Affiliations
Department of Medicine, Denver Health Medical Center
Given name(s)
Rebecca
Family name
Allyn
Degrees
MD

Cognitive Biases Influence Decision-Making Regarding Postacute Care in a Skilled Nursing Facility

Article Type
Changed
Fri, 03/19/2021 - 15:31

The combination of decreasing hospital lengths of stay and increasing age and comorbidity of the United States population is a principal driver of the increased use of postacute care in the US.1-3 Postacute care refers to care in long-term acute care hospitals, inpatient rehabilitation facilities, skilled nursing facilities (SNFs), and care provided by home health agencies after an acute hospitalization. In 2016, 43% of Medicare beneficiaries received postacute care after hospital discharge at the cost of $60 billion annually; nearly half of these received care in an SNF.4 Increasing recognition of the significant cost and poor outcomes of postacute care led to payment reforms, such as bundled payments, that incentivized less expensive forms of postacute care and improvements in outcomes.5-9 Early evaluations suggested that hospitals are sensitive to these reforms and responded by significantly decreasing SNF utilization.10,11 It remains unclear whether this was safe and effective.

In this context, increased attention to how hospital clinicians and hospitalized patients decide whether to use postacute care (and what form to use) is appropriate since the effect of payment reforms could negatively impact vulnerable populations of older adults without adequate protection.12 Suboptimal decision-making can drive both overuse and inappropriate underuse of this expensive medical resource. Initial evidence suggests that patients and clinicians are poorly equipped to make high-quality decisions about postacute care, with significant deficits in both the decision-making process and content.13-16 While these gaps are important to address, they may only be part of the problem. The fields of cognitive psychology and behavioral economics have revealed new insights into decision-making, demonstrating that people deviate from rational decision-making in predictable ways, termed decision heuristics, or cognitive biases.17 This growing field of research suggests heuristics or biases play important roles in decision-making and determining behavior, particularly in situations where there may be little information provided and the patient is stressed, tired, and ill—precisely like deciding on postacute care.18 However, it is currently unknown whether cognitive biases are at play when making hospital discharge decisions.

We sought to identify the most salient heuristics or cognitive biases patients may utilize when making decisions about postacute care at the end of their hospitalization and ways clinicians may contribute to these biases. The overall goal was to derive insights for improving postacute care decision-making.

 

 

METHODS

Study Design

We conducted a secondary analysis on interviews with hospital and SNF clinicians as well as patients and their caregivers who were either leaving the hospital for an SNF or newly arrived in an SNF from the hospital to understand if cognitive biases were present and how they manifested themselves in a real-world clinical context.19 These interviews were part of a larger qualitative study that sought to understand how clinicians, patients, and their caregivers made decisions about postacute care, particularly related to SNFs.13,14 This study represents the analysis of all our interviews, specifically examining decision-making bias. Participating sites, clinical roles, and both patient and caregiver characteristics (Table 1) in our cohort have been previously described.13,14

jhm015010022_t1.jpg

Analysis

We used a team-based approach to framework analysis, which has been used in other decision-making studies14, including those measuring cognitive bias.20 A limitation in cognitive bias research is the lack of a standardized list or categorization of cognitive biases. We reviewed prior systematic17,21 and narrative reviews18,22, as well as prior studies describing examples of cognitive biases playing a role in decision-making about therapy20 to construct a list of possible cognitive biases to evaluate and narrow these a priori to potential biases relevant to the decision about postacute care based on our prior work (Table 2).

burke01510821e_t2.jpg

We applied this framework to analyze transcripts through an iterative process of deductive coding and reviewing across four reviewers (ML, RA, AL, CL) and a hospitalist physician with expertise leading qualitative studies (REB).

Intercoder consensus was built through team discussion by resolving points of disagreement.23 Consistency of coding was regularly checked by having more than one investigator code individual manuscripts and comparing coding, and discrepancies were resolved through team discussion. We triangulated the data (shared our preliminary results) using a larger study team, including an expert in behavioral economics (SRG), physicians at study sites (EC, RA), and an anthropologist with expertise in qualitative methods (CL). We did this to ensure credibility (to what extent the findings are credible or believable) and confirmability of findings (ensuring the findings are based on participant narratives rather than researcher biases).

RESULTS


We reviewed a total of 105 interviews with 25 hospital clinicians, 20 SNF clinicians, 21 patients and 14 caregivers in the hospital, and 15 patients and 10 caregivers in the SNF setting (Table 1). We found authority bias/halo effect; default/status quo bias, anchoring bias, and framing was commonly present in decision-making about postacute care in a SNF, whereas there were few if any examples of ambiguity aversion, availability heuristic, confirmation bias, optimism bias, or false consensus effect (Table 2).

Authority Bias/Halo Effect

While most patients deferred to their inpatient teams when it came to decision-making, this effect seemed to differ across VA and non-VA settings. Veterans expressed a higher degree of potential authority bias regarding the VA as an institution, whereas older adults in non-VA settings saw physicians as the authority figure making decisions in their best interests.

Veterans expressed confidence in the VA regarding both whether to go to a SNF and where to go:

 

 

“The VA wouldn’t license [an SNF] if they didn’t have a good reputation for care, cleanliness, things of that nature” (Veteran, VA CLC)

“I just knew the VA would have my best interests at heart” (Veteran, VA CLC)

Their caregivers expressed similar confidence:

“I’m not gonna decide [on whether the patient they care for goes to postacute care], like I told you, that’s totally up to the VA. I have trust and faith in them…so wherever they send him, that’s where he’s going” (Caregiver, VA hospital)

In some cases, this perspective was closer to the halo effect: a positive experience with the care provider or the care team led the decision-makers to believe that their recommendations about postacute care would be similarly positive.

“I think we were very trusting in the sense that whatever happened the last time around, he survived it…they took care of him…he got back home, and he started his life again, you know, so why would we question what they’re telling us to do? (Caregiver, VA hospital)

In contrast to Veterans, non-Veteran patients seemed to experience authority bias when it came to the inpatient team.

“Well, I’d like to know more about the PTs [Physical Therapists] there, but I assume since they were recommended, they will be good.” (Patient, University hospital)

This perspective was especially apparent when it came to physicians:

“The level of trust that they [patients] put in their doctor is gonna outweigh what anyone else would say.” (Clinical liaison, SNF)

“[In response to a question about influences on the decision to go to rehab] I don’t…that’s not my decision to make, that’s the doctor’s decision.” (Patient, University hospital)

“They said so…[the doctor] said I needed to go to rehab, so I guess I do because it’s the doctor’s decision.” (Patient, University hospital)

Default/Status quo Bias

In a related way, patients and caregivers with exposure to a SNF seemed to default to the same SNF with which they had previous experience. This bias seems to be primarily related to knowing what to expect.

“He thinks it’s [a particular SNF] the right place for him now…he was there before and he knew, again, it was the right place for him to be” (Caregiver, VA hospital)

“It’s the only one I’ve ever been in…but they have a lot of activities; you have a lot of freedom, staff was good” (Patient, VA hospital)

“I’ve been [to this SNF] before and I kind of know what the program involves…so it was kind of like going home, not, going home is the wrong way to put it…I mean coming here is like something I know, you know, I didn’t need anybody to explain it to me.” (Patient, VA hospital)

“Anybody that’s been to [SNF], that would be their choice to go back to, and I guess I must’ve liked it that first time because I asked to go back again.” (Patient, University hospital)

Anchoring Bias

While anchoring bias was less frequent, it came up in two domains: first, related to costs of care, and second, related to facility characteristics. Costs came up most frequently for Veterans who preferred to move their care to the VA for cost reasons, which appeared in these cases to overshadow other considerations:

 

 

“I kept emphasizing that the VA could do all the same things at a lot more reasonable price. The whole purpose of having the VA is for the Veteran, so that…we can get the healthcare that we need at a more reasonable [sic] or a reasonable price.” (Veteran, CLC)

“I think the CLC [VA SNF] is going to take care of her probably the same way any other facility of its type would, unless she were in a private facility, but you know, that costs a lot more money.” (Caregiver, VA hospital)

Patients occasionally had striking responses to particular characteristics of SNFs, regardless of whether this was a central feature or related to their rehabilitation:

“The social worker comes and talks to me about the nursing home where cats are running around, you know, to infect my leg or spin their little cat hairs into my lungs and make my asthma worse…I’m going to have to beg the nurses or the aides or the family or somebody to clean the cat…” (Veteran, VA hospital)

Framing

Framing was the strongest theme among clinician interviews in our sample. Clinicians most frequently described the SNF as a place where patients could recover function (a positive frame), explaining risks (eg, rehospitalization) associated with alternative postacute care options besides the SNF in great detail.

“Aside from explaining the benefits of going and…having that 24-hour care, having the therapies provided to them [the patients], talking about them getting stronger, phrasing it in such a way that patients sometimes are more agreeable, like not calling it a skilled nursing facility, calling it a rehab you know, for them to get physically stronger so they can be the most independent that they can once they do go home, and also explaining … we think that this would be the best plan to prevent them from coming back to the hospital, so those are some of the things that we’ll mention to patients to try and educate them and get them to be agreeable for placement.” (Social worker, University hospital)

Clinicians avoided negative associations with “nursing home” (even though all SNFs are nursing homes) and tended to use more positive frames such as “rehabilitation facility.”

“Use the word rehab….we definitely use the word rehab, to get more therapy, to go home; it’s not a, we really emphasize it’s not a nursing home, it’s not to go to stay forever.” (Physical therapist, safety-net hospital)

Clinicians used a frame of “safety” when discussing the SNF and used a frame of “risk” when discussing alternative postacute care options such as returning home. We did not find examples of clinicians discussing similar risks in going to a SNF even for risks, such as falling, which exist in both settings.

“I’ve talked to them primarily on an avenue of safety because I think people want and they value independence, they value making sure they can get home, but you know, a lot of the times they understand safety is, it can be a concern and outlining that our goal is to make sure that they’re safe and they stay home, and I tend to broach the subject saying that our therapists believe that they might not be safe at home in the moment, but they have potential goals to be safe later on if we continue therapy. I really highlight safety being the major driver of our discussion.” (Physician, VA hospital)

 

 

In some cases, framing was so overt that other risk-mitigating options (eg, home healthcare) are not discussed.

“I definitely tend to explain the ideal first. I’m not going to bring up home care when we really think somebody should go to rehab, however, once people say I don’t want to do that, I’m not going, then that’s when I’m like OK, well, let’s talk to the doctors, but we can see about other supports in the home.” (Social worker, VA hospital)

DISCUSSION

In a large sample of patients and their caregivers, as well as multidisciplinary clinicians at three different hospitals and three SNFs, we found authority bias/halo effect and framing biases were most common and seemed most impactful. Default/status quo bias and anchoring bias were also present in decision-making about a SNF. The combination of authority bias/halo effect and framing biases could synergistically interact to augment the likelihood of patients accepting a SNF for postacute care. Patients who had been to a SNF before seemed more likely to choose the SNF they had experienced previously even if they had no other postacute care experiences, and could be highly influenced by isolated characteristics of that facility (such as the physical environment or cost of care).

It is important to mention that cognitive biases do not necessarily have a negative impact: indeed, as Kahneman and Tversky point out, these are useful heuristics from “fast” thinking that are often effective.24 For example, clinicians may be trying to act in the best interests of the patient when framing the decision in terms of regaining function and averting loss of safety and independence. However, the evidence base regarding the outcomes of an SNF versus other postacute options is not robust, and this decision-making is complex. While this decision was most commonly framed in terms of rehabilitation and returning home, the fact that only about half of patients have returned to the community by 100 days4 was not discussed in any interview. In fact, initial evidence suggests replacing the SNF with home healthcare in patients with hip and knee arthroplasty may reduce costs without worsening clinical outcomes.6 However, across a broader population, SNFs significantly reduce 30-day readmissions when directly compared with home healthcare, but other clinical outcomes are similar.25 This evidence suggests that the “right” postacute care option for an individual patient is not clear, highlighting a key role biases may play in decision-making. Further, the nebulous concept of “safety” could introduce potential disparities related to social determinants of health.12 The observed inclination to accept an SNF with which the individual had prior experience may be influenced by the acceptability of this choice because of personal factors or prior research, even if it also represents a bias by limiting the consideration of current alternatives.

Our findings complement those of others in the literature which have also identified profound gaps in discharge decision-making among patients and clinicians,13-16,26-31 though to our knowledge the role of cognitive biases in these decisions has not been explored. This study also addresses gaps in the cognitive bias literature, including the need for real-world data rather than hypothetical vignettes,17 and evaluation of treatment and management decisions rather than diagnoses, which have been more commonly studied.21

These findings have implications for both individual clinicians and healthcare institutions. In the immediate term, these findings may serve as a call to discharging clinicians to modulate language and “debias” their conversations with patients about care after discharge.18,22 Shared decision-making requires an informed choice by patients based on their goals and values; framing a decision in a way that puts the clinician’s goals or values (eg, safety) ahead of patient values (eg, independence and autonomy) or limits disclosure (eg, a “rehab” is a nursing home) in the hope of influencing choice may be more consistent with framing bias and less with shared decision-making.14 Although controversy exists about the best way to “debias” oneself,32 self-awareness of bias is increasingly recognized across healthcare venues as critical to improving care for vulnerable populations.33 The use of data rather than vignettes may be a useful debiasing strategy, although the limitations of currently available data (eg, capturing nursing home quality) are increasingly recognized.34 From a policy and health system perspective, cognitive biases should be integrated into the development of decision aids to facilitate informed, shared, and high-quality decision-making that incorporates patient values, and perhaps “nudges” from behavioral economics to assist patients in choosing the right postdischarge care for them. Such nudges use principles of framing to influence care without restricting choice.35 As the science informing best practice regarding postacute care improves, identifying the “right” postdischarge care may become easier and recommendations more evidence-based.36

Strengths of the study include a large, diverse sample of patients, caregivers, and clinicians in both the hospital and SNF setting. Also, we used a team-based analysis with an experienced team and a deep knowledge of the data, including triangulation with clinicians to verify results. However, all hospitals and SNFs were located in a single metropolitan area, and responses may vary by region or population density. All three hospitals have housestaff teaching programs, and at the time of the interviews all three community SNFs were “five-star” facilities on the Nursing Home Compare website; results may be different at community hospitals or other SNFs. Hospitalists were the only physician group sampled in the hospital as they provide the majority of inpatient care to older adults; geriatricians, in particular, may have had different perspectives. Since we intended to explore whether cognitive biases were present overall, we did not evaluate whether cognitive biases differed by role or subgroup (by clinician type, patient, or caregiver), but this may be a promising area to explore in future work. Many cognitive biases have been described, and there are likely additional biases we did not identify. To confirm the generalizability of these findings, they should be studied in a larger, more generalizable sample of respondents in future work.

Cognitive biases play an important role in patient decision-making about postacute care, particularly regarding SNF care. As postacute care undergoes a transformation spurred by payment reforms, it is more important than ever to ensure that patients understand their choices at hospital discharge and can make a high-quality decision consistent with their goals.

 

 

References

1. Burke RE, Juarez-Colunga E, Levy C, Prochazka AV, Coleman EA, Ginde AA. Rise of post-acute care facilities as a discharge destination of US hospitalizations. JAMA Intern Med. 2015;175(2):295-296. https://doi.org/10.1001/jamainternmed.2014.6383.
2. Burke RE, Juarez-Colunga E, Levy C, Prochazka AV, Coleman EA, Ginde AA. Patient and hospitalization characteristics associated with increased postacute care facility discharges from US hospitals. Med Care. 2015;53(6):492-500. https://doi.org/10.1097/MLR.0000000000000359.
3. Werner RM, Konetzka RT. Trends in post-acute care use among medicare beneficiaries: 2000 to 2015. JAMA. 2018;319(15):1616-1617. https://doi.org/10.1001/jama.2018.2408.
4. Medicare Payment Advisory Commission June 2018 Report to Congress. http://www.medpac.gov/docs/default-source/reports/jun18_ch5_medpacreport_sec.pdf?sfvrsn=0. Accessed November 9, 2018.
5. Burke RE, Cumbler E, Coleman EA, Levy C. Post-acute care reform: implications and opportunities for hospitalists. J Hosp Med. 2017;12(1):46-51. https://doi.org/10.1002/jhm.2673.
6. Dummit LA, Kahvecioglu D, Marrufo G, et al. Association between hospital participation in a medicare bundled payment initiative and payments and quality outcomes for lower extremity joint replacement episodes. JAMA. 2016;316(12):1267-1278. https://doi.org/10.1001/jama.2016.12717.
7. Navathe AS, Troxel AB, Liao JM, et al. Cost of joint replacement using bundled payment models. JAMA Intern Med. 2017;177(2):214-222. https://doi.org/10.1001/jamainternmed.2016.8263.
8. Kennedy G, Lewis VA, Kundu S, Mousqués J, Colla CH. Accountable care organizations and post-acute care: a focus on preferred SNF networks. Med Care Res Rev MCRR. 2018;1077558718781117. https://doi.org/10.1177/1077558718781117.
9. Chandra A, Dalton MA, Holmes J. Large increases in spending on postacute care in Medicare point to the potential for cost savings in these settings. Health Aff Proj Hope. 2013;32(5):864-872. https://doi.org/10.1377/hlthaff.2012.1262.
10. McWilliams JM, Gilstrap LG, Stevenson DG, Chernew ME, Huskamp HA, Grabowski DC. Changes in postacute care in the Medicare shared savings program. JAMA Intern Med. 2017;177(4):518-526. https://doi.org/10.1001/jamainternmed.2016.9115.
11. Zhu JM, Patel V, Shea JA, Neuman MD, Werner RM. Hospitals using bundled payment report reducing skilled nursing facility use and improving care integration. Health Aff Proj Hope. 2018;37(8):1282-1289. https://doi.org/10.1377/hlthaff.2018.0257.
12. Burke RE, Ibrahim SA. Discharge destination and disparities in postoperative care. JAMA. 2018;319(16):1653-1654. https://doi.org/10.1001/jama.2017.21884.
13. Burke RE, Lawrence E, Ladebue A, et al. How hospital clinicians select patients for skilled nursing facilities. J Am Geriatr Soc. 2017;65(11):2466-2472. https://doi.org/10.1111/jgs.14954.
14. Burke RE, Jones J, Lawrence E, et al. Evaluating the quality of patient decision-making regarding post-acute care. J Gen Intern Med. 2018;33(5):678-684. https://doi.org/10.1007/s11606-017-4298-1.
15. Gadbois EA, Tyler DA, Mor V. Selecting a skilled nursing facility for postacute care: individual and family perspectives. J Am Geriatr Soc. 2017;65(11):2459-2465. https://doi.org/10.1111/jgs.14988.
16. Tyler DA, Gadbois EA, McHugh JP, Shield RR, Winblad U, Mor V. Patients are not given quality-of-care data about skilled nursing facilities when discharged from hospitals. Health Aff. 2017;36(8):1385-1391. https://doi.org/10.1377/hlthaff.2017.0155.
17. Blumenthal-Barby JS, Krieger H. Cognitive biases and heuristics in medical decision making: a critical review using a systematic search strategy. Med Decis Mak Int J Soc Med Decis Mak. 2015;35(4):539-557. https://doi.org/10.1177/0272989X14547740.
18. Croskerry P, Singhal G, Mamede S. Cognitive debiasing 1: origins of bias and theory of debiasing. BMJ Qual Saf. 2013;22 Suppl 2:ii58-ii64. https://doi.org/10.1136/bmjqs-2012-001712.
19. Hinds PS, Vogel RJ, Clarke-Steffen L. The possibilities and pitfalls of doing a secondary analysis of a qualitative data set. Qual Health Res. 1997;7(3):408-424. https://doi.org/10.1177/104973239700700306.
20. Magid M, Mcllvennan CK, Jones J, et al. Exploring cognitive bias in destination therapy left ventricular assist device decision making: a retrospective qualitative framework analysis. Am Heart J. 2016;180:64-73. https://doi.org/10.1016/j.ahj.2016.06.024.
21. Saposnik G, Redelmeier D, Ruff CC, Tobler PN. Cognitive biases associated with medical decisions: a systematic review. BMC Med Inform Decis Mak. 2016;16(1):138. https://doi.org/10.1186/s12911-016-0377-1.
22. Croskerry P, Singhal G, Mamede S. Cognitive debiasing 2: impediments to and strategies for change. BMJ Qual Saf. 2013;22 Suppl 2:ii65-ii72. https://doi.org/10.1136/bmjqs-2012-001713.
23. Bradley EH, Curry LA, Devers KJ. Qualitative data analysis for health services research: developing taxonomy, themes, and theory. Health Serv Res. 2007;42(4):1758-1772. https://doi.org/10.1111/j.1475-6773.2006.00684.x.
24. Thinking, Fast and Slow. Daniel Kahneman. Macmillan. US Macmillan. https://us.macmillan.com/thinkingfastandslow/danielkahneman/9780374533557. Accessed February 5, 2019.
25. Werner RM, Konetzka RT, Coe NB. Does type of post-acute care matter? The effect of hospital discharge to home with home health care versus to skilled nursing facility. JAMA Intern Med. In press.
26. Jones J, Lawrence E, Ladebue A, Leonard C, Ayele R, Burke RE. Nurses’ role in managing “The Fit” of older adults in skilled nursing facilities. J Gerontol Nurs. 2017;43(12):11-20. https://doi.org/10.3928/00989134-20171110-06.
27. Lawrence E, Casler J-J, Jones J, et al. Variability in skilled nursing facility screening and admission processes: implications for value-based purchasing. Health Care Manage Rev. 2018. https://doi.org/10.1097/HMR.0000000000000225.
28. Ayele R, Jones J, Ladebue A, et al. Perceived costs of care influence post-acute care choices by clinicians, patients, and caregivers. J Am Geriatr Soc. 2019. https://doi.org/10.1111/jgs.15768.
29. Sefcik JS, Nock RH, Flores EJ, et al. Patient preferences for information on post-acute care services. Res Gerontol Nurs. 2016;9(4):175-182. https://doi.org/10.3928/19404921-20160120-01.
30. Konetzka RT, Perraillon MC. Use of nursing home compare website appears limited by lack of awareness and initial mistrust of the data. Health Aff Proj Hope. 2016;35(4):706-713. https://doi.org/10.1377/hlthaff.2015.1377.
31. Schapira MM, Shea JA, Duey KA, Kleiman C, Werner RM. The nursing home compare report card: perceptions of residents and caregivers regarding quality ratings and nursing home choice. Health Serv Res. 2016;51 Suppl 2:1212-1228. https://doi.org/10.1111/1475-6773.12458.
32. Dhaliwal G. Premature closure? Not so fast. BMJ Qual Saf. 2017;26(2):87-89. https://doi.org/10.1136/bmjqs-2016-005267.
33. Masters C, Robinson D, Faulkner S, Patterson E, McIlraith T, Ansari A. Addressing biases in patient care with the 5Rs of cultural humility, a clinician coaching tool. J Gen Intern Med. 2019;34(4):627-630. https://doi.org/10.1007/s11606-018-4814-y.
34. Burke RE, Werner RM. Quality measurement and nursing homes: measuring what matters. BMJ Qual Saf. 2019;28(7);520-523. https://doi.org/10.1136/bmjqs-2019-009447.
35. Patel MS, Volpp KG, Asch DA. Nudge units to improve the delivery of health care. N Engl J Med. 2018;378(3):214-216. https://doi.org/10.1056/NEJMp1712984.
36. Jenq GY, Tinetti ME. Post–acute care: who belongs where? JAMA Intern Med. 2015;175(2):296-297. https://doi.org/10.1001/jamainternmed.2014.4298.

Article PDF
Author and Disclosure Information

1Center for Health Equity Research and Promotion (CHERP); Corporal Crescenz VA Medical Center, Philadelphia, Pennsylvania; 2Hospital Medicine Section, Division of General Internal Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania; 3Center of Innovation for Veteran-Centered and Value-Driven Care; Denver VA Medical Center, Aurora, Colorado; 4Division of Hospital Medicine, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado; 5Department of Medicine, Denver Health and Hospital Authority, Denver, Colorado.

Disclosures

Dr. Burke is funded by a VA HSR&D Career Development Award, Dr. Greysen is funded by NIA K23 (AG045338). The authors have no conflicts of interest relevant to the presented work. All views are those of the authors and not necessarily those of the US Department of Veterans Affairs.

Issue
Journal of Hospital Medicine 15(1)
Publications
Topics
Page Number
22-27. Published online first August 21, 2019
Sections
Author and Disclosure Information

1Center for Health Equity Research and Promotion (CHERP); Corporal Crescenz VA Medical Center, Philadelphia, Pennsylvania; 2Hospital Medicine Section, Division of General Internal Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania; 3Center of Innovation for Veteran-Centered and Value-Driven Care; Denver VA Medical Center, Aurora, Colorado; 4Division of Hospital Medicine, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado; 5Department of Medicine, Denver Health and Hospital Authority, Denver, Colorado.

Disclosures

Dr. Burke is funded by a VA HSR&D Career Development Award, Dr. Greysen is funded by NIA K23 (AG045338). The authors have no conflicts of interest relevant to the presented work. All views are those of the authors and not necessarily those of the US Department of Veterans Affairs.

Author and Disclosure Information

1Center for Health Equity Research and Promotion (CHERP); Corporal Crescenz VA Medical Center, Philadelphia, Pennsylvania; 2Hospital Medicine Section, Division of General Internal Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania; 3Center of Innovation for Veteran-Centered and Value-Driven Care; Denver VA Medical Center, Aurora, Colorado; 4Division of Hospital Medicine, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado; 5Department of Medicine, Denver Health and Hospital Authority, Denver, Colorado.

Disclosures

Dr. Burke is funded by a VA HSR&D Career Development Award, Dr. Greysen is funded by NIA K23 (AG045338). The authors have no conflicts of interest relevant to the presented work. All views are those of the authors and not necessarily those of the US Department of Veterans Affairs.

Article PDF
Article PDF
Related Articles

The combination of decreasing hospital lengths of stay and increasing age and comorbidity of the United States population is a principal driver of the increased use of postacute care in the US.1-3 Postacute care refers to care in long-term acute care hospitals, inpatient rehabilitation facilities, skilled nursing facilities (SNFs), and care provided by home health agencies after an acute hospitalization. In 2016, 43% of Medicare beneficiaries received postacute care after hospital discharge at the cost of $60 billion annually; nearly half of these received care in an SNF.4 Increasing recognition of the significant cost and poor outcomes of postacute care led to payment reforms, such as bundled payments, that incentivized less expensive forms of postacute care and improvements in outcomes.5-9 Early evaluations suggested that hospitals are sensitive to these reforms and responded by significantly decreasing SNF utilization.10,11 It remains unclear whether this was safe and effective.

In this context, increased attention to how hospital clinicians and hospitalized patients decide whether to use postacute care (and what form to use) is appropriate since the effect of payment reforms could negatively impact vulnerable populations of older adults without adequate protection.12 Suboptimal decision-making can drive both overuse and inappropriate underuse of this expensive medical resource. Initial evidence suggests that patients and clinicians are poorly equipped to make high-quality decisions about postacute care, with significant deficits in both the decision-making process and content.13-16 While these gaps are important to address, they may only be part of the problem. The fields of cognitive psychology and behavioral economics have revealed new insights into decision-making, demonstrating that people deviate from rational decision-making in predictable ways, termed decision heuristics, or cognitive biases.17 This growing field of research suggests heuristics or biases play important roles in decision-making and determining behavior, particularly in situations where there may be little information provided and the patient is stressed, tired, and ill—precisely like deciding on postacute care.18 However, it is currently unknown whether cognitive biases are at play when making hospital discharge decisions.

We sought to identify the most salient heuristics or cognitive biases patients may utilize when making decisions about postacute care at the end of their hospitalization and ways clinicians may contribute to these biases. The overall goal was to derive insights for improving postacute care decision-making.

 

 

METHODS

Study Design

We conducted a secondary analysis on interviews with hospital and SNF clinicians as well as patients and their caregivers who were either leaving the hospital for an SNF or newly arrived in an SNF from the hospital to understand if cognitive biases were present and how they manifested themselves in a real-world clinical context.19 These interviews were part of a larger qualitative study that sought to understand how clinicians, patients, and their caregivers made decisions about postacute care, particularly related to SNFs.13,14 This study represents the analysis of all our interviews, specifically examining decision-making bias. Participating sites, clinical roles, and both patient and caregiver characteristics (Table 1) in our cohort have been previously described.13,14

jhm015010022_t1.jpg

Analysis

We used a team-based approach to framework analysis, which has been used in other decision-making studies14, including those measuring cognitive bias.20 A limitation in cognitive bias research is the lack of a standardized list or categorization of cognitive biases. We reviewed prior systematic17,21 and narrative reviews18,22, as well as prior studies describing examples of cognitive biases playing a role in decision-making about therapy20 to construct a list of possible cognitive biases to evaluate and narrow these a priori to potential biases relevant to the decision about postacute care based on our prior work (Table 2).

burke01510821e_t2.jpg

We applied this framework to analyze transcripts through an iterative process of deductive coding and reviewing across four reviewers (ML, RA, AL, CL) and a hospitalist physician with expertise leading qualitative studies (REB).

Intercoder consensus was built through team discussion by resolving points of disagreement.23 Consistency of coding was regularly checked by having more than one investigator code individual manuscripts and comparing coding, and discrepancies were resolved through team discussion. We triangulated the data (shared our preliminary results) using a larger study team, including an expert in behavioral economics (SRG), physicians at study sites (EC, RA), and an anthropologist with expertise in qualitative methods (CL). We did this to ensure credibility (to what extent the findings are credible or believable) and confirmability of findings (ensuring the findings are based on participant narratives rather than researcher biases).

RESULTS


We reviewed a total of 105 interviews with 25 hospital clinicians, 20 SNF clinicians, 21 patients and 14 caregivers in the hospital, and 15 patients and 10 caregivers in the SNF setting (Table 1). We found authority bias/halo effect; default/status quo bias, anchoring bias, and framing was commonly present in decision-making about postacute care in a SNF, whereas there were few if any examples of ambiguity aversion, availability heuristic, confirmation bias, optimism bias, or false consensus effect (Table 2).

Authority Bias/Halo Effect

While most patients deferred to their inpatient teams when it came to decision-making, this effect seemed to differ across VA and non-VA settings. Veterans expressed a higher degree of potential authority bias regarding the VA as an institution, whereas older adults in non-VA settings saw physicians as the authority figure making decisions in their best interests.

Veterans expressed confidence in the VA regarding both whether to go to a SNF and where to go:

 

 

“The VA wouldn’t license [an SNF] if they didn’t have a good reputation for care, cleanliness, things of that nature” (Veteran, VA CLC)

“I just knew the VA would have my best interests at heart” (Veteran, VA CLC)

Their caregivers expressed similar confidence:

“I’m not gonna decide [on whether the patient they care for goes to postacute care], like I told you, that’s totally up to the VA. I have trust and faith in them…so wherever they send him, that’s where he’s going” (Caregiver, VA hospital)

In some cases, this perspective was closer to the halo effect: a positive experience with the care provider or the care team led the decision-makers to believe that their recommendations about postacute care would be similarly positive.

“I think we were very trusting in the sense that whatever happened the last time around, he survived it…they took care of him…he got back home, and he started his life again, you know, so why would we question what they’re telling us to do? (Caregiver, VA hospital)

In contrast to Veterans, non-Veteran patients seemed to experience authority bias when it came to the inpatient team.

“Well, I’d like to know more about the PTs [Physical Therapists] there, but I assume since they were recommended, they will be good.” (Patient, University hospital)

This perspective was especially apparent when it came to physicians:

“The level of trust that they [patients] put in their doctor is gonna outweigh what anyone else would say.” (Clinical liaison, SNF)

“[In response to a question about influences on the decision to go to rehab] I don’t…that’s not my decision to make, that’s the doctor’s decision.” (Patient, University hospital)

“They said so…[the doctor] said I needed to go to rehab, so I guess I do because it’s the doctor’s decision.” (Patient, University hospital)

Default/Status quo Bias

In a related way, patients and caregivers with exposure to a SNF seemed to default to the same SNF with which they had previous experience. This bias seems to be primarily related to knowing what to expect.

