgery1.451.251.68
Neuro/psych/chem dep1.351.151.58
OB/GYN2.431.923.08
Ordering caregiver  
Resident1.00Reference
NP/PA1.631.421.88
IM hospitalist1.241.021.50
Attending1.831.542.18
Fellow1.410.982.03
Nurse1.921.442.57
Medical student1.170.701.95
Pharmacist3.080.6714.03
Medication factors  
Nonparenteral1.00Reference
Parenteral1.791.592.03
HighAlert Medication status (no. of meds)[30]
Not on ISMP list1.00Reference
On ISMP list0.370.320.43
No. of warnings per medication 
110621331.00Reference
46810342.301.902.79
1704442.251.852.73
11694.103.424.92
Warning type  
Duplicate1.00Reference
Interaction0.240.210.28
Allergy2.281.942.68
Adverse reaction9.244.5218.90

One thousand five hundred fifty‐four warnings were erased (ie, accepted by clinicians [4%]). In univariate analysis, only patient gender was not associated with warning acceptance. Patient age, LOS, hospital unit at the time of order entry, ordering caregiver type, day and time the medication was ordered, administration route, presence on the ISMP list, warning frequency, and warning type were all significantly associated with warning acceptance (Table 2).

Older patient age, longer LOS, presence of the medication on the ISMP list, and interaction warning type were all negatively associated with warning acceptance in multivariable analysis. Warning acceptance was positively associated with male patient gender, being on a service other than medicine, being a caregiver other than a resident, parenteral medications, lower warning frequency, and allergy or adverse reaction warning types (Table 3).

The 20 medications that generated the most single warnings are shown in Table 4. Medications on the ISMP list accounted for 8 of these top 20 medications. For most of them, duplicate and interaction warnings accounted for most of the warnings generated, except for parenteral hydromorphone, oral oxycodone, parenteral morphine, and oral hydromorphone, which each had more allergy than interaction warnings.

Top 20 Medications Generating Single Warnings and Warning Type Distribution for Each
MedicationISMP ListbNo. of WarningsDuplicate, No. (%)cInteraction, No. (%)cAllergy, No. (%)cAdverse Reaction, No. (%)c
  • NOTE: Abbreviations: ISMP, Institute for Safe Medication Practices.

  • Medications not noted as injectable should be presumed not parenteral.

  • SMP List of High‐Alert Medications.[30]

  • Total may not add up to 100% due to rounding.

Hydromorphone injectableYes2,1331,584 (74.3)127 (6.0)422 (19.8) 
Metoprolol 1,432550 (38.4)870 (60.8)12 (0.8) 
Aspirin 1,375212 (15.4)1,096 (79.7)67 (4.9) 
OxycodoneYes1,360987 (72.6) 364 (26.8)9 (0.7)
Potassium chloride 1,296379 (29.2)917 (70.8)  
Ondansetron injectable 1,1671,013 (86.8)153 (13.1)1 (0.1) 
Aspart insulin injectableYes1,106643 (58.1)463 (41.9)  
WarfarinYes1,034298 (28.8)736 (71.2)  
Heparin injectableYes1,030205 (19.9)816 (79.2)9 (0.3) 
Furosemide injectable 980438 (45.0)542 (55.3)  
Lisinopril 926225 (24.3)698 (75.4)3 (0.3) 
Acetaminophen 860686 (79.8)118 (13.7)54 (6.3)2 (0.2)
Morphine injectableYes804467 (58.1)100 (12.4)233 (29.0)4 (0.5)
Diazepam 786731 (93.0)41 (5.2)14 (1.8) 
Glargine insulin injectableYes746268 (35.9)478 (64.1)  
Ibuprofen 713125 (17.5)529 (74.2)54 (7.6)5 (0.7)
HydromorphoneYes594372 (62.6)31 (5.2)187 (31.5)4 (0.7)
Furosemide 586273 (46.6)312 (53.2)1 (0.2) 
Ketorolac injectable 48739 (8.0)423 (86.9)23 (4.7)2 (0.4)
Prednisone 468166 (35.5)297 (63.5)5 (1.1) 

DISCUSSION

Medication warnings in our study were frequently overridden, particularly when encountered by residents, for patients with a long LOS and on the internal medicine service, and for medications generating the most warnings and on the ISMP list. Disturbingly, this means that potentially important warnings for medications with the highest potential for causing harm, for possibly the sickest and most complex patients, were those that were most often ignored by young physicians in training who should have had the most to gain from them. Of course, this is not entirely surprising. Despite our hope that a culture of safety would influence young physicians' actions when caring for these patients and prescribing these medications, these patients and medications are those for whom the most warnings are generated, and these physicians are the ones entering the most orders. Only 13% of the medications studied were on the ISMP list, but they generated 32% of the warnings. We controlled for number of warnings and ISMP list status, but not for warning validity. Most likely, high‐risk medications have been set up with more warnings, many of them of lower quality, in an errant but well‐intentioned effort to make them safer. If developers of CPOE systems want to gain serious traction in using decision support to promote prescribing safe medications, they must take substantial action to increase attention to important warnings and decrease the number of clinically insignificant, low‐value warnings encountered by active caregivers on a daily basis.

Only 2 prior studies, both by Seidling et al., have specifically looked at provider response to warnings for high risk medications. Interaction warnings were rarely accepted in 1,[18] as in our study; however, in contrast to our findings, warning acceptance in both studies was higher for drugs with dose‐dependent toxicity.[18, 26] The effect of physician experience on warning acceptance has been addressed in 2 prior studies. In Weingart et al., residents were more likely than staff physicians to erase medication orders when presented with allergy and interaction warnings in a primary care setting.[20] Long et al. found that physicians younger than 40 years were less likely than older physicians to accept duplicate warnings, but those who had been at the study hospital for a longer period of time were more likely to accept them.[23] The influence of patient LOS and service on warning acceptance has not previously been described. Further study is needed looking at each of these factors.

Individual hospitals tend to avoid making modifications to order entry warning systems, because monitoring and maintaining these changes is labor intensive. Some institutions may make the decision to turn off certain categories of alerts, such as intermediate interaction warnings, to minimize the noise their providers encounter. There are even tools for disabling individual alerts or groups of alerts, such as that available for purchase from our interaction database vendor.[31] However, institutions may fear litigation should an adverse event be attributed to a disabled warning.[15, 16] Clearly, a comprehensive, health system‐wide approach is warranted.[13, 15] To date, published efforts describing ways to improve the effectiveness of medication warning systems have focused on either heightening the clinical significance of alerts[14, 21, 22, 32, 33, 34, 35, 36] or altering their presentation and how providers experience them.[21, 36, 37, 38, 39, 40, 41, 42, 43] The single medication warnings our providers receive are all presented in an identical font, and presumably response to each would be different if they were better distinguished from each other. We also found that a small but significant number of warnings were repeated for a given patient and even a given provider. If the providers knew they would only be presented with warnings the first time they occurred for a given patient and medication, they might be more attuned to the remaining warnings. Previous studies describe context‐specific decision support for medication ordering[44, 45, 46]; however, only 1 has described the use of patient context factors to modify when or how warnings are presented to providers.[47] None have described tailoring allergy, duplicate, and interaction warnings according to medication or provider types. If further study confirms our findings, modulating basic warning systems according to severity of illness, provider experience, and medication risk could powerfully increase their effectiveness. Of course, this would be extremely challenging to achieve, and is likely outside the capabilities of most, if not all, CPOE systems, at least for now.

Our study has some limitations. First, it was limited to medications that generated a single warning. We did this for ease of analysis and so that we could ensure understanding of provider response to each warning type without bias from simultaneously occurring warnings; however, caregiver response to multiple warnings appearing simultaneously for a particular medication order might be quite different. Second, we did not include any assessment of the number of medications ordered by each provider type or for each patient, either of which could significantly affect provider response to warnings. Third, as previously noted, we did not include any assessment of the validity of the warnings, beyond the 4 main categories described, which could also significantly affect provider response. However, it should be noted that although the validity of interaction warnings varies significantly from 1 medication to another, the validity of duplicate, allergy, and adverse reaction warnings in the described system are essentially the same for all medications. Fourth, it is possible that providers did modify or even erase their orders even after selecting override in response to the warning; it is also possible that providers reentered the same order after choosing erase. Unfortunately auditing for actions such as these would be extremely laborious. Finally, the study was conducted at a single medical center using a single order‐entry system. The system in use at our medical center is in use at one‐third of the 6000 hospitals in the United States, though certainly not all are using our version. Even if a hospital was using the same CPOE version and interaction database as our institution, variations in patient population and local decisions modifying how the database interacts with the warning presentation system might affect reproducibility at that institution.

Commonly encountered medication warnings are overridden at extremely high rates, and in our study this was particularly so for medications on the ISMP list, when ordered by physicians in training. Warnings of little clinical significance must be identified and eliminated, the most important warnings need to be visually distinct to increase user attention, and further research should be done into the patient, provider, setting, and medication factors that affect user responses to warnings, so that they may be customized accordingly and their significance increased. Doing so will enable us to reap the maximum possible potential from our CPOE systems, and increase the CPOE's power to protect our most vulnerable patients from our most dangerous medications, particularly when cared for by our most inexperienced physicians.

Acknowledgements

The authors thank, in particular, Scott Carey, Research Informatics Manager, for assistance with data collection. Additional thanks go to Olga Sherman and Kathleen Ancinich for assistance with data collection and management.

Disclosures: This research was supported in part by the Johns Hopkins Institute for Clinical and Translational Research. All listed authors contributed substantially to the study conception and design, analysis and interpretation of data, drafting the article or revising it critically for important intellectual content, and final approval of the version to be published. No one who fulfills these criteria has been excluded from authorship. This research received no specific grant from any funding agency in the public, commercial, or not‐for‐profit sectors. The authors have no competing interests to declare.

Many computerized provider order entry (CPOE) systems suffer from having too much of a good thing. Few would question the beneficial effect of CPOE on medication order clarity, completeness, and transmission.[1, 2] When mechanisms for basic decision support have been added, however, such as allergy, interaction, and duplicate warnings, reductions in medication errors and adverse events have not been consistently achieved.[3, 4, 5, 6, 7] This is likely due in part to the fact that ordering providers override medication warnings at staggeringly high rates.[8, 9] Clinicians acknowledge that they are ignoring potentially valuable warnings,[10, 11] but suffer from alert fatigue due to the sheer number of messages, many of them judged by clinicians to be of low‐value.[11, 12]

Redesign of medication alert systems to increase their signal‐to‐noise ratio is badly needed,[13, 14, 15, 16] and will need to consider the clinical significance of alerts, their presentation, and context‐specific factors that potentially contribute to warning effectiveness.[17, 18, 19] Relatively few studies, however, have objectively looked at context factors such as the characteristics of providers, patients, medications, and warnings that are associated with provider responses to warnings,[9, 20, 21, 22, 23, 24, 25] and only 2 have studied how warning acceptance is associated with medication risk.[18, 26] We wished to explore these factors further. Warning acceptance has been shown to be higher, at least in the outpatient setting, when orders are entered by low‐volume prescribers for infrequently encountered warnings,[24] and there is some evidence that patients receive higher‐quality care during the day.[27] Significant attention has been placed in recent years on inappropriate prescribing in older patients,[28] and on creating a culture of safety in healthcare.[29] We therefore hypothesized that our providers would be more cautious, and medication warning acceptance rates would be higher, when orders were entered for patients who were older or with more complex medical problems, when they were entered during the day by caregivers who entered few orders, when the medications ordered were potentially associated with greater risk, and when the warnings themselves were infrequently encountered.

METHODS

Setting and Caregivers

Johns Hopkins Bayview Medical Center (JHBMC) is a 400‐bed academic medical center serving southeastern Baltimore, Maryland. Prescribing caregivers include residents and fellows who rotate to both JHBMC and Johns Hopkins Hospital, internal medicine hospitalists, other attending physicians (including teaching attendings for all departments, and hospitalists and clinical associates for departments other than internal medicine), and nurse practitioners and physician assistants from most JHBMC departments. Nearly 100% of patients on the surgery, obstetrics/gynecology, neurology, psychiatry, and chemical dependence services are hospitalized on units dedicated to their respective specialty, and the same is true for approximately 95% of medicine patients.

Order Entry

JHBMC began using a client‐server order entry system by MEDITECH (Westwood, MA) in July 2003. Provider order entry was phased in beginning in October 2003 and completed by the end of 2004. MEDITECH version 5.64 was being used during the study period. Medications may generate duplicate, interaction, allergy, adverse reaction, and dose warnings during a patient ordering session each time they are ordered. Duplicate warnings are generated when the same medication (no matter what route) is ordered that is either on their active medication list, was on the list in the preceding 24 hours, or that is being ordered simultaneously. A drug‐interaction database licensed from First DataBank (South San Francisco, CA) is utilized, and updated monthly, which classifies potential drug‐drug interactions as contraindicated, severe, intermediate, and mild. Those classified as contraindicated by First DataBank are included in the severe category in MEDITECH 5.64. During the study period, JHBMC's version of MEDITECH was configured so that providers were warned of potential severe and intermediate drug‐drug interactions, but not mild. No other customizations had been made. Patients' histories of allergies and other adverse responses to medications can be entered by any credentialed staff member. They are maintained together in an allergies section of the electronic medical record, but are identified as either allergy or adverse reactions at the time they are entered, and each generates its own warnings.

When more than 1 duplicate, interaction, allergy, or adverse reaction warning is generated for a particular medication, all appear listed on a single screen in identical fonts. No visual distinction is made between severe and intermediate drug‐drug interactions; for these, the category of medication ordered is followed by the category of the medication for which there is a potential interaction. A details button can be selected to learn specifically which medications are involved and the severity and nature of the potential interactions identified. In response to the warnings, providers can choose to either override them, erase the order, or replace the order by clicking 1 of 3 buttons at the bottom of the screen. Warnings are not repeated unless the medication is reordered for that patient. Dose warnings appear on a subsequent screen and are not addressed in this article.

Nurses are discouraged from entering verbal orders but do have the capacity to do so, at which time they encounter and must respond to the standard medication warnings, if any. Medical students are able to enter orders, at which time they also encounter and must respond to the standard medication warnings; their orders must then be cosigned by a licensed provider before they can be processed. Warnings encountered by nurses and medical students are not repeated at the time of cosignature by a licensed provider.

Data Collection

We collected data regarding all medication orders placed in our CPOE system from October 1, 2009 to April 20, 2010 for all adult patients. Intensive care unit (ICU) patients were excluded, in anticipation of a separate analysis. Hospitalizations under observation were also excluded. We then ran a report showing all medications that generated any number of warnings of any type (duplicate, interaction, allergy, or adverse reaction) for the same population. Warnings generated during readmissions that occurred at any point during the study period (ranging from 1 to 21 times) were excluded, because these patients likely had many, if not all, of the same medications ordered during their readmissions as during their initial hospitalization, which would unduly influence the analysis if retained.

There was wide variation in the number of warnings generated per medication and in the number of each warning type per medication that generated multiple warnings. Therefore, for ease of analysis and to ensure that we could accurately determine varying response to each individual warning type, we thereafter focused on the medications that generated single warnings during the study period. For each single warning we obtained patient name, account number, event date and time, hospital unit at the time of the event, ordered medication, ordering staff member, warning type, and staff member response to the warning (eg, override warning or erase order [accept the warning]). The response replace was used very infrequently, and therefore warnings that resulted in this response were excluded. Medications available in more than 1 form included the route of administration in their name, and from this they were categorized as parenteral or nonparenteral. All nonparenteral or parenteral forms of a given medication were grouped together as 1 medication (eg, morphine sustained release and morphine elixir were classified as a single‐medication, nonparenteral morphine). Medications were further categorized according to whether or not they were on the Institute for Safe Medication Practice (ISMP) List of High‐Alert Medications.[30]

The study was approved by the Johns Hopkins Institutional Review Board.

