User login
Association Between DCBN and LOS
Slow hospital throughputthe process whereby a patient is admitted, placed in a room, and eventually dischargedcan worsen outcomes if admitted patients are boarded in emergency rooms or postanesthesia units.[1] One potential method to improve throughput is to discharge patients earlier in the day,[2] freeing up available beds and conceivably reducing hospital length of stay (LOS).
To quantify throughput, hospitals are beginning to measure the proportion of patients discharged before noon (DCBN). One study, looking at discharges on a single medical floor in an urban academic medical center, suggested that increasing the percentage of patients discharged by noon decreased observed‐to‐expected LOS in hospitalized medicine patients,[3] and a follow‐up study demonstrated that it was associated with admissions from the emergency department occurring earlier in the day.[4] However, these studies did not adjust for changes in case mix index (CMI) and other patient‐level characteristics that may also have affected these outcomes. Concerns persist that more efforts to discharge patients by noon could inadvertently increase LOS if staff chose to keep patients overnight for an early discharge the following day.
We undertook a retrospective analysis of data from patients discharged from a large academic medical center where an institution‐wide emphasis was placed on discharging more patients by noon. Using these data, we examined the association between discharges before noon and LOS in medical and surgical inpatients.
METHODS
Site and Subjects
Our study was based at the University of California, San Francisco (UCSF) Medical Center, a 400‐bed academic hospital located in San Francisco, California. We examined adult medical and surgical discharges from July 2012 through April 2015. Patients who stayed less than 24 hours or more than 20 days were excluded. Discharges from the hospital medicine service and the following surgical services were included in the analysis: cardiac surgery, colorectal surgery, cardiothoracic surgery, general surgery, gynecologic oncology, gynecology, neurosurgery, orthopedics, otolaryngology, head and neck surgery, plastic surgery, thoracic surgery, urology, and vascular surgery. No exclusions were made based on patient status (eg, observation vs inpatient). UCSF's institutional review board approved our study.
During the time of our study, discharges before noon time became an institutional priority. To this end, rates of DCBN were tracked using retrospective data, and various units undertook efforts such as informal afternoon meetings to prompt planning for the next morning's discharges. These efforts did not differentially affect medical or surgical units or emergent or nonemergent admissions, and no financial incentives or other changes in workflow were in place to increase DCBN rates.
Data Sources
We used the cost accounting system at UCSF (Enterprise Performance System Inc. [EPSI], Chicago, IL) to collect demographic information about each patient, including age, sex, primary race, and primary ethnicity. This system was also used to collect characteristics of each hospitalization including LOS (calculated from admission date time and discharge date time), hospital service at discharge, the discharge attending, discharge disposition of the patient, and the CMI, a marker of the severity of illness of the patient during that hospitalization. EPSI was also used to collect data on the admission type of all patients, either emergent, urgent, or routine, and the insurance status of the patient during that hospitalization.
Data on time of discharge were entered by the discharging nurse or unit assistant to reflect the time the patient left the hospital. Using these data, we defined a before‐noon discharge as one taking place between 8:00 am and 12:00 pm.
Statistical Analysis
Wilcoxon rank sum test and 2 statistics were used to compare baseline characteristics of hospitalizations of patients discharged before and after noon.
We used generalized linear models to assess the association of a discharge before noon on the LOS with gamma models. We accounted for clustering of discharge attendings using generalized estimating equations with exchangeable working correlation and robust standard errors. After the initial unadjusted analyses, covariates were included in the adjusted analysis if they were associated with an LOS at P < 0.05 or reasons of face validity. These variables are shown in Table 1. Because an effort to increase the discharges before noon was started in the 2014 academic year, we added an interaction term between the date of discharge and whether a discharge occurred before noon. The interaction term was included by dividing the study period into time periods corresponding to sequential 6‐month intervals. A new variable was defined by a categorical variable that indicated in which of these time periods a discharge occurred.