“He thinks it’s [a particular SNF] the right place for him now…he was there before and he knew, again, it was the right place for him to be” (Caregiver, VA hospital)

“It’s the only one I’ve ever been in…but they have a lot of activities; you have a lot of freedom, staff was good” (Patient, VA hospital)

“I’ve been [to this SNF] before and I kind of know what the program involves…so it was kind of like going home, not, going home is the wrong way to put it…I mean coming here is like something I know, you know, I didn’t need anybody to explain it to me.” (Patient, VA hospital)

“Anybody that’s been to [SNF], that would be their choice to go back to, and I guess I must’ve liked it that first time because I asked to go back again.” (Patient, University hospital)

Anchoring Bias

While anchoring bias was less frequent, it came up in two domains: first, related to costs of care, and second, related to facility characteristics. Costs came up most frequently for Veterans who preferred to move their care to the VA for cost reasons, which appeared in these cases to overshadow other considerations:

 

 

“I kept emphasizing that the VA could do all the same things at a lot more reasonable price. The whole purpose of having the VA is for the Veteran, so that…we can get the healthcare that we need at a more reasonable [sic] or a reasonable price.” (Veteran, CLC)

“I think the CLC [VA SNF] is going to take care of her probably the same way any other facility of its type would, unless she were in a private facility, but you know, that costs a lot more money.” (Caregiver, VA hospital)

Patients occasionally had striking responses to particular characteristics of SNFs, regardless of whether this was a central feature or related to their rehabilitation:

“The social worker comes and talks to me about the nursing home where cats are running around, you know, to infect my leg or spin their little cat hairs into my lungs and make my asthma worse…I’m going to have to beg the nurses or the aides or the family or somebody to clean the cat…” (Veteran, VA hospital)

Framing

Framing was the strongest theme among clinician interviews in our sample. Clinicians most frequently described the SNF as a place where patients could recover function (a positive frame), explaining risks (eg, rehospitalization) associated with alternative postacute care options besides the SNF in great detail.

“Aside from explaining the benefits of going and…having that 24-hour care, having the therapies provided to them [the patients], talking about them getting stronger, phrasing it in such a way that patients sometimes are more agreeable, like not calling it a skilled nursing facility, calling it a rehab you know, for them to get physically stronger so they can be the most independent that they can once they do go home, and also explaining … we think that this would be the best plan to prevent them from coming back to the hospital, so those are some of the things that we’ll mention to patients to try and educate them and get them to be agreeable for placement.” (Social worker, University hospital)

Clinicians avoided negative associations with “nursing home” (even though all SNFs are nursing homes) and tended to use more positive frames such as “rehabilitation facility.”

“Use the word rehab….we definitely use the word rehab, to get more therapy, to go home; it’s not a, we really emphasize it’s not a nursing home, it’s not to go to stay forever.” (Physical therapist, safety-net hospital)

Clinicians used a frame of “safety” when discussing the SNF and used a frame of “risk” when discussing alternative postacute care options such as returning home. We did not find examples of clinicians discussing similar risks in going to a SNF even for risks, such as falling, which exist in both settings.

“I’ve talked to them primarily on an avenue of safety because I think people want and they value independence, they value making sure they can get home, but you know, a lot of the times they understand safety is, it can be a concern and outlining that our goal is to make sure that they’re safe and they stay home, and I tend to broach the subject saying that our therapists believe that they might not be safe at home in the moment, but they have potential goals to be safe later on if we continue therapy. I really highlight safety being the major driver of our discussion.” (Physician, VA hospital)

 

 

In some cases, framing was so overt that other risk-mitigating options (eg, home healthcare) are not discussed.

“I definitely tend to explain the ideal first. I’m not going to bring up home care when we really think somebody should go to rehab, however, once people say I don’t want to do that, I’m not going, then that’s when I’m like OK, well, let’s talk to the doctors, but we can see about other supports in the home.” (Social worker, VA hospital)

DISCUSSION

In a large sample of patients and their caregivers, as well as multidisciplinary clinicians at three different hospitals and three SNFs, we found authority bias/halo effect and framing biases were most common and seemed most impactful. Default/status quo bias and anchoring bias were also present in decision-making about a SNF. The combination of authority bias/halo effect and framing biases could synergistically interact to augment the likelihood of patients accepting a SNF for postacute care. Patients who had been to a SNF before seemed more likely to choose the SNF they had experienced previously even if they had no other postacute care experiences, and could be highly influenced by isolated characteristics of that facility (such as the physical environment or cost of care).

It is important to mention that cognitive biases do not necessarily have a negative impact: indeed, as Kahneman and Tversky point out, these are useful heuristics from “fast” thinking that are often effective.24 For example, clinicians may be trying to act in the best interests of the patient when framing the decision in terms of regaining function and averting loss of safety and independence. However, the evidence base regarding the outcomes of an SNF versus other postacute options is not robust, and this decision-making is complex. While this decision was most commonly framed in terms of rehabilitation and returning home, the fact that only about half of patients have returned to the community by 100 days4 was not discussed in any interview. In fact, initial evidence suggests replacing the SNF with home healthcare in patients with hip and knee arthroplasty may reduce costs without worsening clinical outcomes.6 However, across a broader population, SNFs significantly reduce 30-day readmissions when directly compared with home healthcare, but other clinical outcomes are similar.25 This evidence suggests that the “right” postacute care option for an individual patient is not clear, highlighting a key role biases may play in decision-making. Further, the nebulous concept of “safety” could introduce potential disparities related to social determinants of health.12 The observed inclination to accept an SNF with which the individual had prior experience may be influenced by the acceptability of this choice because of personal factors or prior research, even if it also represents a bias by limiting the consideration of current alternatives.

Our findings complement those of others in the literature which have also identified profound gaps in discharge decision-making among patients and clinicians,13-16,26-31 though to our knowledge the role of cognitive biases in these decisions has not been explored. This study also addresses gaps in the cognitive bias literature, including the need for real-world data rather than hypothetical vignettes,17 and evaluation of treatment and management decisions rather than diagnoses, which have been more commonly studied.21

These findings have implications for both individual clinicians and healthcare institutions. In the immediate term, these findings may serve as a call to discharging clinicians to modulate language and “debias” their conversations with patients about care after discharge.18,22 Shared decision-making requires an informed choice by patients based on their goals and values; framing a decision in a way that puts the clinician’s goals or values (eg, safety) ahead of patient values (eg, independence and autonomy) or limits disclosure (eg, a “rehab” is a nursing home) in the hope of influencing choice may be more consistent with framing bias and less with shared decision-making.14 Although controversy exists about the best way to “debias” oneself,32 self-awareness of bias is increasingly recognized across healthcare venues as critical to improving care for vulnerable populations.33 The use of data rather than vignettes may be a useful debiasing strategy, although the limitations of currently available data (eg, capturing nursing home quality) are increasingly recognized.34 From a policy and health system perspective, cognitive biases should be integrated into the development of decision aids to facilitate informed, shared, and high-quality decision-making that incorporates patient values, and perhaps “nudges” from behavioral economics to assist patients in choosing the right postdischarge care for them. Such nudges use principles of framing to influence care without restricting choice.35 As the science informing best practice regarding postacute care improves, identifying the “right” postdischarge care may become easier and recommendations more evidence-based.36

Strengths of the study include a large, diverse sample of patients, caregivers, and clinicians in both the hospital and SNF setting. Also, we used a team-based analysis with an experienced team and a deep knowledge of the data, including triangulation with clinicians to verify results. However, all hospitals and SNFs were located in a single metropolitan area, and responses may vary by region or population density. All three hospitals have housestaff teaching programs, and at the time of the interviews all three community SNFs were “five-star” facilities on the Nursing Home Compare website; results may be different at community hospitals or other SNFs. Hospitalists were the only physician group sampled in the hospital as they provide the majority of inpatient care to older adults; geriatricians, in particular, may have had different perspectives. Since we intended to explore whether cognitive biases were present overall, we did not evaluate whether cognitive biases differed by role or subgroup (by clinician type, patient, or caregiver), but this may be a promising area to explore in future work. Many cognitive biases have been described, and there are likely additional biases we did not identify. To confirm the generalizability of these findings, they should be studied in a larger, more generalizable sample of respondents in future work.

Cognitive biases play an important role in patient decision-making about postacute care, particularly regarding SNF care. As postacute care undergoes a transformation spurred by payment reforms, it is more important than ever to ensure that patients understand their choices at hospital discharge and can make a high-quality decision consistent with their goals.

 

 

The combination of decreasing hospital lengths of stay and increasing age and comorbidity of the United States population is a principal driver of the increased use of postacute care in the US.1-3 Postacute care refers to care in long-term acute care hospitals, inpatient rehabilitation facilities, skilled nursing facilities (SNFs), and care provided by home health agencies after an acute hospitalization. In 2016, 43% of Medicare beneficiaries received postacute care after hospital discharge at the cost of $60 billion annually; nearly half of these received care in an SNF.4 Increasing recognition of the significant cost and poor outcomes of postacute care led to payment reforms, such as bundled payments, that incentivized less expensive forms of postacute care and improvements in outcomes.5-9 Early evaluations suggested that hospitals are sensitive to these reforms and responded by significantly decreasing SNF utilization.10,11 It remains unclear whether this was safe and effective.

In this context, increased attention to how hospital clinicians and hospitalized patients decide whether to use postacute care (and what form to use) is appropriate since the effect of payment reforms could negatively impact vulnerable populations of older adults without adequate protection.12 Suboptimal decision-making can drive both overuse and inappropriate underuse of this expensive medical resource. Initial evidence suggests that patients and clinicians are poorly equipped to make high-quality decisions about postacute care, with significant deficits in both the decision-making process and content.13-16 While these gaps are important to address, they may only be part of the problem. The fields of cognitive psychology and behavioral economics have revealed new insights into decision-making, demonstrating that people deviate from rational decision-making in predictable ways, termed decision heuristics, or cognitive biases.17 This growing field of research suggests heuristics or biases play important roles in decision-making and determining behavior, particularly in situations where there may be little information provided and the patient is stressed, tired, and ill—precisely like deciding on postacute care.18 However, it is currently unknown whether cognitive biases are at play when making hospital discharge decisions.

We sought to identify the most salient heuristics or cognitive biases patients may utilize when making decisions about postacute care at the end of their hospitalization and ways clinicians may contribute to these biases. The overall goal was to derive insights for improving postacute care decision-making.

 

 

METHODS

Study Design

We conducted a secondary analysis on interviews with hospital and SNF clinicians as well as patients and their caregivers who were either leaving the hospital for an SNF or newly arrived in an SNF from the hospital to understand if cognitive biases were present and how they manifested themselves in a real-world clinical context.19 These interviews were part of a larger qualitative study that sought to understand how clinicians, patients, and their caregivers made decisions about postacute care, particularly related to SNFs.13,14 This study represents the analysis of all our interviews, specifically examining decision-making bias. Participating sites, clinical roles, and both patient and caregiver characteristics (Table 1) in our cohort have been previously described.13,14

jhm015010022_t1.jpg

Analysis

We used a team-based approach to framework analysis, which has been used in other decision-making studies14, including those measuring cognitive bias.20 A limitation in cognitive bias research is the lack of a standardized list or categorization of cognitive biases. We reviewed prior systematic17,21 and narrative reviews18,22, as well as prior studies describing examples of cognitive biases playing a role in decision-making about therapy20 to construct a list of possible cognitive biases to evaluate and narrow these a priori to potential biases relevant to the decision about postacute care based on our prior work (Table 2).

burke01510821e_t2.jpg

We applied this framework to analyze transcripts through an iterative process of deductive coding and reviewing across four reviewers (ML, RA, AL, CL) and a hospitalist physician with expertise leading qualitative studies (REB).

Intercoder consensus was built through team discussion by resolving points of disagreement.23 Consistency of coding was regularly checked by having more than one investigator code individual manuscripts and comparing coding, and discrepancies were resolved through team discussion. We triangulated the data (shared our preliminary results) using a larger study team, including an expert in behavioral economics (SRG), physicians at study sites (EC, RA), and an anthropologist with expertise in qualitative methods (CL). We did this to ensure credibility (to what extent the findings are credible or believable) and confirmability of findings (ensuring the findings are based on participant narratives rather than researcher biases).

RESULTS


We reviewed a total of 105 interviews with 25 hospital clinicians, 20 SNF clinicians, 21 patients and 14 caregivers in the hospital, and 15 patients and 10 caregivers in the SNF setting (Table 1). We found authority bias/halo effect; default/status quo bias, anchoring bias, and framing was commonly present in decision-making about postacute care in a SNF, whereas there were few if any examples of ambiguity aversion, availability heuristic, confirmation bias, optimism bias, or false consensus effect (Table 2).

Authority Bias/Halo Effect

While most patients deferred to their inpatient teams when it came to decision-making, this effect seemed to differ across VA and non-VA settings. Veterans expressed a higher degree of potential authority bias regarding the VA as an institution, whereas older adults in non-VA settings saw physicians as the authority figure making decisions in their best interests.

Veterans expressed confidence in the VA regarding both whether to go to a SNF and where to go:

 

 

“The VA wouldn’t license [an SNF] if they didn’t have a good reputation for care, cleanliness, things of that nature” (Veteran, VA CLC)

“I just knew the VA would have my best interests at heart” (Veteran, VA CLC)

Their caregivers expressed similar confidence:

“I’m not gonna decide [on whether the patient they care for goes to postacute care], like I told you, that’s totally up to the VA. I have trust and faith in them…so wherever they send him, that’s where he’s going” (Caregiver, VA hospital)

In some cases, this perspective was closer to the halo effect: a positive experience with the care provider or the care team led the decision-makers to believe that their recommendations about postacute care would be similarly positive.

“I think we were very trusting in the sense that whatever happened the last time around, he survived it…they took care of him…he got back home, and he started his life again, you know, so why would we question what they’re telling us to do? (Caregiver, VA hospital)

In contrast to Veterans, non-Veteran patients seemed to experience authority bias when it came to the inpatient team.

“Well, I’d like to know more about the PTs [Physical Therapists] there, but I assume since they were recommended, they will be good.” (Patient, University hospital)

This perspective was especially apparent when it came to physicians:

“The level of trust that they [patients] put in their doctor is gonna outweigh what anyone else would say.” (Clinical liaison, SNF)

“[In response to a question about influences on the decision to go to rehab] I don’t…that’s not my decision to make, that’s the doctor’s decision.” (Patient, University hospital)

“They said so…[the doctor] said I needed to go to rehab, so I guess I do because it’s the doctor’s decision.” (Patient, University hospital)

Default/Status quo Bias

In a related way, patients and caregivers with exposure to a SNF seemed to default to the same SNF with which they had previous experience. This bias seems to be primarily related to knowing what to expect.

“He thinks it’s [a particular SNF] the right place for him now…he was there before and he knew, again, it was the right place for him to be” (Caregiver, VA hospital)

“It’s the only one I’ve ever been in…but they have a lot of activities; you have a lot of freedom, staff was good” (Patient, VA hospital)

“I’ve been [to this SNF] before and I kind of know what the program involves…so it was kind of like going home, not, going home is the wrong way to put it…I mean coming here is like something I know, you know, I didn’t need anybody to explain it to me.” (Patient, VA hospital)

“Anybody that’s been to [SNF], that would be their choice to go back to, and I guess I must’ve liked it that first time because I asked to go back again.” (Patient, University hospital)

Anchoring Bias

While anchoring bias was less frequent, it came up in two domains: first, related to costs of care, and second, related to facility characteristics. Costs came up most frequently for Veterans who preferred to move their care to the VA for cost reasons, which appeared in these cases to overshadow other considerations:

 

 

“I kept emphasizing that the VA could do all the same things at a lot more reasonable price. The whole purpose of having the VA is for the Veteran, so that…we can get the healthcare that we need at a more reasonable [sic] or a reasonable price.” (Veteran, CLC)

“I think the CLC [VA SNF] is going to take care of her probably the same way any other facility of its type would, unless she were in a private facility, but you know, that costs a lot more money.” (Caregiver, VA hospital)

Patients occasionally had striking responses to particular characteristics of SNFs, regardless of whether this was a central feature or related to their rehabilitation:

“The social worker comes and talks to me about the nursing home where cats are running around, you know, to infect my leg or spin their little cat hairs into my lungs and make my asthma worse…I’m going to have to beg the nurses or the aides or the family or somebody to clean the cat…” (Veteran, VA hospital)

Framing

Framing was the strongest theme among clinician interviews in our sample. Clinicians most frequently described the SNF as a place where patients could recover function (a positive frame), explaining risks (eg, rehospitalization) associated with alternative postacute care options besides the SNF in great detail.

“Aside from explaining the benefits of going and…having that 24-hour care, having the therapies provided to them [the patients], talking about them getting stronger, phrasing it in such a way that patients sometimes are more agreeable, like not calling it a skilled nursing facility, calling it a rehab you know, for them to get physically stronger so they can be the most independent that they can once they do go home, and also explaining … we think that this would be the best plan to prevent them from coming back to the hospital, so those are some of the things that we’ll mention to patients to try and educate them and get them to be agreeable for placement.” (Social worker, University hospital)

Clinicians avoided negative associations with “nursing home” (even though all SNFs are nursing homes) and tended to use more positive frames such as “rehabilitation facility.”

“Use the word rehab….we definitely use the word rehab, to get more therapy, to go home; it’s not a, we really emphasize it’s not a nursing home, it’s not to go to stay forever.” (Physical therapist, safety-net hospital)

Clinicians used a frame of “safety” when discussing the SNF and used a frame of “risk” when discussing alternative postacute care options such as returning home. We did not find examples of clinicians discussing similar risks in going to a SNF even for risks, such as falling, which exist in both settings.

“I’ve talked to them primarily on an avenue of safety because I think people want and they value independence, they value making sure they can get home, but you know, a lot of the times they understand safety is, it can be a concern and outlining that our goal is to make sure that they’re safe and they stay home, and I tend to broach the subject saying that our therapists believe that they might not be safe at home in the moment, but they have potential goals to be safe later on if we continue therapy. I really highlight safety being the major driver of our discussion.” (Physician, VA hospital)

 

 

In some cases, framing was so overt that other risk-mitigating options (eg, home healthcare) are not discussed.

“I definitely tend to explain the ideal first. I’m not going to bring up home care when we really think somebody should go to rehab, however, once people say I don’t want to do that, I’m not going, then that’s when I’m like OK, well, let’s talk to the doctors, but we can see about other supports in the home.” (Social worker, VA hospital)

DISCUSSION

In a large sample of patients and their caregivers, as well as multidisciplinary clinicians at three different hospitals and three SNFs, we found authority bias/halo effect and framing biases were most common and seemed most impactful. Default/status quo bias and anchoring bias were also present in decision-making about a SNF. The combination of authority bias/halo effect and framing biases could synergistically interact to augment the likelihood of patients accepting a SNF for postacute care. Patients who had been to a SNF before seemed more likely to choose the SNF they had experienced previously even if they had no other postacute care experiences, and could be highly influenced by isolated characteristics of that facility (such as the physical environment or cost of care).

It is important to mention that cognitive biases do not necessarily have a negative impact: indeed, as Kahneman and Tversky point out, these are useful heuristics from “fast” thinking that are often effective.24 For example, clinicians may be trying to act in the best interests of the patient when framing the decision in terms of regaining function and averting loss of safety and independence. However, the evidence base regarding the outcomes of an SNF versus other postacute options is not robust, and this decision-making is complex. While this decision was most commonly framed in terms of rehabilitation and returning home, the fact that only about half of patients have returned to the community by 100 days4 was not discussed in any interview. In fact, initial evidence suggests replacing the SNF with home healthcare in patients with hip and knee arthroplasty may reduce costs without worsening clinical outcomes.6 However, across a broader population, SNFs significantly reduce 30-day readmissions when directly compared with home healthcare, but other clinical outcomes are similar.25 This evidence suggests that the “right” postacute care option for an individual patient is not clear, highlighting a key role biases may play in decision-making. Further, the nebulous concept of “safety” could introduce potential disparities related to social determinants of health.12 The observed inclination to accept an SNF with which the individual had prior experience may be influenced by the acceptability of this choice because of personal factors or prior research, even if it also represents a bias by limiting the consideration of current alternatives.

Our findings complement those of others in the literature which have also identified profound gaps in discharge decision-making among patients and clinicians,13-16,26-31 though to our knowledge the role of cognitive biases in these decisions has not been explored. This study also addresses gaps in the cognitive bias literature, including the need for real-world data rather than hypothetical vignettes,17 and evaluation of treatment and management decisions rather than diagnoses, which have been more commonly studied.21

These findings have implications for both individual clinicians and healthcare institutions. In the immediate term, these findings may serve as a call to discharging clinicians to modulate language and “debias” their conversations with patients about care after discharge.18,22 Shared decision-making requires an informed choice by patients based on their goals and values; framing a decision in a way that puts the clinician’s goals or values (eg, safety) ahead of patient values (eg, independence and autonomy) or limits disclosure (eg, a “rehab” is a nursing home) in the hope of influencing choice may be more consistent with framing bias and less with shared decision-making.14 Although controversy exists about the best way to “debias” oneself,32 self-awareness of bias is increasingly recognized across healthcare venues as critical to improving care for vulnerable populations.33 The use of data rather than vignettes may be a useful debiasing strategy, although the limitations of currently available data (eg, capturing nursing home quality) are increasingly recognized.34 From a policy and health system perspective, cognitive biases should be integrated into the development of decision aids to facilitate informed, shared, and high-quality decision-making that incorporates patient values, and perhaps “nudges” from behavioral economics to assist patients in choosing the right postdischarge care for them. Such nudges use principles of framing to influence care without restricting choice.35 As the science informing best practice regarding postacute care improves, identifying the “right” postdischarge care may become easier and recommendations more evidence-based.36

Strengths of the study include a large, diverse sample of patients, caregivers, and clinicians in both the hospital and SNF setting. Also, we used a team-based analysis with an experienced team and a deep knowledge of the data, including triangulation with clinicians to verify results. However, all hospitals and SNFs were located in a single metropolitan area, and responses may vary by region or population density. All three hospitals have housestaff teaching programs, and at the time of the interviews all three community SNFs were “five-star” facilities on the Nursing Home Compare website; results may be different at community hospitals or other SNFs. Hospitalists were the only physician group sampled in the hospital as they provide the majority of inpatient care to older adults; geriatricians, in particular, may have had different perspectives. Since we intended to explore whether cognitive biases were present overall, we did not evaluate whether cognitive biases differed by role or subgroup (by clinician type, patient, or caregiver), but this may be a promising area to explore in future work. Many cognitive biases have been described, and there are likely additional biases we did not identify. To confirm the generalizability of these findings, they should be studied in a larger, more generalizable sample of respondents in future work.

Cognitive biases play an important role in patient decision-making about postacute care, particularly regarding SNF care. As postacute care undergoes a transformation spurred by payment reforms, it is more important than ever to ensure that patients understand their choices at hospital discharge and can make a high-quality decision consistent with their goals.

 

 

References

1. Burke RE, Juarez-Colunga E, Levy C, Prochazka AV, Coleman EA, Ginde AA. Rise of post-acute care facilities as a discharge destination of US hospitalizations. JAMA Intern Med. 2015;175(2):295-296. https://doi.org/10.1001/jamainternmed.2014.6383.
2. Burke RE, Juarez-Colunga E, Levy C, Prochazka AV, Coleman EA, Ginde AA. Patient and hospitalization characteristics associated with increased postacute care facility discharges from US hospitals. Med Care. 2015;53(6):492-500. https://doi.org/10.1097/MLR.0000000000000359.
3. Werner RM, Konetzka RT. Trends in post-acute care use among medicare beneficiaries: 2000 to 2015. JAMA. 2018;319(15):1616-1617. https://doi.org/10.1001/jama.2018.2408.
4. Medicare Payment Advisory Commission June 2018 Report to Congress. http://www.medpac.gov/docs/default-source/reports/jun18_ch5_medpacreport_sec.pdf?sfvrsn=0. Accessed November 9, 2018.
5. Burke RE, Cumbler E, Coleman EA, Levy C. Post-acute care reform: implications and opportunities for hospitalists. J Hosp Med. 2017;12(1):46-51. https://doi.org/10.1002/jhm.2673.
6. Dummit LA, Kahvecioglu D, Marrufo G, et al. Association between hospital participation in a medicare bundled payment initiative and payments and quality outcomes for lower extremity joint replacement episodes. JAMA. 2016;316(12):1267-1278. https://doi.org/10.1001/jama.2016.12717.
7. Navathe AS, Troxel AB, Liao JM, et al. Cost of joint replacement using bundled payment models. JAMA Intern Med. 2017;177(2):214-222. https://doi.org/10.1001/jamainternmed.2016.8263.
8. Kennedy G, Lewis VA, Kundu S, Mousqués J, Colla CH. Accountable care organizations and post-acute care: a focus on preferred SNF networks. Med Care Res Rev MCRR. 2018;1077558718781117. https://doi.org/10.1177/1077558718781117.
9. Chandra A, Dalton MA, Holmes J. Large increases in spending on postacute care in Medicare point to the potential for cost savings in these settings. Health Aff Proj Hope. 2013;32(5):864-872. https://doi.org/10.1377/hlthaff.2012.1262.
10. McWilliams JM, Gilstrap LG, Stevenson DG, Chernew ME, Huskamp HA, Grabowski DC. Changes in postacute care in the Medicare shared savings program. JAMA Intern Med. 2017;177(4):518-526. https://doi.org/10.1001/jamainternmed.2016.9115.
11. Zhu JM, Patel V, Shea JA, Neuman MD, Werner RM. Hospitals using bundled payment report reducing skilled nursing facility use and improving care integration. Health Aff Proj Hope. 2018;37(8):1282-1289. https://doi.org/10.1377/hlthaff.2018.0257.
12. Burke RE, Ibrahim SA. Discharge destination and disparities in postoperative care. JAMA. 2018;319(16):1653-1654. https://doi.org/10.1001/jama.2017.21884.
13. Burke RE, Lawrence E, Ladebue A, et al. How hospital clinicians select patients for skilled nursing facilities. J Am Geriatr Soc. 2017;65(11):2466-2472. https://doi.org/10.1111/jgs.14954.
14. Burke RE, Jones J, Lawrence E, et al. Evaluating the quality of patient decision-making regarding post-acute care. J Gen Intern Med. 2018;33(5):678-684. https://doi.org/10.1007/s11606-017-4298-1.
15. Gadbois EA, Tyler DA, Mor V. Selecting a skilled nursing facility for postacute care: individual and family perspectives. J Am Geriatr Soc. 2017;65(11):2459-2465. https://doi.org/10.1111/jgs.14988.
16. Tyler DA, Gadbois EA, McHugh JP, Shield RR, Winblad U, Mor V. Patients are not given quality-of-care data about skilled nursing facilities when discharged from hospitals. Health Aff. 2017;36(8):1385-1391. https://doi.org/10.1377/hlthaff.2017.0155.
17. Blumenthal-Barby JS, Krieger H. Cognitive biases and heuristics in medical decision making: a critical review using a systematic search strategy. Med Decis Mak Int J Soc Med Decis Mak. 2015;35(4):539-557. https://doi.org/10.1177/0272989X14547740.
18. Croskerry P, Singhal G, Mamede S. Cognitive debiasing 1: origins of bias and theory of debiasing. BMJ Qual Saf. 2013;22 Suppl 2:ii58-ii64. https://doi.org/10.1136/bmjqs-2012-001712.
19. Hinds PS, Vogel RJ, Clarke-Steffen L. The possibilities and pitfalls of doing a secondary analysis of a qualitative data set. Qual Health Res. 1997;7(3):408-424. https://doi.org/10.1177/104973239700700306.
20. Magid M, Mcllvennan CK, Jones J, et al. Exploring cognitive bias in destination therapy left ventricular assist device decision making: a retrospective qualitative framework analysis. Am Heart J. 2016;180:64-73. https://doi.org/10.1016/j.ahj.2016.06.024.
21. Saposnik G, Redelmeier D, Ruff CC, Tobler PN. Cognitive biases associated with medical decisions: a systematic review. BMC Med Inform Decis Mak. 2016;16(1):138. https://doi.org/10.1186/s12911-016-0377-1.
22. Croskerry P, Singhal G, Mamede S. Cognitive debiasing 2: impediments to and strategies for change. BMJ Qual Saf. 2013;22 Suppl 2:ii65-ii72. https://doi.org/10.1136/bmjqs-2012-001713.
23. Bradley EH, Curry LA, Devers KJ. Qualitative data analysis for health services research: developing taxonomy, themes, and theory. Health Serv Res. 2007;42(4):1758-1772. https://doi.org/10.1111/j.1475-6773.2006.00684.x.
24. Thinking, Fast and Slow. Daniel Kahneman. Macmillan. US Macmillan. https://us.macmillan.com/thinkingfastandslow/danielkahneman/9780374533557. Accessed February 5, 2019.
25. Werner RM, Konetzka RT, Coe NB. Does type of post-acute care matter? The effect of hospital discharge to home with home health care versus to skilled nursing facility. JAMA Intern Med. In press.
26. Jones J, Lawrence E, Ladebue A, Leonard C, Ayele R, Burke RE. Nurses’ role in managing “The Fit” of older adults in skilled nursing facilities. J Gerontol Nurs. 2017;43(12):11-20. https://doi.org/10.3928/00989134-20171110-06.
27. Lawrence E, Casler J-J, Jones J, et al. Variability in skilled nursing facility screening and admission processes: implications for value-based purchasing. Health Care Manage Rev. 2018. https://doi.org/10.1097/HMR.0000000000000225.
28. Ayele R, Jones J, Ladebue A, et al. Perceived costs of care influence post-acute care choices by clinicians, patients, and caregivers. J Am Geriatr Soc. 2019. https://doi.org/10.1111/jgs.15768.
29. Sefcik JS, Nock RH, Flores EJ, et al. Patient preferences for information on post-acute care services. Res Gerontol Nurs. 2016;9(4):175-182. https://doi.org/10.3928/19404921-20160120-01.
30. Konetzka RT, Perraillon MC. Use of nursing home compare website appears limited by lack of awareness and initial mistrust of the data. Health Aff Proj Hope. 2016;35(4):706-713. https://doi.org/10.1377/hlthaff.2015.1377.
31. Schapira MM, Shea JA, Duey KA, Kleiman C, Werner RM. The nursing home compare report card: perceptions of residents and caregivers regarding quality ratings and nursing home choice. Health Serv Res. 2016;51 Suppl 2:1212-1228. https://doi.org/10.1111/1475-6773.12458.
32. Dhaliwal G. Premature closure? Not so fast. BMJ Qual Saf. 2017;26(2):87-89. https://doi.org/10.1136/bmjqs-2016-005267.
33. Masters C, Robinson D, Faulkner S, Patterson E, McIlraith T, Ansari A. Addressing biases in patient care with the 5Rs of cultural humility, a clinician coaching tool. J Gen Intern Med. 2019;34(4):627-630. https://doi.org/10.1007/s11606-018-4814-y.
34. Burke RE, Werner RM. Quality measurement and nursing homes: measuring what matters. BMJ Qual Saf. 2019;28(7);520-523. https://doi.org/10.1136/bmjqs-2019-009447.
35. Patel MS, Volpp KG, Asch DA. Nudge units to improve the delivery of health care. N Engl J Med. 2018;378(3):214-216. https://doi.org/10.1056/NEJMp1712984.
36. Jenq GY, Tinetti ME. Post–acute care: who belongs where? JAMA Intern Med. 2015;175(2):296-297. https://doi.org/10.1001/jamainternmed.2014.4298.