Analysis

We collected descriptive data about patients and providers. Age and length of stay (LOS) at the time of the event were determined based on the patients' admit date and date of birth, and grouped into quartiles. Hospital units were grouped according to which service or services they primarily served. Medications were grouped into quartiles according to the total number of warnings they generated during the study period. Warnings were dichotomously categorized according to whether they were overridden or accepted. Unpaired t tests were used to compare continuous variables for the 2 groups, and [2] tests were used to compare categorical variables. A multivariate logistic regression was then performed, using variables with a P value of <0.10 in the univariate analysis, to control for confounders and identify independent predictors of medication warning acceptance. All analyses were performed using Intercooled Stata 12 (StataCorp, College Station, TX).

RESULTS

A total of 259,656 medication orders were placed for adult non‐ICU patients during the 7‐month study period. Of those orders, 45,835 generated some number of medication warnings.[1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20] The median number of warnings per patient was 4 (interquartile range [IQR]=28; mean=5.9, standard deviation [SD]=6.2), with a range from 1 to 84. The median number of warnings generated per provider during the study period was 36 (IQR=6106, mean=87.4, SD=133.7), with a range of 1 to 1096.

There were 40,391 orders placed for 454 medications for adult non‐ICU patients, which generated a single‐medication warning (excluding those with the response replace, which was used 20 times) during the 7‐month study period. Data regarding the patients and providers associated with the orders generating single warnings are shown in Table 1. Most patients were on medicine units, and most orders were entered by residents. Patients' LOS at the time the orders were placed ranged from 0 to 118 days (median=1, IQR=04; mean=4.0, SD=7.2). The median number of single warnings per patient was 4 (IQR=28; mean=6.1, SD=6.5), with a range from 1 to 84. The median number of single warnings generated per provider during the study period was 15 (IQR=373; mean=61.7, SD=109.6), with a range of 1 to 1057.

Patient and Provider Features
 No. (%)
  • NOTE: Abbreviations: GYN, gynecology; IM, internal medicine; Neuro/psych/chem dep, neurology/psychiatry/chemical dependence; NP, nurse practitioner; OB, obstetrics; PA, physician assistant.

  • Hospital unit at the time of order entry.

  • Total is >100% due to rounding.

Patients (N=6,646) 
Age 
1545 years2,048 (31%)
4657 years1,610 (24%)
5872 years1,520 (23%)
73104 years1,468 (22%)
Gender 
Male2,934 (44%)
Hospital unita 
Medicine2,992 (45%)
Surgery1,836 (28%)
Neuro/psych/chem dep1,337 (20%)
OB/GYN481 (7%)
Caregivers (N=655) 
Resident248 (38%)b
Nurse154 (24%)
Attending or other97 (15%)
NP/PA69 (11%)
IM hospitalist31 (5%)
Fellow27 (4%)
Medical student23 (4%)
Pharmacist6 (1%)

Patient and caregiver characteristics for the medication orders that generated single warnings are shown in Table 2. The majority of medications were nonparenteral and not on the ISMP list (Table 3). Most warnings generated were either duplicate (47%) or interaction warnings (47%). Warnings of a particular type were repeated 14.5% of the time for a particular medication and patient (from 2 to 24 times, median=2, IQR=22, mean=2.7, SD=1.4), and 9.8% of the time for a particular caregiver, medication, and patient (from 2 to 18 times, median=2, IQR=22, mean=2.4, SD=1.1).

Characteristics of Patients, Caregivers, Orders, Medications, and Warnings for Medication Orders Generating Single Warnings, and Association With Warning Acceptance
VariableNo. of Warnings (%)aNo. of Warnings Accepted (%)aP
  • NOTE: Abbreviations: GYN, gynecology; IM, internal medicine; ISMP, Institute for Safe Medication Practices; Neuro/psych/chem dep, neurology/psychiatry/chemical dependence; NP, nurse practitioner; OB, obstetrics; PA, physician assistant.

  • Totals may not equal 100% due to rounding.

  • Total number of medications is >454 because many medications generated more than 1 warning type.

Patient age   
1545 years10,881 (27)602 (5.5%)<0.001
4657 years9,733 (24)382 (3.9%) 
5872 years10,000 (25)308 (3.1%) 
73104 years9,777 (24)262 (2.7%) 
Patient gender   
Female23,395 (58)866 (3.7%)0.074
Male16,996 (42)688 (4.1%) 
Patient length of stay   
<1 day10,721 (27)660 (6.2%)<0.001
1 day10,854 (27)385 (3.5%) 
24 days10,424 (26)277 (2.7%) 
5118 days8,392 (21)232 (2.8%) 
Patient hospital unit   
Medicine20,057 (50)519 (2.6%)<0.001
Surgery10,274 (25)477 (4.6%) 
Neuro/psych/chem dep8,279 (21)417 (5.0%) 
OB/GYN1,781 (4)141 (7.9%) 
Ordering caregiver   
Resident22,523 (56)700 (3.1%)<0.001
NP/PA7,534 (19)369 (4.9%) 
IM hospitalist5,048 (13)155 (3.1%) 
Attending3225 (8)219 (6.8%) 
Fellow910 (2)34 (3.7%) 
Nurse865 (2)58 (6.7%) 
Medical student265 (<1)17 (6.4%) 
Pharmacist21 (<1)2 (9.5%) 
Day ordered   
Weekday31,499 (78%)1276 (4.1%)<0.001
Weekend8,892 (22%)278 (3.1%) 
Time ordered   
000005594,231 (11%)117 (2.8%)<0.001
0600115911,696 (29%)348 (3.0%) 
1200175915,879 (39%)722 (4.6%) 
180023598,585 (21%)367 (4.3%) 
Administration route (no. of meds)  
Nonparenteral (339)27,086 (67%)956 (3.5%)<0.001
Parenteral (115)13,305 (33%)598 (4.5%) 
ISMP List of High‐Alert Medications status (no. of meds)[30]  
Not on ISMP list (394)27,503 (68%)1251 (4.5%)<0.001
On ISMP list (60)12,888 (32%)303 (2.4%) 
No. of warnings per med (no. of meds)  
11062133 (7)9,869 (24%)191 (1.9%)<0.001
4681034 (13)10,014 (25%)331 (3.3%) 
170444 (40)10,182 (25%)314 (3.1%) 
1169 (394)10,326 (26%)718 (7.0%) 
Warning type (no. of meds)b  
Duplicate (369)19,083 (47%)1041 (5.5%)<0.001
Interaction (315)18,894 (47%)254 (1.3%) 
Allergy (138)2,371 (6%)243 (10.0%) 
Adverse reaction (14)43 (0.1%)16 (37%) 
Multivariate Analysis of Factors Associated With Acceptance of Medication Warnings
VariableAdjusted OR95% CI
  • NOTE: Abbreviations: CI, confidence interval; GYN, gynecology; IM, internal medicine; ISMP, Institute for Safe Medication Practices; Neuro/psych/chem dep, neurology/psychiatry/chemical dependence; NP, nurse practitioner; OB, obstetrics; OR, odds ratio; PA, physician assistant.

  • Day ordered and time of order entry were included but were not significant in the multivariate model.

Patient age  
1545 years1.00Reference
4657 years0.890.771.02
5872 years0.850.730.99
73104 years0.910.771.08
Patient gender  
Female1.00Reference
Male1.261.131.41
Patient length of stay 
<1 day1.00Reference
1 day0.650.550.76
24 days0.490.420.58
5118 days0.490.410.58
Patient hospital unit  
Medicine1.00Reference
Surgery1.451.251.68
Neuro/psych/chem dep1.351.151.58
OB/GYN2.431.923.08
Ordering caregiver  
Resident1.00Reference
NP/PA1.631.421.88
IM hospitalist1.241.021.50
Attending1.831.542.18
Fellow1.410.982.03
Nurse1.921.442.57
Medical student1.170.701.95
Pharmacist3.080.6714.03
Medication factors  
Nonparenteral1.00Reference
Parenteral1.791.592.03
HighAlert Medication status (no. of meds)[30]
Not on ISMP list1.00Reference
On ISMP list0.370.320.43
No. of warnings per medication 
110621331.00Reference
46810342.301.902.79
1704442.251.852.73
11694.103.424.92
Warning type  
Duplicate1.00Reference
Interaction0.240.210.28
Allergy2.281.942.68
Adverse reaction9.244.5218.90

One thousand five hundred fifty‐four warnings were erased (ie, accepted by clinicians [4%]). In univariate analysis, only patient gender was not associated with warning acceptance. Patient age, LOS, hospital unit at the time of order entry, ordering caregiver type, day and time the medication was ordered, administration route, presence on the ISMP list, warning frequency, and warning type were all significantly associated with warning acceptance (Table 2).

Older patient age, longer LOS, presence of the medication on the ISMP list, and interaction warning type were all negatively associated with warning acceptance in multivariable analysis. Warning acceptance was positively associated with male patient gender, being on a service other than medicine, being a caregiver other than a resident, parenteral medications, lower warning frequency, and allergy or adverse reaction warning types (Table 3).

The 20 medications that generated the most single warnings are shown in Table 4. Medications on the ISMP list accounted for 8 of these top 20 medications. For most of them, duplicate and interaction warnings accounted for most of the warnings generated, except for parenteral hydromorphone, oral oxycodone, parenteral morphine, and oral hydromorphone, which each had more allergy than interaction warnings.

Top 20 Medications Generating Single Warnings and Warning Type Distribution for Each
MedicationISMP ListbNo. of WarningsDuplicate, No. (%)cInteraction, No. (%)cAllergy, No. (%)cAdverse Reaction, No. (%)c
  • NOTE: Abbreviations: ISMP, Institute for Safe Medication Practices.

  • Medications not noted as injectable should be presumed not parenteral.

  • SMP List of High‐Alert Medications.[30]

  • Total may not add up to 100% due to rounding.

Hydromorphone injectableYes2,1331,584 (74.3)127 (6.0)422 (19.8) 
Metoprolol 1,432550 (38.4)870 (60.8)12 (0.8) 
Aspirin 1,375212 (15.4)1,096 (79.7)67 (4.9) 
OxycodoneYes1,360987 (72.6) 364 (26.8)9 (0.7)
Potassium chloride 1,296379 (29.2)917 (70.8)  
Ondansetron injectable 1,1671,013 (86.8)153 (13.1)1 (0.1) 
Aspart insulin injectableYes1,106643 (58.1)463 (41.9)  
WarfarinYes1,034298 (28.8)736 (71.2)  
Heparin injectableYes1,030205 (19.9)816 (79.2)9 (0.3) 
Furosemide injectable 980438 (45.0)542 (55.3)  
Lisinopril 926225 (24.3)698 (75.4)3 (0.3) 
Acetaminophen 860686 (79.8)118 (13.7)54 (6.3)2 (0.2)
Morphine injectableYes804467 (58.1)100 (12.4)233 (29.0)4 (0.5)
Diazepam 786731 (93.0)41 (5.2)14 (1.8) 
Glargine insulin injectableYes746268 (35.9)478 (64.1)  
Ibuprofen 713125 (17.5)529 (74.2)54 (7.6)5 (0.7)
HydromorphoneYes594372 (62.6)31 (5.2)187 (31.5)4 (0.7)
Furosemide 586273 (46.6)312 (53.2)1 (0.2) 
Ketorolac injectable 48739 (8.0)423 (86.9)23 (4.7)2 (0.4)
Prednisone 468166 (35.5)297 (63.5)5 (1.1) 

DISCUSSION

Medication warnings in our study were frequently overridden, particularly when encountered by residents, for patients with a long LOS and on the internal medicine service, and for medications generating the most warnings and on the ISMP list. Disturbingly, this means that potentially important warnings for medications with the highest potential for causing harm, for possibly the sickest and most complex patients, were those that were most often ignored by young physicians in training who should have had the most to gain from them. Of course, this is not entirely surprising. Despite our hope that a culture of safety would influence young physicians' actions when caring for these patients and prescribing these medications, these patients and medications are those for whom the most warnings are generated, and these physicians are the ones entering the most orders. Only 13% of the medications studied were on the ISMP list, but they generated 32% of the warnings. We controlled for number of warnings and ISMP list status, but not for warning validity. Most likely, high‐risk medications have been set up with more warnings, many of them of lower quality, in an errant but well‐intentioned effort to make them safer. If developers of CPOE systems want to gain serious traction in using decision support to promote prescribing safe medications, they must take substantial action to increase attention to important warnings and decrease the number of clinically insignificant, low‐value warnings encountered by active caregivers on a daily basis.

Only 2 prior studies, both by Seidling et al., have specifically looked at provider response to warnings for high risk medications. Interaction warnings were rarely accepted in 1,[18] as in our study; however, in contrast to our findings, warning acceptance in both studies was higher for drugs with dose‐dependent toxicity.[18, 26] The effect of physician experience on warning acceptance has been addressed in 2 prior studies. In Weingart et al., residents were more likely than staff physicians to erase medication orders when presented with allergy and interaction warnings in a primary care setting.[20] Long et al. found that physicians younger than 40 years were less likely than older physicians to accept duplicate warnings, but those who had been at the study hospital for a longer period of time were more likely to accept them.[23] The influence of patient LOS and service on warning acceptance has not previously been described. Further study is needed looking at each of these factors.

Individual hospitals tend to avoid making modifications to order entry warning systems, because monitoring and maintaining these changes is labor intensive. Some institutions may make the decision to turn off certain categories of alerts, such as intermediate interaction warnings, to minimize the noise their providers encounter. There are even tools for disabling individual alerts or groups of alerts, such as that available for purchase from our interaction database vendor.[31] However, institutions may fear litigation should an adverse event be attributed to a disabled warning.[15, 16] Clearly, a comprehensive, health system‐wide approach is warranted.[13, 15] To date, published efforts describing ways to improve the effectiveness of medication warning systems have focused on either heightening the clinical significance of alerts[14, 21, 22, 32, 33, 34, 35, 36] or altering their presentation and how providers experience them.[21, 36, 37, 38, 39, 40, 41, 42, 43] The single medication warnings our providers receive are all presented in an identical font, and presumably response to each would be different if they were better distinguished from each other. We also found that a small but significant number of warnings were repeated for a given patient and even a given provider. If the providers knew they would only be presented with warnings the first time they occurred for a given patient and medication, they might be more attuned to the remaining warnings. Previous studies describe context‐specific decision support for medication ordering[44, 45, 46]; however, only 1 has described the use of patient context factors to modify when or how warnings are presented to providers.[47] None have described tailoring allergy, duplicate, and interaction warnings according to medication or provider types. If further study confirms our findings, modulating basic warning systems according to severity of illness, provider experience, and medication risk could powerfully increase their effectiveness. Of course, this would be extremely challenging to achieve, and is likely outside the capabilities of most, if not all, CPOE systems, at least for now.

Our study has some limitations. First, it was limited to medications that generated a single warning. We did this for ease of analysis and so that we could ensure understanding of provider response to each warning type without bias from simultaneously occurring warnings; however, caregiver response to multiple warnings appearing simultaneously for a particular medication order might be quite different. Second, we did not include any assessment of the number of medications ordered by each provider type or for each patient, either of which could significantly affect provider response to warnings. Third, as previously noted, we did not include any assessment of the validity of the warnings, beyond the 4 main categories described, which could also significantly affect provider response. However, it should be noted that although the validity of interaction warnings varies significantly from 1 medication to another, the validity of duplicate, allergy, and adverse reaction warnings in the described system are essentially the same for all medications. Fourth, it is possible that providers did modify or even erase their orders even after selecting override in response to the warning; it is also possible that providers reentered the same order after choosing erase. Unfortunately auditing for actions such as these would be extremely laborious. Finally, the study was conducted at a single medical center using a single order‐entry system. The system in use at our medical center is in use at one‐third of the 6000 hospitals in the United States, though certainly not all are using our version. Even if a hospital was using the same CPOE version and interaction database as our institution, variations in patient population and local decisions modifying how the database interacts with the warning presentation system might affect reproducibility at that institution.