Discharged Before Noon | Discharged After Noon | P Value | |
---|---|---|---|
| |||
Median LOS (IQR) | 3.4 (2.25.9) | 3.7 (2.36.3) | <0.0005 |
Median CMI (IQR) | 1.8 (1.12.4) | 1.7 (1.12.5) | 0.006 |
Service type, N (%) | |||
Hospital medicine | 1,919 (29.6) | 11,290 (35.4) | |
Surgical services | 4,565 (70.4) | 20,591 (64.6) | <0.0005 |
Discharged before noon, N (%) | 6,484 (16.9) | 31,881 (83.1) | |
Discharged on weekend, N (%) | |||
Yes | 1,543 (23.8) | 7,411 (23.3) | |
No | 4,941 (76.2) | 24,470 (76.8) | 0.34 |
Discharge disposition, N (%) | |||
Home with home health | 748 (11.5) | 5,774 (18.1) | |
Home without home health | 3,997 (61.6) | 17,862 (56.0) | |
SNF | 837 (12.9) | 3,082 (9.7) | |
Other | 902 (13.9) | 5,163 (16.2) | <0.0005 |
6‐month interval, N (%) | |||
JulyDecember 2012 | 993 (15.3) | 5,596 (17.6) | |
JanuaryJune 2013 | 980 (15.1) | 5,721 (17.9) | |
JulyDecember 2013 | 1,088 (16.8) | 5,690 (17.9) | |
JanuaryJune 2014 | 1,288 (19.9) | 5,441 (17.1) | |
JulyDecember 2014 | 1,275 (19.7) | 5,656 (17.7) | |
JanuaryApril 2015 | 860 (13.3) | 3,777 (11.9) | <0.0005 |
Age category, N (%) | |||
1864 years | 4,177 (64.4) | 20,044 (62.9) | |
65+ years | 2,307 (35.6) | 11,837 (37.1) | 0.02 |
Male, N (%) | 3,274 (50.5) | 15,596 (48.9) | |
Female, N (%) | 3,210 (49.5) | 16,284 (51.1) | 0.06 |
Race, N (%) | |||
White or Caucasian | 4,133 (63.7) | 18,798 (59.0) | |
African American | 518 (8.0) | 3,020 (9.5) | |
Asian | 703 (10.8) | 4,052 (12.7) | |
Other | 1,130 (17.4) | 6,011 (18.9) | <0.0005 |
Ethnicity, N (%) | |||
Hispanic or Latino | 691 (10.7) | 3,713 (11.7) | |
Not Hispanic or Latino | 5,597 (86.3) | 27,209 (85.4) | |
Unknown/declined | 196 (3.0) | 959 (3.0) | 0.07 |
Admission type, N (%) | |||
Elective | 3,494 (53.9) | 13,881 (43.5) | |
Emergency | 2,047 (31.6) | 12,145 (38.1) | |
Urgent | 889 (13.7) | 5,459 (17.1) | |
Other | 54 (0.8) | 396 (1.2) | <0.0005 |
Payor class, N (%) | |||
Medicare | 2,648 (40.8) | 13,808 (43.3) | |
Medi‐Cal | 1,060 (16.4) | 5,913 (18.6) | |
Commercial | 2,633 (40.6) | 11,242 (35.3) | |
Other | 143 (2.2) | 918 (2.9) | <0.0005 |
We conducted a sensitivity analysis using propensity scores. The propensity score was based on demographic and clinical variables (as listed in Table 1) that exhibited P < 0.2 in bivariate analysis between the variable and being discharged before noon. We then used the propensity score as a covariate in a generalized linear model of the LOS with a gamma distribution and with generalized estimating equations as described above.
Finally, we performed prespecified secondary subset analyses of patients admitted emergently and nonemergently.
Statistical modeling and analysis was completed using Stata version 13 (StataCorp, College Station, TX).
RESULTS
Patient Demographics and Discharge Before Noon
Our study population comprised 27,983 patients for a total of 38,365 hospitalizations with a median LOS of 3.7 days. We observed 6484 discharges before noon (16.9%) and 31,881 discharges after noon (83.1%). The characteristics of the hospitalizations are shown in Table 1.
Patients who were discharged before noon tended to be younger, white, and discharged with a disposition to home without home health. The median CMI was slightly higher in discharges before noon (1.81, P = 0.006), and elective admissions were more likely than emergent to be discharged before noon (53.9% vs 31.6%, P < 0.0005).
Multivariable Analysis
A discharge before noon was associated with a 4.3% increase in LOS (adjusted odds ratio [OR]: 1.043, 95% confidence interval [CI]: 1.003‐1.086), adjusting for CMI, the service type, discharge on the weekend, discharge disposition, age, sex, ethnicity, race, urgency of admission, payor class, and a full interaction with the date of discharge (in 6‐month intervals). In preplanned subset analyses, the association between longer LOS and DCBN was more pronounced in patients admitted emergently (adjusted OR: 1.14, 95% CI: 1.033‐1.249) and less pronounced for patients not admitted emergently (adjusted OR: 1.03, 95% CI: 0.988‐1.074), although the latter did not meet statistical significance. In patients admitted emergently, this corresponds to approximately a 12‐hour increase in LOS. The interaction term of discharge date and DCBN was significant in the model. In further subset analyses, the association between longer LOS and DCBN was more pronounced in medicine patients (adjusted OR: 1.116, 95% CI: 1.014‐1.228) than in surgical patients (adjusted OR: 1.030, 95% CI: 0.989‐1.074), although the relationship in surgical patients did not meet statistical significance.
We also undertook sensitivity analyses utilizing propensity scores as a covariate in our base multivariable models. Results from these analyses did not differ from the base models and are not presented here. Results also did not differ when comparing discharges before and after the initiation of an attending only service.