References

1. Burke RE, Juarez-Colunga E, Levy C, Prochazka AV, Coleman EA, Ginde AA. Rise of post-acute care facilities as a discharge destination of US hospitalizations. JAMA Intern Med. 2015;175(2):295-296. https://doi.org/10.1001/jamainternmed.2014.6383.
2. Burke RE, Juarez-Colunga E, Levy C, Prochazka AV, Coleman EA, Ginde AA. Patient and hospitalization characteristics associated with increased postacute care facility discharges from US hospitals. Med Care. 2015;53(6):492-500. https://doi.org/10.1097/MLR.0000000000000359.
3. Werner RM, Konetzka RT. Trends in post-acute care use among medicare beneficiaries: 2000 to 2015. JAMA. 2018;319(15):1616-1617. https://doi.org/10.1001/jama.2018.2408.
4. Medicare Payment Advisory Commission June 2018 Report to Congress. http://www.medpac.gov/docs/default-source/reports/jun18_ch5_medpacreport_sec.pdf?sfvrsn=0. Accessed November 9, 2018.
5. Burke RE, Cumbler E, Coleman EA, Levy C. Post-acute care reform: implications and opportunities for hospitalists. J Hosp Med. 2017;12(1):46-51. https://doi.org/10.1002/jhm.2673.
6. Dummit LA, Kahvecioglu D, Marrufo G, et al. Association between hospital participation in a medicare bundled payment initiative and payments and quality outcomes for lower extremity joint replacement episodes. JAMA. 2016;316(12):1267-1278. https://doi.org/10.1001/jama.2016.12717.
7. Navathe AS, Troxel AB, Liao JM, et al. Cost of joint replacement using bundled payment models. JAMA Intern Med. 2017;177(2):214-222. https://doi.org/10.1001/jamainternmed.2016.8263.
8. Kennedy G, Lewis VA, Kundu S, Mousqués J, Colla CH. Accountable care organizations and post-acute care: a focus on preferred SNF networks. Med Care Res Rev MCRR. 2018;1077558718781117. https://doi.org/10.1177/1077558718781117.
9. Chandra A, Dalton MA, Holmes J. Large increases in spending on postacute care in Medicare point to the potential for cost savings in these settings. Health Aff Proj Hope. 2013;32(5):864-872. https://doi.org/10.1377/hlthaff.2012.1262.
10. McWilliams JM, Gilstrap LG, Stevenson DG, Chernew ME, Huskamp HA, Grabowski DC. Changes in postacute care in the Medicare shared savings program. JAMA Intern Med. 2017;177(4):518-526. https://doi.org/10.1001/jamainternmed.2016.9115.
11. Zhu JM, Patel V, Shea JA, Neuman MD, Werner RM. Hospitals using bundled payment report reducing skilled nursing facility use and improving care integration. Health Aff Proj Hope. 2018;37(8):1282-1289. https://doi.org/10.1377/hlthaff.2018.0257.
12. Burke RE, Ibrahim SA. Discharge destination and disparities in postoperative care. JAMA. 2018;319(16):1653-1654. https://doi.org/10.1001/jama.2017.21884.
13. Burke RE, Lawrence E, Ladebue A, et al. How hospital clinicians select patients for skilled nursing facilities. J Am Geriatr Soc. 2017;65(11):2466-2472. https://doi.org/10.1111/jgs.14954.
14. Burke RE, Jones J, Lawrence E, et al. Evaluating the quality of patient decision-making regarding post-acute care. J Gen Intern Med. 2018;33(5):678-684. https://doi.org/10.1007/s11606-017-4298-1.
15. Gadbois EA, Tyler DA, Mor V. Selecting a skilled nursing facility for postacute care: individual and family perspectives. J Am Geriatr Soc. 2017;65(11):2459-2465. https://doi.org/10.1111/jgs.14988.
16. Tyler DA, Gadbois EA, McHugh JP, Shield RR, Winblad U, Mor V. Patients are not given quality-of-care data about skilled nursing facilities when discharged from hospitals. Health Aff. 2017;36(8):1385-1391. https://doi.org/10.1377/hlthaff.2017.0155.
17. Blumenthal-Barby JS, Krieger H. Cognitive biases and heuristics in medical decision making: a critical review using a systematic search strategy. Med Decis Mak Int J Soc Med Decis Mak. 2015;35(4):539-557. https://doi.org/10.1177/0272989X14547740.
18. Croskerry P, Singhal G, Mamede S. Cognitive debiasing 1: origins of bias and theory of debiasing. BMJ Qual Saf. 2013;22 Suppl 2:ii58-ii64. https://doi.org/10.1136/bmjqs-2012-001712.
19. Hinds PS, Vogel RJ, Clarke-Steffen L. The possibilities and pitfalls of doing a secondary analysis of a qualitative data set. Qual Health Res. 1997;7(3):408-424. https://doi.org/10.1177/104973239700700306.
20. Magid M, Mcllvennan CK, Jones J, et al. Exploring cognitive bias in destination therapy left ventricular assist device decision making: a retrospective qualitative framework analysis. Am Heart J. 2016;180:64-73. https://doi.org/10.1016/j.ahj.2016.06.024.
21. Saposnik G, Redelmeier D, Ruff CC, Tobler PN. Cognitive biases associated with medical decisions: a systematic review. BMC Med Inform Decis Mak. 2016;16(1):138. https://doi.org/10.1186/s12911-016-0377-1.
22. Croskerry P, Singhal G, Mamede S. Cognitive debiasing 2: impediments to and strategies for change. BMJ Qual Saf. 2013;22 Suppl 2:ii65-ii72. https://doi.org/10.1136/bmjqs-2012-001713.
23. Bradley EH, Curry LA, Devers KJ. Qualitative data analysis for health services research: developing taxonomy, themes, and theory. Health Serv Res. 2007;42(4):1758-1772. https://doi.org/10.1111/j.1475-6773.2006.00684.x.
24. Thinking, Fast and Slow. Daniel Kahneman. Macmillan. US Macmillan. https://us.macmillan.com/thinkingfastandslow/danielkahneman/9780374533557. Accessed February 5, 2019.
25. Werner RM, Konetzka RT, Coe NB. Does type of post-acute care matter? The effect of hospital discharge to home with home health care versus to skilled nursing facility. JAMA Intern Med. In press.
26. Jones J, Lawrence E, Ladebue A, Leonard C, Ayele R, Burke RE. Nurses’ role in managing “The Fit” of older adults in skilled nursing facilities. J Gerontol Nurs. 2017;43(12):11-20. https://doi.org/10.3928/00989134-20171110-06.
27. Lawrence E, Casler J-J, Jones J, et al. Variability in skilled nursing facility screening and admission processes: implications for value-based purchasing. Health Care Manage Rev. 2018. https://doi.org/10.1097/HMR.0000000000000225.
28. Ayele R, Jones J, Ladebue A, et al. Perceived costs of care influence post-acute care choices by clinicians, patients, and caregivers. J Am Geriatr Soc. 2019. https://doi.org/10.1111/jgs.15768.
29. Sefcik JS, Nock RH, Flores EJ, et al. Patient preferences for information on post-acute care services. Res Gerontol Nurs. 2016;9(4):175-182. https://doi.org/10.3928/19404921-20160120-01.
30. Konetzka RT, Perraillon MC. Use of nursing home compare website appears limited by lack of awareness and initial mistrust of the data. Health Aff Proj Hope. 2016;35(4):706-713. https://doi.org/10.1377/hlthaff.2015.1377.
31. Schapira MM, Shea JA, Duey KA, Kleiman C, Werner RM. The nursing home compare report card: perceptions of residents and caregivers regarding quality ratings and nursing home choice. Health Serv Res. 2016;51 Suppl 2:1212-1228. https://doi.org/10.1111/1475-6773.12458.
32. Dhaliwal G. Premature closure? Not so fast. BMJ Qual Saf. 2017;26(2):87-89. https://doi.org/10.1136/bmjqs-2016-005267.
33. Masters C, Robinson D, Faulkner S, Patterson E, McIlraith T, Ansari A. Addressing biases in patient care with the 5Rs of cultural humility, a clinician coaching tool. J Gen Intern Med. 2019;34(4):627-630. https://doi.org/10.1007/s11606-018-4814-y.
34. Burke RE, Werner RM. Quality measurement and nursing homes: measuring what matters. BMJ Qual Saf. 2019;28(7);520-523. https://doi.org/10.1136/bmjqs-2019-009447.
35. Patel MS, Volpp KG, Asch DA. Nudge units to improve the delivery of health care. N Engl J Med. 2018;378(3):214-216. https://doi.org/10.1056/NEJMp1712984.
36. Jenq GY, Tinetti ME. Post–acute care: who belongs where? JAMA Intern Med. 2015;175(2):296-297. https://doi.org/10.1001/jamainternmed.2014.4298.

Issue
Journal of Hospital Medicine 15(1)
Issue
Journal of Hospital Medicine 15(1)
Page Number
22-27. Published online first August 21, 2019
Page Number
22-27. Published online first August 21, 2019
Publications
Publications
Topics
Article Type
Sections
Article Source

© 2020 Society of Hospital Medicine

Disallow All Ads
Correspondence Location
Robert E. Burke MD, MS; E-mail: Robert.Burke2@uphs.upenn.edu; Telephone: 215-573-3205; Twitter: @BBurkeMD
Content Gating
Gated (full article locked unless allowed per User)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Gating Strategy
First Peek Free
Medscape Article
Display survey writer
Reuters content
Article PDF Media

Problems Identified by Advice Line Calls

Article Type
Changed
Sun, 05/21/2017 - 13:47
Display Headline
Postdischarge problems identified by telephone calls to an advice line

The period immediately following hospital discharge is particularly hazardous for patients.[1, 2, 3, 4, 5] Problems occurring after discharge may result in high rates of rehospitalization and unscheduled visits to healthcare providers.[6, 7, 8, 9, 10] Numerous investigators have tried to identify patients who are at increased risk for rehospitalizations within 30 days of discharge, and many studies have examined whether various interventions could decrease these adverse events (summarized in Hansen et al.[11]). An increasing fraction of patients discharged by medicine and surgery services have some or all of their care supervised by hospitalists. Thus, hospitals increasingly look to hospitalists for ways to reduce rehospitalizations.

Patients discharged from our hospital are instructed to call an advice line (AL) if and when questions or concerns arise. Accordingly, we examined when these calls were made and what issues were raised, with the idea that the information collected might identify aspects of our discharge processes that needed improvement.

METHODS

Study Design

We conducted a prospective study of a cohort consisting of all unduplicated patients with a matching medical record number in our data warehouse who called our AL between September 1, 2011 and September 1, 2012, and reported being hospitalized or having surgery (inpatient or outpatient) within 30 days preceding their call. We excluded patients who were incarcerated, those who were transferred from other hospitals, those admitted for routine chemotherapy or emergent dialysis, and those discharged to a skilled nursing facility or hospice. The study involved no intervention. It was approved by the Colorado Multiple Institutional Review Board.

Setting

The study was conducted at Denver Health Medical Center, a 525‐bed, university‐affiliated, public safety‐net hospital. At the time of discharge, all patients were given paperwork that listed the telephone number of the AL and written instructions in English or Spanish telling them to call the AL or their primary care physician if they had any of a list of symptoms that was selected by their discharging physician as being relevant to that specific patient's condition(s).

The AL was established in 1997 to provide medical triage to patients of Denver Health. It operates 24 hours a day, 7 days per week, and receives approximately 100,000 calls per year. A language line service is used with nonEnglish‐speaking callers. Calls are handled by a nurse who, with the assistance of a commercial software program (E‐Centaurus; LVM Systems, Phoenix, AZ) containing clinical algorithms (Schmitt‐Thompson Clinical Content, Windsor, CO), makes a triage recommendation. Nurses rarely contact hospital or clinic physicians to assist with triage decisions.

Variables Assessed

We categorized the nature of the callers' reported problem(s) to the AL using the taxonomy summarized in the online appendix (see Supporting Appendix in the online version of this article). We then queried our data warehouse for each patient's demographic information, patient‐level comorbidities, discharging service, discharge date and diagnoses, hospital length of stay, discharge disposition, and whether they had been hospitalized or sought care in our urgent care center or emergency department within 30 days of discharge. The same variables were collected for all unduplicated patients who met the same inclusion and exclusion criteria and were discharged from Denver Health during the same time period but did not call the AL.

Statistics

Data were analyzed using SAS Enterprise Guide 4.1 (SAS Institute, Inc., Cary, NC). Because we made multiple statistical comparisons, we applied the Bonferroni correction when comparing patients calling the AL with those who did not, such that P<0.004 indicated statistical significance. A Student t test or a Wilcoxon rank sum test was used to compare continuous variables depending on results of normality tests. 2 tests were used to compare categorical variables. The intervals between hospital discharge and the call to the AL for patients discharged from medicine versus surgery services were compared using a log‐rank test, with P<0.05 indicating statistical significance.

RESULTS

During the 1‐year study period, 19,303 unique patients were discharged home with instructions regarding the use of the AL. A total of 310 patients called the AL and reported being hospitalized or having surgery within the preceding 30 days. Of these, 2 were excluded (1 who was incarcerated and 1 who was discharged to a skilled nursing facility), leaving 308 patients in the cohort. This represented 1.5% of the total number of unduplicated patients discharged during this same time period (minus the exclusions described above). The large majority of the calls (277/308, 90%) came directly from patients. The remaining 10% came from a proxy, usually a patient's family member. Compared with patients who were discharged during the same time period who did not call the AL, those who called were more likely to speak English, less likely to speak Spanish, more likely to be medically indigent, had slightly longer lengths of stays for their index hospitalization, and were more likely to be discharged from surgery than medicine services (particularly following inpatient surgery) (Table 1).

Patient Characteristics
Patient CharacteristicsPatients Calling Advice Line After Discharge, N=308Patients Not Calling Advice Line After Discharge, N=18,995P Valuea
  • NOTE: Abbreviations: IQR, interquartile range; SD, standard deviation.

  • Bonferroni correction for multiple comparisons was applied, with a P<0.004 indicating significance.

  • Defined as uninsured, ineligible for Medicaid, and unable to purchase private insurance.

  • Defined as 1 or more visits to a primary care provider within 3 years of index hospitalization.

Age, y (meanSD)421739210.0210
Gender, female, n (%)162 (53)10,655 (56) 
Race/ethnicity, n (%)  0.1208
Hispanic/Latino/Spanish129 (42)8,896 (47) 
African American44 (14)2,674 (14) 
White125 (41)6,569 (35) 
Language, n (%)  <0.0001
English273 (89)14,236 (79) 
Spanish32 (10)3,744 (21) 
Payer, n (%)   
Medicare45 (15)3,013 (16) 
Medicaid105 (34)7,777 (41)0.0152
Commercial49 (16)2,863 (15) 
Medically indigentb93 (30)3,442 (18)<0.0001
Self‐pay5 (1)1,070 (5) 
Primary care provider, n (%)c168 (55)10,136 (53)0.6794
Psychiatric comorbidity, n (%)81 (26)4,528 (24)0.3149
Alcohol or substance abuse comorbidity, n (%)65 (21)3,178 (17)0.0417
Discharging service, n (%)  <0.0001
Surgery193 (63)7,247 (38) 
Inpatient123 (40)3,425 (18) 
Ambulatory70 (23)3,822 (20) 
Medicine93 (30)6,038 (32) 
Pediatric4 (1)1,315 (7) 
Obstetric11 (4)3,333 (18) 
Length of stay, median (IQR)2 (04.5)1 (03)0.0003
Inpatient medicine4 (26)3 (15)0.0020
Inpatient surgery3 (16)2 (14)0.0019
Charlson Comorbidity Index, median (IQR)
Inpatient medicine1 (04)1 (02)0.0435
Inpatient surgery0 (01)0 (01)0.0240

The median time from hospital discharge to the call was 3 days (interquartile range [IQR], 16), but 31% and 47% of calls occurred within 24 or 48 hours of discharge, respectively. Ten percent of patients called the AL the same day of discharge (Figure 1). We found no difference in timing of the calls as a function of discharging service.

jhm2252-fig-0001-m.png
Timing of calls relative to discharge.

The 308 patients reported a total of 612 problems or concerns (meanstandard deviation number of complaints per caller=21), the large majority of which (71%) were symptom‐related (Table 2). The most common symptom was uncontrolled pain, reported by 33% and 40% of patients discharged from medicine and surgery services, respectively. The next most common symptoms related to the gastrointestinal system and to surgical site issues in medicine and surgery patients, respectively (data not shown).

Frequency of Patient‐Reported Concerns
 Total Cohort, n (%)Patients Discharged From Medicine, n (%)Patients Discharged From Surgery, n (%)
PatientsComplaintsPatientsComplaintsPatientsComplaints
Symptom related280 (91)433 (71)89 (96)166 (77)171 (89)234 (66)
Discharge instructions65 (21)81 (13)18 (19)21 (10)43 (22)56 (16)
Medication related65 (21)87 (14)19 (20)25 (11)39 (20)54 (15)
Other10 (3)11 (2)4 (4)4 (2)6 (3)7 (2)
Total 612 (100) 216 (100) 351 (100)

Sixty‐five patients, representing 21% of the cohort, reported 81 problems understanding or executing discharge instructions. No difference was observed between the fraction of these problems reported by patients from medicine versus surgery (19% and 22%, respectively, P=0.54).

Sixty‐five patients, again representing 21% of the cohort, reported 87 medication‐related problems, 20% from both the medicine and surgery services (P=0.99). Medicine patients more frequently reported difficulties understanding their medication instructions, whereas surgery patients more frequently reported lack of efficacy of medications, particularly with respect to pain control (data not shown).

Thirty percent of patients who called the AL were advised by the nurse to go to the emergency department immediately. Medicine patients were more likely to be triaged to the emergency department compared with surgery patients (45% vs 22%, P<0.0001).

The 30‐day readmission rates and the rates of unscheduled urgent or emergent care visits were higher for patients calling the AL compared with those who did not call (46/308, 15% vs 706/18,995, 4%, and 92/308, 30% vs 1303/18,995, 7%, respectively, both P<0.0001). Similar differences were found for patients discharged from medicine or surgery services who called the AL compared with those who did not (data not shown, both P<0.0001). The median number of days between AL call and rehospitalization was 0 (IQR, 02) and 1 (IQR, 08) for medicine and surgery patients, respectively. Ninety‐three percent of rehospitalizations were related to the index hospitalization, and 78% of patients who were readmitted had no outpatient encounter in the interim between discharge and rehospitalization.

DISCUSSION

We investigated the source and nature of patient telephone calls to an AL following a hospitalization or surgery, and our data revealed the following important findings: (1) nearly one‐half of the calls to the AL occurred within the first 48 hours following discharge; (2) the majority of the calls came from surgery patients, and a greater fraction of patients discharged from surgery services called the AL than patients discharged from medicine services; (3) the most common issues were uncontrolled pain, questions about medications, and problems understanding or executing aftercare instructions (particularly pertaining to the care of surgical wounds); and (4) patients calling the AL had higher rates of 30‐day rehospitalization and of unscheduled urgent or emergent care visits.

The utilization of our patient‐initiated call line was only 1.5%, which was on the low end of the 1% to 10% reported in the literature.[7, 12] This can be attributed to a number of issues that are specific to our system. First, the discharge instructions provided to our patients stated that they should call their primary care provider or the AL if they had questions. Accordingly, because approximately 50% of our patients had a primary care provider in our system, some may have preferentially contacted their primary care provider rather than the AL. Second, the instructions stated that the patients should call if they were experiencing the symptoms listed on the instruction sheet, so those with other problems/complaints may not have called. Third, AL personnel identified patients as being in our cohort by asking if they had been discharged or underwent a surgical procedure within 30‐days of their call. This may have resulted in the under‐reporting of patients who were hospitalized or had outpatient surgical procedures. Fourth, there may have been a number of characteristics specific to patients in our system that reduced the frequency with which they utilized the AL (eg, access to telephones or other community providers).

Most previous studies of patient‐initiated call lines have included them as part of multi‐intervention pre‐ and/or postdischarge strategies.[7, 8, 9, 10, 11, 12, 13] One prior small study compared the information reported by 37 patients who called an AL with that elicited by nurse‐initiated patient contact.[12] The most frequently reported problems in this study were medication‐related issues (43%). However, this study only included medicine patients and did not document the proportion of calls occurring at various time intervals.

The problems we identified (in both medicine and surgery patients) have previously been described,[2, 3, 4, 13, 14, 15, 16] but all of the studies reporting these problems utilized calls that were initiated by health care providers to patients at various fixed intervals following discharge (ie, 730 days). Most of these used a scripted approach seeking responses to specific questions or outcomes, and the specific timing at which the problems arose was not addressed. In contrast, we examined unsolicited concerns expressed by patients calling an AL following discharge whenever they felt sufficient urgency to address whatever problems or questions arose. We found that a large fraction of calls occurred on the day of or within the first 48 hours following discharge, much earlier than when provider‐initiated calls in the studies cited above occurred. Accordingly, our results cannot be used to compare the utility of patient‐ versus provider‐initiated calls, or to suggest that other hospitals should create an AL system. Rather, we suggest that our findings might be complementary to those reported in studies of provider‐initiated calls and only propose that by examining calls placed by patients to ALs, problems with hospital discharge processes (some of which may result in increased rates of readmission) may be discovered.

The observation that such a large fraction of calls to our AL occurred within the first 48 hours following discharge, together with the fact that many of the questions asked or concerns raised pertained to issues that should have been discussed during the discharge process (eg, pain control, care of surgical wounds), suggests that suboptimal patient education was occurring prior to discharge as was suggested by Henderson and Zernike.[17] This finding has led us to expand our patient education processes prior to discharge on both medicine and surgery services. Because our hospitalists care for approximately 90% of the patients admitted to medicine services and are increasingly involved in the care of patients on surgery services, they are integrally involved with such quality improvement initiatives.

To our knowledge this is the first study in the literature that describes both medicine and surgery patients who call an AL because of problems or questions following hospital discharge, categorizes these problems, determines when the patients called following their discharge, and identifies those who called as being at increased risk for early rehospitalizations and unscheduled urgent or emergent care visits. Given the financial penalties issued to hospitals with high 30‐day readmission rates, these patients may warrant more attention than is customarily available from telephone call lines or during routine outpatient follow‐up. The majority of patients who called our AL had Medicare, Medicaid, or a commercial insurance, and, accordingly, may have been eligible for additional services such as home visits and/or expedited follow‐up appointments.

Our study has a number of limitations. First, it is a single‐center study, so the results might not generalize to other institutions. Second, because the study was performed in a university‐affiliated, public safety‐net hospital, patient characteristics and the rates and types of postdischarge concerns that we observed might differ from those encountered in different types of hospitals and/or from those in nonteaching institutions. We would suggest, however, that the idea of using concerns raised by patients discharged from any type of hospital in calls to ALs may similarly identify problems with that specific hospital's discharge processes. Third, the information collected from the AL came from summaries provided by nurses answering the calls rather than from actual transcripts. This could have resulted in insufficient or incorrect information pertaining to some of the variables assessed in Table 2. The information presented in Table 1, however, was obtained from our data warehouse after matching medical record numbers. Fourth, we could have underestimated the number of patients who had 30‐day rehospitalizations and/or unplanned for urgent or emergent care visits if patients sought care at other hospitals. Fifth, the number of patients calling the AL was too small to allow us to do any type of robust matching or multivariable analysis. Accordingly, the differences that appeared between patients who called and those who did not (ie, English speakers, being medically indigent, the length of stay for the index hospitalization and the discharging service) could be the result of inadequate matching or interactions among the variables. Although matching or multivariate analysis might have yielded different associations between patients who called the AL versus those who did not, those who called the AL still had an increased risk of readmission and urgent or emergent visits and may still benefit from targeted interventions. Finally, the fact that only 1.5% of unique patients who were discharged called the AL could have biased our results. Because only 55% and 53% of the patients who did or did not call the AL, respectively, saw primary care physicians within our system within the 3 years prior to their index hospitalization (P=0.679), the frequency of calls to the AL that we observed could have underestimated the frequency with which patients had contact with other care providers in the community.

In summary, information collected from patient‐initiated calls to our AL identified several aspects of our discharge processes that needed improvement. We concluded that our predischarge educational processes for both medicine and surgery services needed modification, especially with respect to pain management, which problems to expect after hospitalization or surgery, and how to deal with them. The high rates of 30‐day rehospitalization and of unscheduled urgent or emergent care visits among patients calling the AL identifies them as being at increased risk for these outcomes, although the likelihood of these events may be related to factors other than just calling the AL.

Files
References
  1. Parrish MM, O'Malley K, Adams RI, Adams SR, Coleman EA. Implementation of the care transitions intervention: sustainability and lessons learned. Prof Case Manag. 2009;14(6):282293.
  2. Arora VM, Prochaska ML, Farnan JM, et al. Problems after discharge and understanding of communication with their primary care physicians among hospitalized seniors: a mixed methods study. J Hosp Med. 2010;5(7):385391.
  3. Forster AJ, Clark HD, Menard A, et al. Adverse events among medical patients after discharge from hospital. CMAJ. 2004;170(3):345349.
  4. Forster AJ, Murff HJ, Peterson JF, Gandhi TK, Bates DW. The incidence and severity of adverse events affecting patients after discharge from the hospital. Ann Intern Med. 2003;138(3):161167.
  5. Misky GJ, Wald HL, Coleman EA. Post‐hospitalization transitions: examining the effects of timing of primary care provider follow‐up. J Hosp Med. 2010;5(7):392397.
  6. Bostrom J, Caldwell J, McGuire K, Everson D. Telephone follow‐up after discharge from the hospital: does it make a difference? Appl Nurs Res. 1996;9(2) 4752.
  7. Sorknaes AD, Bech M, Madsen H, et al. The effect of real‐time teleconsultations between hospital‐based nurses and patients with severe COPD discharged after an exacerbation. J Telemed Telecare. 2013;19(8):466474.
  8. Kwok T, Lum CM, Chan HS, Ma HM, Lee D, Woo J. A randomized, controlled trial of an intensive community nurse‐supported discharge program in preventing hospital readmissions of older patients with chronic lung disease. J Am Geriatr Soc. 2004;52(8):12401246.
  9. Jaarsma T, Halfens R, Huijer Abu‐Saad H, et al. Effects of education and support on self‐care and resource utilization in patients with heart failure. Eur Heart J. 1999;20(9):673682.
  10. Naylor MD, Brooten D, Campbell R, et al. Comprehensive discharge planning and home follow‐up of hospitalized elders: a randomized clinical trial. JAMA. 1999;281(7):613620.
  11. Hansen LO, Young RS, Hinami K, Leung A, Williams MV. Interventions to reduce 30‐day rehospitalization: a systematic review. Ann Intern Med. 2011;155(8):520528.
  12. Rennke S, Kesh S, Neeman N, Sehgal NL. Complementary telephone strategies to improve postdischarge communication. Am J Med. 2012;125(1):2830.
  13. Shu CC, Hsu NC, Lin YF, Wang JY, Lin JW, Ko WJ. Integrated postdischarge transitional care in a hospitalist system to improve discharge outcome: an experimental study. BMC Med. 2011;9:96.
  14. Hinami K, Bilimoria KY, Kallas PG, Simons YM, Christensen NP, Williams MV. Patient experiences after hospitalizations for elective surgery. Am J Surg. 2014;207(6):855862.
  15. Kable A, Gibberd R, Spigelman A. Complications after discharge for surgical patients. ANZ J Surg. 2004;74(3):9297.
  16. Visser A, Ubbink DT, Gouma DJ, Goslings JC. Surgeons are overlooking post‐discharge complications: a prospective cohort study. World J Surg. 2014;38(5):10191025.
  17. Henderson A, Zernike W. A study of the impact of discharge information for surgical patients. J Adv Nurs. 2001;35(3):435441.
Article PDF
Issue
Journal of Hospital Medicine - 9(11)
Publications
Page Number
695-699
Sections
Files
Files
Article PDF
Article PDF

The period immediately following hospital discharge is particularly hazardous for patients.[1, 2, 3, 4, 5] Problems occurring after discharge may result in high rates of rehospitalization and unscheduled visits to healthcare providers.[6, 7, 8, 9, 10] Numerous investigators have tried to identify patients who are at increased risk for rehospitalizations within 30 days of discharge, and many studies have examined whether various interventions could decrease these adverse events (summarized in Hansen et al.[11]). An increasing fraction of patients discharged by medicine and surgery services have some or all of their care supervised by hospitalists. Thus, hospitals increasingly look to hospitalists for ways to reduce rehospitalizations.

Patients discharged from our hospital are instructed to call an advice line (AL) if and when questions or concerns arise. Accordingly, we examined when these calls were made and what issues were raised, with the idea that the information collected might identify aspects of our discharge processes that needed improvement.

METHODS

Study Design

We conducted a prospective study of a cohort consisting of all unduplicated patients with a matching medical record number in our data warehouse who called our AL between September 1, 2011 and September 1, 2012, and reported being hospitalized or having surgery (inpatient or outpatient) within 30 days preceding their call. We excluded patients who were incarcerated, those who were transferred from other hospitals, those admitted for routine chemotherapy or emergent dialysis, and those discharged to a skilled nursing facility or hospice. The study involved no intervention. It was approved by the Colorado Multiple Institutional Review Board.

Setting

The study was conducted at Denver Health Medical Center, a 525‐bed, university‐affiliated, public safety‐net hospital. At the time of discharge, all patients were given paperwork that listed the telephone number of the AL and written instructions in English or Spanish telling them to call the AL or their primary care physician if they had any of a list of symptoms that was selected by their discharging physician as being relevant to that specific patient's condition(s).

The AL was established in 1997 to provide medical triage to patients of Denver Health. It operates 24 hours a day, 7 days per week, and receives approximately 100,000 calls per year. A language line service is used with nonEnglish‐speaking callers. Calls are handled by a nurse who, with the assistance of a commercial software program (E‐Centaurus; LVM Systems, Phoenix, AZ) containing clinical algorithms (Schmitt‐Thompson Clinical Content, Windsor, CO), makes a triage recommendation. Nurses rarely contact hospital or clinic physicians to assist with triage decisions.

Variables Assessed

We categorized the nature of the callers' reported problem(s) to the AL using the taxonomy summarized in the online appendix (see Supporting Appendix in the online version of this article). We then queried our data warehouse for each patient's demographic information, patient‐level comorbidities, discharging service, discharge date and diagnoses, hospital length of stay, discharge disposition, and whether they had been hospitalized or sought care in our urgent care center or emergency department within 30 days of discharge. The same variables were collected for all unduplicated patients who met the same inclusion and exclusion criteria and were discharged from Denver Health during the same time period but did not call the AL.

Statistics

Data were analyzed using SAS Enterprise Guide 4.1 (SAS Institute, Inc., Cary, NC). Because we made multiple statistical comparisons, we applied the Bonferroni correction when comparing patients calling the AL with those who did not, such that P<0.004 indicated statistical significance. A Student t test or a Wilcoxon rank sum test was used to compare continuous variables depending on results of normality tests. 2 tests were used to compare categorical variables. The intervals between hospital discharge and the call to the AL for patients discharged from medicine versus surgery services were compared using a log‐rank test, with P<0.05 indicating statistical significance.

RESULTS

During the 1‐year study period, 19,303 unique patients were discharged home with instructions regarding the use of the AL. A total of 310 patients called the AL and reported being hospitalized or having surgery within the preceding 30 days. Of these, 2 were excluded (1 who was incarcerated and 1 who was discharged to a skilled nursing facility), leaving 308 patients in the cohort. This represented 1.5% of the total number of unduplicated patients discharged during this same time period (minus the exclusions described above). The large majority of the calls (277/308, 90%) came directly from patients. The remaining 10% came from a proxy, usually a patient's family member. Compared with patients who were discharged during the same time period who did not call the AL, those who called were more likely to speak English, less likely to speak Spanish, more likely to be medically indigent, had slightly longer lengths of stays for their index hospitalization, and were more likely to be discharged from surgery than medicine services (particularly following inpatient surgery) (Table 1).

Patient Characteristics
Patient CharacteristicsPatients Calling Advice Line After Discharge, N=308Patients Not Calling Advice Line After Discharge, N=18,995P Valuea
  • NOTE: Abbreviations: IQR, interquartile range; SD, standard deviation.

  • Bonferroni correction for multiple comparisons was applied, with a P<0.004 indicating significance.

  • Defined as uninsured, ineligible for Medicaid, and unable to purchase private insurance.

  • Defined as 1 or more visits to a primary care provider within 3 years of index hospitalization.

Age, y (meanSD)421739210.0210
Gender, female, n (%)162 (53)10,655 (56) 
Race/ethnicity, n (%)  0.1208
Hispanic/Latino/Spanish129 (42)8,896 (47) 
African American44 (14)2,674 (14) 
White125 (41)6,569 (35) 
Language, n (%)  <0.0001
English273 (89)14,236 (79) 
Spanish32 (10)3,744 (21) 
Payer, n (%)   
Medicare45 (15)3,013 (16) 
Medicaid105 (34)7,777 (41)0.0152
Commercial49 (16)2,863 (15) 
Medically indigentb93 (30)3,442 (18)<0.0001
Self‐pay5 (1)1,070 (5) 
Primary care provider, n (%)c168 (55)10,136 (53)0.6794
Psychiatric comorbidity, n (%)81 (26)4,528 (24)0.3149
Alcohol or substance abuse comorbidity, n (%)65 (21)3,178 (17)0.0417
Discharging service, n (%)  <0.0001
Surgery193 (63)7,247 (38) 
Inpatient123 (40)3,425 (18) 
Ambulatory70 (23)3,822 (20) 
Medicine93 (30)6,038 (32) 
Pediatric4 (1)1,315 (7) 
Obstetric11 (4)3,333 (18) 
Length of stay, median (IQR)2 (04.5)1 (03)0.0003
Inpatient medicine4 (26)3 (15)0.0020
Inpatient surgery3 (16)2 (14)0.0019
Charlson Comorbidity Index, median (IQR)
Inpatient medicine1 (04)1 (02)0.0435
Inpatient surgery0 (01)0 (01)0.0240

The median time from hospital discharge to the call was 3 days (interquartile range [IQR], 16), but 31% and 47% of calls occurred within 24 or 48 hours of discharge, respectively. Ten percent of patients called the AL the same day of discharge (Figure 1). We found no difference in timing of the calls as a function of discharging service.

jhm2252-fig-0001-m.png
Timing of calls relative to discharge.

The 308 patients reported a total of 612 problems or concerns (meanstandard deviation number of complaints per caller=21), the large majority of which (71%) were symptom‐related (Table 2). The most common symptom was uncontrolled pain, reported by 33% and 40% of patients discharged from medicine and surgery services, respectively. The next most common symptoms related to the gastrointestinal system and to surgical site issues in medicine and surgery patients, respectively (data not shown).