Commonly encountered medication warnings are overridden at extremely high rates, and in our study this was particularly so for medications on the ISMP list, when ordered by physicians in training. Warnings of little clinical significance must be identified and eliminated, the most important warnings need to be visually distinct to increase user attention, and further research should be done into the patient, provider, setting, and medication factors that affect user responses to warnings, so that they may be customized accordingly and their significance increased. Doing so will enable us to reap the maximum possible potential from our CPOE systems, and increase the CPOE's power to protect our most vulnerable patients from our most dangerous medications, particularly when cared for by our most inexperienced physicians.

Acknowledgements

The authors thank, in particular, Scott Carey, Research Informatics Manager, for assistance with data collection. Additional thanks go to Olga Sherman and Kathleen Ancinich for assistance with data collection and management.

Disclosures: This research was supported in part by the Johns Hopkins Institute for Clinical and Translational Research. All listed authors contributed substantially to the study conception and design, analysis and interpretation of data, drafting the article or revising it critically for important intellectual content, and final approval of the version to be published. No one who fulfills these criteria has been excluded from authorship. This research received no specific grant from any funding agency in the public, commercial, or not‐for‐profit sectors. The authors have no competing interests to declare.

References
  1. Bates DW, Leape L, Cullen DJ, et al., Effect of computerized physician order entry and a team intervention on prevention of serious medication errors. JAMA. 1998;280:13111316.
  2. Teich JM, Merchia PR, Schmiz JL, Kuperman GJ, Spurr CD, Bates DW. Effects of computerized provider order entry on prescribing practices. Arch Intern Med. 2000;160:27412747.
  3. Garg AX, Adhikari NKJ, McDonald H, et al. Effects of computerized clinician decision support systems on practitioner performance and patient outcomes: a systematic review. JAMA. 2005;293:12231238.
  4. Wolfstadt JI, Gurwitz JH, Field TS, et al. The effect of computerized physician order entry with clinical decision support on the rates of adverse drug events: a systematic review. J Gen Intern Med. 2008;23:451458.
  5. Eslami S, Keizer NF, Abu‐Hanna A. The impact of computerized physician medication order entry in hospitalized patients—a systematic review. Int J Med Inform. 2008;77:365376.
  6. Schedlbauer A, Prasad V, Mulvaney C, et al. What evidence supports the use of computerized alerts and prompts to improve clinicians' prescribing behavior? J Am Med Inform Assoc. 2009;16:531538.
  7. Reckmann MH, Westbrook JI, Koh Y, Lo C, Day RO. Does computerized provider order entry reduce prescribing errors for hospital inpatients? A systematic review. J Am Med Inform Assoc. 2009;16:613623.
  8. Sijs H, Aarts J, Vulto A, Berg M. Overriding of drug safety alerts in computerized physician order entry. J Am Med Inform Assoc. 2006;13:138147.
  9. Lin CP, Payne TH, Nichol WP, Hoey PJ, Anderson CL, Gennari JH. Evaluating clinical decision support systems: monitoring CPOE order check override rates in the Department of Veterans Affairs' Computerized Patient Record System. J Am Med Inform Assoc. 2008;15:620626.
  10. Magnus D, Rodger S, Avery AJ. GPs' views on computerized drug interaction alerts: questionnaire survey. J Clin Pharm Ther. 2002;27:377382.
  11. Weingart SN, Simchowitz B, Shiman L, et al. Clinicians' assessments of electronic medication safety alerts in ambulatory care. Arch Intern Med. 2009;169:16271632.
  12. Lapane KL, Waring ME, Schneider KL, Dube C, Quilliam BJ. A mixed method study of the merits of e‐prescribing drug alerts in primary care. J Gen Intern Med. 2008;23:442446.
  13. Bates DW. CPOE and clinical decision support in hospitals: getting the benefits: comment on “Unintended effects of a computerized physician order entry nearly hard‐stop alert to prevent a drug interaction.” Arch Intern Med. 2010;170:15831584.
  14. Classen DC, Phansalkar S, Bates DW. Critical drug‐drug interactions for use in electronic health records systems with computerized physician order entry: review of leading approaches. J Patient Saf. 2011;7:6165.
  15. Kesselheim AS, Cresswell K, Phansalkar S, Bates DW, Sheikh A. Clinical decision support systems could be modified to reduce 'alert fatigue' while still minimizing the risk of litigation. Health Aff (Millwood). 2011;30:23102317.
  16. Hines LE, Murphy JE, Grizzle AJ, Malone DC. Critical issues associated with drug‐drug interactions: highlights of a multistakeholder conference. Am J Health Syst Pharm. 2011;68:941946.
  17. Riedmann D, Jung M, Hackl WO, Stuhlinger W, der Sijs H, Ammenwerth E. Development of a context model to prioritize drug safety alerts in CPOE systems. BMC Med Inform Decis Mak. 2011;11:35.
  18. Seidling HM, Phansalkar S, Seger DL, et al. Factors influencing alert acceptance: a novel approach for predicting the success of clinical decision support. J Am Med Inform Assoc. 2011;18:479484.
  19. Riedmann D, Jung M, Hackl WO, Ammenwerth E. How to improve the delivery of medication alerts within computerized physician order entry systems: an international Delphi study. J Am Med Inform Assoc. 2011;18:760766.
  20. Weingart SN, Toth M, Sands DZ, Aronson MD, Davis RB, Phillips RS. Physicians' decisions to override computerized drug alerts in primary care. Arch Intern Med. 2003;163:26252631.
  21. Shah NR, Seger AC, Seger DL, et al. Improving acceptance of computerized prescribing alerts in ambulatory care. J Am Med Inform Assoc. 2006;13:511.
  22. Stutman HR, Fineman R, Meyer K, Jones D. Optimizing the acceptance of medication‐based alerts by physicians during CPOE implementation in a community hospital environment. AMIA Annu Symp Proc. 2007:701705.
  23. Long AJ, Chang P, Li YC, Chiu WT. The use of a CPOE log for the analysis of physicians' behavior when responding to drug‐duplication reminders. Int J Med Inform. 2008;77:499506.
  24. Isaac T, Weissman JS, Davis RB, et al. Overrides of medication alerts in ambulatory care. Arch Intern Med. 2009;169:305311.
  25. der Sijs H, Mulder A, Gelder T, Aarts J, Berg M, Vulto A. Drug safety alert generation and overriding in a large Dutch university medical centre. Pharmacoepidemiol Drug Saf. 2009;18:941947.
  26. Seidling HM, Schmitt SP, Bruckner T, et al. Patient‐specific electronic decision support reduces prescription of excessive doses. Qual Saf Health Care. 2010;19:e15.
  27. Peberdy MA, Ornato JP, Larkin GL, et al. Survival from in‐hospital cardiac arrest during nights and weekends. JAMA. 2008;299:785792.
  28. Steinman MA, Hanlon JT. Managing medications in clinically complex elders: “There's got to be a happy medium.” JAMA. 2010;304:15921601.
  29. Agency for Healthcare Research and Quality. Safety culture. Available at: http://psnet.ahrq.gov/primer.aspx?primerID=5. Accessed October 29, 2013.
  30. Institute for Safe Medication Practice. List of High‐Alert Medications. Available at: http://www.ismp.org/Tools/highalertmedications.pdf. Accessed June 18, 2013.
  31. First Databank. FDB AlertSpace. Available at: http://www.fdbhealth.com/solutions/fdb‐alertspace. Accessed July 3, 2014.
  32. Abookire SA, Teich JM, Sandige H, et al. Improving allergy alerting in a computerized physician order entry system. Proc AMIA Symp. 2000:26.
  33. Boussadi A, Caruba T, Zapletal E, Sabatier B, Durieux P, Degoulet P. A clinical data warehouse‐based process for refining medication orders alerts. J Am Med Inform Assoc. 2012;19:782785.
  34. Phansalkar S, der Sijs H, Tucker AD, et al. Drug‐drug interactions that should be non‐interruptive in order to reduce alert fatigue in electronic health records. J Am Med Inform Assoc. 2013;20:489493.
  35. Phansalkar S, Desai AA, Bell D, et al. High‐priority drug‐drug interactions for use in electronic health records. J Am Med Inform Assoc. 2012;19:735743.
  36. Horsky J, Phansalkar S, Desai A, Bell D, Middleton B. Design of decision support interventions for medication prescribing. Int J Med Inform. 2013;82:492503.
  37. Tamblyn R, Huang A, Taylor L, et al. A randomized trial of the effectiveness of on‐demand versus computer‐triggered drug decision support in primary care. J Am Med Inform Assoc. 2008;15:430438.
  38. Paterno MD, Maviglia SM, Gorman PN, et al. Tiering drug‐drug interaction alerts by severity increases compliance rates. J Am Med Inform Assoc. 2009;16:4046.
  39. Phansalkar S, Edworthy J, Hellier E, et al. A review of human factors principles for the design and implementation of medication safety alerts in clinical information systems. J Am Med Inform Assoc. 2010;17:493501.
  40. Strom BL, Schinnar R, Aberra F, et al. Unintended effects of a computerized physician order entry nearly hard‐stop alert to prevent a drug interaction: a randomized controlled trial. Arch Intern Med. 2010;170:15781583.
  41. Strom BL, Schinnar R, Bilker W, Hennessy S, Leonard CE, Pifer E. Randomized clinical trial of a customized electronic alert requiring an affirmative response compared to a control group receiving a commercial passive CPOE alert: NSAID—warfarin co‐prescribing as a test case. J Am Med Inform Assoc. 2010;17:411415.
  42. Scott GP, Shah P, Wyatt JC, Makubate B, Cross FW. Making electronic prescribing alerts more effective: scenario‐based experimental study in junior doctors. J Am Med Inform Assoc. 2011;18:789798.
  43. Zachariah M, Phansalkar S, Seidling HM, et al. Development and preliminary evidence for the validity of an instrument assessing implementation of human‐factors principles in medication‐related decision‐support systems—I‐MeDeSA. J Am Med Inform Assoc. 2011;18(suppl 1):i62i72.
  44. Kuperman GJ, Bobb A, Payne TH, et al. Medication‐related clinical decision support in computerized provider order entry systems: a review. J Am Med Inform Assoc. 2007;14:2940.
  45. Jung M, Riedmann D, Hackl WO, et al. Physicians' perceptions on the usefulness of contextual information for prioritizing and presenting alerts in Computerized Physician Order Entry systems. BMC Med Inform Decis Mak. 2012;12:111.
  46. Hemens BJ, Holbrook A, Tonkin M, et al. Computerized clinical decision support systems for drug prescribing and management: a decision‐maker‐researcher partnership systematic review. Implement Sci. 2011;6:89.
  47. Duke JD, Bolchini D. A successful model and visual design for creating context‐aware drug‐drug interaction alerts. AMIA Annu Symp Proc. 2011;2011:339348.
References
  1. Bates DW, Leape L, Cullen DJ, et al., Effect of computerized physician order entry and a team intervention on prevention of serious medication errors. JAMA. 1998;280:13111316.
  2. Teich JM, Merchia PR, Schmiz JL, Kuperman GJ, Spurr CD, Bates DW. Effects of computerized provider order entry on prescribing practices. Arch Intern Med. 2000;160:27412747.
  3. Garg AX, Adhikari NKJ, McDonald H, et al. Effects of computerized clinician decision support systems on practitioner performance and patient outcomes: a systematic review. JAMA. 2005;293:12231238.
  4. Wolfstadt JI, Gurwitz JH, Field TS, et al. The effect of computerized physician order entry with clinical decision support on the rates of adverse drug events: a systematic review. J Gen Intern Med. 2008;23:451458.
  5. Eslami S, Keizer NF, Abu‐Hanna A. The impact of computerized physician medication order entry in hospitalized patients—a systematic review. Int J Med Inform. 2008;77:365376.
  6. Schedlbauer A, Prasad V, Mulvaney C, et al. What evidence supports the use of computerized alerts and prompts to improve clinicians' prescribing behavior? J Am Med Inform Assoc. 2009;16:531538.
  7. Reckmann MH, Westbrook JI, Koh Y, Lo C, Day RO. Does computerized provider order entry reduce prescribing errors for hospital inpatients? A systematic review. J Am Med Inform Assoc. 2009;16:613623.
  8. Sijs H, Aarts J, Vulto A, Berg M. Overriding of drug safety alerts in computerized physician order entry. J Am Med Inform Assoc. 2006;13:138147.
  9. Lin CP, Payne TH, Nichol WP, Hoey PJ, Anderson CL, Gennari JH. Evaluating clinical decision support systems: monitoring CPOE order check override rates in the Department of Veterans Affairs' Computerized Patient Record System. J Am Med Inform Assoc. 2008;15:620626.
  10. Magnus D, Rodger S, Avery AJ. GPs' views on computerized drug interaction alerts: questionnaire survey. J Clin Pharm Ther. 2002;27:377382.
  11. Weingart SN, Simchowitz B, Shiman L, et al. Clinicians' assessments of electronic medication safety alerts in ambulatory care. Arch Intern Med. 2009;169:16271632.
  12. Lapane KL, Waring ME, Schneider KL, Dube C, Quilliam BJ. A mixed method study of the merits of e‐prescribing drug alerts in primary care. J Gen Intern Med. 2008;23:442446.
  13. Bates DW. CPOE and clinical decision support in hospitals: getting the benefits: comment on “Unintended effects of a computerized physician order entry nearly hard‐stop alert to prevent a drug interaction.” Arch Intern Med. 2010;170:15831584.
  14. Classen DC, Phansalkar S, Bates DW. Critical drug‐drug interactions for use in electronic health records systems with computerized physician order entry: review of leading approaches. J Patient Saf. 2011;7:6165.
  15. Kesselheim AS, Cresswell K, Phansalkar S, Bates DW, Sheikh A. Clinical decision support systems could be modified to reduce 'alert fatigue' while still minimizing the risk of litigation. Health Aff (Millwood). 2011;30:23102317.
  16. Hines LE, Murphy JE, Grizzle AJ, Malone DC. Critical issues associated with drug‐drug interactions: highlights of a multistakeholder conference. Am J Health Syst Pharm. 2011;68:941946.
  17. Riedmann D, Jung M, Hackl WO, Stuhlinger W, der Sijs H, Ammenwerth E. Development of a context model to prioritize drug safety alerts in CPOE systems. BMC Med Inform Decis Mak. 2011;11:35.
  18. Seidling HM, Phansalkar S, Seger DL, et al. Factors influencing alert acceptance: a novel approach for predicting the success of clinical decision support. J Am Med Inform Assoc. 2011;18:479484.
  19. Riedmann D, Jung M, Hackl WO, Ammenwerth E. How to improve the delivery of medication alerts within computerized physician order entry systems: an international Delphi study. J Am Med Inform Assoc. 2011;18:760766.
  20. Weingart SN, Toth M, Sands DZ, Aronson MD, Davis RB, Phillips RS. Physicians' decisions to override computerized drug alerts in primary care. Arch Intern Med. 2003;163:26252631.
  21. Shah NR, Seger AC, Seger DL, et al. Improving acceptance of computerized prescribing alerts in ambulatory care. J Am Med Inform Assoc. 2006;13:511.
  22. Stutman HR, Fineman R, Meyer K, Jones D. Optimizing the acceptance of medication‐based alerts by physicians during CPOE implementation in a community hospital environment. AMIA Annu Symp Proc. 2007:701705.
  23. Long AJ, Chang P, Li YC, Chiu WT. The use of a CPOE log for the analysis of physicians' behavior when responding to drug‐duplication reminders. Int J Med Inform. 2008;77:499506.
  24. Isaac T, Weissman JS, Davis RB, et al. Overrides of medication alerts in ambulatory care. Arch Intern Med. 2009;169:305311.
  25. der Sijs H, Mulder A, Gelder T, Aarts J, Berg M, Vulto A. Drug safety alert generation and overriding in a large Dutch university medical centre. Pharmacoepidemiol Drug Saf. 2009;18:941947.
  26. Seidling HM, Schmitt SP, Bruckner T, et al. Patient‐specific electronic decision support reduces prescription of excessive doses. Qual Saf Health Care. 2010;19:e15.
  27. Peberdy MA, Ornato JP, Larkin GL, et al. Survival from in‐hospital cardiac arrest during nights and weekends. JAMA. 2008;299:785792.
  28. Steinman MA, Hanlon JT. Managing medications in clinically complex elders: “There's got to be a happy medium.” JAMA. 2010;304:15921601.
  29. Agency for Healthcare Research and Quality. Safety culture. Available at: http://psnet.ahrq.gov/primer.aspx?primerID=5. Accessed October 29, 2013.
  30. Institute for Safe Medication Practice. List of High‐Alert Medications. Available at: http://www.ismp.org/Tools/highalertmedications.pdf. Accessed June 18, 2013.
  31. First Databank. FDB AlertSpace. Available at: http://www.fdbhealth.com/solutions/fdb‐alertspace. Accessed July 3, 2014.
  32. Abookire SA, Teich JM, Sandige H, et al. Improving allergy alerting in a computerized physician order entry system. Proc AMIA Symp. 2000:26.
  33. Boussadi A, Caruba T, Zapletal E, Sabatier B, Durieux P, Degoulet P. A clinical data warehouse‐based process for refining medication orders alerts. J Am Med Inform Assoc. 2012;19:782785.
  34. Phansalkar S, der Sijs H, Tucker AD, et al. Drug‐drug interactions that should be non‐interruptive in order to reduce alert fatigue in electronic health records. J Am Med Inform Assoc. 2013;20:489493.
  35. Phansalkar S, Desai AA, Bell D, et al. High‐priority drug‐drug interactions for use in electronic health records. J Am Med Inform Assoc. 2012;19:735743.
  36. Horsky J, Phansalkar S, Desai A, Bell D, Middleton B. Design of decision support interventions for medication prescribing. Int J Med Inform. 2013;82:492503.
  37. Tamblyn R, Huang A, Taylor L, et al. A randomized trial of the effectiveness of on‐demand versus computer‐triggered drug decision support in primary care. J Am Med Inform Assoc. 2008;15:430438.
  38. Paterno MD, Maviglia SM, Gorman PN, et al. Tiering drug‐drug interaction alerts by severity increases compliance rates. J Am Med Inform Assoc. 2009;16:4046.
  39. Phansalkar S, Edworthy J, Hellier E, et al. A review of human factors principles for the design and implementation of medication safety alerts in clinical information systems. J Am Med Inform Assoc. 2010;17:493501.
  40. Strom BL, Schinnar R, Aberra F, et al. Unintended effects of a computerized physician order entry nearly hard‐stop alert to prevent a drug interaction: a randomized controlled trial. Arch Intern Med. 2010;170:15781583.
  41. Strom BL, Schinnar R, Bilker W, Hennessy S, Leonard CE, Pifer E. Randomized clinical trial of a customized electronic alert requiring an affirmative response compared to a control group receiving a commercial passive CPOE alert: NSAID—warfarin co‐prescribing as a test case. J Am Med Inform Assoc. 2010;17:411415.
  42. Scott GP, Shah P, Wyatt JC, Makubate B, Cross FW. Making electronic prescribing alerts more effective: scenario‐based experimental study in junior doctors. J Am Med Inform Assoc. 2011;18:789798.
  43. Zachariah M, Phansalkar S, Seidling HM, et al. Development and preliminary evidence for the validity of an instrument assessing implementation of human‐factors principles in medication‐related decision‐support systems—I‐MeDeSA. J Am Med Inform Assoc. 2011;18(suppl 1):i62i72.
  44. Kuperman GJ, Bobb A, Payne TH, et al. Medication‐related clinical decision support in computerized provider order entry systems: a review. J Am Med Inform Assoc. 2007;14:2940.
  45. Jung M, Riedmann D, Hackl WO, et al. Physicians' perceptions on the usefulness of contextual information for prioritizing and presenting alerts in Computerized Physician Order Entry systems. BMC Med Inform Decis Mak. 2012;12:111.
  46. Hemens BJ, Holbrook A, Tonkin M, et al. Computerized clinical decision support systems for drug prescribing and management: a decision‐maker‐researcher partnership systematic review. Implement Sci. 2011;6:89.
  47. Duke JD, Bolchini D. A successful model and visual design for creating context‐aware drug‐drug interaction alerts. AMIA Annu Symp Proc. 2011;2011:339348.
Issue
Journal of Hospital Medicine - 10(1)
Issue
Journal of Hospital Medicine - 10(1)
Page Number
19-25
Page Number
19-25
Publications
Publications
Article Type
Display Headline
Factors associated with medication warning acceptance for hospitalized adults
Display Headline
Factors associated with medication warning acceptance for hospitalized adults
Sections
Article Source