Frequency of Patient‐Reported Concerns
 Total Cohort, n (%)Patients Discharged From Medicine, n (%)Patients Discharged From Surgery, n (%)
PatientsComplaintsPatientsComplaintsPatientsComplaints
Symptom related280 (91)433 (71)89 (96)166 (77)171 (89)234 (66)
Discharge instructions65 (21)81 (13)18 (19)21 (10)43 (22)56 (16)
Medication related65 (21)87 (14)19 (20)25 (11)39 (20)54 (15)
Other10 (3)11 (2)4 (4)4 (2)6 (3)7 (2)
Total 612 (100) 216 (100) 351 (100)

Sixty‐five patients, representing 21% of the cohort, reported 81 problems understanding or executing discharge instructions. No difference was observed between the fraction of these problems reported by patients from medicine versus surgery (19% and 22%, respectively, P=0.54).

Sixty‐five patients, again representing 21% of the cohort, reported 87 medication‐related problems, 20% from both the medicine and surgery services (P=0.99). Medicine patients more frequently reported difficulties understanding their medication instructions, whereas surgery patients more frequently reported lack of efficacy of medications, particularly with respect to pain control (data not shown).

Thirty percent of patients who called the AL were advised by the nurse to go to the emergency department immediately. Medicine patients were more likely to be triaged to the emergency department compared with surgery patients (45% vs 22%, P<0.0001).

The 30‐day readmission rates and the rates of unscheduled urgent or emergent care visits were higher for patients calling the AL compared with those who did not call (46/308, 15% vs 706/18,995, 4%, and 92/308, 30% vs 1303/18,995, 7%, respectively, both P<0.0001). Similar differences were found for patients discharged from medicine or surgery services who called the AL compared with those who did not (data not shown, both P<0.0001). The median number of days between AL call and rehospitalization was 0 (IQR, 02) and 1 (IQR, 08) for medicine and surgery patients, respectively. Ninety‐three percent of rehospitalizations were related to the index hospitalization, and 78% of patients who were readmitted had no outpatient encounter in the interim between discharge and rehospitalization.

DISCUSSION

We investigated the source and nature of patient telephone calls to an AL following a hospitalization or surgery, and our data revealed the following important findings: (1) nearly one‐half of the calls to the AL occurred within the first 48 hours following discharge; (2) the majority of the calls came from surgery patients, and a greater fraction of patients discharged from surgery services called the AL than patients discharged from medicine services; (3) the most common issues were uncontrolled pain, questions about medications, and problems understanding or executing aftercare instructions (particularly pertaining to the care of surgical wounds); and (4) patients calling the AL had higher rates of 30‐day rehospitalization and of unscheduled urgent or emergent care visits.

The utilization of our patient‐initiated call line was only 1.5%, which was on the low end of the 1% to 10% reported in the literature.[7, 12] This can be attributed to a number of issues that are specific to our system. First, the discharge instructions provided to our patients stated that they should call their primary care provider or the AL if they had questions. Accordingly, because approximately 50% of our patients had a primary care provider in our system, some may have preferentially contacted their primary care provider rather than the AL. Second, the instructions stated that the patients should call if they were experiencing the symptoms listed on the instruction sheet, so those with other problems/complaints may not have called. Third, AL personnel identified patients as being in our cohort by asking if they had been discharged or underwent a surgical procedure within 30‐days of their call. This may have resulted in the under‐reporting of patients who were hospitalized or had outpatient surgical procedures. Fourth, there may have been a number of characteristics specific to patients in our system that reduced the frequency with which they utilized the AL (eg, access to telephones or other community providers).

Most previous studies of patient‐initiated call lines have included them as part of multi‐intervention pre‐ and/or postdischarge strategies.[7, 8, 9, 10, 11, 12, 13] One prior small study compared the information reported by 37 patients who called an AL with that elicited by nurse‐initiated patient contact.[12] The most frequently reported problems in this study were medication‐related issues (43%). However, this study only included medicine patients and did not document the proportion of calls occurring at various time intervals.

The problems we identified (in both medicine and surgery patients) have previously been described,[2, 3, 4, 13, 14, 15, 16] but all of the studies reporting these problems utilized calls that were initiated by health care providers to patients at various fixed intervals following discharge (ie, 730 days). Most of these used a scripted approach seeking responses to specific questions or outcomes, and the specific timing at which the problems arose was not addressed. In contrast, we examined unsolicited concerns expressed by patients calling an AL following discharge whenever they felt sufficient urgency to address whatever problems or questions arose. We found that a large fraction of calls occurred on the day of or within the first 48 hours following discharge, much earlier than when provider‐initiated calls in the studies cited above occurred. Accordingly, our results cannot be used to compare the utility of patient‐ versus provider‐initiated calls, or to suggest that other hospitals should create an AL system. Rather, we suggest that our findings might be complementary to those reported in studies of provider‐initiated calls and only propose that by examining calls placed by patients to ALs, problems with hospital discharge processes (some of which may result in increased rates of readmission) may be discovered.

The observation that such a large fraction of calls to our AL occurred within the first 48 hours following discharge, together with the fact that many of the questions asked or concerns raised pertained to issues that should have been discussed during the discharge process (eg, pain control, care of surgical wounds), suggests that suboptimal patient education was occurring prior to discharge as was suggested by Henderson and Zernike.[17] This finding has led us to expand our patient education processes prior to discharge on both medicine and surgery services. Because our hospitalists care for approximately 90% of the patients admitted to medicine services and are increasingly involved in the care of patients on surgery services, they are integrally involved with such quality improvement initiatives.

To our knowledge this is the first study in the literature that describes both medicine and surgery patients who call an AL because of problems or questions following hospital discharge, categorizes these problems, determines when the patients called following their discharge, and identifies those who called as being at increased risk for early rehospitalizations and unscheduled urgent or emergent care visits. Given the financial penalties issued to hospitals with high 30‐day readmission rates, these patients may warrant more attention than is customarily available from telephone call lines or during routine outpatient follow‐up. The majority of patients who called our AL had Medicare, Medicaid, or a commercial insurance, and, accordingly, may have been eligible for additional services such as home visits and/or expedited follow‐up appointments.

Our study has a number of limitations. First, it is a single‐center study, so the results might not generalize to other institutions. Second, because the study was performed in a university‐affiliated, public safety‐net hospital, patient characteristics and the rates and types of postdischarge concerns that we observed might differ from those encountered in different types of hospitals and/or from those in nonteaching institutions. We would suggest, however, that the idea of using concerns raised by patients discharged from any type of hospital in calls to ALs may similarly identify problems with that specific hospital's discharge processes. Third, the information collected from the AL came from summaries provided by nurses answering the calls rather than from actual transcripts. This could have resulted in insufficient or incorrect information pertaining to some of the variables assessed in Table 2. The information presented in Table 1, however, was obtained from our data warehouse after matching medical record numbers. Fourth, we could have underestimated the number of patients who had 30‐day rehospitalizations and/or unplanned for urgent or emergent care visits if patients sought care at other hospitals. Fifth, the number of patients calling the AL was too small to allow us to do any type of robust matching or multivariable analysis. Accordingly, the differences that appeared between patients who called and those who did not (ie, English speakers, being medically indigent, the length of stay for the index hospitalization and the discharging service) could be the result of inadequate matching or interactions among the variables. Although matching or multivariate analysis might have yielded different associations between patients who called the AL versus those who did not, those who called the AL still had an increased risk of readmission and urgent or emergent visits and may still benefit from targeted interventions. Finally, the fact that only 1.5% of unique patients who were discharged called the AL could have biased our results. Because only 55% and 53% of the patients who did or did not call the AL, respectively, saw primary care physicians within our system within the 3 years prior to their index hospitalization (P=0.679), the frequency of calls to the AL that we observed could have underestimated the frequency with which patients had contact with other care providers in the community.

In summary, information collected from patient‐initiated calls to our AL identified several aspects of our discharge processes that needed improvement. We concluded that our predischarge educational processes for both medicine and surgery services needed modification, especially with respect to pain management, which problems to expect after hospitalization or surgery, and how to deal with them. The high rates of 30‐day rehospitalization and of unscheduled urgent or emergent care visits among patients calling the AL identifies them as being at increased risk for these outcomes, although the likelihood of these events may be related to factors other than just calling the AL.

The period immediately following hospital discharge is particularly hazardous for patients.[1, 2, 3, 4, 5] Problems occurring after discharge may result in high rates of rehospitalization and unscheduled visits to healthcare providers.[6, 7, 8, 9, 10] Numerous investigators have tried to identify patients who are at increased risk for rehospitalizations within 30 days of discharge, and many studies have examined whether various interventions could decrease these adverse events (summarized in Hansen et al.[11]). An increasing fraction of patients discharged by medicine and surgery services have some or all of their care supervised by hospitalists. Thus, hospitals increasingly look to hospitalists for ways to reduce rehospitalizations.

Patients discharged from our hospital are instructed to call an advice line (AL) if and when questions or concerns arise. Accordingly, we examined when these calls were made and what issues were raised, with the idea that the information collected might identify aspects of our discharge processes that needed improvement.

METHODS

Study Design

We conducted a prospective study of a cohort consisting of all unduplicated patients with a matching medical record number in our data warehouse who called our AL between September 1, 2011 and September 1, 2012, and reported being hospitalized or having surgery (inpatient or outpatient) within 30 days preceding their call. We excluded patients who were incarcerated, those who were transferred from other hospitals, those admitted for routine chemotherapy or emergent dialysis, and those discharged to a skilled nursing facility or hospice. The study involved no intervention. It was approved by the Colorado Multiple Institutional Review Board.

Setting

The study was conducted at Denver Health Medical Center, a 525‐bed, university‐affiliated, public safety‐net hospital. At the time of discharge, all patients were given paperwork that listed the telephone number of the AL and written instructions in English or Spanish telling them to call the AL or their primary care physician if they had any of a list of symptoms that was selected by their discharging physician as being relevant to that specific patient's condition(s).

The AL was established in 1997 to provide medical triage to patients of Denver Health. It operates 24 hours a day, 7 days per week, and receives approximately 100,000 calls per year. A language line service is used with nonEnglish‐speaking callers. Calls are handled by a nurse who, with the assistance of a commercial software program (E‐Centaurus; LVM Systems, Phoenix, AZ) containing clinical algorithms (Schmitt‐Thompson Clinical Content, Windsor, CO), makes a triage recommendation. Nurses rarely contact hospital or clinic physicians to assist with triage decisions.

Variables Assessed

We categorized the nature of the callers' reported problem(s) to the AL using the taxonomy summarized in the online appendix (see Supporting Appendix in the online version of this article). We then queried our data warehouse for each patient's demographic information, patient‐level comorbidities, discharging service, discharge date and diagnoses, hospital length of stay, discharge disposition, and whether they had been hospitalized or sought care in our urgent care center or emergency department within 30 days of discharge. The same variables were collected for all unduplicated patients who met the same inclusion and exclusion criteria and were discharged from Denver Health during the same time period but did not call the AL.

Statistics

Data were analyzed using SAS Enterprise Guide 4.1 (SAS Institute, Inc., Cary, NC). Because we made multiple statistical comparisons, we applied the Bonferroni correction when comparing patients calling the AL with those who did not, such that P<0.004 indicated statistical significance. A Student t test or a Wilcoxon rank sum test was used to compare continuous variables depending on results of normality tests. 2 tests were used to compare categorical variables. The intervals between hospital discharge and the call to the AL for patients discharged from medicine versus surgery services were compared using a log‐rank test, with P<0.05 indicating statistical significance.

RESULTS

During the 1‐year study period, 19,303 unique patients were discharged home with instructions regarding the use of the AL. A total of 310 patients called the AL and reported being hospitalized or having surgery within the preceding 30 days. Of these, 2 were excluded (1 who was incarcerated and 1 who was discharged to a skilled nursing facility), leaving 308 patients in the cohort. This represented 1.5% of the total number of unduplicated patients discharged during this same time period (minus the exclusions described above). The large majority of the calls (277/308, 90%) came directly from patients. The remaining 10% came from a proxy, usually a patient's family member. Compared with patients who were discharged during the same time period who did not call the AL, those who called were more likely to speak English, less likely to speak Spanish, more likely to be medically indigent, had slightly longer lengths of stays for their index hospitalization, and were more likely to be discharged from surgery than medicine services (particularly following inpatient surgery) (Table 1).

Patient Characteristics
Patient CharacteristicsPatients Calling Advice Line After Discharge, N=308Patients Not Calling Advice Line After Discharge, N=18,995P Valuea
  • NOTE: Abbreviations: IQR, interquartile range; SD, standard deviation.

  • Bonferroni correction for multiple comparisons was applied, with a P<0.004 indicating significance.

  • Defined as uninsured, ineligible for Medicaid, and unable to purchase private insurance.

  • Defined as 1 or more visits to a primary care provider within 3 years of index hospitalization.

Age, y (meanSD)421739210.0210
Gender, female, n (%)162 (53)10,655 (56) 
Race/ethnicity, n (%)  0.1208
Hispanic/Latino/Spanish129 (42)8,896 (47) 
African American44 (14)2,674 (14) 
White125 (41)6,569 (35) 
Language, n (%)  <0.0001
English273 (89)14,236 (79) 
Spanish32 (10)3,744 (21) 
Payer, n (%)   
Medicare45 (15)3,013 (16) 
Medicaid105 (34)7,777 (41)0.0152
Commercial49 (16)2,863 (15) 
Medically indigentb93 (30)3,442 (18)<0.0001
Self‐pay5 (1)1,070 (5) 
Primary care provider, n (%)c168 (55)10,136 (53)0.6794
Psychiatric comorbidity, n (%)81 (26)4,528 (24)0.3149
Alcohol or substance abuse comorbidity, n (%)65 (21)3,178 (17)0.0417
Discharging service, n (%)  <0.0001
Surgery193 (63)7,247 (38) 
Inpatient123 (40)3,425 (18) 
Ambulatory70 (23)3,822 (20) 
Medicine93 (30)6,038 (32) 
Pediatric4 (1)1,315 (7) 
Obstetric11 (4)3,333 (18) 
Length of stay, median (IQR)2 (04.5)1 (03)0.0003
Inpatient medicine4 (26)3 (15)0.0020
Inpatient surgery3 (16)2 (14)0.0019
Charlson Comorbidity Index, median (IQR)
Inpatient medicine1 (04)1 (02)0.0435
Inpatient surgery0 (01)0 (01)0.0240

The median time from hospital discharge to the call was 3 days (interquartile range [IQR], 16), but 31% and 47% of calls occurred within 24 or 48 hours of discharge, respectively. Ten percent of patients called the AL the same day of discharge (Figure 1). We found no difference in timing of the calls as a function of discharging service.

jhm2252-fig-0001-m.png
Timing of calls relative to discharge.

The 308 patients reported a total of 612 problems or concerns (meanstandard deviation number of complaints per caller=21), the large majority of which (71%) were symptom‐related (Table 2). The most common symptom was uncontrolled pain, reported by 33% and 40% of patients discharged from medicine and surgery services, respectively. The next most common symptoms related to the gastrointestinal system and to surgical site issues in medicine and surgery patients, respectively (data not shown).

Frequency of Patient‐Reported Concerns
 Total Cohort, n (%)Patients Discharged From Medicine, n (%)Patients Discharged From Surgery, n (%)
PatientsComplaintsPatientsComplaintsPatientsComplaints
Symptom related280 (91)433 (71)89 (96)166 (77)171 (89)234 (66)
Discharge instructions65 (21)81 (13)18 (19)21 (10)43 (22)56 (16)
Medication related65 (21)87 (14)19 (20)25 (11)39 (20)54 (15)
Other10 (3)11 (2)4 (4)4 (2)6 (3)7 (2)
Total 612 (100) 216 (100) 351 (100)

Sixty‐five patients, representing 21% of the cohort, reported 81 problems understanding or executing discharge instructions. No difference was observed between the fraction of these problems reported by patients from medicine versus surgery (19% and 22%, respectively, P=0.54).

Sixty‐five patients, again representing 21% of the cohort, reported 87 medication‐related problems, 20% from both the medicine and surgery services (P=0.99). Medicine patients more frequently reported difficulties understanding their medication instructions, whereas surgery patients more frequently reported lack of efficacy of medications, particularly with respect to pain control (data not shown).

Thirty percent of patients who called the AL were advised by the nurse to go to the emergency department immediately. Medicine patients were more likely to be triaged to the emergency department compared with surgery patients (45% vs 22%, P<0.0001).

The 30‐day readmission rates and the rates of unscheduled urgent or emergent care visits were higher for patients calling the AL compared with those who did not call (46/308, 15% vs 706/18,995, 4%, and 92/308, 30% vs 1303/18,995, 7%, respectively, both P<0.0001). Similar differences were found for patients discharged from medicine or surgery services who called the AL compared with those who did not (data not shown, both P<0.0001). The median number of days between AL call and rehospitalization was 0 (IQR, 02) and 1 (IQR, 08) for medicine and surgery patients, respectively. Ninety‐three percent of rehospitalizations were related to the index hospitalization, and 78% of patients who were readmitted had no outpatient encounter in the interim between discharge and rehospitalization.

DISCUSSION

We investigated the source and nature of patient telephone calls to an AL following a hospitalization or surgery, and our data revealed the following important findings: (1) nearly one‐half of the calls to the AL occurred within the first 48 hours following discharge; (2) the majority of the calls came from surgery patients, and a greater fraction of patients discharged from surgery services called the AL than patients discharged from medicine services; (3) the most common issues were uncontrolled pain, questions about medications, and problems understanding or executing aftercare instructions (particularly pertaining to the care of surgical wounds); and (4) patients calling the AL had higher rates of 30‐day rehospitalization and of unscheduled urgent or emergent care visits.

The utilization of our patient‐initiated call line was only 1.5%, which was on the low end of the 1% to 10% reported in the literature.[7, 12] This can be attributed to a number of issues that are specific to our system. First, the discharge instructions provided to our patients stated that they should call their primary care provider or the AL if they had questions. Accordingly, because approximately 50% of our patients had a primary care provider in our system, some may have preferentially contacted their primary care provider rather than the AL. Second, the instructions stated that the patients should call if they were experiencing the symptoms listed on the instruction sheet, so those with other problems/complaints may not have called. Third, AL personnel identified patients as being in our cohort by asking if they had been discharged or underwent a surgical procedure within 30‐days of their call. This may have resulted in the under‐reporting of patients who were hospitalized or had outpatient surgical procedures. Fourth, there may have been a number of characteristics specific to patients in our system that reduced the frequency with which they utilized the AL (eg, access to telephones or other community providers).

Most previous studies of patient‐initiated call lines have included them as part of multi‐intervention pre‐ and/or postdischarge strategies.[7, 8, 9, 10, 11, 12, 13] One prior small study compared the information reported by 37 patients who called an AL with that elicited by nurse‐initiated patient contact.[12] The most frequently reported problems in this study were medication‐related issues (43%). However, this study only included medicine patients and did not document the proportion of calls occurring at various time intervals.

The problems we identified (in both medicine and surgery patients) have previously been described,[2, 3, 4, 13, 14, 15, 16] but all of the studies reporting these problems utilized calls that were initiated by health care providers to patients at various fixed intervals following discharge (ie, 730 days). Most of these used a scripted approach seeking responses to specific questions or outcomes, and the specific timing at which the problems arose was not addressed. In contrast, we examined unsolicited concerns expressed by patients calling an AL following discharge whenever they felt sufficient urgency to address whatever problems or questions arose. We found that a large fraction of calls occurred on the day of or within the first 48 hours following discharge, much earlier than when provider‐initiated calls in the studies cited above occurred. Accordingly, our results cannot be used to compare the utility of patient‐ versus provider‐initiated calls, or to suggest that other hospitals should create an AL system. Rather, we suggest that our findings might be complementary to those reported in studies of provider‐initiated calls and only propose that by examining calls placed by patients to ALs, problems with hospital discharge processes (some of which may result in increased rates of readmission) may be discovered.

The observation that such a large fraction of calls to our AL occurred within the first 48 hours following discharge, together with the fact that many of the questions asked or concerns raised pertained to issues that should have been discussed during the discharge process (eg, pain control, care of surgical wounds), suggests that suboptimal patient education was occurring prior to discharge as was suggested by Henderson and Zernike.[17] This finding has led us to expand our patient education processes prior to discharge on both medicine and surgery services. Because our hospitalists care for approximately 90% of the patients admitted to medicine services and are increasingly involved in the care of patients on surgery services, they are integrally involved with such quality improvement initiatives.

To our knowledge this is the first study in the literature that describes both medicine and surgery patients who call an AL because of problems or questions following hospital discharge, categorizes these problems, determines when the patients called following their discharge, and identifies those who called as being at increased risk for early rehospitalizations and unscheduled urgent or emergent care visits. Given the financial penalties issued to hospitals with high 30‐day readmission rates, these patients may warrant more attention than is customarily available from telephone call lines or during routine outpatient follow‐up. The majority of patients who called our AL had Medicare, Medicaid, or a commercial insurance, and, accordingly, may have been eligible for additional services such as home visits and/or expedited follow‐up appointments.

Our study has a number of limitations. First, it is a single‐center study, so the results might not generalize to other institutions. Second, because the study was performed in a university‐affiliated, public safety‐net hospital, patient characteristics and the rates and types of postdischarge concerns that we observed might differ from those encountered in different types of hospitals and/or from those in nonteaching institutions. We would suggest, however, that the idea of using concerns raised by patients discharged from any type of hospital in calls to ALs may similarly identify problems with that specific hospital's discharge processes. Third, the information collected from the AL came from summaries provided by nurses answering the calls rather than from actual transcripts. This could have resulted in insufficient or incorrect information pertaining to some of the variables assessed in Table 2. The information presented in Table 1, however, was obtained from our data warehouse after matching medical record numbers. Fourth, we could have underestimated the number of patients who had 30‐day rehospitalizations and/or unplanned for urgent or emergent care visits if patients sought care at other hospitals. Fifth, the number of patients calling the AL was too small to allow us to do any type of robust matching or multivariable analysis. Accordingly, the differences that appeared between patients who called and those who did not (ie, English speakers, being medically indigent, the length of stay for the index hospitalization and the discharging service) could be the result of inadequate matching or interactions among the variables. Although matching or multivariate analysis might have yielded different associations between patients who called the AL versus those who did not, those who called the AL still had an increased risk of readmission and urgent or emergent visits and may still benefit from targeted interventions. Finally, the fact that only 1.5% of unique patients who were discharged called the AL could have biased our results. Because only 55% and 53% of the patients who did or did not call the AL, respectively, saw primary care physicians within our system within the 3 years prior to their index hospitalization (P=0.679), the frequency of calls to the AL that we observed could have underestimated the frequency with which patients had contact with other care providers in the community.

In summary, information collected from patient‐initiated calls to our AL identified several aspects of our discharge processes that needed improvement. We concluded that our predischarge educational processes for both medicine and surgery services needed modification, especially with respect to pain management, which problems to expect after hospitalization or surgery, and how to deal with them. The high rates of 30‐day rehospitalization and of unscheduled urgent or emergent care visits among patients calling the AL identifies them as being at increased risk for these outcomes, although the likelihood of these events may be related to factors other than just calling the AL.

References
  1. Parrish MM, O'Malley K, Adams RI, Adams SR, Coleman EA. Implementation of the care transitions intervention: sustainability and lessons learned. Prof Case Manag. 2009;14(6):282293.
  2. Arora VM, Prochaska ML, Farnan JM, et al. Problems after discharge and understanding of communication with their primary care physicians among hospitalized seniors: a mixed methods study. J Hosp Med. 2010;5(7):385391.
  3. Forster AJ, Clark HD, Menard A, et al. Adverse events among medical patients after discharge from hospital. CMAJ. 2004;170(3):345349.
  4. Forster AJ, Murff HJ, Peterson JF, Gandhi TK, Bates DW. The incidence and severity of adverse events affecting patients after discharge from the hospital. Ann Intern Med. 2003;138(3):161167.
  5. Misky GJ, Wald HL, Coleman EA. Post‐hospitalization transitions: examining the effects of timing of primary care provider follow‐up. J Hosp Med. 2010;5(7):392397.
  6. Bostrom J, Caldwell J, McGuire K, Everson D. Telephone follow‐up after discharge from the hospital: does it make a difference? Appl Nurs Res. 1996;9(2) 4752.
  7. Sorknaes AD, Bech M, Madsen H, et al. The effect of real‐time teleconsultations between hospital‐based nurses and patients with severe COPD discharged after an exacerbation. J Telemed Telecare. 2013;19(8):466474.
  8. Kwok T, Lum CM, Chan HS, Ma HM, Lee D, Woo J. A randomized, controlled trial of an intensive community nurse‐supported discharge program in preventing hospital readmissions of older patients with chronic lung disease. J Am Geriatr Soc. 2004;52(8):12401246.
  9. Jaarsma T, Halfens R, Huijer Abu‐Saad H, et al. Effects of education and support on self‐care and resource utilization in patients with heart failure. Eur Heart J. 1999;20(9):673682.
  10. Naylor MD, Brooten D, Campbell R, et al. Comprehensive discharge planning and home follow‐up of hospitalized elders: a randomized clinical trial. JAMA. 1999;281(7):613620.
  11. Hansen LO, Young RS, Hinami K, Leung A, Williams MV. Interventions to reduce 30‐day rehospitalization: a systematic review. Ann Intern Med. 2011;155(8):520528.
  12. Rennke S, Kesh S, Neeman N, Sehgal NL. Complementary telephone strategies to improve postdischarge communication. Am J Med. 2012;125(1):2830.
  13. Shu CC, Hsu NC, Lin YF, Wang JY, Lin JW, Ko WJ. Integrated postdischarge transitional care in a hospitalist system to improve discharge outcome: an experimental study. BMC Med. 2011;9:96.
  14. Hinami K, Bilimoria KY, Kallas PG, Simons YM, Christensen NP, Williams MV. Patient experiences after hospitalizations for elective surgery. Am J Surg. 2014;207(6):855862.
  15. Kable A, Gibberd R, Spigelman A. Complications after discharge for surgical patients. ANZ J Surg. 2004;74(3):9297.
  16. Visser A, Ubbink DT, Gouma DJ, Goslings JC. Surgeons are overlooking post‐discharge complications: a prospective cohort study. World J Surg. 2014;38(5):10191025.
  17. Henderson A, Zernike W. A study of the impact of discharge information for surgical patients. J Adv Nurs. 2001;35(3):435441.
References
  1. Parrish MM, O'Malley K, Adams RI, Adams SR, Coleman EA. Implementation of the care transitions intervention: sustainability and lessons learned. Prof Case Manag. 2009;14(6):282293.
  2. Arora VM, Prochaska ML, Farnan JM, et al. Problems after discharge and understanding of communication with their primary care physicians among hospitalized seniors: a mixed methods study. J Hosp Med. 2010;5(7):385391.
  3. Forster AJ, Clark HD, Menard A, et al. Adverse events among medical patients after discharge from hospital. CMAJ. 2004;170(3):345349.
  4. Forster AJ, Murff HJ, Peterson JF, Gandhi TK, Bates DW. The incidence and severity of adverse events affecting patients after discharge from the hospital. Ann Intern Med. 2003;138(3):161167.
  5. Misky GJ, Wald HL, Coleman EA. Post‐hospitalization transitions: examining the effects of timing of primary care provider follow‐up. J Hosp Med. 2010;5(7):392397.
  6. Bostrom J, Caldwell J, McGuire K, Everson D. Telephone follow‐up after discharge from the hospital: does it make a difference? Appl Nurs Res. 1996;9(2) 4752.
  7. Sorknaes AD, Bech M, Madsen H, et al. The effect of real‐time teleconsultations between hospital‐based nurses and patients with severe COPD discharged after an exacerbation. J Telemed Telecare. 2013;19(8):466474.
  8. Kwok T, Lum CM, Chan HS, Ma HM, Lee D, Woo J. A randomized, controlled trial of an intensive community nurse‐supported discharge program in preventing hospital readmissions of older patients with chronic lung disease. J Am Geriatr Soc. 2004;52(8):12401246.
  9. Jaarsma T, Halfens R, Huijer Abu‐Saad H, et al. Effects of education and support on self‐care and resource utilization in patients with heart failure. Eur Heart J. 1999;20(9):673682.
  10. Naylor MD, Brooten D, Campbell R, et al. Comprehensive discharge planning and home follow‐up of hospitalized elders: a randomized clinical trial. JAMA. 1999;281(7):613620.
  11. Hansen LO, Young RS, Hinami K, Leung A, Williams MV. Interventions to reduce 30‐day rehospitalization: a systematic review. Ann Intern Med. 2011;155(8):520528.
  12. Rennke S, Kesh S, Neeman N, Sehgal NL. Complementary telephone strategies to improve postdischarge communication. Am J Med. 2012;125(1):2830.
  13. Shu CC, Hsu NC, Lin YF, Wang JY, Lin JW, Ko WJ. Integrated postdischarge transitional care in a hospitalist system to improve discharge outcome: an experimental study. BMC Med. 2011;9:96.
  14. Hinami K, Bilimoria KY, Kallas PG, Simons YM, Christensen NP, Williams MV. Patient experiences after hospitalizations for elective surgery. Am J Surg. 2014;207(6):855862.
  15. Kable A, Gibberd R, Spigelman A. Complications after discharge for surgical patients. ANZ J Surg. 2004;74(3):9297.
  16. Visser A, Ubbink DT, Gouma DJ, Goslings JC. Surgeons are overlooking post‐discharge complications: a prospective cohort study. World J Surg. 2014;38(5):10191025.
  17. Henderson A, Zernike W. A study of the impact of discharge information for surgical patients. J Adv Nurs. 2001;35(3):435441.
Issue
Journal of Hospital Medicine - 9(11)
Issue
Journal of Hospital Medicine - 9(11)
Page Number
695-699
Page Number
695-699
Publications
Publications
Article Type
Display Headline
Postdischarge problems identified by telephone calls to an advice line
Display Headline
Postdischarge problems identified by telephone calls to an advice line
Sections
Article Source

© 2014 Society of Hospital Medicine

Disallow All Ads
Correspondence Location
Address for correspondence and reprint requests: Sarah A. Stella, MD, Denver Health, 777 Bannock, MC 4000, Denver, CO 80204; Telephone: 303‐596‐1511; Fax: 303‐602‐5056; E‐mail: sarah.stella@dhha.org
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Article PDF Media
Image
Disable zoom
Off
Media Files
Image
Disable zoom
Off

Caring About Prognosis

Article Type
Changed
Sun, 05/21/2017 - 17:42
Display Headline
Caring about prognosis: A validation study of the caring criteria to identify hospitalized patients at high risk for death at 1 year

Prognostication continues to be a challenge to the clinician despite over 100 prognostic indices that have been developed during the past few decades to inform clinical practice and medical decision making.[1] Physicians are not accurate in prognostication of patients' risk of death and tend to overestimate survival.[2, 3] In addition, many physicians do not feel comfortable offering a prognosis to patients, despite patients' wishes to be informed.[4, 5] Regardless of the prevalence in the literature and value in improving physicians' prognostic accuracy, prognostic indices of survival are not regularly utilized in the hospital setting. Prognostic tools available for providers are often complicated and may require data about patients that are not readily available.[6, 7, 8] Prognostic indices may be too specific to a patient population, too difficult to remember, or too time consuming to use. A simple, rapid, and practical prognostic index is important in the hospital setting to assist in identifying patients at high risk of death so that primary palliative interventions can be incorporated into the plan of care early in the hospital stay. Patient and family education, advance care planning, formulating the plan of care based on patientfamily goals, and improved resource utilization could be better executed by more accurate risk of death prediction on hospital admission.

The CARING criteria are the only prognostic index to our knowledge that evaluates a patient's risk of death in the next year, with information readily available at the time of hospital admission (Table 1).[9] The CARING criteria are a unique prognostic tool: (1) CARING is a mnemonic acronym, making it more user friendly to the clinician. (2) The 5 prognostic indicators are readily available from the patient's chart on admission; gathering further data by patient or caretaker interviews or by obtaining laboratory data is not needed. (3) The timing for application of the tool on admission to the hospital is an ideal opportunity to intervene and introduce palliative interventions early on the hospital stay. The CARING criteria were developed and validated in a Veteran's Administration hospital setting by Fischer et al.[9] We sought to validate the CARING criteria in a broader patient populationmedical and surgical patients from a tertiary referral university hospital setting and a safety‐net hospital setting.

METHODS

Study Design

This study was a retrospective observational cohort study. The study was approved by the Colorado Multiple Institutional Review Board and the University of Colorado Hospital Research Review Committee.

Study Purpose

To validate the CARING criteria in a tertiary referral university hospital (University of Colorado Hospital [UCH]) and safety‐net hospital (Denver Health and Hospitals [DHH]) setting using similar methodology to that employed by the original CARING criteria study.[9]

Study Setting/Population

All adults (18 years of age) admitted as inpatients to the medical and surgical services of internal medicine, hospitalist, pulmonary, cardiology, hematology/oncology, hepatology, surgery, intensive care unit, and intermediary care unit at UCH and DHH during the study period of July 2005 through August 2005. The only exclusion criteria were those patients who were prisoners or pregnant. Administrative admission data from July 2005 to August 2005 were used to identify names of all persons admitted to the medicine and surgical services of the study hospitals during the specified time period.