© 2015 Society of Hospital Medicine

Disallow All Ads
Correspondence Location
Address for correspondence and reprint requests: Amy M. Knight, MD, Division of Hospital Medicine, Johns Hopkins Bayview Medical Center, 5200 Eastern Ave., Mason F. Lord West Tower, 6th Floor, Baltimore, MD 21224; Telephone: 410‐550‐5018; Fax: 410‐550‐2972; E‐mail: aknight@jhmi.edu
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Article PDF Media
Image
Disable zoom
Off
Media Files
Image
Disable zoom
Off

Sepsis Outcomes Across Settings

Article Type
Changed
Mon, 05/22/2017 - 18:30
Display Headline
Does sepsis treatment differ between primary and overflow intensive care units?

Sepsis is a major cause of death in hospitalized patients.13 It is recommended that patients with sepsis be treated with early appropriate antibiotics, as well as early goal‐directed therapy including fluid and vasopressor support according to evidence‐based guidelines.46 Following such evidence‐based protocols and process‐of‐care interventions has been shown to be associated with better patient outcomes, including decreased mortality.7, 8

Most patients with severe sepsis are cared for in intensive care units (ICUs). At times, there are no beds available in the primary ICU and patients presenting to the hospital with sepsis are cared for in other units. Patients admitted to a non‐preferred clinical inpatient setting are sometimes referred to as overflow.9 ICUs can differ significantly in staffing patterns, equipment, and training.10 It is not known if overflow sepsis patients receive similar care when admitted to non‐primary ICUs.

At our hospital, we have an active bed management system led by the hospitalist division.11 This system includes protocols to place sepsis patients in the overflow ICU if the primary ICU is full. We hypothesized that process‐of‐care interventions would be more strictly adhered to when sepsis patients were in the primary ICU rather than in the overflow unit at our institution.

METHODS

Design

This was a retrospective cohort study of all patients with sepsis admitted to either the primary medical intensive care unit (MICU) or the overflow cardiac intensive care unit (CICU) at our hospital between July 2009 and February 2010. We reviewed the admission database starting with the month of February 2010 and proceeded backwards, month by month, until we reached the target number of patients.

Setting

The study was conducted at our 320‐bed, university‐affiliated academic medical center in Baltimore, MD. The MICU and the CICU are closed units that are located adjacent to each other and have 12 beds each. They are staffed by separate pools of attending physicians trained in pulmonary/critical care medicine and cardiovascular diseases, respectively, and no attending physician attends in both units. During the study period, there were 10 unique MICU and 14 unique CICU attending physicians; while most attending physicians covered the unit for 14 days, none of the physicians were on service more than 2 of the 2‐week blocks (28 days). Each unit is additionally staffed by fellows of the respective specialties, and internal medicine residents and interns belonging to the same residency program (who rotate through both ICUs). Residents and fellows are generally assigned to these ICUs for 4 continuous weeks. The assignment of specific attendings, fellows, and residents to either ICU is performed by individual division administrators on a rotational basis based on residency, fellowship, and faculty service requirements. The teams in each ICU function independently of each other. Clinical care of patients requiring the assistance of the other specialty (pulmonary medicine or cardiology) have guidance conferred via an official consultation. Orders on patients in both ICUs are written by the residents using the same computerized order entry system (CPOE) under the supervision of their attending physicians. The nursing staff is exclusive to each ICU. The respiratory therapists spend time in both units. The nursing and respiratory therapy staff in both ICUs are similarly trained and certified, and have the same patient‐to‐nursing ratios.

Subjects

All patients admitted with a possible diagnosis of sepsis to either the MICU or CICU were identified by querying the hospital electronic triage database called etriage. This Web‐based application is used to admit patients to all the Medicine services at our hospital. We employed a wide case‐finding net using keywords that included pneumonia, sepsis, hypotension, high lactate, hypoxia, UTI (urinary tract infection)/urosepsis, SIRS (systemic inflammatory response syndrome), hypothermia, and respiratory failure. A total of 197 adult patients were identified. The charts and the electronic medical record (EMR) of these patients were then reviewed to determine the presence of a sepsis diagnosis using standard consensus criteria.12 Severe sepsis was defined by sepsis associated with organ dysfunction, hypoperfusion, or hypotension using criteria described by Bone et al.12

Fifty‐six did not meet the criteria for sepsis and were excluded from the analysis. A total of 141 patients were included in the study. This being a pilot study, we did not have any preliminary data regarding adherence to sepsis guidelines in overflow ICUs to calculate appropriate sample size. However, in 2 recent studies of dedicated ICUs (Ferrer et al13 and Castellanos‐Ortega et al14), the averaged adherence to a single measure like checking of lactate level was 27% pre‐intervention and 62% post‐intervention. With alpha level 0.05 and 80% power, one would need 31 patients in each unit to detect such differences with respect to this intervention. Although this data does not necessarily apply to overflow ICUs or for combination of processes, we used a goal of having at least 31 patients in each ICU.

The study was approved by the Johns Hopkins Institutional Review Board. The need for informed consent was waived given the retrospective nature of the study.

Data Extraction Process and Procedures

The clinical data was extracted from the EMR and patient charts using a standardized data extraction instrument, modified from a case report form (CRF) used and validated in previous studies.15, 16 The following procedures were used for the data extraction:

  • The data extractors included 4 physicians and 1 research assistant and were trained and tested by a single expert in data review and extraction.

  • Lab data was transcribed directly from the EMR. Calculation of acute physiology and chronic health evaluation (APACHE II) scores were done using the website http://www.sfar.org/subores2/apache22.html (Socit Franaise d'Anesthsie et de Ranimation). Sepsis‐related organ failure assessment (SOFA) scores were calculated using usual criteria.17

  • Delivery of specific treatments and interventions, including their timing, was extracted from the EMR.

  • The attending physicians' notes were used as the final source to assign diagnoses such as presence of acute lung injury, site of infection, and record interventions.

 

Data Analysis

Analyses focused primarily on assessing whether patients were treated differently between the MICU and CICU. The primary exposure variables were the process‐of‐care measures. We specifically used measurement of central venous saturation, checking of lactate level, and administration of antibiotics within 60 minutes in patients with severe sepsis as our primary process‐of‐care measures.13 Continuous variables were reported as mean standard deviation, and Student's t tests were used to compare the 2 groups. Categorical data were expressed as frequency distributions, and chi‐square tests were used to identify differences between the 2 groups. All tests were 2‐tailed with statistical significance set at 0.05. Statistical analysis was performed using SPSS version 19.0. (IBM, Armonk, NY).

To overcome data constraints, we created a dichotomous variable for each of the 3 primary processes‐of‐care (indicating receipt of process or not) and then combined them into 1 dichotomous variable indicating whether or not the patients with severe sepsis received all 3 primary processes‐of‐care. The combined variable was the key independent variable in the model.

We performed logistic regression analysis on patients with severe sepsis. The equation Logit [P(ICU Type = CICU)] = + 1Combined + 2Age describes the framework of the model, with ICU type being the dependent variable, and the combined variable of patients receiving all primary measures being the independent variable and controlled for age. Logistic regression was performed using JMP (SAS Institute, Inc, Cary, NC).

We additionally performed a secondary analysis to explore possible predictors of mortality using a logistic regression model, with the event of death as the dependent variable, and age, APACHE II scores, combined processes‐of‐care, and ICU type included as independent variables.

RESULTS

There were 100 patients admitted to the MICU and 41 patients admitted to the CICU during the study period (Table 1). The majority of the patients were admitted to the ICUs directly from the emergency department (ED) (n = 129), with a small number of patients who were transferred from the Medicine floors (n = 12).

Baseline Patient Characteristics for the 141 Patients Admitted to Intensive Care Units With Sepsis During the Study Period
 MICU (N =100)CICU (N =41)P Value
  • Abbreviations: CICU, cardiac intensive care unit; MICU, medical intensive care unit; APACHE II, acute physiology and chronic health evaluation; SOFA, sepsis‐related organ failure assessment.

Age in years, mean SD67 14.872 15.10.11
Female, n (%)57 (57)27 (66)0.33
Patients with chronic organ insufficiency, n (%)59 (59)22 (54)0.56
Patients with severe sepsis, n (%)88 (88)21 (51)<0.001
Patients needing mechanical ventilation, n (%)43 (43)14 (34)0.33
APACHE II score, mean SD25.53 9.1124.37 9.530.50
SOFA score on day 1, mean SD7.09 3.556.71 4.570.60
Patients with acute lung injury on presentation, n (%)8 (8)2 (5)0.50

There were no significant differences between the 2 study groups in terms of age, sex, primary site of infection, mean APACHE II score, SOFA scores on day 1, chronic organ insufficiency, immune suppression, or need for mechanical ventilation (Table 1). The most common site of infection was lung. There were significantly more patients with severe sepsis in the MICU (88% vs 51%, P <0.001).

Sepsis Process‐of‐Care Measures

There were no significant differences in the proportion of severe sepsis patients who had central venous saturation checked (MICU: 46% vs CICU: 41%, P = 0.67), lactate level checked (95% vs 100%, P = 0.37), or received antibiotics within 60 minutes of presentation (75% vs 69%, P = 0.59) (Table 2). Multiple other processes and treatments were delivered similarly, as shown in Table 2.

ICU Treatments and Processes‐of‐Care for Patients With Sepsis During the Study Period
Primary Process‐of‐Care Measures (Severe Sepsis Patients)MICU (N = 88)CICU (N = 21)P Value
  • Abbreviations: CICU, cardiac intensive care unit; DVT, deep vein thrombosis; GI, gastrointestinal; ICU, intensive care unit; MICU, medical intensive care unit; RBC, red blood cell; SD, standard deviation. * Missing data causes percentages to be other than what might be suspected if it were available for all patients.

Patients with central venous oxygen saturation checked, n (%)*31 (46)7 (41)0.67
Patients with lactate level checked, n (%)*58 (95)16 (100)0.37
Received antibiotics within 60 min, n (%)*46 (75)11 (69)0.59
Patients who had all 3 above processes and treatments, n (%)19 (22)4 (19)0.79
Received vasopressor, n (%)25 (28)8 (38)0.55
ICU Treatments and Processes (All Sepsis Patients)(N =100)(N = 41) 
Fluid balance 24 h after admission in liters, mean SD1.96 2.421.42 2.630.24
Patients who received stress dose steroids, n (%)11 (11)4 (10)0.83
Patients who received Drotrecogin alfa, n (%)0 (0)0 (0) 
Morning glucose 24 h after admission in mg/dL, mean SD161 111144 800.38
Received DVT prophylaxis within 24 h of admission, n (%)74 (74)20 (49)0.004
Received GI prophylaxis within 24 h of admission, n (%)68 (68)18 (44)0.012
Received RBC transfusion within 24 h of admission, n (%)8 (8)7 (17)0.11
Received renal replacement therapy, n (%)13 (13)3 (7)0.33
Received a spontaneous breathing trial within 24 h of admission, n (%)*4 (11)4 (33)0.07

Logistic regression analysis examining the receipt of all 3 primary processes‐of‐care while controlling for age revealed that the odds of the being in one of the ICUs was not significantly different (P = 0.85). The secondary analysis regression models revealed that only the APACHE II score (odds ratio [OR] = 1.21; confidence interval [CI], 1.121.31) was significantly associated with higher odds of mortality. ICU‐type [MICU vs CICU] (OR = 1.85; CI, 0.428.20), age (OR = 1.01; CI, 0.971.06), and combined processes of care (OR = 0.26; CI, 0.071.01) did not have significant associations with odds of mortality.

A review of microbiologic sensitivities revealed a trend towards significance that the cultured microorganism(s) was likely to be resistant to the initial antibiotics administered in MICU vs CICU (15% vs 5%, respectively, P = 0.09).

Mechanical Ventilation Parameters

The majority of the ventilated patients were admitted to each ICU in assist control (AC) mode. There were no significant differences in categories of mean tidal volume (TV) (P = 0.3), mean plateau pressures (P = 0.12), mean fraction of inspired oxygen (FiO2) (P = 0.95), and mean positive end‐expiratory pressures (PEEP) (P = 0.98) noted across the 2 units at the time of ICU admission, and also 24 hours after ICU admission. Further comparison of measurements of tidal volumes and plateau pressures over 7 days of ICU stay revealed no significant differences in the 2 ICUs (P = 0.40 and 0.57, respectively, on day 7 of ICU admission). There was a trend towards significance in fewer patients in the MICU receiving spontaneous breathing trial within 24 hours of ICU admission (11% vs 33%, P = 0.07) (Table 2).

Patient Outcomes

There were no significant differences in ICU mortality (MICU 19% vs CICU 10%, P = 0.18), or hospital mortality (21% vs 15%, P = 0.38) across the units (Table 3). Mean ICU and hospital length of stay (LOS) and proportion of patients discharged home with unassisted breathing were similar (Table 3).

Patient Outcomes for the 141 Patients Admitted to the Intensive Care Units With Sepsis During the Study Period
Patient OutcomesMICU (N = 100)CICU (N = 41)P Value
  • Abbreviations: CICU, cardiac intensive care unit; ICU, intensive care unit; MICU, medical intensive care unit; SD, standard deviation.

ICU mortality, n (%)19 (19)4 (10)0.18
Hospital mortality, n (%)21 (21)6 (15)0.38
Discharged home with unassisted breathing, n (%)33 (33)19 (46)0.14
ICU length of stay in days, mean SD4.78 6.244.92 6.320.97
Hospital length of stay in days, mean SD9.68 9.229.73 9.330.98

DISCUSSION

Since sepsis is more commonly treated in the medical ICU and some data suggests that specialty ICUs may be better at providing desired care,18, 19 we believed that patients treated in the MICU would be more likely to receive guideline‐concordant care. The study refutes our a priori hypothesis and reveals that evidence‐based processes‐of‐care associated with improved outcomes for sepsis are similarly implemented at our institution in the primary and overflow ICU. These findings are important, as ICU bed availability is a frequent problem and many hospitals overflow patients to non‐primary ICUs.9, 20

The observed equivalence in the care delivered may be a function of the relatively high number of patients with sepsis treated in the overflow unit, thereby giving the delivery teams enough experience to provide the desired care. An alternative explanation could be that the residents in CICU brought with them the experience from having previously trained in the MICU. Although, some of the care processes for sepsis patients are influenced by the CPOE (with embedded order sets and protocols), it is unlikely that CPOE can fully account for similarity in care because many processes and therapies (like use of steroids, amount of fluid delivered in first 24 hours, packed red blood cells [PRBC] transfusion, and spontaneous breathing trials) are not embedded within order sets.

The significant difference noted in the areas of deep vein thrombosis (DVT) and gastrointestinal (GI) prophylaxis within 24 hours of ICU admission was unexpected. These preventive therapies are included in initial order sets in the CPOE, which prompt physicians to order them as standard‐of‐care. With respect to DVT prophylaxis, we suspect that some of the difference might be attributable to specific contraindications to its use, which could have been more common in one of the units. There were more patients in MICU on mechanical ventilation (although not statistically significant) and with severe sepsis (statistically significant) at time of admission, which might have contributed to the difference noted in use of GI prophylaxis. It is also plausible that these differences might have disappeared if they were reassessed beyond 24 hours into the ICU admission. We cannot rule out the presence of unit‐ and physician‐level differences that contributed to this. Likewise, there was an unexpected trend towards significance, wherein more patients in CICU had spontaneous breathing trials within 24 hours of admission. This might also be explained by the higher number of patients with severe sepsis in the MICU (preempting any weaning attempts). These caveats aside, it is reassuring that, at our institution, admitting septic patients to the first available ICU bed does not adversely affect important processes‐of‐care.

One might ask whether this study's data should reassure other sites who are boarding septic patients in non‐primary ICUs. Irrespective of the number of patients studied or the degree of statistical significance of the associations, an observational study design cannot prove that boarding septic patients in non‐primary ICUs is either safe or unsafe. However, we hope that readers reflect on, and take inventory of, systems issues that may be different between unitswith an eye towards eliminating variation such that all units managing septic patients are primed to deliver guideline‐concordant care. Other hospitals that use CPOE with sepsis order sets, have protocols for sepsis care, and who train nursing and respiratory therapists to meet high standards might be pleased to see that the patients in our study received comparable, high‐quality care across the 2 units. While our data suggests that boarding patients in overflow units may be safe, these findings would need to be replicated at other sites using prospective designs to prove safety.

Length of emergency room stay prior to admission is associated with higher mortality rates.2123 At many hospitals, critical care beds are a scarce resource such that most hospitals have a policy for the triage of patients to critical care beds.24, 25 Lundberg and colleagues' study demonstrated that patients who developed septic shock on the medical wards experienced delays in receipt of intravenous fluids, inotropic agents and transfer to a critical care setting.26 Thus, rather than waiting in the ED or on the medical service for an MICU bed to become available, it may be most wise to admit a critically sick septic patient to the first available ICU bed, even to an overflow ICU. In a recent study by Sidlow and Aggarwal, 1104 patients discharged from the coronary care unit (CCU) with a non‐cardiac primary diagnosis were compared to patients admitted to the MICU in the same hospital.27 The study found no differences in patient mortality, 30‐day readmission rate, hospital LOS, ICU LOS, and safety outcomes of ventilator‐associated pneumonia and catheter‐associated bloodstream infections between ICUs. However, their study did not examine processes‐of‐care delivered between the primary ICU and the overflow unit, and did not validate the primary diagnoses of patients admitted to the ICU.

Several limitations of this study should be considered. First, this study was conducted at a single center. Second, we used a retrospective study design; however, a prospective study randomizing patients to 1 of the 2 units would likely never be possible. Third, the relatively small number of patients limited the power of the study to detect mortality differences between the units. However, this was a pilot study focused on processes of care as opposed to clinical outcomes. Fourth, it is possible that we did not capture every single patient with sepsis with our keyword search. Our use of a previously validated screening process should have limited the number of missed cases.15, 16 Fifth, although the 2 ICUs have exclusive nursing staff and attending physicians, the housestaff and respiratory therapists do rotate between the 2 ICUs and place orders in the common CPOE. The rotating housestaff may certainly represent a source for confounding, but the large numbers (>30) of evenly spread housestaff over the study period minimizes the potential for any trainee to be responsible for a large proportion of observed practice. Sixth, ICU attendings are the physicians of record and could influence the results. Because no attending physician was on service for more than 4 weeks during the study period, and patients were equally spread over this same time, concerns about clustering and biases this may have created should be minimal but cannot be ruled out. Seventh, some interventions and processes, such as antibiotic administration and measurement of lactate, may have been initiated in the ED, thereby decreasing the potential for differences between the groups. Additionally, we cannot rule out the possibility that factors other than bed availability drove the admission process (we found that the relative proportion of patients admitted to overflow ICU during hours of ambulance diversion was similar to the overflow ICU admissions during non‐ambulance diversion hours). It is possible that some selection bias by the hospitalist assigning patients to specific ICUs influenced their triage decisionsalthough all triaging doctors go through the same process of training in active bed management.11 While more patients admitted to the MICU had severe sepsis, there were no differences between groups in APACHE II or SOFA scores. However, we cannot rule out that there were other residual confounders. Finally, in a small number of cases (4/41, 10%), the CICU team consulted the MICU attending for assistance. This input had the potential to reduce disparities in care between the units.

Overflowing patients to non‐primary ICUs occurs in many hospitals. Our study demonstrates that sepsis treatment for overflow patients may be similar to that received in the primary ICU. While a large multicentered and randomized trial could determine whether significant management and outcome differences exist between primary and overflow ICUs, feasibility concerns make it unlikely that such a study will ever be conducted.

Acknowledgements

Disclosure: Dr Wright is a Miller‐Coulson Family Scholar and this work is supported by the Miller‐Coulson family through the Johns Hopkins Center for Innovative Medicine. Dr Sevransky was supported with a grant from National Institute of General Medical Sciences, NIGMS K‐23‐1399. All other authors disclose no relevant or financial conflicts of interest.

Files
References
  1. Angus DC,Linde‐Zwirble WT,Lidicker J,Clermont G,Carcillo J,Pinsky MR.Epidemiology of severe sepsis in the United States: analysis of incidence, outcome, and associated costs of care.Crit Care Med.2001;29(7):13031310.
  2. Kumar G,Kumar N,Taneja A, et al;for the Milwaukee Initiative in Critical Care Outcomes Research (MICCOR) Group of Investigators.Nationwide trends of severe sepsis in the twenty first century (2000–2007).Chest.2011;140(5):12231231.
  3. Dombrovskiy VY,Martin AA,Sunderram J,Paz HL.Rapid increase in hospitalization and mortality rates for severe sepsis in the United States: a trend analysis from 1993 to 2003.Crit Care Med.2007;35(5):12441250.
  4. Dellinger RP,Levy MM,Carlet JM, et al.Surviving sepsis campaign: international guidelines for management of severe sepsis and septic shock: 2008.Crit Care Med.2008;36(1):296327.
  5. Jones AE,Shapiro NI,Trzeciak S, et al.Lactate clearance vs central venous oxygen saturation as goals of early sepsis therapy: a randomized clinical trial.JAMA.2010;303(8):739746.
  6. 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(19):13681377.
  7. Nguyen HB,Corbett SW,Steele R, et al.Implementation of a bundle of quality indicators for the early management of severe sepsis and septic shock is associated with decreased mortality.Crit Care Med.2007;35(4):11051112.
  8. Kumar A,Zarychanski R,Light B, et al.Early combination antibiotic therapy yields improved survival compared with monotherapy in septic shock: a propensity‐matched analysis.Crit Care Med.2010;38(9):17731785.
  9. Johannes MS.A new dimension of the PACU: the dilemma of the ICU overflow patient.J Post Anesth Nurs.1994;9(5):297300.
  10. Groeger JS,Strosberg MA,Halpern NA, et al.Descriptive analysis of critical care units in the United States.Crit Care Med.1992;20(6):846863.
  11. Howell E,Bessman E,Kravet S,Kolodner K,Marshall R,Wright S.Active bed management by hospitalists and emergency department throughput.Ann Intern Med.2008;149(11):804811.
  12. 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):16441655.
  13. 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):22942303.
  14. Castellanos‐Ortega A,Suberviola B,Garcia‐Astudillo LA, et al.Impact of the surviving sepsis campaign protocols on hospital length of stay and mortality in septic shock patients: results of a three‐year follow‐up quasi‐experimental study.Crit Care Med.2010;38(4):10361043.
  15. Needham DM,Dennison CR,Dowdy DW, et al.Study protocol: the improving care of acute lung injury patients (ICAP) study.Crit Care.2006;10(1):R9.
  16. Ali N,Gutteridge D,Shahul S,Checkley W,Sevransky J,Martin G.Critical illness outcome study: an observational study of protocols and mortality in intensive care units.Open Access J Clin Trials.2011;3(September):5565.
  17. Vincent JL,Moreno R,Takala J, et al.The SOFA (sepsis‐related organ failure assessment) score to describe organ dysfunction/failure: on behalf of the Working Group on Sepsis‐Related Problems of the European Society of Intensive Care Medicine.Intensive Care Med.1996;22(7):707710.
  18. Pronovost PJ,Angus DC,Dorman T,Robinson KA,Dremsizov TT,Young TL.Physician staffing patterns and clinical outcomes in critically ill patients: a systematic review.JAMA.2002;288(17):21512162.
  19. Fuchs RJ,Berenholtz SM,Dorman T.Do intensivists in ICU improve outcome?Best Pract Res Clin Anaesthesiol.2005;19(1):125135.
  20. Lindsay M.Is the postanesthesia care unit becoming an intensive care unit?J Perianesth Nurs.1999;14(2):7377.
  21. Chalfin DB,Trzeciak S,Likourezos A,Baumann BM,Dellinger RP;for the DELAY‐ED Study Group.Impact of delayed transfer of critically ill patients from the emergency department to the intensive care unit.Crit Care Med.2007;35(6):14771483.
  22. Renaud B,Santin A,Coma E, et al.Association between timing of intensive care unit admission and outcomes for emergency department patients with community‐acquired pneumonia.Crit Care Med.2009;37(11):28672874.
  23. Shen YC,Hsia RY.Association between ambulance diversion and survival among patients with acute myocardial infarction.JAMA.2011;305(23):24402447.
  24. Teres D.Civilian triage in the intensive care unit: the ritual of the last bed.Crit Care Med.1993;21(4):598606.
  25. Sinuff T,Kahnamoui K,Cook DJ,Luce JM,Levy MM;for the Values Ethics and Rationing in Critical Care Task Force.Rationing critical care beds: a systematic review.Crit Care Med.2004;32(7):15881597.
  26. Lundberg JS,Perl TM,Wiblin T, et al.Septic shock: an analysis of outcomes for patients with onset on hospital wards versus intensive care units.Crit Care Med.1998;26(6):10201024.
  27. Sidlow R,Aggarwal V.“The MICU is full”: one hospital's experience with an overflow triage policy.Jt Comm J Qual Patient Saf.2011;37(10):456460.
Article PDF
Issue
Journal of Hospital Medicine - 7(8)
Publications
Page Number
600-605
Sections
Files
Files
Article PDF
Article PDF

Sepsis is a major cause of death in hospitalized patients.13 It is recommended that patients with sepsis be treated with early appropriate antibiotics, as well as early goal‐directed therapy including fluid and vasopressor support according to evidence‐based guidelines.46 Following such evidence‐based protocols and process‐of‐care interventions has been shown to be associated with better patient outcomes, including decreased mortality.7, 8

Most patients with severe sepsis are cared for in intensive care units (ICUs). At times, there are no beds available in the primary ICU and patients presenting to the hospital with sepsis are cared for in other units. Patients admitted to a non‐preferred clinical inpatient setting are sometimes referred to as overflow.9 ICUs can differ significantly in staffing patterns, equipment, and training.10 It is not known if overflow sepsis patients receive similar care when admitted to non‐primary ICUs.

At our hospital, we have an active bed management system led by the hospitalist division.11 This system includes protocols to place sepsis patients in the overflow ICU if the primary ICU is full. We hypothesized that process‐of‐care interventions would be more strictly adhered to when sepsis patients were in the primary ICU rather than in the overflow unit at our institution.

METHODS

Design

This was a retrospective cohort study of all patients with sepsis admitted to either the primary medical intensive care unit (MICU) or the overflow cardiac intensive care unit (CICU) at our hospital between July 2009 and February 2010. We reviewed the admission database starting with the month of February 2010 and proceeded backwards, month by month, until we reached the target number of patients.