The 2 study hospitals, UCH and DHH, provide a range of patients who vary in ethnicity, socioeconomic status, and medical illness. This variability allows for greater generalizability of the results. Both hospitals are affiliated with the University of Colorado School of Medicine internal medicine residency training program and are located in Denver, Colorado.

At the time of the study, UCH was a licensed 550‐bed tertiary referral, academic hospital serving the Denver metropolitan area and the Rocky Mountain region as a specialty care and referral center. DHH was a 398‐bed, academic, safety‐net hospital serving primarily the Denver metropolitan area. DHH provides 42% of the care for the uninsured in Denver and 26% of the uninsured care for the state of Colorado.

Measures

The CARING criteria were developed and validated in a Veteran's Administration (VA) hospital setting by Fischer et al.[9] The purpose of the CARING criteria is to identify patients, at the time of hospital admission, who are at higher risk of death in the following year. The prognostic index uses 5 predictors that can be abstracted from the chart at time of admission. The CARING criteria were developed a priori, and patients were evaluated using only the medical data available at the time of admission. The criteria include items that are already part of the routine physician admission notes and do not require additional data collection or assessments. The criteria include: C=primary diagnosis of cancer, A=2 admissions to the hospital for a chronic illness within the last year; R=resident in a nursing home; I=intensive care unit (ICU) admission with multiorgan failure (MOF), NG=noncancer hospice guidelines (meeting 2 of the National Hospice and Palliative Care Organization's [NHPCO] guidelines).

Patients were identified using name, date of birth, social security number, address, and phone number. This identifying information was then used for tracing death records 1 year after hospital admission.

Mortality at 1 year following the index hospitalization was the primary end point. To minimize missing data and the number of subjects lost to follow‐up, 3 determinants of mortality were used. First, electronic medical records of the 2 participating hospitals and their outpatient clinics were reviewed to determine if a follow‐up appointment had occurred past the study's end point of 1 year (August 2006). For those without a confirmed follow‐up visit, death records from the Colorado Department of Public Health and Vital Records were obtained. For those patients residing outside of Colorado or whose mortality status was still unclear, the National Death Index was accessed.

Medical Record Review

Medical records for all study participants were reviewed by J.Y. (UCH) and B.C. (DHH). Data collection was completed using direct data entry into a Microsoft Access (Microsoft Corp., Redmond, WA) database utilizing a data entry form linked with the database table. This form utilized skip patterns and input masks to ensure quality of data entry and minimize missing or invalid data. Inter‐rater reliability was assessed by an independent rereview (S.F.) of 5% of the total charts. Demographic variables were collected using hospital administrative data. These included personal identifiers of the participants for purposes of mortality follow‐up. Clinical data including the 5 CARING variables and additional descriptive variables were abstracted from the paper hospital chart and the electronic record of the chart (together these constitute the medical record).

Death Follow‐up

A search of Colorado death records was conducted in February 2011 for all subjects. Death records were used to determine mortality and time to death from the index hospitalization. The National Death Index was then searched for any subjects without or record of death in Colorado.

Analysis

All analyses were conducted using the statistical application software SAS for Windows version 9.3 (SAS Institute, Cary, NC). Simple frequencies and means ( standard deviation) were used to describe the baseline characteristics. Multiple logistic regression models were used to model 1‐year mortality. The models were fitted using all of the CARING variables and age. As the aim of the study was to validate the CARING criteria, the variables for the models were selected a priori based on the original index. Two hospital cohorts (DHH and UCH) were modeled separately and as a combined sample. Kaplan‐Meier survival analysis was conducted to compare those subjects who met 1 of the CARING criteria with those who did not through the entire period of mortality follow‐up (20052011). Finally, using the probabilities from the logistic regression models, we again developed a scoring rule appropriate for a non‐VA setting to allow clinicians to easily identify patient risk for 1‐year mortality at the time of hospital admission.

RESULTS

There were a total of 1064 patients admitted to the medical and surgical services during the study period568 patients at DHH and 496 patients at UCH. Sample characteristics of each individual hospital cohort and the entire combined study cohort are detailed in Table 2. Overall, slightly over half the population were male, with a mean age of 50 years, and the ethnic breakdown roughly reflects the population in Denver. A total of 36.5% (n=388) of the study population met 1 of the CARING criteria, and 12.6% (n=134 among 1063 excluding 1 without an admit date) died within 1 year of the index hospitalization. These were younger and healthier patients compared to the VA sample used in developing the CARING criteria.

CARING Criteria
  • NOTE: The CARING criteria must be applied to patients who are hospitalized on the first day after admission (ie, they met the criteria on the day of admission). It is unknown if the CARING criteria are predictive of high mortality when applied to patients who are either not in the hospital or later in the hospital stay. Cancer: Is there a primary diagnosis of cancer? This includes patients who are admitted for chemotherapy (most chemotherapy is administered as an outpatient, and patients who require hospitalization for administration of chemotherapy are likely more ill or have more aggressive cancers requiring more intensive monitoring) due to complications from their chemotherapy (ie, neutropenic fever), or for aggressive symptom management. What is important about this criterion is that cancer must be the primary reason they are admitted. A person with colon cancer admitted for suspected angina would not qualify. Admitted to the hospital for 2 times in the past year for a chronic illness. For example, a man is admitted with pneumonia and COPD exacerbation, and looking back at his chart you discover that he was also admitted for a COPD exacerbation 2 months ago. That would add up to 2 hospital admissions in the past year for a chronic illness; therefore, the patient would meet this criterion. A patient admitted twice in the past year for musculoskeletal chest pain would not meet the criterion. Resident in a nursing home: A patient admitted from either a long‐term care facility or a skilled nursing facility would meet this criterion. It is essentially a proxy for poor functional status. ICU admission with multiorgan failure: An example would be a patient admitted to the ICU requiring mechanical ventilation (pulmonary system in failure) and on pressors or in renal failure (either requiring dialysis or nearing that point; a small increase in the creatinine would not qualify for organ failure). Noncancer hospice guidelines: Patient must meet at least 2 items in any given category. Abbreviations: AIDS, acquired immunodeficiency syndrome; BiPAP, bilevel positive airway pressure; BP, blood pressure; CARING, C=primary diagnosis of cancer, A=2 admissions to the hospital for a chronic illness within the last year; R=resident in a nursing home; I=intensive care unit (ICU) admission with multiorgan failure, NG=noncancer hospice guidelines (meeting 2 of the National Hospice and Palliative Care Organization's guidelines) CHF, congestive heart failure; CNS, central nervous system; COPD, chronic obstructive pulmonary disease; CVA, cardiovascular accident; ER, emergency room; HIV, human immunodeficiency virus; ICU, intensive care unit.

RenalDementia
Stop/decline dialysisUnable to ambulate independently
Not candidate for transplantUrinary or fecal incontinence
Urine output < 40cc/24 hoursUnable to speak with more than single words
Creatinine > 8.0 (>6.0 for diabetics)Unable to bathe independently
Creatinine clearance 10cc/minUnable to dress independently
UremiaCo‐morbid conditions:
Persistent serum K + > 7.0Aspiration pneumonia
Co‐morbid conditions:Pyelonephritis
Cancer CHFDecubitus ulcer
Chronic lung disease AIDS/HIVDifficulty swallowing or refusal to eat
Sepsis Cirrhosis 
CardiacPulmonary
Ejection fraction < 20%Dyspnea at rest
Symptomatic with diuretics and vasodilatorsFEV1 < 30%
Not candidate for transplantFrequent ER or hospital admits for pulmonary infections or respiratory distress
History of cardiac arrestCor pulmonale or right heart failure
History of syncope02 sat < 88% on 02
Systolic BP < 120mmHGPC02 > 50
CVA cardiac originResting tachycardia > 100/min
Co‐morbid conditions as listed in RenalCo‐morbid conditions as listed in Renal
LiverStroke/CVA
End stage cirrhosisComa at onset
Not candidate for transplantComa >3 days
Protime > 5sec and albumin <2.5Limb paralysis
Ascites unresponsive to treatmentUrinary/fecal incontinence
Hepatorenal syndromeImpaired sitting balance
Hepatic encephalopathyKarnofsky < 50%
Spontaneous bacterial peritonitisRecurrent aspiration
Recurrent variceal bleedAge > 70
Co‐morbid conditions as listed in RenalCo‐morbid conditions as listed in Renal
HIV/AIDSNeuromuscular
Persistent decline in functionDiminished respiratory function
Chronic diarrhea 1 yearChosen not to receive BiPAP/vent
Decision to stop treatmentDifficulty swallowing
CNS lymphomaDiminished functional status
MAC‐untreatedIncontinence
Systemic lymphomaCo‐morbid conditions as listed in Renal
Dilated cardiomyopathy 
CD4 < 25 with disease progression 
Viral load > 100,000 
Validation Study Cohort Characteristics
 Safety‐Net Hospital Cohort, N=568Academic Center Cohort, N=496Study Cohort,N=1064Original CARING Cohort, N=8739
  • NOTE: Cases with missing data were negligible (<4%). Abbreviations: CARING, C=primary diagnosis of cancer, A=2 admissions to the hospital for a chronic illness within the last year; R=resident in a nursing home; I=intensive care unit (ICU) admission with multiorgan failure (MOF), NG=noncancer hospice guidelines (meeting 2 of the National Hospice and Palliative Care Organization's [NHPCO] guidelines); SD, standard deviation.

Mean age ( SD), y47.8 (16.5)54.4 (17.5)50.9 (17.3)63 (13)
Male gender59.5% (338)50.1% (248)55.1% (586)98% (856)
Ethnicity    
African American14.1% (80)13.5% (65)13.8% (145)13% (114)
Asian0.4% (2)1.5% (7)0.9% (9)Not reported
Caucasian41.7% (237)66.3% (318)53.0 % (555)69% (602)
Latino41.9% (238)9.6% (46)27.1% (284)8% (70)
Native American0.5% (3)0.4% (2)0.5% (5)Not reported
Other0.5% (3)0.6% (3)0.6% (6)10% (87)
Unknown0.9% (5)8.1% (39)4.2% (44)Not reported
CARING criteria    
Cancer6.2% (35)19.4% (96)12.3% (131)23% (201)
Admissions to the hospital 2 in past year13.6% (77)42.7% (212)27.2% (289)36% (314)
Resident in a nursing home1.8% (10)3.4% (17)2.5% (27)3% (26)
ICU with MOF3.7% (21)1.2% (6)2.5% (27)2% (17)
NHPCO (2) noncancer guidelines1.6% (9)5.9% (29)3.6% (38)8% (70)

Reliability testing demonstrated excellent inter‐rater reliability. Kappa for each criterion is as follows: (1) primary diagnosis of cancer=1.0, (2) 2 admissions to the hospital in the past year=0.91, (3) resident in a nursing home=1.0, (4) ICU admission with MOF=1.0, and (5) 2 noncancer hospice guidelines=0.78.

This study aimed to validate the CARING criteria9; therefore, all original individual CARING criterion were included in the validation logistic regression models. The 1 exception to this was in the university hospital study cohort, where the ICU criterion was excluded from the model due to small sample size and quasiseparation in the model. The model results are presented in Table 3 for the individual hospitals and combined study cohort.

Prediction of 1‐Year Mortality Using CARING Criteria
 Safety Net Hospital Cohort, C Index=0.76Academic Center Cohort, C Index=0.76Combined Hospital Cohort, C Index=0.79
 EstimateOdds Ratio (95% CI)EstimateOdds Ratio (95% CI)EstimateOdds Ratio (95% CI)
  • NOTE: Abbreviations: CARING, C=primary diagnosis of cancer, A=2 admissions to the hospital for a chronic illness within the last year; R=resident in a nursing home; I=intensive care unit (ICU) admission with multiorgan failure (MOF), N=noncancer hospice guidelines (meeting 2 of the National Hospice and Palliative Care Organization's [NHPCO] guidelines); CI, confidence interval.

  • Odds ratio is statistically significant as evidenced by the CI that does not cross 1.0.

  • Age was divided into 4 categories: 55 years, 5665 years, 6675 years, and >75 years.

Cancer1.926.85 (2.83‐16.59)a1.856.36 (3.54‐11.41)a1.987.23 (4.45‐11.75)a
Admissions to the hospital 2 in past year0.551.74 (0.76‐3.97)0.140.87 (0.51‐1.49)0.201.22 (0.78‐1.91)
Resident in a nursing home0.490.61 (0.06‐6.56)0.271.31 (0.37‐4.66)0.091.09 (0.36‐3.32)
ICU with MOF1.856.34 (2.0219.90)a  1.946.97 (2.75‐17.68)a
NHPCO (2) noncancer guidelines3.0420.86 (4.25102.32)a2.6213.73 (5.86‐32.15)a2.7415.55 (7.2833.23)a
Ageb0.381.46 (1.05‐2.03)a0.451.56 (1.23‐1.98)a0.471.60 (1.32‐1.93)a

In the safety‐net hospital, admission to the hospital with a primary diagnosis related to cancer, 2 noncancer hospice guidelines, ICU admission with MOF, and age by category all were significant predictors of 1‐year mortality. In the university hospital cohort, primary diagnosis of cancer, 2 noncancer hospice guidelines, and age by category were predictive of 1‐year mortality. Finally, in the entire study cohort, primary diagnosis of cancer, ICU with MOF, 2 noncancer hospice guidelines, and age were all predictive of 1‐year mortality. Parameter estimates were similar in 3 of the criteria compared to the VA setting. Differences in patient characteristics may have caused the differences in the estimates. Gender was additionally tested but not significant in any model. One‐year survival was significantly lower for those who met 1 of the CARING criteria versus those who did not (Figure 1).

jhm2107-fig-0001-m.png
Survival plot for those subjects who did (CARING ≥1) or did not (CARING = 0) meet at least 1 of the CARING criteria. Abbreviations: CARING, C = primary diagnosis of cancer, A = ≥2 admissions to the hospital for a chronic illness within the last year; R = resident in a nursing home; I = intensive care unit (ICU) admission with multiorgan failure, N = noncancer hospice guidelines (meeting ≥2 of the National Hospice and Palliative Care Organization's guidelines).

Based on the framework from the original CARING criteria analysis, a scoring rule was developed using the regression results of this validation cohort. To predict a high probability of 1‐year mortality, sensitivity was set to 58% and specificity was set at 86% (error rate=17%). Medium to high probability was set with a sensitivity of 73% and specificity of 72% (error rate=28%). The coefficients from the regression model of the entire study cohort were converted to scores for each of the CARING criteria. The scores are as follows: 0.5 points for admission from a nursing home, 1 point for 2 hospital admissions in the past year for a chronic illness, 10 points for primary diagnosis of cancer, 10 points for ICU admission with MOF, and 14 points for 2 noncancer hospice guidelines. For every age category increase, 2 points are assigned so that 0 points for age <55 years, 2 points for ages 56 to 65 years, 4 points for ages 66 to 75 years, and 6 points for >75 years. Points for individual risk factors were proportional to s (ie, log odds) in the logistic regression model for death at 1 year. Although no linear transformation exists between s and probabilities (of death at 1 year), the aggregated points for combinations of risk factors shown in Table 4 follow the probabilities in an approximately linear fashion, so that different degrees of risk of death can be represented contiguously (as highlighted by differently shaded regions in the scoring matrix) (Table 4). The scoring matrix allows for quick identification for patients at high risk for 1‐year mortality. In this non‐VA setting with healthier patients, low risk is defined at a lower probability threshold (0.1) compared to the VA setting (0.175).

Score of Risk of Death at 1 Year for the CARING Criteria by Age Group
 CARING Criteria Components
 NoneResident in a Nursing HomeAdmitted to the Hospital 2 Times in the Past YearResident in a Nursing Home Admitted to the Hospital 2 Times in the Past YearPrimary Diagnosis of CancerICU Admission With MOFNoncancer Hospice Guidelines
  • NOTE: Abbreviations: CARING, C=primary diagnosis of cancer, A=2 admissions to the hospital for a chronic illness within the last year; R=resident in a nursing home; I=intensive care unit (ICU) admission with multiorgan failure (MOF), N=noncancer hospice guidelines (meeting 2 of the National Hospice and Palliative Care Organization's guidelines).

Age       
55 years00.511.5 10 
5565 years22.533.5 10 
6675 years44.555.5 10 
>75 years66.577.5 10 
Risk       
Low 3.5Probability<0.1  
Medium 46.50.1probability <0.175  
High 7Probability0.175  

DISCUSSION

The CARING criteria are a practical prognostic tool that can be easily and rapidly applied to patients admitted to the hospital to estimate risk of death in 1 year, with the goal of identifying patients who may benefit most from incorporating palliative interventions into their plan of care. This study validated the CARING criteria in a tertiary referral university hospital and safety‐net hospital setting, demonstrating applicability in a much broader population than the VA hospital of the original CARING criteria study. The population studied represented a younger population by over 10 years, a more equitable proportion of males to females, a broader ethnic diversity, and lower 1‐year deaths rates than the original study. Despite the broader representation of the population, the significance of each of the individual CARING criterion was maintained except for 2 hospital admissions in the past year for a chronic illness (admission from a nursing home did not meet significance in either study as a sole criterion). As with the original study, meeting 2 of the NHPCO noncancer hospice guidelines demonstrated the highest risk of 1‐year mortality following index hospitalization, followed by primary diagnosis of cancer and ICU admission with MOF. Advancing age, also similar to the original study, conferred increased risk across the criterion.

Hospitalists could be an effective target for utilizing the CARING criteria because they are frequently the first‐line providers in the hospital setting. With the national shortage of palliative care specialists, hospitalists need to be able to identify when a patient has a limited life expectancy so they will be better equipped to make clinical decisions that are aligned with their patients' values, preferences, and goals of care. With the realization that not addressing advance care planning and patient goals of care may be considered medical errors, primary palliative care skills become alarmingly more important as priorities for hospitalists to obtain and feel comfortable using in daily practice.

The CARING criteria are directly applicable to patients who are seen by hospitalists. Other prognostic indices have focused on select patient populations, such as the elderly,[10, 11, 12] require collection of data that are not readily available on admission or would not otherwise be obtained,[10, 13] or apply to patients post‐hospital discharge, thereby missing the opportunity to make an impact earlier in the disease trajectory and incorporate palliative care into the hospital plan of care when key discussions about goals of care and preferences should be encouraged.

Additionally, the CARING criteria could easily be incorporated as a trigger for palliative care consults on hospital admission. Palliative care consults tend to happen late in a hospital stay, limiting the effectiveness of the palliative care team. A trigger system for hospitalists and other primary providers on hospital admission would lend to more effective timing of palliative measures being incorporated into the plan of care. Palliative care consults would not only be initiated earlier, but could be targeted for the more complex and sick patients with the highest risk of death in the next year.

In the time‐pressured environment, the presence of any 1 of the CARING criteria can act as a trigger to begin incorporating primary palliative care measures into the plan of care. The admitting hospitalist provider (ie, physician, nurse practitioner, physician assistant) could access the CARING criteria through an electronic health record prompt when admitting patients. When a more detailed assessment of mortality risk is helpful, the hospitalist can use the scoring matrix, which combines age with the individual criterion to calculate patients at medium or high risk of death within 1 year. Limited resources can then be directed to the patients with the greatest need. Patients with a focused care need, such as advance care planning or hospice referral, can be directed to the social worker or case manager. More complicated patients may be referred to a specialty palliative care team.

Several limitations to this study are recognized, including the small sample size of patients meeting criterion for ICU with MOF in the academic center study cohort. The patient data were collected during a transition time when the university hospital moved to a new campus, resulting in an ICU at each campus that housed patients with differing levels of illness severity, which may have contributed to the lower acuity ICU patient observed. Although we advocate the simplicity of the CARING criteria, the NHPCO noncancer hospice guidelines are more complicated, as they incorporates 8 broad categories of chronic illness. The hospice guidelines may not be general knowledge to the hospitalist or other primary providers. ePrognosis (http://eprognosis.ucsf.edu/) has a Web‐based calculator for the CARING criteria, including a link referencing the NHPCO noncancer hospice guidelines. Alternatively, providing a pocket card, smart phone or tablet app, or electronic health record tool containing the NHPCO criteria and CARING criteria could easily overcome this gap in knowledge. Finally, the reviewer agreement was not 100% for each criterion due to personal interpretation differences in the criterion. NHPCO criterion had the lowest kappa, yet it still was 0.78 and achieved a highly acceptable level of agreement.

CONCLUSION

The CARING criteria are a simple, practical prognostic tool predictive of death within 1 year that has been validated in a broad population of hospitalized patients. The criteria hold up in a younger, healthier population that is more diverse by age, gender, and ethnicity than the VA population. With ready access to critical prognostic information on hospital admission, clinicians will be better informed to make decisions that are aligned with their patients' values, preferences, and goals of care.

Disclosure

Nothing to report.

Files
References
  1. Siontis GC, Tzoulaki I, Ioannidis JP. Predicting death: an empirical evaluation of predictive tools for mortality. Arch Intern Med. 2011;171:17211726.
  2. Christakis NA, Lamont EB. Extent and determinants of error in physicians' prognoses in terminally ill patients: prospective cohort study. West J Med. 2000;172:310313.
  3. Glare P, Virik K, Jones M, et al. A systematic review of physicians' survival predictions in terminally ill cancer patients. BMJ. 2003;327:195198.
  4. Christakis NA, Iwashyna TJ. Attitude and self‐reported practice regarding prognostication in a national sample of internists. Arch Intern Med. 1998;158:23892395.
  5. Campbell TC, Carey EC, Jackson VA, et al. Discussing prognosis: balancing hope and realism. Cancer J. 2010;16:461466.
  6. Zimmerman JE, Kramer AA, McNair DS, Malila FM. Acute Physiology and Chronic Health Evaluation (APACHE) IV: hospital mortality assessment for today's critically ill patients. Crit Care Med. 2006;34:12971310.
  7. Ledoux D, Canivet JL, Preiser JC, Lefrancq J, Damas P. SAPS 3 admission score: an external validation in a general intensive care population. Intensive Care Med. 2008;34:18731877.
  8. Higgins TL, Kramer AA, Nathanson BH, Copes W, Stark M, Teres D. Prospective validation of the intensive care unit admission Mortality Probability Model (MPM0‐III). Crit Care Med. 2009;37:16191623.
  9. Fischer SM, Gozansky W, Sauaia A, Min SJ, Kutner JS, Kramer A. A practical tool to identify patients who may benefit from a palliative approach: the CARING criteria. J Pain Symptom Manage. 2006;31:285292.
  10. Teno JM, Harrell FE, Knaus W, et al. Prediction of survival for older hospitalized patients: the HELP survival model. J Am Geriatr Soc. 2000;48:S16S24.
  11. Pilotto A, Ferrucci L, Franceschi M, et al. Development and validation of a multidimensional prognostic index for one‐year mortality from comprehensive geriatric assessment in hospitalized older patients. Rejuvenation Res. 2008;11:151161.
  12. Inouye SK, Bogardus ST, Vitagliano G, et al. Burden of illness score for elderly persons: risk adjustment incorporating the cumulative impact of diseases, physiologic abnormalities, and functional impairments. Med Care. 2003;41:7083.
  13. Knaus WA, Harrell FE, Lynn J, et al. The SUPPORT prognostic model. Objective estimates of survival for seriously ill hospitalized adults. Study to understand prognoses and preferences for outcomes and risks of treatments. Ann Intern Med. 1995;122:191203.
Article PDF
Issue
Journal of Hospital Medicine - 8(12)
Publications
Page Number
696-701
Sections
Files
Files
Article PDF
Article PDF

Prognostication continues to be a challenge to the clinician despite over 100 prognostic indices that have been developed during the past few decades to inform clinical practice and medical decision making.[1] Physicians are not accurate in prognostication of patients' risk of death and tend to overestimate survival.[2, 3] In addition, many physicians do not feel comfortable offering a prognosis to patients, despite patients' wishes to be informed.[4, 5] Regardless of the prevalence in the literature and value in improving physicians' prognostic accuracy, prognostic indices of survival are not regularly utilized in the hospital setting. Prognostic tools available for providers are often complicated and may require data about patients that are not readily available.[6, 7, 8] Prognostic indices may be too specific to a patient population, too difficult to remember, or too time consuming to use. A simple, rapid, and practical prognostic index is important in the hospital setting to assist in identifying patients at high risk of death so that primary palliative interventions can be incorporated into the plan of care early in the hospital stay. Patient and family education, advance care planning, formulating the plan of care based on patientfamily goals, and improved resource utilization could be better executed by more accurate risk of death prediction on hospital admission.

The CARING criteria are the only prognostic index to our knowledge that evaluates a patient's risk of death in the next year, with information readily available at the time of hospital admission (Table 1).[9] The CARING criteria are a unique prognostic tool: (1) CARING is a mnemonic acronym, making it more user friendly to the clinician. (2) The 5 prognostic indicators are readily available from the patient's chart on admission; gathering further data by patient or caretaker interviews or by obtaining laboratory data is not needed. (3) The timing for application of the tool on admission to the hospital is an ideal opportunity to intervene and introduce palliative interventions early on the hospital stay. The CARING criteria were developed and validated in a Veteran's Administration hospital setting by Fischer et al.[9] We sought to validate the CARING criteria in a broader patient populationmedical and surgical patients from a tertiary referral university hospital setting and a safety‐net hospital setting.

METHODS

Study Design

This study was a retrospective observational cohort study. The study was approved by the Colorado Multiple Institutional Review Board and the University of Colorado Hospital Research Review Committee.

Study Purpose

To validate the CARING criteria in a tertiary referral university hospital (University of Colorado Hospital [UCH]) and safety‐net hospital (Denver Health and Hospitals [DHH]) setting using similar methodology to that employed by the original CARING criteria study.[9]

Study Setting/Population

All adults (18 years of age) admitted as inpatients to the medical and surgical services of internal medicine, hospitalist, pulmonary, cardiology, hematology/oncology, hepatology, surgery, intensive care unit, and intermediary care unit at UCH and DHH during the study period of July 2005 through August 2005. The only exclusion criteria were those patients who were prisoners or pregnant. Administrative admission data from July 2005 to August 2005 were used to identify names of all persons admitted to the medicine and surgical services of the study hospitals during the specified time period.

The 2 study hospitals, UCH and DHH, provide a range of patients who vary in ethnicity, socioeconomic status, and medical illness. This variability allows for greater generalizability of the results. Both hospitals are affiliated with the University of Colorado School of Medicine internal medicine residency training program and are located in Denver, Colorado.

At the time of the study, UCH was a licensed 550‐bed tertiary referral, academic hospital serving the Denver metropolitan area and the Rocky Mountain region as a specialty care and referral center. DHH was a 398‐bed, academic, safety‐net hospital serving primarily the Denver metropolitan area. DHH provides 42% of the care for the uninsured in Denver and 26% of the uninsured care for the state of Colorado.

Measures

The CARING criteria were developed and validated in a Veteran's Administration (VA) hospital setting by Fischer et al.[9] The purpose of the CARING criteria is to identify patients, at the time of hospital admission, who are at higher risk of death in the following year. The prognostic index uses 5 predictors that can be abstracted from the chart at time of admission. The CARING criteria were developed a priori, and patients were evaluated using only the medical data available at the time of admission. The criteria include items that are already part of the routine physician admission notes and do not require additional data collection or assessments. The criteria include: C=primary diagnosis of cancer, A=2 admissions to the hospital for a chronic illness within the last year; R=resident in a nursing home; I=intensive care unit (ICU) admission with multiorgan failure (MOF), NG=noncancer hospice guidelines (meeting 2 of the National Hospice and Palliative Care Organization's [NHPCO] guidelines).

Patients were identified using name, date of birth, social security number, address, and phone number. This identifying information was then used for tracing death records 1 year after hospital admission.

Mortality at 1 year following the index hospitalization was the primary end point. To minimize missing data and the number of subjects lost to follow‐up, 3 determinants of mortality were used. First, electronic medical records of the 2 participating hospitals and their outpatient clinics were reviewed to determine if a follow‐up appointment had occurred past the study's end point of 1 year (August 2006). For those without a confirmed follow‐up visit, death records from the Colorado Department of Public Health and Vital Records were obtained. For those patients residing outside of Colorado or whose mortality status was still unclear, the National Death Index was accessed.

Medical Record Review

Medical records for all study participants were reviewed by J.Y. (UCH) and B.C. (DHH). Data collection was completed using direct data entry into a Microsoft Access (Microsoft Corp., Redmond, WA) database utilizing a data entry form linked with the database table. This form utilized skip patterns and input masks to ensure quality of data entry and minimize missing or invalid data. Inter‐rater reliability was assessed by an independent rereview (S.F.) of 5% of the total charts. Demographic variables were collected using hospital administrative data. These included personal identifiers of the participants for purposes of mortality follow‐up. Clinical data including the 5 CARING variables and additional descriptive variables were abstracted from the paper hospital chart and the electronic record of the chart (together these constitute the medical record).

Death Follow‐up

A search of Colorado death records was conducted in February 2011 for all subjects. Death records were used to determine mortality and time to death from the index hospitalization. The National Death Index was then searched for any subjects without or record of death in Colorado.

Analysis

All analyses were conducted using the statistical application software SAS for Windows version 9.3 (SAS Institute, Cary, NC). Simple frequencies and means ( standard deviation) were used to describe the baseline characteristics. Multiple logistic regression models were used to model 1‐year mortality. The models were fitted using all of the CARING variables and age. As the aim of the study was to validate the CARING criteria, the variables for the models were selected a priori based on the original index. Two hospital cohorts (DHH and UCH) were modeled separately and as a combined sample. Kaplan‐Meier survival analysis was conducted to compare those subjects who met 1 of the CARING criteria with those who did not through the entire period of mortality follow‐up (20052011). Finally, using the probabilities from the logistic regression models, we again developed a scoring rule appropriate for a non‐VA setting to allow clinicians to easily identify patient risk for 1‐year mortality at the time of hospital admission.

RESULTS

There were a total of 1064 patients admitted to the medical and surgical services during the study period568 patients at DHH and 496 patients at UCH. Sample characteristics of each individual hospital cohort and the entire combined study cohort are detailed in Table 2. Overall, slightly over half the population were male, with a mean age of 50 years, and the ethnic breakdown roughly reflects the population in Denver. A total of 36.5% (n=388) of the study population met 1 of the CARING criteria, and 12.6% (n=134 among 1063 excluding 1 without an admit date) died within 1 year of the index hospitalization. These were younger and healthier patients compared to the VA sample used in developing the CARING criteria.

CARING Criteria
  • NOTE: The CARING criteria must be applied to patients who are hospitalized on the first day after admission (ie, they met the criteria on the day of admission). It is unknown if the CARING criteria are predictive of high mortality when applied to patients who are either not in the hospital or later in the hospital stay. Cancer: Is there a primary diagnosis of cancer? This includes patients who are admitted for chemotherapy (most chemotherapy is administered as an outpatient, and patients who require hospitalization for administration of chemotherapy are likely more ill or have more aggressive cancers requiring more intensive monitoring) due to complications from their chemotherapy (ie, neutropenic fever), or for aggressive symptom management. What is important about this criterion is that cancer must be the primary reason they are admitted. A person with colon cancer admitted for suspected angina would not qualify. Admitted to the hospital for 2 times in the past year for a chronic illness. For example, a man is admitted with pneumonia and COPD exacerbation, and looking back at his chart you discover that he was also admitted for a COPD exacerbation 2 months ago. That would add up to 2 hospital admissions in the past year for a chronic illness; therefore, the patient would meet this criterion. A patient admitted twice in the past year for musculoskeletal chest pain would not meet the criterion. Resident in a nursing home: A patient admitted from either a long‐term care facility or a skilled nursing facility would meet this criterion. It is essentially a proxy for poor functional status. ICU admission with multiorgan failure: An example would be a patient admitted to the ICU requiring mechanical ventilation (pulmonary system in failure) and on pressors or in renal failure (either requiring dialysis or nearing that point; a small increase in the creatinine would not qualify for organ failure). Noncancer hospice guidelines: Patient must meet at least 2 items in any given category. Abbreviations: AIDS, acquired immunodeficiency syndrome; BiPAP, bilevel positive airway pressure; BP, blood pressure; CARING, C=primary diagnosis of cancer, A=2 admissions to the hospital for a chronic illness within the last year; R=resident in a nursing home; I=intensive care unit (ICU) admission with multiorgan failure, NG=noncancer hospice guidelines (meeting 2 of the National Hospice and Palliative Care Organization's guidelines) CHF, congestive heart failure; CNS, central nervous system; COPD, chronic obstructive pulmonary disease; CVA, cardiovascular accident; ER, emergency room; HIV, human immunodeficiency virus; ICU, intensive care unit.

RenalDementia
Stop/decline dialysisUnable to ambulate independently
Not candidate for transplantUrinary or fecal incontinence
Urine output < 40cc/24 hoursUnable to speak with more than single words
Creatinine > 8.0 (>6.0 for diabetics)Unable to bathe independently
Creatinine clearance 10cc/minUnable to dress independently
UremiaCo‐morbid conditions:
Persistent serum K + > 7.0Aspiration pneumonia
Co‐morbid conditions:Pyelonephritis
Cancer CHFDecubitus ulcer
Chronic lung disease AIDS/HIVDifficulty swallowing or refusal to eat
Sepsis Cirrhosis 
CardiacPulmonary
Ejection fraction < 20%Dyspnea at rest
Symptomatic with diuretics and vasodilatorsFEV1 < 30%
Not candidate for transplantFrequent ER or hospital admits for pulmonary infections or respiratory distress
History of cardiac arrestCor pulmonale or right heart failure
History of syncope02 sat < 88% on 02
Systolic BP < 120mmHGPC02 > 50
CVA cardiac originResting tachycardia > 100/min
Co‐morbid conditions as listed in RenalCo‐morbid conditions as listed in Renal
LiverStroke/CVA
End stage cirrhosisComa at onset
Not candidate for transplantComa >3 days
Protime > 5sec and albumin <2.5Limb paralysis
Ascites unresponsive to treatmentUrinary/fecal incontinence
Hepatorenal syndromeImpaired sitting balance
Hepatic encephalopathyKarnofsky < 50%
Spontaneous bacterial peritonitisRecurrent aspiration
Recurrent variceal bleedAge > 70
Co‐morbid conditions as listed in RenalCo‐morbid conditions as listed in Renal
HIV/AIDSNeuromuscular
Persistent decline in functionDiminished respiratory function
Chronic diarrhea 1 yearChosen not to receive BiPAP/vent
Decision to stop treatmentDifficulty swallowing
CNS lymphomaDiminished functional status
MAC‐untreatedIncontinence
Systemic lymphomaCo‐morbid conditions as listed in Renal
Dilated cardiomyopathy 
CD4 < 25 with disease progression 
Viral load > 100,000 
Validation Study Cohort Characteristics
 Safety‐Net Hospital Cohort, N=568Academic Center Cohort, N=496Study Cohort,N=1064Original CARING Cohort, N=8739
  • NOTE: Cases with missing data were negligible (<4%). Abbreviations: CARING, C=primary diagnosis of cancer, A=2 admissions to the hospital for a chronic illness within the last year; R=resident in a nursing home; I=intensive care unit (ICU) admission with multiorgan failure (MOF), NG=noncancer hospice guidelines (meeting 2 of the National Hospice and Palliative Care Organization's [NHPCO] guidelines); SD, standard deviation.

Mean age ( SD), y47.8 (16.5)54.4 (17.5)50.9 (17.3)63 (13)
Male gender59.5% (338)50.1% (248)55.1% (586)98% (856)
Ethnicity    
African American14.1% (80)13.5% (65)13.8% (145)13% (114)
Asian0.4% (2)1.5% (7)0.9% (9)Not reported
Caucasian41.7% (237)66.3% (318)53.0 % (555)69% (602)
Latino41.9% (238)9.6% (46)27.1% (284)8% (70)
Native American0.5% (3)0.4% (2)0.5% (5)Not reported
Other0.5% (3)0.6% (3)0.6% (6)10% (87)
Unknown0.9% (5)8.1% (39)4.2% (44)Not reported
CARING criteria    
Cancer6.2% (35)19.4% (96)12.3% (131)23% (201)
Admissions to the hospital 2 in past year13.6% (77)42.7% (212)27.2% (289)36% (314)
Resident in a nursing home1.8% (10)3.4% (17)2.5% (27)3% (26)
ICU with MOF3.7% (21)1.2% (6)2.5% (27)2% (17)
NHPCO (2) noncancer guidelines1.6% (9)5.9% (29)3.6% (38)8% (70)

Reliability testing demonstrated excellent inter‐rater reliability. Kappa for each criterion is as follows: (1) primary diagnosis of cancer=1.0, (2) 2 admissions to the hospital in the past year=0.91, (3) resident in a nursing home=1.0, (4) ICU admission with MOF=1.0, and (5) 2 noncancer hospice guidelines=0.78.

This study aimed to validate the CARING criteria9; therefore, all original individual CARING criterion were included in the validation logistic regression models. The 1 exception to this was in the university hospital study cohort, where the ICU criterion was excluded from the model due to small sample size and quasiseparation in the model. The model results are presented in Table 3 for the individual hospitals and combined study cohort.

Prediction of 1‐Year Mortality Using CARING Criteria
 Safety Net Hospital Cohort, C Index=0.76Academic Center Cohort, C Index=0.76Combined Hospital Cohort, C Index=0.79
 EstimateOdds Ratio (95% CI)EstimateOdds Ratio (95% CI)EstimateOdds Ratio (95% CI)
  • NOTE: Abbreviations: CARING, C=primary diagnosis of cancer, A=2 admissions to the hospital for a chronic illness within the last year; R=resident in a nursing home; I=intensive care unit (ICU) admission with multiorgan failure (MOF), N=noncancer hospice guidelines (meeting 2 of the National Hospice and Palliative Care Organization's [NHPCO] guidelines); CI, confidence interval.

  • Odds ratio is statistically significant as evidenced by the CI that does not cross 1.0.

  • Age was divided into 4 categories: 55 years, 5665 years, 6675 years, and >75 years.

Cancer1.926.85 (2.83‐16.59)a1.856.36 (3.54‐11.41)a1.987.23 (4.45‐11.75)a
Admissions to the hospital 2 in past year0.551.74 (0.76‐3.97)0.140.87 (0.51‐1.49)0.201.22 (0.78‐1.91)
Resident in a nursing home0.490.61 (0.06‐6.56)0.271.31 (0.37‐4.66)0.091.09 (0.36‐3.32)
ICU with MOF1.856.34 (2.0219.90)a  1.946.97 (2.75‐17.68)a
NHPCO (2) noncancer guidelines3.0420.86 (4.25102.32)a2.6213.73 (5.86‐32.15)a2.7415.55 (7.2833.23)a
Ageb0.381.46 (1.05‐2.03)a0.451.56 (1.23‐1.98)a0.471.60 (1.32‐1.93)a

In the safety‐net hospital, admission to the hospital with a primary diagnosis related to cancer, 2 noncancer hospice guidelines, ICU admission with MOF, and age by category all were significant predictors of 1‐year mortality. In the university hospital cohort, primary diagnosis of cancer, 2 noncancer hospice guidelines, and age by category were predictive of 1‐year mortality. Finally, in the entire study cohort, primary diagnosis of cancer, ICU with MOF, 2 noncancer hospice guidelines, and age were all predictive of 1‐year mortality. Parameter estimates were similar in 3 of the criteria compared to the VA setting. Differences in patient characteristics may have caused the differences in the estimates. Gender was additionally tested but not significant in any model. One‐year survival was significantly lower for those who met 1 of the CARING criteria versus those who did not (Figure 1).

jhm2107-fig-0001-m.png
Survival plot for those subjects who did (CARING ≥1) or did not (CARING = 0) meet at least 1 of the CARING criteria. Abbreviations: CARING, C = primary diagnosis of cancer, A = ≥2 admissions to the hospital for a chronic illness within the last year; R = resident in a nursing home; I = intensive care unit (ICU) admission with multiorgan failure, N = noncancer hospice guidelines (meeting ≥2 of the National Hospice and Palliative Care Organization's guidelines).

Based on the framework from the original CARING criteria analysis, a scoring rule was developed using the regression results of this validation cohort. To predict a high probability of 1‐year mortality, sensitivity was set to 58% and specificity was set at 86% (error rate=17%). Medium to high probability was set with a sensitivity of 73% and specificity of 72% (error rate=28%). The coefficients from the regression model of the entire study cohort were converted to scores for each of the CARING criteria. The scores are as follows: 0.5 points for admission from a nursing home, 1 point for 2 hospital admissions in the past year for a chronic illness, 10 points for primary diagnosis of cancer, 10 points for ICU admission with MOF, and 14 points for 2 noncancer hospice guidelines. For every age category increase, 2 points are assigned so that 0 points for age <55 years, 2 points for ages 56 to 65 years, 4 points for ages 66 to 75 years, and 6 points for >75 years. Points for individual risk factors were proportional to s (ie, log odds) in the logistic regression model for death at 1 year. Although no linear transformation exists between s and probabilities (of death at 1 year), the aggregated points for combinations of risk factors shown in Table 4 follow the probabilities in an approximately linear fashion, so that different degrees of risk of death can be represented contiguously (as highlighted by differently shaded regions in the scoring matrix) (Table 4). The scoring matrix allows for quick identification for patients at high risk for 1‐year mortality. In this non‐VA setting with healthier patients, low risk is defined at a lower probability threshold (0.1) compared to the VA setting (0.175).

Score of Risk of Death at 1 Year for the CARING Criteria by Age Group
 CARING Criteria Components
 NoneResident in a Nursing HomeAdmitted to the Hospital 2 Times in the Past YearResident in a Nursing Home Admitted to the Hospital 2 Times in the Past YearPrimary Diagnosis of CancerICU Admission With MOFNoncancer Hospice Guidelines
  • NOTE: Abbreviations: CARING, C=primary diagnosis of cancer, A=2 admissions to the hospital for a chronic illness within the last year; R=resident in a nursing home; I=intensive care unit (ICU) admission with multiorgan failure (MOF), N=noncancer hospice guidelines (meeting 2 of the National Hospice and Palliative Care Organization's guidelines).

Age       
55 years00.511.5 10 
5565 years22.533.5 10 
6675 years44.555.5 10 
>75 years66.577.5 10 
Risk       
Low 3.5Probability<0.1  
Medium 46.50.1probability <0.175  
High 7Probability0.175  

DISCUSSION

The CARING criteria are a practical prognostic tool that can be easily and rapidly applied to patients admitted to the hospital to estimate risk of death in 1 year, with the goal of identifying patients who may benefit most from incorporating palliative interventions into their plan of care. This study validated the CARING criteria in a tertiary referral university hospital and safety‐net hospital setting, demonstrating applicability in a much broader population than the VA hospital of the original CARING criteria study. The population studied represented a younger population by over 10 years, a more equitable proportion of males to females, a broader ethnic diversity, and lower 1‐year deaths rates than the original study. Despite the broader representation of the population, the significance of each of the individual CARING criterion was maintained except for 2 hospital admissions in the past year for a chronic illness (admission from a nursing home did not meet significance in either study as a sole criterion). As with the original study, meeting 2 of the NHPCO noncancer hospice guidelines demonstrated the highest risk of 1‐year mortality following index hospitalization, followed by primary diagnosis of cancer and ICU admission with MOF. Advancing age, also similar to the original study, conferred increased risk across the criterion.

Hospitalists could be an effective target for utilizing the CARING criteria because they are frequently the first‐line providers in the hospital setting. With the national shortage of palliative care specialists, hospitalists need to be able to identify when a patient has a limited life expectancy so they will be better equipped to make clinical decisions that are aligned with their patients' values, preferences, and goals of care. With the realization that not addressing advance care planning and patient goals of care may be considered medical errors, primary palliative care skills become alarmingly more important as priorities for hospitalists to obtain and feel comfortable using in daily practice.

The CARING criteria are directly applicable to patients who are seen by hospitalists. Other prognostic indices have focused on select patient populations, such as the elderly,[10, 11, 12] require collection of data that are not readily available on admission or would not otherwise be obtained,[10, 13] or apply to patients post‐hospital discharge, thereby missing the opportunity to make an impact earlier in the disease trajectory and incorporate palliative care into the hospital plan of care when key discussions about goals of care and preferences should be encouraged.

Additionally, the CARING criteria could easily be incorporated as a trigger for palliative care consults on hospital admission. Palliative care consults tend to happen late in a hospital stay, limiting the effectiveness of the palliative care team. A trigger system for hospitalists and other primary providers on hospital admission would lend to more effective timing of palliative measures being incorporated into the plan of care. Palliative care consults would not only be initiated earlier, but could be targeted for the more complex and sick patients with the highest risk of death in the next year.

In the time‐pressured environment, the presence of any 1 of the CARING criteria can act as a trigger to begin incorporating primary palliative care measures into the plan of care. The admitting hospitalist provider (ie, physician, nurse practitioner, physician assistant) could access the CARING criteria through an electronic health record prompt when admitting patients. When a more detailed assessment of mortality risk is helpful, the hospitalist can use the scoring matrix, which combines age with the individual criterion to calculate patients at medium or high risk of death within 1 year. Limited resources can then be directed to the patients with the greatest need. Patients with a focused care need, such as advance care planning or hospice referral, can be directed to the social worker or case manager. More complicated patients may be referred to a specialty palliative care team.

Several limitations to this study are recognized, including the small sample size of patients meeting criterion for ICU with MOF in the academic center study cohort. The patient data were collected during a transition time when the university hospital moved to a new campus, resulting in an ICU at each campus that housed patients with differing levels of illness severity, which may have contributed to the lower acuity ICU patient observed. Although we advocate the simplicity of the CARING criteria, the NHPCO noncancer hospice guidelines are more complicated, as they incorporates 8 broad categories of chronic illness. The hospice guidelines may not be general knowledge to the hospitalist or other primary providers. ePrognosis (http://eprognosis.ucsf.edu/) has a Web‐based calculator for the CARING criteria, including a link referencing the NHPCO noncancer hospice guidelines. Alternatively, providing a pocket card, smart phone or tablet app, or electronic health record tool containing the NHPCO criteria and CARING criteria could easily overcome this gap in knowledge. Finally, the reviewer agreement was not 100% for each criterion due to personal interpretation differences in the criterion. NHPCO criterion had the lowest kappa, yet it still was 0.78 and achieved a highly acceptable level of agreement.

CONCLUSION

The CARING criteria are a simple, practical prognostic tool predictive of death within 1 year that has been validated in a broad population of hospitalized patients. The criteria hold up in a younger, healthier population that is more diverse by age, gender, and ethnicity than the VA population. With ready access to critical prognostic information on hospital admission, clinicians will be better informed to make decisions that are aligned with their patients' values, preferences, and goals of care.

Disclosure

Nothing to report.

Prognostication continues to be a challenge to the clinician despite over 100 prognostic indices that have been developed during the past few decades to inform clinical practice and medical decision making.[1] Physicians are not accurate in prognostication of patients' risk of death and tend to overestimate survival.[2, 3] In addition, many physicians do not feel comfortable offering a prognosis to patients, despite patients' wishes to be informed.[4, 5] Regardless of the prevalence in the literature and value in improving physicians' prognostic accuracy, prognostic indices of survival are not regularly utilized in the hospital setting. Prognostic tools available for providers are often complicated and may require data about patients that are not readily available.[6, 7, 8] Prognostic indices may be too specific to a patient population, too difficult to remember, or too time consuming to use. A simple, rapid, and practical prognostic index is important in the hospital setting to assist in identifying patients at high risk of death so that primary palliative interventions can be incorporated into the plan of care early in the hospital stay. Patient and family education, advance care planning, formulating the plan of care based on patientfamily goals, and improved resource utilization could be better executed by more accurate risk of death prediction on hospital admission.

The CARING criteria are the only prognostic index to our knowledge that evaluates a patient's risk of death in the next year, with information readily available at the time of hospital admission (Table 1).[9] The CARING criteria are a unique prognostic tool: (1) CARING is a mnemonic acronym, making it more user friendly to the clinician. (2) The 5 prognostic indicators are readily available from the patient's chart on admission; gathering further data by patient or caretaker interviews or by obtaining laboratory data is not needed. (3) The timing for application of the tool on admission to the hospital is an ideal opportunity to intervene and introduce palliative interventions early on the hospital stay. The CARING criteria were developed and validated in a Veteran's Administration hospital setting by Fischer et al.[9] We sought to validate the CARING criteria in a broader patient populationmedical and surgical patients from a tertiary referral university hospital setting and a safety‐net hospital setting.

METHODS

Study Design

This study was a retrospective observational cohort study. The study was approved by the Colorado Multiple Institutional Review Board and the University of Colorado Hospital Research Review Committee.

Study Purpose

To validate the CARING criteria in a tertiary referral university hospital (University of Colorado Hospital [UCH]) and safety‐net hospital (Denver Health and Hospitals [DHH]) setting using similar methodology to that employed by the original CARING criteria study.[9]

Study Setting/Population

All adults (18 years of age) admitted as inpatients to the medical and surgical services of internal medicine, hospitalist, pulmonary, cardiology, hematology/oncology, hepatology, surgery, intensive care unit, and intermediary care unit at UCH and DHH during the study period of July 2005 through August 2005. The only exclusion criteria were those patients who were prisoners or pregnant. Administrative admission data from July 2005 to August 2005 were used to identify names of all persons admitted to the medicine and surgical services of the study hospitals during the specified time period.

The 2 study hospitals, UCH and DHH, provide a range of patients who vary in ethnicity, socioeconomic status, and medical illness. This variability allows for greater generalizability of the results. Both hospitals are affiliated with the University of Colorado School of Medicine internal medicine residency training program and are located in Denver, Colorado.

At the time of the study, UCH was a licensed 550‐bed tertiary referral, academic hospital serving the Denver metropolitan area and the Rocky Mountain region as a specialty care and referral center. DHH was a 398‐bed, academic, safety‐net hospital serving primarily the Denver metropolitan area. DHH provides 42% of the care for the uninsured in Denver and 26% of the uninsured care for the state of Colorado.

Measures

The CARING criteria were developed and validated in a Veteran's Administration (VA) hospital setting by Fischer et al.[9] The purpose of the CARING criteria is to identify patients, at the time of hospital admission, who are at higher risk of death in the following year. The prognostic index uses 5 predictors that can be abstracted from the chart at time of admission. The CARING criteria were developed a priori, and patients were evaluated using only the medical data available at the time of admission. The criteria include items that are already part of the routine physician admission notes and do not require additional data collection or assessments. The criteria include: C=primary diagnosis of cancer, A=2 admissions to the hospital for a chronic illness within the last year; R=resident in a nursing home; I=intensive care unit (ICU) admission with multiorgan failure (MOF), NG=noncancer hospice guidelines (meeting 2 of the National Hospice and Palliative Care Organization's [NHPCO] guidelines).

Patients were identified using name, date of birth, social security number, address, and phone number. This identifying information was then used for tracing death records 1 year after hospital admission.

Mortality at 1 year following the index hospitalization was the primary end point. To minimize missing data and the number of subjects lost to follow‐up, 3 determinants of mortality were used. First, electronic medical records of the 2 participating hospitals and their outpatient clinics were reviewed to determine if a follow‐up appointment had occurred past the study's end point of 1 year (August 2006). For those without a confirmed follow‐up visit, death records from the Colorado Department of Public Health and Vital Records were obtained. For those patients residing outside of Colorado or whose mortality status was still unclear, the National Death Index was accessed.

Medical Record Review

Medical records for all study participants were reviewed by J.Y. (UCH) and B.C. (DHH). Data collection was completed using direct data entry into a Microsoft Access (Microsoft Corp., Redmond, WA) database utilizing a data entry form linked with the database table. This form utilized skip patterns and input masks to ensure quality of data entry and minimize missing or invalid data. Inter‐rater reliability was assessed by an independent rereview (S.F.) of 5% of the total charts. Demographic variables were collected using hospital administrative data. These included personal identifiers of the participants for purposes of mortality follow‐up. Clinical data including the 5 CARING variables and additional descriptive variables were abstracted from the paper hospital chart and the electronic record of the chart (together these constitute the medical record).

Death Follow‐up

A search of Colorado death records was conducted in February 2011 for all subjects. Death records were used to determine mortality and time to death from the index hospitalization. The National Death Index was then searched for any subjects without or record of death in Colorado.

Analysis

All analyses were conducted using the statistical application software SAS for Windows version 9.3 (SAS Institute, Cary, NC). Simple frequencies and means ( standard deviation) were used to describe the baseline characteristics. Multiple logistic regression models were used to model 1‐year mortality. The models were fitted using all of the CARING variables and age. As the aim of the study was to validate the CARING criteria, the variables for the models were selected a priori based on the original index. Two hospital cohorts (DHH and UCH) were modeled separately and as a combined sample. Kaplan‐Meier survival analysis was conducted to compare those subjects who met 1 of the CARING criteria with those who did not through the entire period of mortality follow‐up (20052011). Finally, using the probabilities from the logistic regression models, we again developed a scoring rule appropriate for a non‐VA setting to allow clinicians to easily identify patient risk for 1‐year mortality at the time of hospital admission.

RESULTS

There were a total of 1064 patients admitted to the medical and surgical services during the study period568 patients at DHH and 496 patients at UCH. Sample characteristics of each individual hospital cohort and the entire combined study cohort are detailed in Table 2. Overall, slightly over half the population were male, with a mean age of 50 years, and the ethnic breakdown roughly reflects the population in Denver. A total of 36.5% (n=388) of the study population met 1 of the CARING criteria, and 12.6% (n=134 among 1063 excluding 1 without an admit date) died within 1 year of the index hospitalization. These were younger and healthier patients compared to the VA sample used in developing the CARING criteria.

CARING Criteria
  • NOTE: The CARING criteria must be applied to patients who are hospitalized on the first day after admission (ie, they met the criteria on the day of admission). It is unknown if the CARING criteria are predictive of high mortality when applied to patients who are either not in the hospital or later in the hospital stay. Cancer: Is there a primary diagnosis of cancer? This includes patients who are admitted for chemotherapy (most chemotherapy is administered as an outpatient, and patients who require hospitalization for administration of chemotherapy are likely more ill or have more aggressive cancers requiring more intensive monitoring) due to complications from their chemotherapy (ie, neutropenic fever), or for aggressive symptom management. What is important about this criterion is that cancer must be the primary reason they are admitted. A person with colon cancer admitted for suspected angina would not qualify. Admitted to the hospital for 2 times in the past year for a chronic illness. For example, a man is admitted with pneumonia and COPD exacerbation, and looking back at his chart you discover that he was also admitted for a COPD exacerbation 2 months ago. That would add up to 2 hospital admissions in the past year for a chronic illness; therefore, the patient would meet this criterion. A patient admitted twice in the past year for musculoskeletal chest pain would not meet the criterion. Resident in a nursing home: A patient admitted from either a long‐term care facility or a skilled nursing facility would meet this criterion. It is essentially a proxy for poor functional status. ICU admission with multiorgan failure: An example would be a patient admitted to the ICU requiring mechanical ventilation (pulmonary system in failure) and on pressors or in renal failure (either requiring dialysis or nearing that point; a small increase in the creatinine would not qualify for organ failure). Noncancer hospice guidelines: Patient must meet at least 2 items in any given category. Abbreviations: AIDS, acquired immunodeficiency syndrome; BiPAP, bilevel positive airway pressure; BP, blood pressure; CARING, C=primary diagnosis of cancer, A=2 admissions to the hospital for a chronic illness within the last year; R=resident in a nursing home; I=intensive care unit (ICU) admission with multiorgan failure, NG=noncancer hospice guidelines (meeting 2 of the National Hospice and Palliative Care Organization's guidelines) CHF, congestive heart failure; CNS, central nervous system; COPD, chronic obstructive pulmonary disease; CVA, cardiovascular accident; ER, emergency room; HIV, human immunodeficiency virus; ICU, intensive care unit.

RenalDementia
Stop/decline dialysisUnable to ambulate independently
Not candidate for transplantUrinary or fecal incontinence
Urine output < 40cc/24 hoursUnable to speak with more than single words
Creatinine > 8.0 (>6.0 for diabetics)Unable to bathe independently
Creatinine clearance 10cc/minUnable to dress independently
UremiaCo‐morbid conditions:
Persistent serum K + > 7.0Aspiration pneumonia
Co‐morbid conditions:Pyelonephritis
Cancer CHFDecubitus ulcer
Chronic lung disease AIDS/HIVDifficulty swallowing or refusal to eat
Sepsis Cirrhosis 
CardiacPulmonary
Ejection fraction < 20%Dyspnea at rest
Symptomatic with diuretics and vasodilatorsFEV1 < 30%
Not candidate for transplantFrequent ER or hospital admits for pulmonary infections or respiratory distress
History of cardiac arrestCor pulmonale or right heart failure
History of syncope02 sat < 88% on 02
Systolic BP < 120mmHGPC02 > 50
CVA cardiac originResting tachycardia > 100/min
Co‐morbid conditions as listed in RenalCo‐morbid conditions as listed in Renal
LiverStroke/CVA
End stage cirrhosisComa at onset
Not candidate for transplantComa >3 days
Protime > 5sec and albumin <2.5Limb paralysis
Ascites unresponsive to treatmentUrinary/fecal incontinence
Hepatorenal syndromeImpaired sitting balance
Hepatic encephalopathyKarnofsky < 50%
Spontaneous bacterial peritonitisRecurrent aspiration
Recurrent variceal bleedAge > 70
Co‐morbid conditions as listed in RenalCo‐morbid conditions as listed in Renal
HIV/AIDSNeuromuscular
Persistent decline in functionDiminished respiratory function
Chronic diarrhea 1 yearChosen not to receive BiPAP/vent
Decision to stop treatmentDifficulty swallowing
CNS lymphomaDiminished functional status
MAC‐untreatedIncontinence
Systemic lymphomaCo‐morbid conditions as listed in Renal
Dilated cardiomyopathy 
CD4 < 25 with disease progression 
Viral load > 100,000 
Validation Study Cohort Characteristics
 Safety‐Net Hospital Cohort, N=568Academic Center Cohort, N=496Study Cohort,N=1064Original CARING Cohort, N=8739
  • NOTE: Cases with missing data were negligible (<4%). Abbreviations: CARING, C=primary diagnosis of cancer, A=2 admissions to the hospital for a chronic illness within the last year; R=resident in a nursing home; I=intensive care unit (ICU) admission with multiorgan failure (MOF), NG=noncancer hospice guidelines (meeting 2 of the National Hospice and Palliative Care Organization's [NHPCO] guidelines); SD, standard deviation.

Mean age ( SD), y47.8 (16.5)54.4 (17.5)50.9 (17.3)63 (13)
Male gender59.5% (338)50.1% (248)55.1% (586)98% (856)
Ethnicity    
African American14.1% (80)13.5% (65)13.8% (145)13% (114)
Asian0.4% (2)1.5% (7)0.9% (9)Not reported
Caucasian41.7% (237)66.3% (318)53.0 % (555)69% (602)
Latino41.9% (238)9.6% (46)27.1% (284)8% (70)
Native American0.5% (3)0.4% (2)0.5% (5)Not reported
Other0.5% (3)0.6% (3)0.6% (6)10% (87)
Unknown0.9% (5)8.1% (39)4.2% (44)Not reported
CARING criteria    
Cancer6.2% (35)19.4% (96)12.3% (131)23% (201)
Admissions to the hospital 2 in past year13.6% (77)42.7% (212)27.2% (289)36% (314)
Resident in a nursing home1.8% (10)3.4% (17)2.5% (27)3% (26)
ICU with MOF3.7% (21)1.2% (6)2.5% (27)2% (17)
NHPCO (2) noncancer guidelines1.6% (9)5.9% (29)3.6% (38)8% (70)

Reliability testing demonstrated excellent inter‐rater reliability. Kappa for each criterion is as follows: (1) primary diagnosis of cancer=1.0, (2) 2 admissions to the hospital in the past year=0.91, (3) resident in a nursing home=1.0, (4) ICU admission with MOF=1.0, and (5) 2 noncancer hospice guidelines=0.78.

This study aimed to validate the CARING criteria9; therefore, all original individual CARING criterion were included in the validation logistic regression models. The 1 exception to this was in the university hospital study cohort, where the ICU criterion was excluded from the model due to small sample size and quasiseparation in the model. The model results are presented in Table 3 for the individual hospitals and combined study cohort.

Prediction of 1‐Year Mortality Using CARING Criteria
 Safety Net Hospital Cohort, C Index=0.76Academic Center Cohort, C Index=0.76Combined Hospital Cohort, C Index=0.79
 EstimateOdds Ratio (95% CI)EstimateOdds Ratio (95% CI)EstimateOdds Ratio (95% CI)
  • NOTE: Abbreviations: CARING, C=primary diagnosis of cancer, A=2 admissions to the hospital for a chronic illness within the last year; R=resident in a nursing home; I=intensive care unit (ICU) admission with multiorgan failure (MOF), N=noncancer hospice guidelines (meeting 2 of the National Hospice and Palliative Care Organization's [NHPCO] guidelines); CI, confidence interval.

  • Odds ratio is statistically significant as evidenced by the CI that does not cross 1.0.

  • Age was divided into 4 categories: 55 years, 5665 years, 6675 years, and >75 years.

Cancer1.926.85 (2.83‐16.59)a1.856.36 (3.54‐11.41)a1.987.23 (4.45‐11.75)a
Admissions to the hospital 2 in past year0.551.74 (0.76‐3.97)0.140.87 (0.51‐1.49)0.201.22 (0.78‐1.91)
Resident in a nursing home0.490.61 (0.06‐6.56)0.271.31 (0.37‐4.66)0.091.09 (0.36‐3.32)
ICU with MOF1.856.34 (2.0219.90)a  1.946.97 (2.75‐17.68)a
NHPCO (2) noncancer guidelines3.0420.86 (4.25102.32)a2.6213.73 (5.86‐32.15)a2.7415.55 (7.2833.23)a
Ageb0.381.46 (1.05‐2.03)a0.451.56 (1.23‐1.98)a0.471.60 (1.32‐1.93)a

In the safety‐net hospital, admission to the hospital with a primary diagnosis related to cancer, 2 noncancer hospice guidelines, ICU admission with MOF, and age by category all were significant predictors of 1‐year mortality. In the university hospital cohort, primary diagnosis of cancer, 2 noncancer hospice guidelines, and age by category were predictive of 1‐year mortality. Finally, in the entire study cohort, primary diagnosis of cancer, ICU with MOF, 2 noncancer hospice guidelines, and age were all predictive of 1‐year mortality. Parameter estimates were similar in 3 of the criteria compared to the VA setting. Differences in patient characteristics may have caused the differences in the estimates. Gender was additionally tested but not significant in any model. One‐year survival was significantly lower for those who met 1 of the CARING criteria versus those who did not (Figure 1).

jhm2107-fig-0001-m.png
Survival plot for those subjects who did (CARING ≥1) or did not (CARING = 0) meet at least 1 of the CARING criteria. Abbreviations: CARING, C = primary diagnosis of cancer, A = ≥2 admissions to the hospital for a chronic illness within the last year; R = resident in a nursing home; I = intensive care unit (ICU) admission with multiorgan failure, N = noncancer hospice guidelines (meeting ≥2 of the National Hospice and Palliative Care Organization's guidelines).

Based on the framework from the original CARING criteria analysis, a scoring rule was developed using the regression results of this validation cohort. To predict a high probability of 1‐year mortality, sensitivity was set to 58% and specificity was set at 86% (error rate=17%). Medium to high probability was set with a sensitivity of 73% and specificity of 72% (error rate=28%). The coefficients from the regression model of the entire study cohort were converted to scores for each of the CARING criteria. The scores are as follows: 0.5 points for admission from a nursing home, 1 point for 2 hospital admissions in the past year for a chronic illness, 10 points for primary diagnosis of cancer, 10 points for ICU admission with MOF, and 14 points for 2 noncancer hospice guidelines. For every age category increase, 2 points are assigned so that 0 points for age <55 years, 2 points for ages 56 to 65 years, 4 points for ages 66 to 75 years, and 6 points for >75 years. Points for individual risk factors were proportional to s (ie, log odds) in the logistic regression model for death at 1 year. Although no linear transformation exists between s and probabilities (of death at 1 year), the aggregated points for combinations of risk factors shown in Table 4 follow the probabilities in an approximately linear fashion, so that different degrees of risk of death can be represented contiguously (as highlighted by differently shaded regions in the scoring matrix) (Table 4). The scoring matrix allows for quick identification for patients at high risk for 1‐year mortality. In this non‐VA setting with healthier patients, low risk is defined at a lower probability threshold (0.1) compared to the VA setting (0.175).

Score of Risk of Death at 1 Year for the CARING Criteria by Age Group
 CARING Criteria Components
 NoneResident in a Nursing HomeAdmitted to the Hospital 2 Times in the Past YearResident in a Nursing Home Admitted to the Hospital 2 Times in the Past YearPrimary Diagnosis of CancerICU Admission With MOFNoncancer Hospice Guidelines
  • NOTE: Abbreviations: CARING, C=primary diagnosis of cancer, A=2 admissions to the hospital for a chronic illness within the last year; R=resident in a nursing home; I=intensive care unit (ICU) admission with multiorgan failure (MOF), N=noncancer hospice guidelines (meeting 2 of the National Hospice and Palliative Care Organization's guidelines).