Setting

The study was conducted at our 320‐bed, university‐affiliated academic medical center in Baltimore, MD. The MICU and the CICU are closed units that are located adjacent to each other and have 12 beds each. They are staffed by separate pools of attending physicians trained in pulmonary/critical care medicine and cardiovascular diseases, respectively, and no attending physician attends in both units. During the study period, there were 10 unique MICU and 14 unique CICU attending physicians; while most attending physicians covered the unit for 14 days, none of the physicians were on service more than 2 of the 2‐week blocks (28 days). Each unit is additionally staffed by fellows of the respective specialties, and internal medicine residents and interns belonging to the same residency program (who rotate through both ICUs). Residents and fellows are generally assigned to these ICUs for 4 continuous weeks. The assignment of specific attendings, fellows, and residents to either ICU is performed by individual division administrators on a rotational basis based on residency, fellowship, and faculty service requirements. The teams in each ICU function independently of each other. Clinical care of patients requiring the assistance of the other specialty (pulmonary medicine or cardiology) have guidance conferred via an official consultation. Orders on patients in both ICUs are written by the residents using the same computerized order entry system (CPOE) under the supervision of their attending physicians. The nursing staff is exclusive to each ICU. The respiratory therapists spend time in both units. The nursing and respiratory therapy staff in both ICUs are similarly trained and certified, and have the same patient‐to‐nursing ratios.

Subjects

All patients admitted with a possible diagnosis of sepsis to either the MICU or CICU were identified by querying the hospital electronic triage database called etriage. This Web‐based application is used to admit patients to all the Medicine services at our hospital. We employed a wide case‐finding net using keywords that included pneumonia, sepsis, hypotension, high lactate, hypoxia, UTI (urinary tract infection)/urosepsis, SIRS (systemic inflammatory response syndrome), hypothermia, and respiratory failure. A total of 197 adult patients were identified. The charts and the electronic medical record (EMR) of these patients were then reviewed to determine the presence of a sepsis diagnosis using standard consensus criteria.12 Severe sepsis was defined by sepsis associated with organ dysfunction, hypoperfusion, or hypotension using criteria described by Bone et al.12

Fifty‐six did not meet the criteria for sepsis and were excluded from the analysis. A total of 141 patients were included in the study. This being a pilot study, we did not have any preliminary data regarding adherence to sepsis guidelines in overflow ICUs to calculate appropriate sample size. However, in 2 recent studies of dedicated ICUs (Ferrer et al13 and Castellanos‐Ortega et al14), the averaged adherence to a single measure like checking of lactate level was 27% pre‐intervention and 62% post‐intervention. With alpha level 0.05 and 80% power, one would need 31 patients in each unit to detect such differences with respect to this intervention. Although this data does not necessarily apply to overflow ICUs or for combination of processes, we used a goal of having at least 31 patients in each ICU.

The study was approved by the Johns Hopkins Institutional Review Board. The need for informed consent was waived given the retrospective nature of the study.

Data Extraction Process and Procedures

The clinical data was extracted from the EMR and patient charts using a standardized data extraction instrument, modified from a case report form (CRF) used and validated in previous studies.15, 16 The following procedures were used for the data extraction:

  • The data extractors included 4 physicians and 1 research assistant and were trained and tested by a single expert in data review and extraction.

  • Lab data was transcribed directly from the EMR. Calculation of acute physiology and chronic health evaluation (APACHE II) scores were done using the website http://www.sfar.org/subores2/apache22.html (Socit Franaise d'Anesthsie et de Ranimation). Sepsis‐related organ failure assessment (SOFA) scores were calculated using usual criteria.17

  • Delivery of specific treatments and interventions, including their timing, was extracted from the EMR.

  • The attending physicians' notes were used as the final source to assign diagnoses such as presence of acute lung injury, site of infection, and record interventions.

 

Data Analysis

Analyses focused primarily on assessing whether patients were treated differently between the MICU and CICU. The primary exposure variables were the process‐of‐care measures. We specifically used measurement of central venous saturation, checking of lactate level, and administration of antibiotics within 60 minutes in patients with severe sepsis as our primary process‐of‐care measures.13 Continuous variables were reported as mean standard deviation, and Student's t tests were used to compare the 2 groups. Categorical data were expressed as frequency distributions, and chi‐square tests were used to identify differences between the 2 groups. All tests were 2‐tailed with statistical significance set at 0.05. Statistical analysis was performed using SPSS version 19.0. (IBM, Armonk, NY).

To overcome data constraints, we created a dichotomous variable for each of the 3 primary processes‐of‐care (indicating receipt of process or not) and then combined them into 1 dichotomous variable indicating whether or not the patients with severe sepsis received all 3 primary processes‐of‐care. The combined variable was the key independent variable in the model.

We performed logistic regression analysis on patients with severe sepsis. The equation Logit [P(ICU Type = CICU)] = + 1Combined + 2Age describes the framework of the model, with ICU type being the dependent variable, and the combined variable of patients receiving all primary measures being the independent variable and controlled for age. Logistic regression was performed using JMP (SAS Institute, Inc, Cary, NC).

We additionally performed a secondary analysis to explore possible predictors of mortality using a logistic regression model, with the event of death as the dependent variable, and age, APACHE II scores, combined processes‐of‐care, and ICU type included as independent variables.

RESULTS

There were 100 patients admitted to the MICU and 41 patients admitted to the CICU during the study period (Table 1). The majority of the patients were admitted to the ICUs directly from the emergency department (ED) (n = 129), with a small number of patients who were transferred from the Medicine floors (n = 12).

Baseline Patient Characteristics for the 141 Patients Admitted to Intensive Care Units With Sepsis During the Study Period
 MICU (N =100)CICU (N =41)P Value
  • Abbreviations: CICU, cardiac intensive care unit; MICU, medical intensive care unit; APACHE II, acute physiology and chronic health evaluation; SOFA, sepsis‐related organ failure assessment.

Age in years, mean SD67 14.872 15.10.11
Female, n (%)57 (57)27 (66)0.33
Patients with chronic organ insufficiency, n (%)59 (59)22 (54)0.56
Patients with severe sepsis, n (%)88 (88)21 (51)<0.001
Patients needing mechanical ventilation, n (%)43 (43)14 (34)0.33
APACHE II score, mean SD25.53 9.1124.37 9.530.50
SOFA score on day 1, mean SD7.09 3.556.71 4.570.60
Patients with acute lung injury on presentation, n (%)8 (8)2 (5)0.50

There were no significant differences between the 2 study groups in terms of age, sex, primary site of infection, mean APACHE II score, SOFA scores on day 1, chronic organ insufficiency, immune suppression, or need for mechanical ventilation (Table 1). The most common site of infection was lung. There were significantly more patients with severe sepsis in the MICU (88% vs 51%, P <0.001).

Sepsis Process‐of‐Care Measures

There were no significant differences in the proportion of severe sepsis patients who had central venous saturation checked (MICU: 46% vs CICU: 41%, P = 0.67), lactate level checked (95% vs 100%, P = 0.37), or received antibiotics within 60 minutes of presentation (75% vs 69%, P = 0.59) (Table 2). Multiple other processes and treatments were delivered similarly, as shown in Table 2.

ICU Treatments and Processes‐of‐Care for Patients With Sepsis During the Study Period
Primary Process‐of‐Care Measures (Severe Sepsis Patients)MICU (N = 88)CICU (N = 21)P Value
  • Abbreviations: CICU, cardiac intensive care unit; DVT, deep vein thrombosis; GI, gastrointestinal; ICU, intensive care unit; MICU, medical intensive care unit; RBC, red blood cell; SD, standard deviation. * Missing data causes percentages to be other than what might be suspected if it were available for all patients.

Patients with central venous oxygen saturation checked, n (%)*31 (46)7 (41)0.67
Patients with lactate level checked, n (%)*58 (95)16 (100)0.37
Received antibiotics within 60 min, n (%)*46 (75)11 (69)0.59
Patients who had all 3 above processes and treatments, n (%)19 (22)4 (19)0.79
Received vasopressor, n (%)25 (28)8 (38)0.55
ICU Treatments and Processes (All Sepsis Patients)(N =100)(N = 41) 
Fluid balance 24 h after admission in liters, mean SD1.96 2.421.42 2.630.24
Patients who received stress dose steroids, n (%)11 (11)4 (10)0.83
Patients who received Drotrecogin alfa, n (%)0 (0)0 (0) 
Morning glucose 24 h after admission in mg/dL, mean SD161 111144 800.38
Received DVT prophylaxis within 24 h of admission, n (%)74 (74)20 (49)0.004
Received GI prophylaxis within 24 h of admission, n (%)68 (68)18 (44)0.012
Received RBC transfusion within 24 h of admission, n (%)8 (8)7 (17)0.11
Received renal replacement therapy, n (%)13 (13)3 (7)0.33
Received a spontaneous breathing trial within 24 h of admission, n (%)*4 (11)4 (33)0.07

Logistic regression analysis examining the receipt of all 3 primary processes‐of‐care while controlling for age revealed that the odds of the being in one of the ICUs was not significantly different (P = 0.85). The secondary analysis regression models revealed that only the APACHE II score (odds ratio [OR] = 1.21; confidence interval [CI], 1.121.31) was significantly associated with higher odds of mortality. ICU‐type [MICU vs CICU] (OR = 1.85; CI, 0.428.20), age (OR = 1.01; CI, 0.971.06), and combined processes of care (OR = 0.26; CI, 0.071.01) did not have significant associations with odds of mortality.

A review of microbiologic sensitivities revealed a trend towards significance that the cultured microorganism(s) was likely to be resistant to the initial antibiotics administered in MICU vs CICU (15% vs 5%, respectively, P = 0.09).

Mechanical Ventilation Parameters

The majority of the ventilated patients were admitted to each ICU in assist control (AC) mode. There were no significant differences in categories of mean tidal volume (TV) (P = 0.3), mean plateau pressures (P = 0.12), mean fraction of inspired oxygen (FiO2) (P = 0.95), and mean positive end‐expiratory pressures (PEEP) (P = 0.98) noted across the 2 units at the time of ICU admission, and also 24 hours after ICU admission. Further comparison of measurements of tidal volumes and plateau pressures over 7 days of ICU stay revealed no significant differences in the 2 ICUs (P = 0.40 and 0.57, respectively, on day 7 of ICU admission). There was a trend towards significance in fewer patients in the MICU receiving spontaneous breathing trial within 24 hours of ICU admission (11% vs 33%, P = 0.07) (Table 2).

Patient Outcomes

There were no significant differences in ICU mortality (MICU 19% vs CICU 10%, P = 0.18), or hospital mortality (21% vs 15%, P = 0.38) across the units (Table 3). Mean ICU and hospital length of stay (LOS) and proportion of patients discharged home with unassisted breathing were similar (Table 3).

Patient Outcomes for the 141 Patients Admitted to the Intensive Care Units With Sepsis During the Study Period
Patient OutcomesMICU (N = 100)CICU (N = 41)P Value
  • Abbreviations: CICU, cardiac intensive care unit; ICU, intensive care unit; MICU, medical intensive care unit; SD, standard deviation.

ICU mortality, n (%)19 (19)4 (10)0.18
Hospital mortality, n (%)21 (21)6 (15)0.38
Discharged home with unassisted breathing, n (%)33 (33)19 (46)0.14
ICU length of stay in days, mean SD4.78 6.244.92 6.320.97
Hospital length of stay in days, mean SD9.68 9.229.73 9.330.98

DISCUSSION

Since sepsis is more commonly treated in the medical ICU and some data suggests that specialty ICUs may be better at providing desired care,18, 19 we believed that patients treated in the MICU would be more likely to receive guideline‐concordant care. The study refutes our a priori hypothesis and reveals that evidence‐based processes‐of‐care associated with improved outcomes for sepsis are similarly implemented at our institution in the primary and overflow ICU. These findings are important, as ICU bed availability is a frequent problem and many hospitals overflow patients to non‐primary ICUs.9, 20

The observed equivalence in the care delivered may be a function of the relatively high number of patients with sepsis treated in the overflow unit, thereby giving the delivery teams enough experience to provide the desired care. An alternative explanation could be that the residents in CICU brought with them the experience from having previously trained in the MICU. Although, some of the care processes for sepsis patients are influenced by the CPOE (with embedded order sets and protocols), it is unlikely that CPOE can fully account for similarity in care because many processes and therapies (like use of steroids, amount of fluid delivered in first 24 hours, packed red blood cells [PRBC] transfusion, and spontaneous breathing trials) are not embedded within order sets.

The significant difference noted in the areas of deep vein thrombosis (DVT) and gastrointestinal (GI) prophylaxis within 24 hours of ICU admission was unexpected. These preventive therapies are included in initial order sets in the CPOE, which prompt physicians to order them as standard‐of‐care. With respect to DVT prophylaxis, we suspect that some of the difference might be attributable to specific contraindications to its use, which could have been more common in one of the units. There were more patients in MICU on mechanical ventilation (although not statistically significant) and with severe sepsis (statistically significant) at time of admission, which might have contributed to the difference noted in use of GI prophylaxis. It is also plausible that these differences might have disappeared if they were reassessed beyond 24 hours into the ICU admission. We cannot rule out the presence of unit‐ and physician‐level differences that contributed to this. Likewise, there was an unexpected trend towards significance, wherein more patients in CICU had spontaneous breathing trials within 24 hours of admission. This might also be explained by the higher number of patients with severe sepsis in the MICU (preempting any weaning attempts). These caveats aside, it is reassuring that, at our institution, admitting septic patients to the first available ICU bed does not adversely affect important processes‐of‐care.

One might ask whether this study's data should reassure other sites who are boarding septic patients in non‐primary ICUs. Irrespective of the number of patients studied or the degree of statistical significance of the associations, an observational study design cannot prove that boarding septic patients in non‐primary ICUs is either safe or unsafe. However, we hope that readers reflect on, and take inventory of, systems issues that may be different between unitswith an eye towards eliminating variation such that all units managing septic patients are primed to deliver guideline‐concordant care. Other hospitals that use CPOE with sepsis order sets, have protocols for sepsis care, and who train nursing and respiratory therapists to meet high standards might be pleased to see that the patients in our study received comparable, high‐quality care across the 2 units. While our data suggests that boarding patients in overflow units may be safe, these findings would need to be replicated at other sites using prospective designs to prove safety.

Length of emergency room stay prior to admission is associated with higher mortality rates.2123 At many hospitals, critical care beds are a scarce resource such that most hospitals have a policy for the triage of patients to critical care beds.24, 25 Lundberg and colleagues' study demonstrated that patients who developed septic shock on the medical wards experienced delays in receipt of intravenous fluids, inotropic agents and transfer to a critical care setting.26 Thus, rather than waiting in the ED or on the medical service for an MICU bed to become available, it may be most wise to admit a critically sick septic patient to the first available ICU bed, even to an overflow ICU. In a recent study by Sidlow and Aggarwal, 1104 patients discharged from the coronary care unit (CCU) with a non‐cardiac primary diagnosis were compared to patients admitted to the MICU in the same hospital.27 The study found no differences in patient mortality, 30‐day readmission rate, hospital LOS, ICU LOS, and safety outcomes of ventilator‐associated pneumonia and catheter‐associated bloodstream infections between ICUs. However, their study did not examine processes‐of‐care delivered between the primary ICU and the overflow unit, and did not validate the primary diagnoses of patients admitted to the ICU.