Age       
55 years00.511.5 10 
5565 years22.533.5 10 
6675 years44.555.5 10 
>75 years66.577.5 10 
Risk       
Low 3.5Probability<0.1  
Medium 46.50.1probability <0.175  
High 7Probability0.175  

DISCUSSION

The CARING criteria are a practical prognostic tool that can be easily and rapidly applied to patients admitted to the hospital to estimate risk of death in 1 year, with the goal of identifying patients who may benefit most from incorporating palliative interventions into their plan of care. This study validated the CARING criteria in a tertiary referral university hospital and safety‐net hospital setting, demonstrating applicability in a much broader population than the VA hospital of the original CARING criteria study. The population studied represented a younger population by over 10 years, a more equitable proportion of males to females, a broader ethnic diversity, and lower 1‐year deaths rates than the original study. Despite the broader representation of the population, the significance of each of the individual CARING criterion was maintained except for 2 hospital admissions in the past year for a chronic illness (admission from a nursing home did not meet significance in either study as a sole criterion). As with the original study, meeting 2 of the NHPCO noncancer hospice guidelines demonstrated the highest risk of 1‐year mortality following index hospitalization, followed by primary diagnosis of cancer and ICU admission with MOF. Advancing age, also similar to the original study, conferred increased risk across the criterion.

Hospitalists could be an effective target for utilizing the CARING criteria because they are frequently the first‐line providers in the hospital setting. With the national shortage of palliative care specialists, hospitalists need to be able to identify when a patient has a limited life expectancy so they will be better equipped to make clinical decisions that are aligned with their patients' values, preferences, and goals of care. With the realization that not addressing advance care planning and patient goals of care may be considered medical errors, primary palliative care skills become alarmingly more important as priorities for hospitalists to obtain and feel comfortable using in daily practice.

The CARING criteria are directly applicable to patients who are seen by hospitalists. Other prognostic indices have focused on select patient populations, such as the elderly,[10, 11, 12] require collection of data that are not readily available on admission or would not otherwise be obtained,[10, 13] or apply to patients post‐hospital discharge, thereby missing the opportunity to make an impact earlier in the disease trajectory and incorporate palliative care into the hospital plan of care when key discussions about goals of care and preferences should be encouraged.

Additionally, the CARING criteria could easily be incorporated as a trigger for palliative care consults on hospital admission. Palliative care consults tend to happen late in a hospital stay, limiting the effectiveness of the palliative care team. A trigger system for hospitalists and other primary providers on hospital admission would lend to more effective timing of palliative measures being incorporated into the plan of care. Palliative care consults would not only be initiated earlier, but could be targeted for the more complex and sick patients with the highest risk of death in the next year.

In the time‐pressured environment, the presence of any 1 of the CARING criteria can act as a trigger to begin incorporating primary palliative care measures into the plan of care. The admitting hospitalist provider (ie, physician, nurse practitioner, physician assistant) could access the CARING criteria through an electronic health record prompt when admitting patients. When a more detailed assessment of mortality risk is helpful, the hospitalist can use the scoring matrix, which combines age with the individual criterion to calculate patients at medium or high risk of death within 1 year. Limited resources can then be directed to the patients with the greatest need. Patients with a focused care need, such as advance care planning or hospice referral, can be directed to the social worker or case manager. More complicated patients may be referred to a specialty palliative care team.

Several limitations to this study are recognized, including the small sample size of patients meeting criterion for ICU with MOF in the academic center study cohort. The patient data were collected during a transition time when the university hospital moved to a new campus, resulting in an ICU at each campus that housed patients with differing levels of illness severity, which may have contributed to the lower acuity ICU patient observed. Although we advocate the simplicity of the CARING criteria, the NHPCO noncancer hospice guidelines are more complicated, as they incorporates 8 broad categories of chronic illness. The hospice guidelines may not be general knowledge to the hospitalist or other primary providers. ePrognosis (http://eprognosis.ucsf.edu/) has a Web‐based calculator for the CARING criteria, including a link referencing the NHPCO noncancer hospice guidelines. Alternatively, providing a pocket card, smart phone or tablet app, or electronic health record tool containing the NHPCO criteria and CARING criteria could easily overcome this gap in knowledge. Finally, the reviewer agreement was not 100% for each criterion due to personal interpretation differences in the criterion. NHPCO criterion had the lowest kappa, yet it still was 0.78 and achieved a highly acceptable level of agreement.

CONCLUSION

The CARING criteria are a simple, practical prognostic tool predictive of death within 1 year that has been validated in a broad population of hospitalized patients. The criteria hold up in a younger, healthier population that is more diverse by age, gender, and ethnicity than the VA population. With ready access to critical prognostic information on hospital admission, clinicians will be better informed to make decisions that are aligned with their patients' values, preferences, and goals of care.

Disclosure

Nothing to report.

References
  1. Siontis GC, Tzoulaki I, Ioannidis JP. Predicting death: an empirical evaluation of predictive tools for mortality. Arch Intern Med. 2011;171:17211726.
  2. Christakis NA, Lamont EB. Extent and determinants of error in physicians' prognoses in terminally ill patients: prospective cohort study. West J Med. 2000;172:310313.
  3. Glare P, Virik K, Jones M, et al. A systematic review of physicians' survival predictions in terminally ill cancer patients. BMJ. 2003;327:195198.
  4. Christakis NA, Iwashyna TJ. Attitude and self‐reported practice regarding prognostication in a national sample of internists. Arch Intern Med. 1998;158:23892395.
  5. Campbell TC, Carey EC, Jackson VA, et al. Discussing prognosis: balancing hope and realism. Cancer J. 2010;16:461466.
  6. Zimmerman JE, Kramer AA, McNair DS, Malila FM. Acute Physiology and Chronic Health Evaluation (APACHE) IV: hospital mortality assessment for today's critically ill patients. Crit Care Med. 2006;34:12971310.
  7. Ledoux D, Canivet JL, Preiser JC, Lefrancq J, Damas P. SAPS 3 admission score: an external validation in a general intensive care population. Intensive Care Med. 2008;34:18731877.
  8. Higgins TL, Kramer AA, Nathanson BH, Copes W, Stark M, Teres D. Prospective validation of the intensive care unit admission Mortality Probability Model (MPM0‐III). Crit Care Med. 2009;37:16191623.
  9. Fischer SM, Gozansky W, Sauaia A, Min SJ, Kutner JS, Kramer A. A practical tool to identify patients who may benefit from a palliative approach: the CARING criteria. J Pain Symptom Manage. 2006;31:285292.
  10. Teno JM, Harrell FE, Knaus W, et al. Prediction of survival for older hospitalized patients: the HELP survival model. J Am Geriatr Soc. 2000;48:S16S24.
  11. Pilotto A, Ferrucci L, Franceschi M, et al. Development and validation of a multidimensional prognostic index for one‐year mortality from comprehensive geriatric assessment in hospitalized older patients. Rejuvenation Res. 2008;11:151161.
  12. Inouye SK, Bogardus ST, Vitagliano G, et al. Burden of illness score for elderly persons: risk adjustment incorporating the cumulative impact of diseases, physiologic abnormalities, and functional impairments. Med Care. 2003;41:7083.
  13. Knaus WA, Harrell FE, Lynn J, et al. The SUPPORT prognostic model. Objective estimates of survival for seriously ill hospitalized adults. Study to understand prognoses and preferences for outcomes and risks of treatments. Ann Intern Med. 1995;122:191203.
References
  1. Siontis GC, Tzoulaki I, Ioannidis JP. Predicting death: an empirical evaluation of predictive tools for mortality. Arch Intern Med. 2011;171:17211726.
  2. Christakis NA, Lamont EB. Extent and determinants of error in physicians' prognoses in terminally ill patients: prospective cohort study. West J Med. 2000;172:310313.
  3. Glare P, Virik K, Jones M, et al. A systematic review of physicians' survival predictions in terminally ill cancer patients. BMJ. 2003;327:195198.
  4. Christakis NA, Iwashyna TJ. Attitude and self‐reported practice regarding prognostication in a national sample of internists. Arch Intern Med. 1998;158:23892395.
  5. Campbell TC, Carey EC, Jackson VA, et al. Discussing prognosis: balancing hope and realism. Cancer J. 2010;16:461466.
  6. Zimmerman JE, Kramer AA, McNair DS, Malila FM. Acute Physiology and Chronic Health Evaluation (APACHE) IV: hospital mortality assessment for today's critically ill patients. Crit Care Med. 2006;34:12971310.
  7. Ledoux D, Canivet JL, Preiser JC, Lefrancq J, Damas P. SAPS 3 admission score: an external validation in a general intensive care population. Intensive Care Med. 2008;34:18731877.
  8. Higgins TL, Kramer AA, Nathanson BH, Copes W, Stark M, Teres D. Prospective validation of the intensive care unit admission Mortality Probability Model (MPM0‐III). Crit Care Med. 2009;37:16191623.
  9. Fischer SM, Gozansky W, Sauaia A, Min SJ, Kutner JS, Kramer A. A practical tool to identify patients who may benefit from a palliative approach: the CARING criteria. J Pain Symptom Manage. 2006;31:285292.
  10. Teno JM, Harrell FE, Knaus W, et al. Prediction of survival for older hospitalized patients: the HELP survival model. J Am Geriatr Soc. 2000;48:S16S24.
  11. Pilotto A, Ferrucci L, Franceschi M, et al. Development and validation of a multidimensional prognostic index for one‐year mortality from comprehensive geriatric assessment in hospitalized older patients. Rejuvenation Res. 2008;11:151161.
  12. Inouye SK, Bogardus ST, Vitagliano G, et al. Burden of illness score for elderly persons: risk adjustment incorporating the cumulative impact of diseases, physiologic abnormalities, and functional impairments. Med Care. 2003;41:7083.
  13. Knaus WA, Harrell FE, Lynn J, et al. The SUPPORT prognostic model. Objective estimates of survival for seriously ill hospitalized adults. Study to understand prognoses and preferences for outcomes and risks of treatments. Ann Intern Med. 1995;122:191203.
Issue
Journal of Hospital Medicine - 8(12)
Issue
Journal of Hospital Medicine - 8(12)
Page Number
696-701
Page Number
696-701
Publications
Publications
Article Type
Display Headline
Caring about prognosis: A validation study of the caring criteria to identify hospitalized patients at high risk for death at 1 year
Display Headline
Caring about prognosis: A validation study of the caring criteria to identify hospitalized patients at high risk for death at 1 year
Sections
Article Source

© 2013 Society of Hospital Medicine

Disallow All Ads
Correspondence Location
Address for correspondence and reprint requests: Jeanie Youngwerth, MD, Hospitalist, Assistant Professor of Medicine, University of Colorado School of Medicine, Hospital Medicine Group, 12401 E. 17th Ave., Mail Stop F782, Aurora, CO 80045; Telephone: 720–848‐4289; Fax: 720–848‐4293; E‐mail: jean.youngwerth@ucdenver.edu
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Article PDF Media
Image
Disable zoom
Off
Media Files
Image
Disable zoom
Off

The latest research you need to know

Article Type
Changed
Fri, 09/14/2018 - 12:34
Display Headline
The latest research you need to know

In This Edition

Does a short duration of perioperative smoking cessation lead to a reduction in postoperative complications?

Background: Prior studies have demonstrated a reduction in postoperative complications when patients stop smoking in the perioperative period. However, they have not clearly shown what effect a fairly short duration of cessation, such as a period of only four weeks, has on the frequency of complications.

Study design: Randomized controlled trial.

Setting: Four university-affiliated hospitals in Sweden.

Synopsis: Using 117 patients who were daily smokers for less than one year between the ages of 18-79 who were scheduled for elective general or orthopedic surgery, this study showed that a smoking-cessation intervention initiated as little as four weeks prior to surgery resulted in fewer postoperative complications. The complication rate was reduced from 41% in the control group to 21% in the intervention group, which received cessation counseling and nicotine-replacement therapy. The relative risk reduction was 49% (95% confidence interval, 3-40) with a number needed to treat of five.

Because this was a randomized controlled trial with a large observed benefit, it appears to be reasonable to endorse perioperative smoking cessation as late as four weeks before an elective surgery. The study was limited in its ability to detect a difference in wound infections by the small sample size and the possibility patients might have unblinded themselves to outcome assessors, causing an overestimation of the effect of the intervention on the primary outcome of all complications.

Bottom line: Perioperative smoking cessation reduces postoperative complications even when started just four weeks prior to surgery.

Citation: Lindstrom D, Azodi OS, Wladis A, et al. Effects of a perioperative smoking cessation intervention on postoperative complications. Ann Surg. 2008;248(5):739-745.

Does implantable-defibrillator therapy cause deterioration in quality of life for patients with heart failure?

Background: Patients with depressed left-ventricular function are known to have improved survival after receiving implantable cardioverter defibrillators (ICDs). However, there is concern ICD therapy can prolong survival at the expense of a diminished quality of life.

Study design: Randomized placebo-controlled trial.

Setting: Multiple centers in the U.S., Canada, and New Zealand.

Synopsis: Using 2,479 patients from the Sudden Cardiac Death in Heart Failure trial who were 18 and older and had stable heart failure and depressed left-ventricular function, this study demonstrated no significant quality-of-life difference at 30 months when compared with patients who received ICD, amiodarone, and state-of-the-art medical therapy or an amiodarone placebo and state-of-the-art medical therapy. While functional status did not differ at any time between the three groups, psychological well-being was improved in the ICD group at three months (p=0.01) and 12 months (p=0.03) when compared with the placebo group, but at 30 months there was no difference between the groups.

While the trial was randomized and placebo-controlled, the investigators were unable to blind patients or outcome assessors. Nevertheless, the lack of deterioration of quality of life in ICD patients is reassuring.

 

 

Bottom line: Placement of ICDs in heart failure patients with a high risk of sudden cardiac death does not appear to decrease quality of life.

Citation: Mark DB, Anstrom KJ, Sun JL, et al. Quality of life with defibrillator therapy or amiodarone in heart failure. N Engl J Med. 2008;359:999-1008.

CLINICAL SHORTS

SERIAL 2-POINT ULTRASONOGRAPHY PLUS D-DIMER IS EQUIVALENT TO WHOLE-LEG ULTRASONOGRAPHY FOR DIAGNOSING DVT

Randomized trials show that when comparing serial 2-point ultrasonography plus D-dimer testing with whole-leg ultrasonography, the strategies were equivalent in excluding a first episode of suspected DVT in outpatients.

Citation: Bernardi E, Camporese G, Buller HR, et al. Serial 2-point ultrasonography plus D-dimer vs whole-leg color-coded Doppler ultrasonography for diagnosing suspected symptomatic deep vein thrombosis. JAMA. 2008;300(14):1653-1659.

DAILY HEMODIALYSIS IS COST-EFFECTIVE IN ICU PATIENTS WITH ACUTE KIDNEY INJURY (AKI)

Markov model based on prospective trial data shows daily hemodialysis is cost-effective for AKI in the ICU compared with alternate-day hemodialysis.

Citation: Desai AA, Baras J, Berk BB, et al. Management of acute kidney injury in the intensive care unit. Arch Intern Med. 2008;168(16):1761-1767.

THROMBOLYSIS FOR IN-HOSPITAL STROKE IS SAFE, BUT ASSOCIATED WITH DELAYS

Prospective observational trial shows thrombolysis is safe and effective for in-hospital stroke, but statistically significant delays exist compared with out-of-hospital strokes.

Citation: Masjuan J, Simal P, Fuentes B, et al. In-hospital stroke treated with intravenous tissue plasminogen activator. Stroke. 2008;39:2614-2616.

ALGORITHM CAN IDENTIFY HIGH-RISK HEART FAILURE PATIENTS

Prospective observational study identifies clinical variables for a bedside algorithm, which stratifies the risk of hospitalized heart failure patients for early mortality or readmission to identify those who might benefit from closer follow-up.

Citation: O’Connor CM, Abraham WT, Albert NM, et al. Predictors of mortality after discharge in patients hospitalized with heart failure: an analysis from the Organized Program to Initiate Lifesaving Treatment in Hospitalized Patients with Heart Failure (OPTIMIZE-HF). Am Heart J. 2008;156(4):662-673.

IN-HOSPITAL SMOKING-CESSATION INTERVENTIONS WITH FOLLOW-UP CAN WORK

Meta-analysis of 33 trials shows in-hospital smoking-cessation counseling followed up with more than one month of outpatient support can be effective.

Citation: Rigotti NA, Munafo MR, Stead LF. Smoking cessation interventions for hospitalized smokers. Arch Intern Med. 2008;168(18):1950-1960.

OMISSION OF KEY INFORMATION DURING SIGN-OUT LEADS TO ADVERSE CONSEQUENCES

An audio-taped study of sign-out among internal medicine house staff teams revealed omission of key information during sign-out resulted in delays in diagnosis or treatment.

Citation: Horwitz LI, Moin T, Krumholz HM, Wang L, Bradley EH. Consequences of inadequate sign-out for patient care. Arch Intern Med. 2008;168(16):1755-1760.

HOSPITAL PALLIATIVE CARE CONSULTATION TEAMS ARE ASSOCIATED WITH HOSPITAL COST SAVINGS

Analysis of administrative data from eight diverse hospitals with palliative-care programs revealed consultation with palliative care saved $1,696 (p<0.001) per hospital admission in patients discharged alive, and $4,098 (p=0.003) per hospital admission in patients who died in the hospital.

Citation: Morrison RS, Penrod JD, Cassel JB, et al. Cost savings associated with U.S. hospital palliative care consultation programs. Arch Intern Med. 2008;168(16):1783-1790.

HIGHER EDUCATIONAL DEBT INFLUENCES INTERNAL MEDICINE RESIDENT CAREER PLANS

U.S. medical graduates with a debt of $50,000 to $99,999 are more likely than those with no debt to choose a hospitalist career, and this preference increased with increased debt level.

Citation: McDonald FS, West CP, Popkave C, Kolars JC. Educational debt and reported career plans among internal medicine residents. Ann Intern Med. 2008;149:416-420.

BRAIN IMAGING IMPORTANT IN IDENTIFYING VASCULAR TERRITORY AFTER TIA OR MINOR STROKE

Neurologists were only moderately reliable at identifying the vascular territory of a TIA or motor stroke, highlighting the fact brain imaging is needed to accurately identify the vascular territories of these events.

Citation: Flossmann E, Redgrave JN, Briley D, Rothwell PM. Reliability of clinical diagnosis of the symptomatic vascular territory in patients with recent transient ischemic attack or minor stroke. Stroke. 2008;39:2457-2460.

HIGH-DOSE VITAMIN B SUPPLEMENTATION DOES NOT SLOW COGNITIVE DECLINE IN ALZHEIMER’S DISEASE

Multicenter, randomized, placebo-controlled trial finds no difference in the rate of cognitive decline in patients with Alzheimer’s disease treated with high-dose vitamin B supplements for 18 months.

Citation: Aisen PS, Schneider LS, Sano M, et al. High-dose B vitamin supplementation and cognitive decline in Alzheimer’s disease. JAMA. 2008; 300(15):1774-1783.

 

 

Can a simplified, revised Geneva score retain diagnostic accuracy and clinical utility?

Background: The revised Geneva score is a validated and objective clinical decision rule, but has multiple variables with different weights. This can make the tool cumbersome and difficult to remember, and could lead to inaccurate calculations and misjudgments in patient care.

Study design: Retrospective cohort study.

Setting: Four university-affiliated European hospitals.

Synopsis: Using data from two prior prospective trials involving patients with suspected pulmonary embolism (PE), this study showed re-analysis of these patients with a simplified, revised Geneva score, which gives only one point to each clinical factor, resulted in the same level of diagnostic accuracy. Specifically, data from 1,049 patients was used to construct a receiver-operating characteristic curve analysis comparing the standardized and simplified Geneva score, which showed areas under the curve of 0.75 (95% confidence interval 0.71-0.78) and 0.74 (0.70-0.77), respectively. Additionally, the safety of using this clinical tool to rule out PE was demonstrated when using both a three-level (low-intermediate probability) and a dichotomized scheme (PE unlikely) in combination with a negative D-dimer test.

The retrospective nature of the study was its major limitation. The authors suggest a prospective study to complete validation of the simplified, revised Geneva score.

Bottom line: With prospective analysis, it might be possible to further validate a simplified, revised Geneva score.

Citation: Klok FA, Mos ICM, Nijkeuter M, et al. Simplification of the revised Geneva score for assessing clinical probability of pulmonary embolism. Arch Intern Med. 2008;168(19):2131-2136.

Is the rate of postoperative major adverse cardiac events (MACEs) inversely related to time after percutaneous coronary intervention (PCI) with a drug-eluting stent (DES)?

Background: The American College of Cardiology and the American Heart Association recently released an advisory that included a recommendation to delay elective noncardiac surgery (NCS) for one year after DES placement. However, no large study addresses the timing of NCS after PCI with DES.

Study design: Retrospective observational study.

Setting: Mayo Clinic, Rochester, Minn.

Synopsis: Looking at 520 patients who had NCS after DES at the Mayo Clinic, 5.4% experienced MACEs, but the rate of MACEs was not significantly associated with the time after stent placement to surgery (p=0.337). However, observed rates of MACEs were lower after one year. Elderly patients and those going for emergent surgery are at the highest risk for MACE. Bleeding complications were not associated with antiplatelet use.

Although this study does not provide a clear cutoff time for when it is safe to proceed to NCS after DES, it is somewhat reassuring to see the relatively small number of MACEs and the lack of bleeding complications associated with antiplatelet use. However, careful coordination between hospitalists, cardiologists, anesthesiologists, and surgeons is still needed when coordinating NCS after DES, especially in the elderly or during emergent situations.

Bottom line: While time to noncardiac surgery after drug-eluting stent placement is not associated with major adverse cardiac events, observed rates of events are lower after one year.

Citation: Rabbitts JA, Nuttall GA, Brown MJ, et al. Cardiac risk of non-cardiac surgery after percutaneous coronary intervention with drug-eluting stents. Anesthesiology.2008;109: 596-604.

Is the risk of MACEs and bleeding events for patients undergoing NCS related to the time interval between PCI with bare-metal stent?

Background: In order to prevent thrombosis of bare-metal stents (BMS) placed during percutaneous coronary intervention (PCI), antiplatelet therapy is used. This poses a risk of bleeding, if surgery is needed during the antiplatelet therapy. The American College of Cardiology and the American Heart Association recommends delaying NCS for at least six weeks after PCI with BMS.

 

 

Study design: Retrospective observational study.

Setting: Mayo Clinic, Rochester, Minn.

Synopsis: Looking at 899 patients who had NCS within one year of PCI with BMS at the Mayo Clinic between Jan. 1, 1990, and Jan. 1, 2005, this study found that when NCS was done 30 days or less after PCI with BMS, the MACEs rate was 10.5%, compared with 2.8% when NCS was done 91 or more days after PCI with BMS. After a multivariable analysis, it also was shown bleeding events were not associated with time between PCI with BMS and NCS.

While the American College of Cardiology and the American Heart Association recommends delaying NCS for at least six weeks after PCI with BMS, waiting at least 90 days would permit completion of antiplatelet therapy and re-endothelialization of the stent.

Bottom line: The risk of MACEs with noncardiac surgery is lowest when performed at least 90 days after PCI with bare-metal stent.

Citation: Nuttall GA, Brown MJ, Stombaugh JW, et al. Time and cardiac risk of surgery after bare-metal stent, percutaneous coronary intervention. Anesthesiology. 2008;109: 588-595.

Should we screen extensively for cancer in patients with newly diagnosed venous thromboembolism (VTE)?

Background: It is well known VTE can be the first manifestation of previously undiagnosed cancer. Retrospective studies have suggested “limited” cancer screening, including a history and physical examination, along with basic blood work, adequately identifies malignancy in patients with unexplained VTE. However, more recent prospective studies have suggested more extensive screening, which includes imaging studies or tumor-marker measurement, can increase the rate of cancer detection.

Study design: Systematic review.

Setting: Literature search using MEDLINE, EMBASE, the Cochrane Register of Controlled Trials, and evidence-based medicine reviews.

Synopsis: Thirty-six studies of 9,516 patients with VTE reported the period prevalence of previously undiagnosed cancer from baseline to 12 months was 6.3% (95% confidence interval (CI) of 5.6% to 6.9%) in all patients with VTE, and was even higher in patients with unprovoked VTE, 10% (95% CI 8.6% to 11.3%). Of the 34 articles used for prevalence assessment, an extensive screening strategy using CT scans of the abdomen and pelvis increased the proportion of previously undiagnosed cancer detection from 49.4% (CI, 40.2% to 58.5%; limited screening) to 69.7% (CI, 61.1% to 77.8%) in patients with unprovoked VTE. Ultrasonography of the abdomen and pelvis and tumor-marker screening did not result in a clinically significant increase in the frequency of cancer detection.

Four studies compared the rate of detection of early-stage, previously undiagnosed cancer between the limited and extensive screening strategies. Extensive screening led to an absolute decrease in cancer-related mortality of 1.9%, but this difference was not statistically significant.

In this systematic review, there is a great deal of heterogeneity in the studies. Most of the studies did not look at whether an increase in detection of new malignant conditions resulted in a change in the detection rate of early-stage cancer, or a decrease in cancer-related morbidity, cancer-related mortality, or overall mortality. Furthermore, the studies did not assess the consequences of extensive screening, such as patient anxiety and discomfort, testing complications, burden of additional tests for false-positive results, or cost-effectiveness. However, it is important for hospitalists to recognize undiagnosed cancer is common in unexplained VTE and warrants at least a limited-screening approach with more extensive screening.

Bottom line: Although the prevalence of undiagnosed cancer is common in VTE, extensive screening did not offer a cancer-related mortality benefit. CT of the abdomen and pelvis did, however, lead to a greater number of cancer diagnoses in patients with unexplained VTE.

 

 

Citation: Carrier M, Le Gal G, Wells PS, Fergusson D, Ramsay T, Rodger MA. Systematic review: the Trousseau syndrome revisited: should we screen extensively for cancer in patients with venous thromboembolism? Ann Intern Med. 2008;149: 323-333.

Does the use of preadmission statins decrease the risk of death, bacteremia, and pulmonary complications in patients admitted with pneumonia?

Background: Both experimental and clinical studies have suggested statins improve outcomes in severe infections, such as sepsis. This is thought to be due to the antithrombotic, anti-inflammatory, and immunomodulatory effects of statins. However, previous studies on the effect of statins on pneumonia have conflicting outcomes.

Study design: Population-based cohort study of 29,900 patients.

Setting: Danish Health Registry.

Synopsis: Researchers studied patients ages 15 years and older hospitalized with pneumonia for the first time between January 1997 and December 2004. While statin users had more co-morbidities than nonusers, the 30-day mortality was 10.3% in users, compared with 15.7% in nonusers, corresponding to an adjusted 30-day mortality rate ratio of 0.69 (95% CI of 0.58-0.82). The 90-day mortality ratio was 16.8% in users, compared with 22.4% in nonusers, corresponding to an adjusted 90-day mortality ratio of 0.75 (95% CI of 0.65-0.86). Former use of statins was not associated with a decreased risk of death. The adjusted risk for bacteremia and pulmonary complications was not significantly different between nonusers and users.

Because this was an observational study, a causal relationship cannot be determined. Hospitalists should be alerted to the possibility statins might, in the future, prove to be a standard treatment modality in pneumonia. A randomized, double-blind trial might help further determine the effect of the acute use of statins on pneumonia outcomes.

Bottom line: Preadmission statin use is associated with a decrease in 30- and 90-day mortality in pneumonia.

Citation: Thomsen RW, Riis A, Kornum JB, Christensen S, Johnsen SP, Sorensen HT. Preadmission use of statins and outcomes after hospitalization with pneumonia. Arch Intern Med. 2008;168(19):2081-2087.

Do outcomes differ when patients with acute myocardial infarction (MI) undergo PCI with drug-eluting stents (DES) compared with bare-metal stents?

Background: Randomized trials comparing drug-eluting stents with bare-metal stents in acute MI have been limited in size and duration. Concern exists regarding higher mortality among patients with ST-elevation MI treated with DES.

Study design: Observational, cohort study.

Setting: Patients were identified from a state-mandated database, in which all PCI performed in Massachusetts are reported.

Synopsis: Between April 2003 and September 2004, 7,217 eligible patients underwent stenting for acute MI. They were assigned to either the DES group or the bare-metal stent (BMS) group using propensity score matching. Patients in the DES group had lower mortality at two years, compared to a matched cohort of patients in the BMS group with MI (10.7% vs. 12.8%; absolute risk difference, -2.1%, CI, -3.8% to -0.4%). A statistically significant difference was noted among patients with or without ST-elevation MI.

The rates of target vessel revascularization at two years with MI were significantly lower among patients receiving DES than among those receiving BMS (9.6% vs. 14.5%; risk difference, -4.9%; CI, -6.1% to -3.1%).

The study is limited by its observational nature and residual confounding bias after matching. Importantly, this study was performed to determine if DESs were harmful, and the finding of reduced mortality was unanticipated.

Bottom line: Although patients with acute MI treated with drug-eluting stents had lower mortality and repeat revascularization rates compared with bare-metal stents, this outcome merits confirmation in randomized trials.

Citation: Mauri L, Silbaugh TS, Garg P, et al. Drug-eluting or bare-metal stents for acute myocardial infarction. N Engl J Med. 2008;359 (13):1330-1342.

Issue
The Hospitalist - 2009(04)
Publications
Sections

In This Edition

Does a short duration of perioperative smoking cessation lead to a reduction in postoperative complications?

Background: Prior studies have demonstrated a reduction in postoperative complications when patients stop smoking in the perioperative period. However, they have not clearly shown what effect a fairly short duration of cessation, such as a period of only four weeks, has on the frequency of complications.

Study design: Randomized controlled trial.

Setting: Four university-affiliated hospitals in Sweden.

Synopsis: Using 117 patients who were daily smokers for less than one year between the ages of 18-79 who were scheduled for elective general or orthopedic surgery, this study showed that a smoking-cessation intervention initiated as little as four weeks prior to surgery resulted in fewer postoperative complications. The complication rate was reduced from 41% in the control group to 21% in the intervention group, which received cessation counseling and nicotine-replacement therapy. The relative risk reduction was 49% (95% confidence interval, 3-40) with a number needed to treat of five.

Because this was a randomized controlled trial with a large observed benefit, it appears to be reasonable to endorse perioperative smoking cessation as late as four weeks before an elective surgery. The study was limited in its ability to detect a difference in wound infections by the small sample size and the possibility patients might have unblinded themselves to outcome assessors, causing an overestimation of the effect of the intervention on the primary outcome of all complications.

Bottom line: Perioperative smoking cessation reduces postoperative complications even when started just four weeks prior to surgery.

Citation: Lindstrom D, Azodi OS, Wladis A, et al. Effects of a perioperative smoking cessation intervention on postoperative complications. Ann Surg. 2008;248(5):739-745.

Does implantable-defibrillator therapy cause deterioration in quality of life for patients with heart failure?

Background: Patients with depressed left-ventricular function are known to have improved survival after receiving implantable cardioverter defibrillators (ICDs). However, there is concern ICD therapy can prolong survival at the expense of a diminished quality of life.

Study design: Randomized placebo-controlled trial.

Setting: Multiple centers in the U.S., Canada, and New Zealand.

Synopsis: Using 2,479 patients from the Sudden Cardiac Death in Heart Failure trial who were 18 and older and had stable heart failure and depressed left-ventricular function, this study demonstrated no significant quality-of-life difference at 30 months when compared with patients who received ICD, amiodarone, and state-of-the-art medical therapy or an amiodarone placebo and state-of-the-art medical therapy. While functional status did not differ at any time between the three groups, psychological well-being was improved in the ICD group at three months (p=0.01) and 12 months (p=0.03) when compared with the placebo group, but at 30 months there was no difference between the groups.

While the trial was randomized and placebo-controlled, the investigators were unable to blind patients or outcome assessors. Nevertheless, the lack of deterioration of quality of life in ICD patients is reassuring.

 

 

Bottom line: Placement of ICDs in heart failure patients with a high risk of sudden cardiac death does not appear to decrease quality of life.

Citation: Mark DB, Anstrom KJ, Sun JL, et al. Quality of life with defibrillator therapy or amiodarone in heart failure. N Engl J Med. 2008;359:999-1008.

CLINICAL SHORTS

SERIAL 2-POINT ULTRASONOGRAPHY PLUS D-DIMER IS EQUIVALENT TO WHOLE-LEG ULTRASONOGRAPHY FOR DIAGNOSING DVT

Randomized trials show that when comparing serial 2-point ultrasonography plus D-dimer testing with whole-leg ultrasonography, the strategies were equivalent in excluding a first episode of suspected DVT in outpatients.

Citation: Bernardi E, Camporese G, Buller HR, et al. Serial 2-point ultrasonography plus D-dimer vs whole-leg color-coded Doppler ultrasonography for diagnosing suspected symptomatic deep vein thrombosis. JAMA. 2008;300(14):1653-1659.

DAILY HEMODIALYSIS IS COST-EFFECTIVE IN ICU PATIENTS WITH ACUTE KIDNEY INJURY (AKI)

Markov model based on prospective trial data shows daily hemodialysis is cost-effective for AKI in the ICU compared with alternate-day hemodialysis.

Citation: Desai AA, Baras J, Berk BB, et al. Management of acute kidney injury in the intensive care unit. Arch Intern Med. 2008;168(16):1761-1767.