Several limitations of this study should be considered. First, this study was conducted at a single center. Second, we used a retrospective study design; however, a prospective study randomizing patients to 1 of the 2 units would likely never be possible. Third, the relatively small number of patients limited the power of the study to detect mortality differences between the units. However, this was a pilot study focused on processes of care as opposed to clinical outcomes. Fourth, it is possible that we did not capture every single patient with sepsis with our keyword search. Our use of a previously validated screening process should have limited the number of missed cases.15, 16 Fifth, although the 2 ICUs have exclusive nursing staff and attending physicians, the housestaff and respiratory therapists do rotate between the 2 ICUs and place orders in the common CPOE. The rotating housestaff may certainly represent a source for confounding, but the large numbers (>30) of evenly spread housestaff over the study period minimizes the potential for any trainee to be responsible for a large proportion of observed practice. Sixth, ICU attendings are the physicians of record and could influence the results. Because no attending physician was on service for more than 4 weeks during the study period, and patients were equally spread over this same time, concerns about clustering and biases this may have created should be minimal but cannot be ruled out. Seventh, some interventions and processes, such as antibiotic administration and measurement of lactate, may have been initiated in the ED, thereby decreasing the potential for differences between the groups. Additionally, we cannot rule out the possibility that factors other than bed availability drove the admission process (we found that the relative proportion of patients admitted to overflow ICU during hours of ambulance diversion was similar to the overflow ICU admissions during non‐ambulance diversion hours). It is possible that some selection bias by the hospitalist assigning patients to specific ICUs influenced their triage decisionsalthough all triaging doctors go through the same process of training in active bed management.11 While more patients admitted to the MICU had severe sepsis, there were no differences between groups in APACHE II or SOFA scores. However, we cannot rule out that there were other residual confounders. Finally, in a small number of cases (4/41, 10%), the CICU team consulted the MICU attending for assistance. This input had the potential to reduce disparities in care between the units.

Overflowing patients to non‐primary ICUs occurs in many hospitals. Our study demonstrates that sepsis treatment for overflow patients may be similar to that received in the primary ICU. While a large multicentered and randomized trial could determine whether significant management and outcome differences exist between primary and overflow ICUs, feasibility concerns make it unlikely that such a study will ever be conducted.

Acknowledgements

Disclosure: Dr Wright is a Miller‐Coulson Family Scholar and this work is supported by the Miller‐Coulson family through the Johns Hopkins Center for Innovative Medicine. Dr Sevransky was supported with a grant from National Institute of General Medical Sciences, NIGMS K‐23‐1399. All other authors disclose no relevant or financial conflicts of interest.

Sepsis is a major cause of death in hospitalized patients.13 It is recommended that patients with sepsis be treated with early appropriate antibiotics, as well as early goal‐directed therapy including fluid and vasopressor support according to evidence‐based guidelines.46 Following such evidence‐based protocols and process‐of‐care interventions has been shown to be associated with better patient outcomes, including decreased mortality.7, 8

Most patients with severe sepsis are cared for in intensive care units (ICUs). At times, there are no beds available in the primary ICU and patients presenting to the hospital with sepsis are cared for in other units. Patients admitted to a non‐preferred clinical inpatient setting are sometimes referred to as overflow.9 ICUs can differ significantly in staffing patterns, equipment, and training.10 It is not known if overflow sepsis patients receive similar care when admitted to non‐primary ICUs.

At our hospital, we have an active bed management system led by the hospitalist division.11 This system includes protocols to place sepsis patients in the overflow ICU if the primary ICU is full. We hypothesized that process‐of‐care interventions would be more strictly adhered to when sepsis patients were in the primary ICU rather than in the overflow unit at our institution.

METHODS

Design

This was a retrospective cohort study of all patients with sepsis admitted to either the primary medical intensive care unit (MICU) or the overflow cardiac intensive care unit (CICU) at our hospital between July 2009 and February 2010. We reviewed the admission database starting with the month of February 2010 and proceeded backwards, month by month, until we reached the target number of patients.

Setting

The study was conducted at our 320‐bed, university‐affiliated academic medical center in Baltimore, MD. The MICU and the CICU are closed units that are located adjacent to each other and have 12 beds each. They are staffed by separate pools of attending physicians trained in pulmonary/critical care medicine and cardiovascular diseases, respectively, and no attending physician attends in both units. During the study period, there were 10 unique MICU and 14 unique CICU attending physicians; while most attending physicians covered the unit for 14 days, none of the physicians were on service more than 2 of the 2‐week blocks (28 days). Each unit is additionally staffed by fellows of the respective specialties, and internal medicine residents and interns belonging to the same residency program (who rotate through both ICUs). Residents and fellows are generally assigned to these ICUs for 4 continuous weeks. The assignment of specific attendings, fellows, and residents to either ICU is performed by individual division administrators on a rotational basis based on residency, fellowship, and faculty service requirements. The teams in each ICU function independently of each other. Clinical care of patients requiring the assistance of the other specialty (pulmonary medicine or cardiology) have guidance conferred via an official consultation. Orders on patients in both ICUs are written by the residents using the same computerized order entry system (CPOE) under the supervision of their attending physicians. The nursing staff is exclusive to each ICU. The respiratory therapists spend time in both units. The nursing and respiratory therapy staff in both ICUs are similarly trained and certified, and have the same patient‐to‐nursing ratios.

Subjects

All patients admitted with a possible diagnosis of sepsis to either the MICU or CICU were identified by querying the hospital electronic triage database called etriage. This Web‐based application is used to admit patients to all the Medicine services at our hospital. We employed a wide case‐finding net using keywords that included pneumonia, sepsis, hypotension, high lactate, hypoxia, UTI (urinary tract infection)/urosepsis, SIRS (systemic inflammatory response syndrome), hypothermia, and respiratory failure. A total of 197 adult patients were identified. The charts and the electronic medical record (EMR) of these patients were then reviewed to determine the presence of a sepsis diagnosis using standard consensus criteria.12 Severe sepsis was defined by sepsis associated with organ dysfunction, hypoperfusion, or hypotension using criteria described by Bone et al.12

Fifty‐six did not meet the criteria for sepsis and were excluded from the analysis. A total of 141 patients were included in the study. This being a pilot study, we did not have any preliminary data regarding adherence to sepsis guidelines in overflow ICUs to calculate appropriate sample size. However, in 2 recent studies of dedicated ICUs (Ferrer et al13 and Castellanos‐Ortega et al14), the averaged adherence to a single measure like checking of lactate level was 27% pre‐intervention and 62% post‐intervention. With alpha level 0.05 and 80% power, one would need 31 patients in each unit to detect such differences with respect to this intervention. Although this data does not necessarily apply to overflow ICUs or for combination of processes, we used a goal of having at least 31 patients in each ICU.

The study was approved by the Johns Hopkins Institutional Review Board. The need for informed consent was waived given the retrospective nature of the study.

Data Extraction Process and Procedures

The clinical data was extracted from the EMR and patient charts using a standardized data extraction instrument, modified from a case report form (CRF) used and validated in previous studies.15, 16 The following procedures were used for the data extraction:

  • The data extractors included 4 physicians and 1 research assistant and were trained and tested by a single expert in data review and extraction.

  • Lab data was transcribed directly from the EMR. Calculation of acute physiology and chronic health evaluation (APACHE II) scores were done using the website http://www.sfar.org/subores2/apache22.html (Socit Franaise d'Anesthsie et de Ranimation). Sepsis‐related organ failure assessment (SOFA) scores were calculated using usual criteria.17

  • Delivery of specific treatments and interventions, including their timing, was extracted from the EMR.

  • The attending physicians' notes were used as the final source to assign diagnoses such as presence of acute lung injury, site of infection, and record interventions.

 

Data Analysis

Analyses focused primarily on assessing whether patients were treated differently between the MICU and CICU. The primary exposure variables were the process‐of‐care measures. We specifically used measurement of central venous saturation, checking of lactate level, and administration of antibiotics within 60 minutes in patients with severe sepsis as our primary process‐of‐care measures.13 Continuous variables were reported as mean standard deviation, and Student's t tests were used to compare the 2 groups. Categorical data were expressed as frequency distributions, and chi‐square tests were used to identify differences between the 2 groups. All tests were 2‐tailed with statistical significance set at 0.05. Statistical analysis was performed using SPSS version 19.0. (IBM, Armonk, NY).

To overcome data constraints, we created a dichotomous variable for each of the 3 primary processes‐of‐care (indicating receipt of process or not) and then combined them into 1 dichotomous variable indicating whether or not the patients with severe sepsis received all 3 primary processes‐of‐care. The combined variable was the key independent variable in the model.

We performed logistic regression analysis on patients with severe sepsis. The equation Logit [P(ICU Type = CICU)] = + 1Combined + 2Age describes the framework of the model, with ICU type being the dependent variable, and the combined variable of patients receiving all primary measures being the independent variable and controlled for age. Logistic regression was performed using JMP (SAS Institute, Inc, Cary, NC).

We additionally performed a secondary analysis to explore possible predictors of mortality using a logistic regression model, with the event of death as the dependent variable, and age, APACHE II scores, combined processes‐of‐care, and ICU type included as independent variables.

RESULTS

There were 100 patients admitted to the MICU and 41 patients admitted to the CICU during the study period (Table 1). The majority of the patients were admitted to the ICUs directly from the emergency department (ED) (n = 129), with a small number of patients who were transferred from the Medicine floors (n = 12).

Baseline Patient Characteristics for the 141 Patients Admitted to Intensive Care Units With Sepsis During the Study Period
 MICU (N =100)CICU (N =41)P Value
  • Abbreviations: CICU, cardiac intensive care unit; MICU, medical intensive care unit; APACHE II, acute physiology and chronic health evaluation; SOFA, sepsis‐related organ failure assessment.

Age in years, mean SD67 14.872 15.10.11
Female, n (%)57 (57)27 (66)0.33
Patients with chronic organ insufficiency, n (%)59 (59)22 (54)0.56
Patients with severe sepsis, n (%)88 (88)21 (51)<0.001
Patients needing mechanical ventilation, n (%)43 (43)14 (34)0.33
APACHE II score, mean SD25.53 9.1124.37 9.530.50
SOFA score on day 1, mean SD7.09 3.556.71 4.570.60
Patients with acute lung injury on presentation, n (%)8 (8)2 (5)0.50

There were no significant differences between the 2 study groups in terms of age, sex, primary site of infection, mean APACHE II score, SOFA scores on day 1, chronic organ insufficiency, immune suppression, or need for mechanical ventilation (Table 1). The most common site of infection was lung. There were significantly more patients with severe sepsis in the MICU (88% vs 51%, P <0.001).

Sepsis Process‐of‐Care Measures

There were no significant differences in the proportion of severe sepsis patients who had central venous saturation checked (MICU: 46% vs CICU: 41%, P = 0.67), lactate level checked (95% vs 100%, P = 0.37), or received antibiotics within 60 minutes of presentation (75% vs 69%, P = 0.59) (Table 2). Multiple other processes and treatments were delivered similarly, as shown in Table 2.

ICU Treatments and Processes‐of‐Care for Patients With Sepsis During the Study Period
Primary Process‐of‐Care Measures (Severe Sepsis Patients)MICU (N = 88)CICU (N = 21)P Value
  • Abbreviations: CICU, cardiac intensive care unit; DVT, deep vein thrombosis; GI, gastrointestinal; ICU, intensive care unit; MICU, medical intensive care unit; RBC, red blood cell; SD, standard deviation. * Missing data causes percentages to be other than what might be suspected if it were available for all patients.

Patients with central venous oxygen saturation checked, n (%)*31 (46)7 (41)0.67
Patients with lactate level checked, n (%)*58 (95)16 (100)0.37
Received antibiotics within 60 min, n (%)*46 (75)11 (69)0.59
Patients who had all 3 above processes and treatments, n (%)19 (22)4 (19)0.79
Received vasopressor, n (%)25 (28)8 (38)0.55
ICU Treatments and Processes (All Sepsis Patients)(N =100)(N = 41) 
Fluid balance 24 h after admission in liters, mean SD1.96 2.421.42 2.630.24
Patients who received stress dose steroids, n (%)11 (11)4 (10)0.83
Patients who received Drotrecogin alfa, n (%)0 (0)0 (0) 
Morning glucose 24 h after admission in mg/dL, mean SD161 111144 800.38
Received DVT prophylaxis within 24 h of admission, n (%)74 (74)20 (49)0.004
Received GI prophylaxis within 24 h of admission, n (%)68 (68)18 (44)0.012
Received RBC transfusion within 24 h of admission, n (%)8 (8)7 (17)0.11
Received renal replacement therapy, n (%)13 (13)3 (7)0.33
Received a spontaneous breathing trial within 24 h of admission, n (%)*4 (11)4 (33)0.07

Logistic regression analysis examining the receipt of all 3 primary processes‐of‐care while controlling for age revealed that the odds of the being in one of the ICUs was not significantly different (P = 0.85). The secondary analysis regression models revealed that only the APACHE II score (odds ratio [OR] = 1.21; confidence interval [CI], 1.121.31) was significantly associated with higher odds of mortality. ICU‐type [MICU vs CICU] (OR = 1.85; CI, 0.428.20), age (OR = 1.01; CI, 0.971.06), and combined processes of care (OR = 0.26; CI, 0.071.01) did not have significant associations with odds of mortality.

A review of microbiologic sensitivities revealed a trend towards significance that the cultured microorganism(s) was likely to be resistant to the initial antibiotics administered in MICU vs CICU (15% vs 5%, respectively, P = 0.09).

Mechanical Ventilation Parameters

The majority of the ventilated patients were admitted to each ICU in assist control (AC) mode. There were no significant differences in categories of mean tidal volume (TV) (P = 0.3), mean plateau pressures (P = 0.12), mean fraction of inspired oxygen (FiO2) (P = 0.95), and mean positive end‐expiratory pressures (PEEP) (P = 0.98) noted across the 2 units at the time of ICU admission, and also 24 hours after ICU admission. Further comparison of measurements of tidal volumes and plateau pressures over 7 days of ICU stay revealed no significant differences in the 2 ICUs (P = 0.40 and 0.57, respectively, on day 7 of ICU admission). There was a trend towards significance in fewer patients in the MICU receiving spontaneous breathing trial within 24 hours of ICU admission (11% vs 33%, P = 0.07) (Table 2).

Patient Outcomes

There were no significant differences in ICU mortality (MICU 19% vs CICU 10%, P = 0.18), or hospital mortality (21% vs 15%, P = 0.38) across the units (Table 3). Mean ICU and hospital length of stay (LOS) and proportion of patients discharged home with unassisted breathing were similar (Table 3).

Patient Outcomes for the 141 Patients Admitted to the Intensive Care Units With Sepsis During the Study Period
Patient OutcomesMICU (N = 100)CICU (N = 41)P Value
  • Abbreviations: CICU, cardiac intensive care unit; ICU, intensive care unit; MICU, medical intensive care unit; SD, standard deviation.

ICU mortality, n (%)19 (19)4 (10)0.18
Hospital mortality, n (%)21 (21)6 (15)0.38
Discharged home with unassisted breathing, n (%)33 (33)19 (46)0.14
ICU length of stay in days, mean SD4.78 6.244.92 6.320.97
Hospital length of stay in days, mean SD9.68 9.229.73 9.330.98

DISCUSSION

Since sepsis is more commonly treated in the medical ICU and some data suggests that specialty ICUs may be better at providing desired care,18, 19 we believed that patients treated in the MICU would be more likely to receive guideline‐concordant care. The study refutes our a priori hypothesis and reveals that evidence‐based processes‐of‐care associated with improved outcomes for sepsis are similarly implemented at our institution in the primary and overflow ICU. These findings are important, as ICU bed availability is a frequent problem and many hospitals overflow patients to non‐primary ICUs.9, 20

The observed equivalence in the care delivered may be a function of the relatively high number of patients with sepsis treated in the overflow unit, thereby giving the delivery teams enough experience to provide the desired care. An alternative explanation could be that the residents in CICU brought with them the experience from having previously trained in the MICU. Although, some of the care processes for sepsis patients are influenced by the CPOE (with embedded order sets and protocols), it is unlikely that CPOE can fully account for similarity in care because many processes and therapies (like use of steroids, amount of fluid delivered in first 24 hours, packed red blood cells [PRBC] transfusion, and spontaneous breathing trials) are not embedded within order sets.

The significant difference noted in the areas of deep vein thrombosis (DVT) and gastrointestinal (GI) prophylaxis within 24 hours of ICU admission was unexpected. These preventive therapies are included in initial order sets in the CPOE, which prompt physicians to order them as standard‐of‐care. With respect to DVT prophylaxis, we suspect that some of the difference might be attributable to specific contraindications to its use, which could have been more common in one of the units. There were more patients in MICU on mechanical ventilation (although not statistically significant) and with severe sepsis (statistically significant) at time of admission, which might have contributed to the difference noted in use of GI prophylaxis. It is also plausible that these differences might have disappeared if they were reassessed beyond 24 hours into the ICU admission. We cannot rule out the presence of unit‐ and physician‐level differences that contributed to this. Likewise, there was an unexpected trend towards significance, wherein more patients in CICU had spontaneous breathing trials within 24 hours of admission. This might also be explained by the higher number of patients with severe sepsis in the MICU (preempting any weaning attempts). These caveats aside, it is reassuring that, at our institution, admitting septic patients to the first available ICU bed does not adversely affect important processes‐of‐care.

One might ask whether this study's data should reassure other sites who are boarding septic patients in non‐primary ICUs. Irrespective of the number of patients studied or the degree of statistical significance of the associations, an observational study design cannot prove that boarding septic patients in non‐primary ICUs is either safe or unsafe. However, we hope that readers reflect on, and take inventory of, systems issues that may be different between unitswith an eye towards eliminating variation such that all units managing septic patients are primed to deliver guideline‐concordant care. Other hospitals that use CPOE with sepsis order sets, have protocols for sepsis care, and who train nursing and respiratory therapists to meet high standards might be pleased to see that the patients in our study received comparable, high‐quality care across the 2 units. While our data suggests that boarding patients in overflow units may be safe, these findings would need to be replicated at other sites using prospective designs to prove safety.

Length of emergency room stay prior to admission is associated with higher mortality rates.2123 At many hospitals, critical care beds are a scarce resource such that most hospitals have a policy for the triage of patients to critical care beds.24, 25 Lundberg and colleagues' study demonstrated that patients who developed septic shock on the medical wards experienced delays in receipt of intravenous fluids, inotropic agents and transfer to a critical care setting.26 Thus, rather than waiting in the ED or on the medical service for an MICU bed to become available, it may be most wise to admit a critically sick septic patient to the first available ICU bed, even to an overflow ICU. In a recent study by Sidlow and Aggarwal, 1104 patients discharged from the coronary care unit (CCU) with a non‐cardiac primary diagnosis were compared to patients admitted to the MICU in the same hospital.27 The study found no differences in patient mortality, 30‐day readmission rate, hospital LOS, ICU LOS, and safety outcomes of ventilator‐associated pneumonia and catheter‐associated bloodstream infections between ICUs. However, their study did not examine processes‐of‐care delivered between the primary ICU and the overflow unit, and did not validate the primary diagnoses of patients admitted to the ICU.

Several limitations of this study should be considered. First, this study was conducted at a single center. Second, we used a retrospective study design; however, a prospective study randomizing patients to 1 of the 2 units would likely never be possible. Third, the relatively small number of patients limited the power of the study to detect mortality differences between the units. However, this was a pilot study focused on processes of care as opposed to clinical outcomes. Fourth, it is possible that we did not capture every single patient with sepsis with our keyword search. Our use of a previously validated screening process should have limited the number of missed cases.15, 16 Fifth, although the 2 ICUs have exclusive nursing staff and attending physicians, the housestaff and respiratory therapists do rotate between the 2 ICUs and place orders in the common CPOE. The rotating housestaff may certainly represent a source for confounding, but the large numbers (>30) of evenly spread housestaff over the study period minimizes the potential for any trainee to be responsible for a large proportion of observed practice. Sixth, ICU attendings are the physicians of record and could influence the results. Because no attending physician was on service for more than 4 weeks during the study period, and patients were equally spread over this same time, concerns about clustering and biases this may have created should be minimal but cannot be ruled out. Seventh, some interventions and processes, such as antibiotic administration and measurement of lactate, may have been initiated in the ED, thereby decreasing the potential for differences between the groups. Additionally, we cannot rule out the possibility that factors other than bed availability drove the admission process (we found that the relative proportion of patients admitted to overflow ICU during hours of ambulance diversion was similar to the overflow ICU admissions during non‐ambulance diversion hours). It is possible that some selection bias by the hospitalist assigning patients to specific ICUs influenced their triage decisionsalthough all triaging doctors go through the same process of training in active bed management.11 While more patients admitted to the MICU had severe sepsis, there were no differences between groups in APACHE II or SOFA scores. However, we cannot rule out that there were other residual confounders. Finally, in a small number of cases (4/41, 10%), the CICU team consulted the MICU attending for assistance. This input had the potential to reduce disparities in care between the units.

Overflowing patients to non‐primary ICUs occurs in many hospitals. Our study demonstrates that sepsis treatment for overflow patients may be similar to that received in the primary ICU. While a large multicentered and randomized trial could determine whether significant management and outcome differences exist between primary and overflow ICUs, feasibility concerns make it unlikely that such a study will ever be conducted.

Acknowledgements

Disclosure: Dr Wright is a Miller‐Coulson Family Scholar and this work is supported by the Miller‐Coulson family through the Johns Hopkins Center for Innovative Medicine. Dr Sevransky was supported with a grant from National Institute of General Medical Sciences, NIGMS K‐23‐1399. All other authors disclose no relevant or financial conflicts of interest.

References
  1. Angus DC,Linde‐Zwirble WT,Lidicker J,Clermont G,Carcillo J,Pinsky MR.Epidemiology of severe sepsis in the United States: analysis of incidence, outcome, and associated costs of care.Crit Care Med.2001;29(7):13031310.
  2. Kumar G,Kumar N,Taneja A, et al;for the Milwaukee Initiative in Critical Care Outcomes Research (MICCOR) Group of Investigators.Nationwide trends of severe sepsis in the twenty first century (2000–2007).Chest.2011;140(5):12231231.
  3. Dombrovskiy VY,Martin AA,Sunderram J,Paz HL.Rapid increase in hospitalization and mortality rates for severe sepsis in the United States: a trend analysis from 1993 to 2003.Crit Care Med.2007;35(5):12441250.
  4. Dellinger RP,Levy MM,Carlet JM, et al.Surviving sepsis campaign: international guidelines for management of severe sepsis and septic shock: 2008.Crit Care Med.2008;36(1):296327.
  5. Jones AE,Shapiro NI,Trzeciak S, et al.Lactate clearance vs central venous oxygen saturation as goals of early sepsis therapy: a randomized clinical trial.JAMA.2010;303(8):739746.
  6. 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(19):13681377.
  7. Nguyen HB,Corbett SW,Steele R, et al.Implementation of a bundle of quality indicators for the early management of severe sepsis and septic shock is associated with decreased mortality.Crit Care Med.2007;35(4):11051112.
  8. Kumar A,Zarychanski R,Light B, et al.Early combination antibiotic therapy yields improved survival compared with monotherapy in septic shock: a propensity‐matched analysis.Crit Care Med.2010;38(9):17731785.
  9. Johannes MS.A new dimension of the PACU: the dilemma of the ICU overflow patient.J Post Anesth Nurs.1994;9(5):297300.
  10. Groeger JS,Strosberg MA,Halpern NA, et al.Descriptive analysis of critical care units in the United States.Crit Care Med.1992;20(6):846863.
  11. Howell E,Bessman E,Kravet S,Kolodner K,Marshall R,Wright S.Active bed management by hospitalists and emergency department throughput.Ann Intern Med.2008;149(11):804811.
  12. 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):16441655.
  13. 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):22942303.
  14. Castellanos‐Ortega A,Suberviola B,Garcia‐Astudillo LA, et al.Impact of the surviving sepsis campaign protocols on hospital length of stay and mortality in septic shock patients: results of a three‐year follow‐up quasi‐experimental study.Crit Care Med.2010;38(4):10361043.
  15. Needham DM,Dennison CR,Dowdy DW, et al.Study protocol: the improving care of acute lung injury patients (ICAP) study.Crit Care.2006;10(1):R9.
  16. Ali N,Gutteridge D,Shahul S,Checkley W,Sevransky J,Martin G.Critical illness outcome study: an observational study of protocols and mortality in intensive care units.Open Access J Clin Trials.2011;3(September):5565.
  17. Vincent JL,Moreno R,Takala J, et al.The SOFA (sepsis‐related organ failure assessment) score to describe organ dysfunction/failure: on behalf of the Working Group on Sepsis‐Related Problems of the European Society of Intensive Care Medicine.Intensive Care Med.1996;22(7):707710.
  18. Pronovost PJ,Angus DC,Dorman T,Robinson KA,Dremsizov TT,Young TL.Physician staffing patterns and clinical outcomes in critically ill patients: a systematic review.JAMA.2002;288(17):21512162.
  19. Fuchs RJ,Berenholtz SM,Dorman T.Do intensivists in ICU improve outcome?Best Pract Res Clin Anaesthesiol.2005;19(1):125135.
  20. Lindsay M.Is the postanesthesia care unit becoming an intensive care unit?J Perianesth Nurs.1999;14(2):7377.
  21. Chalfin DB,Trzeciak S,Likourezos A,Baumann BM,Dellinger RP;for the DELAY‐ED Study Group.Impact of delayed transfer of critically ill patients from the emergency department to the intensive care unit.Crit Care Med.2007;35(6):14771483.
  22. Renaud B,Santin A,Coma E, et al.Association between timing of intensive care unit admission and outcomes for emergency department patients with community‐acquired pneumonia.Crit Care Med.2009;37(11):28672874.
  23. Shen YC,Hsia RY.Association between ambulance diversion and survival among patients with acute myocardial infarction.JAMA.2011;305(23):24402447.
  24. Teres D.Civilian triage in the intensive care unit: the ritual of the last bed.Crit Care Med.1993;21(4):598606.
  25. Sinuff T,Kahnamoui K,Cook DJ,Luce JM,Levy MM;for the Values Ethics and Rationing in Critical Care Task Force.Rationing critical care beds: a systematic review.Crit Care Med.2004;32(7):15881597.
  26. Lundberg JS,Perl TM,Wiblin T, et al.Septic shock: an analysis of outcomes for patients with onset on hospital wards versus intensive care units.Crit Care Med.1998;26(6):10201024.
  27. Sidlow R,Aggarwal V.“The MICU is full”: one hospital's experience with an overflow triage policy.Jt Comm J Qual Patient Saf.2011;37(10):456460.
References
  1. Angus DC,Linde‐Zwirble WT,Lidicker J,Clermont G,Carcillo J,Pinsky MR.Epidemiology of severe sepsis in the United States: analysis of incidence, outcome, and associated costs of care.Crit Care Med.2001;29(7):13031310.
  2. Kumar G,Kumar N,Taneja A, et al;for the Milwaukee Initiative in Critical Care Outcomes Research (MICCOR) Group of Investigators.Nationwide trends of severe sepsis in the twenty first century (2000–2007).Chest.2011;140(5):12231231.
  3. Dombrovskiy VY,Martin AA,Sunderram J,Paz HL.Rapid increase in hospitalization and mortality rates for severe sepsis in the United States: a trend analysis from 1993 to 2003.Crit Care Med.2007;35(5):12441250.
  4. Dellinger RP,Levy MM,Carlet JM, et al.Surviving sepsis campaign: international guidelines for management of severe sepsis and septic shock: 2008.Crit Care Med.2008;36(1):296327.
  5. Jones AE,Shapiro NI,Trzeciak S, et al.Lactate clearance vs central venous oxygen saturation as goals of early sepsis therapy: a randomized clinical trial.JAMA.2010;303(8):739746.
  6. 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(19):13681377.
  7. Nguyen HB,Corbett SW,Steele R, et al.Implementation of a bundle of quality indicators for the early management of severe sepsis and septic shock is associated with decreased mortality.Crit Care Med.2007;35(4):11051112.
  8. Kumar A,Zarychanski R,Light B, et al.Early combination antibiotic therapy yields improved survival compared with monotherapy in septic shock: a propensity‐matched analysis.Crit Care Med.2010;38(9):17731785.
  9. Johannes MS.A new dimension of the PACU: the dilemma of the ICU overflow patient.J Post Anesth Nurs.1994;9(5):297300.
  10. Groeger JS,Strosberg MA,Halpern NA, et al.Descriptive analysis of critical care units in the United States.Crit Care Med.1992;20(6):846863.
  11. Howell E,Bessman E,Kravet S,Kolodner K,Marshall R,Wright S.Active bed management by hospitalists and emergency department throughput.Ann Intern Med.2008;149(11):804811.
  12. 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):16441655.
  13. 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):22942303.
  14. Castellanos‐Ortega A,Suberviola B,Garcia‐Astudillo LA, et al.Impact of the surviving sepsis campaign protocols on hospital length of stay and mortality in septic shock patients: results of a three‐year follow‐up quasi‐experimental study.Crit Care Med.2010;38(4):10361043.
  15. Needham DM,Dennison CR,Dowdy DW, et al.Study protocol: the improving care of acute lung injury patients (ICAP) study.Crit Care.2006;10(1):R9.
  16. Ali N,Gutteridge D,Shahul S,Checkley W,Sevransky J,Martin G.Critical illness outcome study: an observational study of protocols and mortality in intensive care units.Open Access J Clin Trials.2011;3(September):5565.
  17. Vincent JL,Moreno R,Takala J, et al.The SOFA (sepsis‐related organ failure assessment) score to describe organ dysfunction/failure: on behalf of the Working Group on Sepsis‐Related Problems of the European Society of Intensive Care Medicine.Intensive Care Med.1996;22(7):707710.
  18. Pronovost PJ,Angus DC,Dorman T,Robinson KA,Dremsizov TT,Young TL.Physician staffing patterns and clinical outcomes in critically ill patients: a systematic review.JAMA.2002;288(17):21512162.
  19. Fuchs RJ,Berenholtz SM,Dorman T.Do intensivists in ICU improve outcome?Best Pract Res Clin Anaesthesiol.2005;19(1):125135.
  20. Lindsay M.Is the postanesthesia care unit becoming an intensive care unit?J Perianesth Nurs.1999;14(2):7377.
  21. Chalfin DB,Trzeciak S,Likourezos A,Baumann BM,Dellinger RP;for the DELAY‐ED Study Group.Impact of delayed transfer of critically ill patients from the emergency department to the intensive care unit.Crit Care Med.2007;35(6):14771483.
  22. Renaud B,Santin A,Coma E, et al.Association between timing of intensive care unit admission and outcomes for emergency department patients with community‐acquired pneumonia.Crit Care Med.2009;37(11):28672874.
  23. Shen YC,Hsia RY.Association between ambulance diversion and survival among patients with acute myocardial infarction.JAMA.2011;305(23):24402447.
  24. Teres D.Civilian triage in the intensive care unit: the ritual of the last bed.Crit Care Med.1993;21(4):598606.
  25. Sinuff T,Kahnamoui K,Cook DJ,Luce JM,Levy MM;for the Values Ethics and Rationing in Critical Care Task Force.Rationing critical care beds: a systematic review.Crit Care Med.2004;32(7):15881597.
  26. Lundberg JS,Perl TM,Wiblin T, et al.Septic shock: an analysis of outcomes for patients with onset on hospital wards versus intensive care units.Crit Care Med.1998;26(6):10201024.
  27. Sidlow R,Aggarwal V.“The MICU is full”: one hospital's experience with an overflow triage policy.Jt Comm J Qual Patient Saf.2011;37(10):456460.
Issue
Journal of Hospital Medicine - 7(8)
Issue
Journal of Hospital Medicine - 7(8)
Page Number
600-605
Page Number
600-605
Publications
Publications
Article Type
Display Headline
Does sepsis treatment differ between primary and overflow intensive care units?
Display Headline
Does sepsis treatment differ between primary and overflow intensive care units?
Sections
Article Source

Copyright © 2012 Society of Hospital Medicine

Disallow All Ads
Correspondence Location
Johns Hopkins Bayview Medical Center, 5200 Eastern Ave, Baltimore, MD 21224
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Article PDF Media
Image
Disable zoom
Off
Media Files
Image
Disable zoom
Off