THROMBOLYSIS FOR IN-HOSPITAL STROKE IS SAFE, BUT ASSOCIATED WITH DELAYS

Prospective observational trial shows thrombolysis is safe and effective for in-hospital stroke, but statistically significant delays exist compared with out-of-hospital strokes.

Citation: Masjuan J, Simal P, Fuentes B, et al. In-hospital stroke treated with intravenous tissue plasminogen activator. Stroke. 2008;39:2614-2616.

ALGORITHM CAN IDENTIFY HIGH-RISK HEART FAILURE PATIENTS

Prospective observational study identifies clinical variables for a bedside algorithm, which stratifies the risk of hospitalized heart failure patients for early mortality or readmission to identify those who might benefit from closer follow-up.

Citation: O’Connor CM, Abraham WT, Albert NM, et al. Predictors of mortality after discharge in patients hospitalized with heart failure: an analysis from the Organized Program to Initiate Lifesaving Treatment in Hospitalized Patients with Heart Failure (OPTIMIZE-HF). Am Heart J. 2008;156(4):662-673.

IN-HOSPITAL SMOKING-CESSATION INTERVENTIONS WITH FOLLOW-UP CAN WORK

Meta-analysis of 33 trials shows in-hospital smoking-cessation counseling followed up with more than one month of outpatient support can be effective.

Citation: Rigotti NA, Munafo MR, Stead LF. Smoking cessation interventions for hospitalized smokers. Arch Intern Med. 2008;168(18):1950-1960.

OMISSION OF KEY INFORMATION DURING SIGN-OUT LEADS TO ADVERSE CONSEQUENCES

An audio-taped study of sign-out among internal medicine house staff teams revealed omission of key information during sign-out resulted in delays in diagnosis or treatment.

Citation: Horwitz LI, Moin T, Krumholz HM, Wang L, Bradley EH. Consequences of inadequate sign-out for patient care. Arch Intern Med. 2008;168(16):1755-1760.

HOSPITAL PALLIATIVE CARE CONSULTATION TEAMS ARE ASSOCIATED WITH HOSPITAL COST SAVINGS

Analysis of administrative data from eight diverse hospitals with palliative-care programs revealed consultation with palliative care saved $1,696 (p<0.001) per hospital admission in patients discharged alive, and $4,098 (p=0.003) per hospital admission in patients who died in the hospital.

Citation: Morrison RS, Penrod JD, Cassel JB, et al. Cost savings associated with U.S. hospital palliative care consultation programs. Arch Intern Med. 2008;168(16):1783-1790.

HIGHER EDUCATIONAL DEBT INFLUENCES INTERNAL MEDICINE RESIDENT CAREER PLANS

U.S. medical graduates with a debt of $50,000 to $99,999 are more likely than those with no debt to choose a hospitalist career, and this preference increased with increased debt level.

Citation: McDonald FS, West CP, Popkave C, Kolars JC. Educational debt and reported career plans among internal medicine residents. Ann Intern Med. 2008;149:416-420.

BRAIN IMAGING IMPORTANT IN IDENTIFYING VASCULAR TERRITORY AFTER TIA OR MINOR STROKE

Neurologists were only moderately reliable at identifying the vascular territory of a TIA or motor stroke, highlighting the fact brain imaging is needed to accurately identify the vascular territories of these events.

Citation: Flossmann E, Redgrave JN, Briley D, Rothwell PM. Reliability of clinical diagnosis of the symptomatic vascular territory in patients with recent transient ischemic attack or minor stroke. Stroke. 2008;39:2457-2460.

HIGH-DOSE VITAMIN B SUPPLEMENTATION DOES NOT SLOW COGNITIVE DECLINE IN ALZHEIMER’S DISEASE

Multicenter, randomized, placebo-controlled trial finds no difference in the rate of cognitive decline in patients with Alzheimer’s disease treated with high-dose vitamin B supplements for 18 months.

Citation: Aisen PS, Schneider LS, Sano M, et al. High-dose B vitamin supplementation and cognitive decline in Alzheimer’s disease. JAMA. 2008; 300(15):1774-1783.

 

 

Can a simplified, revised Geneva score retain diagnostic accuracy and clinical utility?

Background: The revised Geneva score is a validated and objective clinical decision rule, but has multiple variables with different weights. This can make the tool cumbersome and difficult to remember, and could lead to inaccurate calculations and misjudgments in patient care.

Study design: Retrospective cohort study.

Setting: Four university-affiliated European hospitals.

Synopsis: Using data from two prior prospective trials involving patients with suspected pulmonary embolism (PE), this study showed re-analysis of these patients with a simplified, revised Geneva score, which gives only one point to each clinical factor, resulted in the same level of diagnostic accuracy. Specifically, data from 1,049 patients was used to construct a receiver-operating characteristic curve analysis comparing the standardized and simplified Geneva score, which showed areas under the curve of 0.75 (95% confidence interval 0.71-0.78) and 0.74 (0.70-0.77), respectively. Additionally, the safety of using this clinical tool to rule out PE was demonstrated when using both a three-level (low-intermediate probability) and a dichotomized scheme (PE unlikely) in combination with a negative D-dimer test.

The retrospective nature of the study was its major limitation. The authors suggest a prospective study to complete validation of the simplified, revised Geneva score.

Bottom line: With prospective analysis, it might be possible to further validate a simplified, revised Geneva score.

Citation: Klok FA, Mos ICM, Nijkeuter M, et al. Simplification of the revised Geneva score for assessing clinical probability of pulmonary embolism. Arch Intern Med. 2008;168(19):2131-2136.

Is the rate of postoperative major adverse cardiac events (MACEs) inversely related to time after percutaneous coronary intervention (PCI) with a drug-eluting stent (DES)?

Background: The American College of Cardiology and the American Heart Association recently released an advisory that included a recommendation to delay elective noncardiac surgery (NCS) for one year after DES placement. However, no large study addresses the timing of NCS after PCI with DES.

Study design: Retrospective observational study.

Setting: Mayo Clinic, Rochester, Minn.

Synopsis: Looking at 520 patients who had NCS after DES at the Mayo Clinic, 5.4% experienced MACEs, but the rate of MACEs was not significantly associated with the time after stent placement to surgery (p=0.337). However, observed rates of MACEs were lower after one year. Elderly patients and those going for emergent surgery are at the highest risk for MACE. Bleeding complications were not associated with antiplatelet use.

Although this study does not provide a clear cutoff time for when it is safe to proceed to NCS after DES, it is somewhat reassuring to see the relatively small number of MACEs and the lack of bleeding complications associated with antiplatelet use. However, careful coordination between hospitalists, cardiologists, anesthesiologists, and surgeons is still needed when coordinating NCS after DES, especially in the elderly or during emergent situations.

Bottom line: While time to noncardiac surgery after drug-eluting stent placement is not associated with major adverse cardiac events, observed rates of events are lower after one year.

Citation: Rabbitts JA, Nuttall GA, Brown MJ, et al. Cardiac risk of non-cardiac surgery after percutaneous coronary intervention with drug-eluting stents. Anesthesiology.2008;109: 596-604.

Is the risk of MACEs and bleeding events for patients undergoing NCS related to the time interval between PCI with bare-metal stent?

Background: In order to prevent thrombosis of bare-metal stents (BMS) placed during percutaneous coronary intervention (PCI), antiplatelet therapy is used. This poses a risk of bleeding, if surgery is needed during the antiplatelet therapy. The American College of Cardiology and the American Heart Association recommends delaying NCS for at least six weeks after PCI with BMS.

 

 

Study design: Retrospective observational study.

Setting: Mayo Clinic, Rochester, Minn.

Synopsis: Looking at 899 patients who had NCS within one year of PCI with BMS at the Mayo Clinic between Jan. 1, 1990, and Jan. 1, 2005, this study found that when NCS was done 30 days or less after PCI with BMS, the MACEs rate was 10.5%, compared with 2.8% when NCS was done 91 or more days after PCI with BMS. After a multivariable analysis, it also was shown bleeding events were not associated with time between PCI with BMS and NCS.

While the American College of Cardiology and the American Heart Association recommends delaying NCS for at least six weeks after PCI with BMS, waiting at least 90 days would permit completion of antiplatelet therapy and re-endothelialization of the stent.

Bottom line: The risk of MACEs with noncardiac surgery is lowest when performed at least 90 days after PCI with bare-metal stent.

Citation: Nuttall GA, Brown MJ, Stombaugh JW, et al. Time and cardiac risk of surgery after bare-metal stent, percutaneous coronary intervention. Anesthesiology. 2008;109: 588-595.

Should we screen extensively for cancer in patients with newly diagnosed venous thromboembolism (VTE)?

Background: It is well known VTE can be the first manifestation of previously undiagnosed cancer. Retrospective studies have suggested “limited” cancer screening, including a history and physical examination, along with basic blood work, adequately identifies malignancy in patients with unexplained VTE. However, more recent prospective studies have suggested more extensive screening, which includes imaging studies or tumor-marker measurement, can increase the rate of cancer detection.

Study design: Systematic review.

Setting: Literature search using MEDLINE, EMBASE, the Cochrane Register of Controlled Trials, and evidence-based medicine reviews.

Synopsis: Thirty-six studies of 9,516 patients with VTE reported the period prevalence of previously undiagnosed cancer from baseline to 12 months was 6.3% (95% confidence interval (CI) of 5.6% to 6.9%) in all patients with VTE, and was even higher in patients with unprovoked VTE, 10% (95% CI 8.6% to 11.3%). Of the 34 articles used for prevalence assessment, an extensive screening strategy using CT scans of the abdomen and pelvis increased the proportion of previously undiagnosed cancer detection from 49.4% (CI, 40.2% to 58.5%; limited screening) to 69.7% (CI, 61.1% to 77.8%) in patients with unprovoked VTE. Ultrasonography of the abdomen and pelvis and tumor-marker screening did not result in a clinically significant increase in the frequency of cancer detection.

Four studies compared the rate of detection of early-stage, previously undiagnosed cancer between the limited and extensive screening strategies. Extensive screening led to an absolute decrease in cancer-related mortality of 1.9%, but this difference was not statistically significant.

In this systematic review, there is a great deal of heterogeneity in the studies. Most of the studies did not look at whether an increase in detection of new malignant conditions resulted in a change in the detection rate of early-stage cancer, or a decrease in cancer-related morbidity, cancer-related mortality, or overall mortality. Furthermore, the studies did not assess the consequences of extensive screening, such as patient anxiety and discomfort, testing complications, burden of additional tests for false-positive results, or cost-effectiveness. However, it is important for hospitalists to recognize undiagnosed cancer is common in unexplained VTE and warrants at least a limited-screening approach with more extensive screening.

Bottom line: Although the prevalence of undiagnosed cancer is common in VTE, extensive screening did not offer a cancer-related mortality benefit. CT of the abdomen and pelvis did, however, lead to a greater number of cancer diagnoses in patients with unexplained VTE.

 

 

Citation: Carrier M, Le Gal G, Wells PS, Fergusson D, Ramsay T, Rodger MA. Systematic review: the Trousseau syndrome revisited: should we screen extensively for cancer in patients with venous thromboembolism? Ann Intern Med. 2008;149: 323-333.

Does the use of preadmission statins decrease the risk of death, bacteremia, and pulmonary complications in patients admitted with pneumonia?

Background: Both experimental and clinical studies have suggested statins improve outcomes in severe infections, such as sepsis. This is thought to be due to the antithrombotic, anti-inflammatory, and immunomodulatory effects of statins. However, previous studies on the effect of statins on pneumonia have conflicting outcomes.

Study design: Population-based cohort study of 29,900 patients.

Setting: Danish Health Registry.

Synopsis: Researchers studied patients ages 15 years and older hospitalized with pneumonia for the first time between January 1997 and December 2004. While statin users had more co-morbidities than nonusers, the 30-day mortality was 10.3% in users, compared with 15.7% in nonusers, corresponding to an adjusted 30-day mortality rate ratio of 0.69 (95% CI of 0.58-0.82). The 90-day mortality ratio was 16.8% in users, compared with 22.4% in nonusers, corresponding to an adjusted 90-day mortality ratio of 0.75 (95% CI of 0.65-0.86). Former use of statins was not associated with a decreased risk of death. The adjusted risk for bacteremia and pulmonary complications was not significantly different between nonusers and users.

Because this was an observational study, a causal relationship cannot be determined. Hospitalists should be alerted to the possibility statins might, in the future, prove to be a standard treatment modality in pneumonia. A randomized, double-blind trial might help further determine the effect of the acute use of statins on pneumonia outcomes.

Bottom line: Preadmission statin use is associated with a decrease in 30- and 90-day mortality in pneumonia.

Citation: Thomsen RW, Riis A, Kornum JB, Christensen S, Johnsen SP, Sorensen HT. Preadmission use of statins and outcomes after hospitalization with pneumonia. Arch Intern Med. 2008;168(19):2081-2087.

Do outcomes differ when patients with acute myocardial infarction (MI) undergo PCI with drug-eluting stents (DES) compared with bare-metal stents?

Background: Randomized trials comparing drug-eluting stents with bare-metal stents in acute MI have been limited in size and duration. Concern exists regarding higher mortality among patients with ST-elevation MI treated with DES.

Study design: Observational, cohort study.

Setting: Patients were identified from a state-mandated database, in which all PCI performed in Massachusetts are reported.

Synopsis: Between April 2003 and September 2004, 7,217 eligible patients underwent stenting for acute MI. They were assigned to either the DES group or the bare-metal stent (BMS) group using propensity score matching. Patients in the DES group had lower mortality at two years, compared to a matched cohort of patients in the BMS group with MI (10.7% vs. 12.8%; absolute risk difference, -2.1%, CI, -3.8% to -0.4%). A statistically significant difference was noted among patients with or without ST-elevation MI.

The rates of target vessel revascularization at two years with MI were significantly lower among patients receiving DES than among those receiving BMS (9.6% vs. 14.5%; risk difference, -4.9%; CI, -6.1% to -3.1%).

The study is limited by its observational nature and residual confounding bias after matching. Importantly, this study was performed to determine if DESs were harmful, and the finding of reduced mortality was unanticipated.

Bottom line: Although patients with acute MI treated with drug-eluting stents had lower mortality and repeat revascularization rates compared with bare-metal stents, this outcome merits confirmation in randomized trials.

Citation: Mauri L, Silbaugh TS, Garg P, et al. Drug-eluting or bare-metal stents for acute myocardial infarction. N Engl J Med. 2008;359 (13):1330-1342.

In This Edition

Does a short duration of perioperative smoking cessation lead to a reduction in postoperative complications?

Background: Prior studies have demonstrated a reduction in postoperative complications when patients stop smoking in the perioperative period. However, they have not clearly shown what effect a fairly short duration of cessation, such as a period of only four weeks, has on the frequency of complications.

Study design: Randomized controlled trial.

Setting: Four university-affiliated hospitals in Sweden.

Synopsis: Using 117 patients who were daily smokers for less than one year between the ages of 18-79 who were scheduled for elective general or orthopedic surgery, this study showed that a smoking-cessation intervention initiated as little as four weeks prior to surgery resulted in fewer postoperative complications. The complication rate was reduced from 41% in the control group to 21% in the intervention group, which received cessation counseling and nicotine-replacement therapy. The relative risk reduction was 49% (95% confidence interval, 3-40) with a number needed to treat of five.

Because this was a randomized controlled trial with a large observed benefit, it appears to be reasonable to endorse perioperative smoking cessation as late as four weeks before an elective surgery. The study was limited in its ability to detect a difference in wound infections by the small sample size and the possibility patients might have unblinded themselves to outcome assessors, causing an overestimation of the effect of the intervention on the primary outcome of all complications.

Bottom line: Perioperative smoking cessation reduces postoperative complications even when started just four weeks prior to surgery.

Citation: Lindstrom D, Azodi OS, Wladis A, et al. Effects of a perioperative smoking cessation intervention on postoperative complications. Ann Surg. 2008;248(5):739-745.

Does implantable-defibrillator therapy cause deterioration in quality of life for patients with heart failure?

Background: Patients with depressed left-ventricular function are known to have improved survival after receiving implantable cardioverter defibrillators (ICDs). However, there is concern ICD therapy can prolong survival at the expense of a diminished quality of life.

Study design: Randomized placebo-controlled trial.

Setting: Multiple centers in the U.S., Canada, and New Zealand.

Synopsis: Using 2,479 patients from the Sudden Cardiac Death in Heart Failure trial who were 18 and older and had stable heart failure and depressed left-ventricular function, this study demonstrated no significant quality-of-life difference at 30 months when compared with patients who received ICD, amiodarone, and state-of-the-art medical therapy or an amiodarone placebo and state-of-the-art medical therapy. While functional status did not differ at any time between the three groups, psychological well-being was improved in the ICD group at three months (p=0.01) and 12 months (p=0.03) when compared with the placebo group, but at 30 months there was no difference between the groups.

While the trial was randomized and placebo-controlled, the investigators were unable to blind patients or outcome assessors. Nevertheless, the lack of deterioration of quality of life in ICD patients is reassuring.

 

 

Bottom line: Placement of ICDs in heart failure patients with a high risk of sudden cardiac death does not appear to decrease quality of life.

Citation: Mark DB, Anstrom KJ, Sun JL, et al. Quality of life with defibrillator therapy or amiodarone in heart failure. N Engl J Med. 2008;359:999-1008.

CLINICAL SHORTS

SERIAL 2-POINT ULTRASONOGRAPHY PLUS D-DIMER IS EQUIVALENT TO WHOLE-LEG ULTRASONOGRAPHY FOR DIAGNOSING DVT

Randomized trials show that when comparing serial 2-point ultrasonography plus D-dimer testing with whole-leg ultrasonography, the strategies were equivalent in excluding a first episode of suspected DVT in outpatients.

Citation: Bernardi E, Camporese G, Buller HR, et al. Serial 2-point ultrasonography plus D-dimer vs whole-leg color-coded Doppler ultrasonography for diagnosing suspected symptomatic deep vein thrombosis. JAMA. 2008;300(14):1653-1659.

DAILY HEMODIALYSIS IS COST-EFFECTIVE IN ICU PATIENTS WITH ACUTE KIDNEY INJURY (AKI)

Markov model based on prospective trial data shows daily hemodialysis is cost-effective for AKI in the ICU compared with alternate-day hemodialysis.

Citation: Desai AA, Baras J, Berk BB, et al. Management of acute kidney injury in the intensive care unit. Arch Intern Med. 2008;168(16):1761-1767.

THROMBOLYSIS FOR IN-HOSPITAL STROKE IS SAFE, BUT ASSOCIATED WITH DELAYS

Prospective observational trial shows thrombolysis is safe and effective for in-hospital stroke, but statistically significant delays exist compared with out-of-hospital strokes.

Citation: Masjuan J, Simal P, Fuentes B, et al. In-hospital stroke treated with intravenous tissue plasminogen activator. Stroke. 2008;39:2614-2616.

ALGORITHM CAN IDENTIFY HIGH-RISK HEART FAILURE PATIENTS

Prospective observational study identifies clinical variables for a bedside algorithm, which stratifies the risk of hospitalized heart failure patients for early mortality or readmission to identify those who might benefit from closer follow-up.

Citation: O’Connor CM, Abraham WT, Albert NM, et al. Predictors of mortality after discharge in patients hospitalized with heart failure: an analysis from the Organized Program to Initiate Lifesaving Treatment in Hospitalized Patients with Heart Failure (OPTIMIZE-HF). Am Heart J. 2008;156(4):662-673.

IN-HOSPITAL SMOKING-CESSATION INTERVENTIONS WITH FOLLOW-UP CAN WORK

Meta-analysis of 33 trials shows in-hospital smoking-cessation counseling followed up with more than one month of outpatient support can be effective.

Citation: Rigotti NA, Munafo MR, Stead LF. Smoking cessation interventions for hospitalized smokers. Arch Intern Med. 2008;168(18):1950-1960.

OMISSION OF KEY INFORMATION DURING SIGN-OUT LEADS TO ADVERSE CONSEQUENCES

An audio-taped study of sign-out among internal medicine house staff teams revealed omission of key information during sign-out resulted in delays in diagnosis or treatment.

Citation: Horwitz LI, Moin T, Krumholz HM, Wang L, Bradley EH. Consequences of inadequate sign-out for patient care. Arch Intern Med. 2008;168(16):1755-1760.

HOSPITAL PALLIATIVE CARE CONSULTATION TEAMS ARE ASSOCIATED WITH HOSPITAL COST SAVINGS

Analysis of administrative data from eight diverse hospitals with palliative-care programs revealed consultation with palliative care saved $1,696 (p<0.001) per hospital admission in patients discharged alive, and $4,098 (p=0.003) per hospital admission in patients who died in the hospital.

Citation: Morrison RS, Penrod JD, Cassel JB, et al. Cost savings associated with U.S. hospital palliative care consultation programs. Arch Intern Med. 2008;168(16):1783-1790.

HIGHER EDUCATIONAL DEBT INFLUENCES INTERNAL MEDICINE RESIDENT CAREER PLANS

U.S. medical graduates with a debt of $50,000 to $99,999 are more likely than those with no debt to choose a hospitalist career, and this preference increased with increased debt level.

Citation: McDonald FS, West CP, Popkave C, Kolars JC. Educational debt and reported career plans among internal medicine residents. Ann Intern Med. 2008;149:416-420.

BRAIN IMAGING IMPORTANT IN IDENTIFYING VASCULAR TERRITORY AFTER TIA OR MINOR STROKE

Neurologists were only moderately reliable at identifying the vascular territory of a TIA or motor stroke, highlighting the fact brain imaging is needed to accurately identify the vascular territories of these events.

Citation: Flossmann E, Redgrave JN, Briley D, Rothwell PM. Reliability of clinical diagnosis of the symptomatic vascular territory in patients with recent transient ischemic attack or minor stroke. Stroke. 2008;39:2457-2460.

HIGH-DOSE VITAMIN B SUPPLEMENTATION DOES NOT SLOW COGNITIVE DECLINE IN ALZHEIMER’S DISEASE

Multicenter, randomized, placebo-controlled trial finds no difference in the rate of cognitive decline in patients with Alzheimer’s disease treated with high-dose vitamin B supplements for 18 months.

Citation: Aisen PS, Schneider LS, Sano M, et al. High-dose B vitamin supplementation and cognitive decline in Alzheimer’s disease. JAMA. 2008; 300(15):1774-1783.

 

 

Can a simplified, revised Geneva score retain diagnostic accuracy and clinical utility?

Background: The revised Geneva score is a validated and objective clinical decision rule, but has multiple variables with different weights. This can make the tool cumbersome and difficult to remember, and could lead to inaccurate calculations and misjudgments in patient care.

Study design: Retrospective cohort study.

Setting: Four university-affiliated European hospitals.

Synopsis: Using data from two prior prospective trials involving patients with suspected pulmonary embolism (PE), this study showed re-analysis of these patients with a simplified, revised Geneva score, which gives only one point to each clinical factor, resulted in the same level of diagnostic accuracy. Specifically, data from 1,049 patients was used to construct a receiver-operating characteristic curve analysis comparing the standardized and simplified Geneva score, which showed areas under the curve of 0.75 (95% confidence interval 0.71-0.78) and 0.74 (0.70-0.77), respectively. Additionally, the safety of using this clinical tool to rule out PE was demonstrated when using both a three-level (low-intermediate probability) and a dichotomized scheme (PE unlikely) in combination with a negative D-dimer test.

The retrospective nature of the study was its major limitation. The authors suggest a prospective study to complete validation of the simplified, revised Geneva score.

Bottom line: With prospective analysis, it might be possible to further validate a simplified, revised Geneva score.

Citation: Klok FA, Mos ICM, Nijkeuter M, et al. Simplification of the revised Geneva score for assessing clinical probability of pulmonary embolism. Arch Intern Med. 2008;168(19):2131-2136.

Is the rate of postoperative major adverse cardiac events (MACEs) inversely related to time after percutaneous coronary intervention (PCI) with a drug-eluting stent (DES)?

Background: The American College of Cardiology and the American Heart Association recently released an advisory that included a recommendation to delay elective noncardiac surgery (NCS) for one year after DES placement. However, no large study addresses the timing of NCS after PCI with DES.

Study design: Retrospective observational study.

Setting: Mayo Clinic, Rochester, Minn.

Synopsis: Looking at 520 patients who had NCS after DES at the Mayo Clinic, 5.4% experienced MACEs, but the rate of MACEs was not significantly associated with the time after stent placement to surgery (p=0.337). However, observed rates of MACEs were lower after one year. Elderly patients and those going for emergent surgery are at the highest risk for MACE. Bleeding complications were not associated with antiplatelet use.

Although this study does not provide a clear cutoff time for when it is safe to proceed to NCS after DES, it is somewhat reassuring to see the relatively small number of MACEs and the lack of bleeding complications associated with antiplatelet use. However, careful coordination between hospitalists, cardiologists, anesthesiologists, and surgeons is still needed when coordinating NCS after DES, especially in the elderly or during emergent situations.

Bottom line: While time to noncardiac surgery after drug-eluting stent placement is not associated with major adverse cardiac events, observed rates of events are lower after one year.

Citation: Rabbitts JA, Nuttall GA, Brown MJ, et al. Cardiac risk of non-cardiac surgery after percutaneous coronary intervention with drug-eluting stents. Anesthesiology.2008;109: 596-604.

Is the risk of MACEs and bleeding events for patients undergoing NCS related to the time interval between PCI with bare-metal stent?

Background: In order to prevent thrombosis of bare-metal stents (BMS) placed during percutaneous coronary intervention (PCI), antiplatelet therapy is used. This poses a risk of bleeding, if surgery is needed during the antiplatelet therapy. The American College of Cardiology and the American Heart Association recommends delaying NCS for at least six weeks after PCI with BMS.

 

 

Study design: Retrospective observational study.

Setting: Mayo Clinic, Rochester, Minn.

Synopsis: Looking at 899 patients who had NCS within one year of PCI with BMS at the Mayo Clinic between Jan. 1, 1990, and Jan. 1, 2005, this study found that when NCS was done 30 days or less after PCI with BMS, the MACEs rate was 10.5%, compared with 2.8% when NCS was done 91 or more days after PCI with BMS. After a multivariable analysis, it also was shown bleeding events were not associated with time between PCI with BMS and NCS.

While the American College of Cardiology and the American Heart Association recommends delaying NCS for at least six weeks after PCI with BMS, waiting at least 90 days would permit completion of antiplatelet therapy and re-endothelialization of the stent.

Bottom line: The risk of MACEs with noncardiac surgery is lowest when performed at least 90 days after PCI with bare-metal stent.

Citation: Nuttall GA, Brown MJ, Stombaugh JW, et al. Time and cardiac risk of surgery after bare-metal stent, percutaneous coronary intervention. Anesthesiology. 2008;109: 588-595.

Should we screen extensively for cancer in patients with newly diagnosed venous thromboembolism (VTE)?

Background: It is well known VTE can be the first manifestation of previously undiagnosed cancer. Retrospective studies have suggested “limited” cancer screening, including a history and physical examination, along with basic blood work, adequately identifies malignancy in patients with unexplained VTE. However, more recent prospective studies have suggested more extensive screening, which includes imaging studies or tumor-marker measurement, can increase the rate of cancer detection.

Study design: Systematic review.

Setting: Literature search using MEDLINE, EMBASE, the Cochrane Register of Controlled Trials, and evidence-based medicine reviews.

Synopsis: Thirty-six studies of 9,516 patients with VTE reported the period prevalence of previously undiagnosed cancer from baseline to 12 months was 6.3% (95% confidence interval (CI) of 5.6% to 6.9%) in all patients with VTE, and was even higher in patients with unprovoked VTE, 10% (95% CI 8.6% to 11.3%). Of the 34 articles used for prevalence assessment, an extensive screening strategy using CT scans of the abdomen and pelvis increased the proportion of previously undiagnosed cancer detection from 49.4% (CI, 40.2% to 58.5%; limited screening) to 69.7% (CI, 61.1% to 77.8%) in patients with unprovoked VTE. Ultrasonography of the abdomen and pelvis and tumor-marker screening did not result in a clinically significant increase in the frequency of cancer detection.

Four studies compared the rate of detection of early-stage, previously undiagnosed cancer between the limited and extensive screening strategies. Extensive screening led to an absolute decrease in cancer-related mortality of 1.9%, but this difference was not statistically significant.

In this systematic review, there is a great deal of heterogeneity in the studies. Most of the studies did not look at whether an increase in detection of new malignant conditions resulted in a change in the detection rate of early-stage cancer, or a decrease in cancer-related morbidity, cancer-related mortality, or overall mortality. Furthermore, the studies did not assess the consequences of extensive screening, such as patient anxiety and discomfort, testing complications, burden of additional tests for false-positive results, or cost-effectiveness. However, it is important for hospitalists to recognize undiagnosed cancer is common in unexplained VTE and warrants at least a limited-screening approach with more extensive screening.

Bottom line: Although the prevalence of undiagnosed cancer is common in VTE, extensive screening did not offer a cancer-related mortality benefit. CT of the abdomen and pelvis did, however, lead to a greater number of cancer diagnoses in patients with unexplained VTE.

 

 

Citation: Carrier M, Le Gal G, Wells PS, Fergusson D, Ramsay T, Rodger MA. Systematic review: the Trousseau syndrome revisited: should we screen extensively for cancer in patients with venous thromboembolism? Ann Intern Med. 2008;149: 323-333.

Does the use of preadmission statins decrease the risk of death, bacteremia, and pulmonary complications in patients admitted with pneumonia?

Background: Both experimental and clinical studies have suggested statins improve outcomes in severe infections, such as sepsis. This is thought to be due to the antithrombotic, anti-inflammatory, and immunomodulatory effects of statins. However, previous studies on the effect of statins on pneumonia have conflicting outcomes.

Study design: Population-based cohort study of 29,900 patients.

Setting: Danish Health Registry.

Synopsis: Researchers studied patients ages 15 years and older hospitalized with pneumonia for the first time between January 1997 and December 2004. While statin users had more co-morbidities than nonusers, the 30-day mortality was 10.3% in users, compared with 15.7% in nonusers, corresponding to an adjusted 30-day mortality rate ratio of 0.69 (95% CI of 0.58-0.82). The 90-day mortality ratio was 16.8% in users, compared with 22.4% in nonusers, corresponding to an adjusted 90-day mortality ratio of 0.75 (95% CI of 0.65-0.86). Former use of statins was not associated with a decreased risk of death. The adjusted risk for bacteremia and pulmonary complications was not significantly different between nonusers and users.

Because this was an observational study, a causal relationship cannot be determined. Hospitalists should be alerted to the possibility statins might, in the future, prove to be a standard treatment modality in pneumonia. A randomized, double-blind trial might help further determine the effect of the acute use of statins on pneumonia outcomes.

Bottom line: Preadmission statin use is associated with a decrease in 30- and 90-day mortality in pneumonia.

Citation: Thomsen RW, Riis A, Kornum JB, Christensen S, Johnsen SP, Sorensen HT. Preadmission use of statins and outcomes after hospitalization with pneumonia. Arch Intern Med. 2008;168(19):2081-2087.

Do outcomes differ when patients with acute myocardial infarction (MI) undergo PCI with drug-eluting stents (DES) compared with bare-metal stents?

Background: Randomized trials comparing drug-eluting stents with bare-metal stents in acute MI have been limited in size and duration. Concern exists regarding higher mortality among patients with ST-elevation MI treated with DES.

Study design: Observational, cohort study.

Setting: Patients were identified from a state-mandated database, in which all PCI performed in Massachusetts are reported.

Synopsis: Between April 2003 and September 2004, 7,217 eligible patients underwent stenting for acute MI. They were assigned to either the DES group or the bare-metal stent (BMS) group using propensity score matching. Patients in the DES group had lower mortality at two years, compared to a matched cohort of patients in the BMS group with MI (10.7% vs. 12.8%; absolute risk difference, -2.1%, CI, -3.8% to -0.4%). A statistically significant difference was noted among patients with or without ST-elevation MI.

The rates of target vessel revascularization at two years with MI were significantly lower among patients receiving DES than among those receiving BMS (9.6% vs. 14.5%; risk difference, -4.9%; CI, -6.1% to -3.1%).

The study is limited by its observational nature and residual confounding bias after matching. Importantly, this study was performed to determine if DESs were harmful, and the finding of reduced mortality was unanticipated.

Bottom line: Although patients with acute MI treated with drug-eluting stents had lower mortality and repeat revascularization rates compared with bare-metal stents, this outcome merits confirmation in randomized trials.

Citation: Mauri L, Silbaugh TS, Garg P, et al. Drug-eluting or bare-metal stents for acute myocardial infarction. N Engl J Med. 2008;359 (13):1330-1342.

Issue
The Hospitalist - 2009(04)
Issue
The Hospitalist - 2009(04)
Publications
Publications
Article Type
Display Headline
The latest research you need to know
Display Headline
The latest research you need to know
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)