DISCUSSION AND CONCLUSION
In our retrospective study of patients discharged from an academic medical center, discharge before noon was associated with a longer LOS, with the effect more pronounced in patients admitted emergently in the hospital. Our results suggest that efforts to discharge patients earlier in the day may have varying degrees of success depending on patient characteristics. Conceivably, elective admissions recover according to predictable plans, allowing for discharges earlier in the day. In contrast, patients discharged from emergent hospitalizations may have ongoing evolution of their care plan, making plans for discharging before noon more challenging.
Our results differ from a previous study,[3] which suggested that increasing the proportion of before‐noon discharges was associated with a fall in observed‐to‐expected LOS. However, observational studies of DCBN are challenging, because the association between early discharge and LOS is potentially bidirectional. One interpretation, for example, is that patients were kept longer in order to be discharged by noon the following day, which for the subgroups of patients admitted emergently corresponded to a roughly 12‐hour increase in LOS. However, it is also plausible that patients who stayed longer also had more time to plan for an early discharge. In either scenario, the ability of managers to utilize LOS as a key metric of throughput efforts may be flawed, and suggests that alternatives (eg, number of patients waiting for beds off unit) may be a more reasonable measure of throughput. Our results have several limitations. As in any observational study, our results are vulnerable to biases from unmeasured covariates that confound the analysis. We caution that a causal relationship between a discharge before noon and LOS cannot be determined from the nature of the study. Our results are also limited in that we were unable to adjust for day‐to‐day hospital capacity and other variables that affect LOS including caregiver and transportation availability, bed capacity at receiving care facilities, and patient consent to discharge. Finally, as a single‐site study, our findings may not be applicable to nonacademic settings.
In conclusion, our observational study discerned an association between discharging patients before noon and longer LOS. We believe our findings suggest a rationale for alternate approaches to measuring an early discharge program's effectiveness, namely, that the evaluation of the success of an early discharge initiative should consider multiple evaluation metrics including the effect on emergency department wait times, intensive care unit or postanesthesia transitions, and on patient reported experiences of care transitions.
Disclosures
Andrew Auerbach, MD, is supported by a K24 grant from the National Heart, Lung, and Blood Institute: K24HL098372. The authors report no conflicts of interest.
- The effect of emergency department crowding on clinically oriented outcomes. Acad Emerg Med. 2009;16(1):1–10. , , , et al.
- Centers for Medicare 2013.
- Discharge before noon: an achievable hospital goal. J Hosp Med. 2014;9(4):210–214. , , , et al.
- Discharge before noon: effect on throughput and sustainability. J Hosp Med. 2015;10(10):664–669. , , , et al.
Slow hospital throughputthe process whereby a patient is admitted, placed in a room, and eventually dischargedcan worsen outcomes if admitted patients are boarded in emergency rooms or postanesthesia units.[1] One potential method to improve throughput is to discharge patients earlier in the day,[2] freeing up available beds and conceivably reducing hospital length of stay (LOS).
To quantify throughput, hospitals are beginning to measure the proportion of patients discharged before noon (DCBN). One study, looking at discharges on a single medical floor in an urban academic medical center, suggested that increasing the percentage of patients discharged by noon decreased observed‐to‐expected LOS in hospitalized medicine patients,[3] and a follow‐up study demonstrated that it was associated with admissions from the emergency department occurring earlier in the day.[4] However, these studies did not adjust for changes in case mix index (CMI) and other patient‐level characteristics that may also have affected these outcomes. Concerns persist that more efforts to discharge patients by noon could inadvertently increase LOS if staff chose to keep patients overnight for an early discharge the following day.
We undertook a retrospective analysis of data from patients discharged from a large academic medical center where an institution‐wide emphasis was placed on discharging more patients by noon. Using these data, we examined the association between discharges before noon and LOS in medical and surgical inpatients.
METHODS
Site and Subjects
Our study was based at the University of California, San Francisco (UCSF) Medical Center, a 400‐bed academic hospital located in San Francisco, California. We examined adult medical and surgical discharges from July 2012 through April 2015. Patients who stayed less than 24 hours or more than 20 days were excluded. Discharges from the hospital medicine service and the following surgical services were included in the analysis: cardiac surgery, colorectal surgery, cardiothoracic surgery, general surgery, gynecologic oncology, gynecology, neurosurgery, orthopedics, otolaryngology, head and neck surgery, plastic surgery, thoracic surgery, urology, and vascular surgery. No exclusions were made based on patient status (eg, observation vs inpatient). UCSF's institutional review board approved our study.
During the time of our study, discharges before noon time became an institutional priority. To this end, rates of DCBN were tracked using retrospective data, and various units undertook efforts such as informal afternoon meetings to prompt planning for the next morning's discharges. These efforts did not differentially affect medical or surgical units or emergent or nonemergent admissions, and no financial incentives or other changes in workflow were in place to increase DCBN rates.
Data Sources
We used the cost accounting system at UCSF (Enterprise Performance System Inc. [EPSI], Chicago, IL) to collect demographic information about each patient, including age, sex, primary race, and primary ethnicity. This system was also used to collect characteristics of each hospitalization including LOS (calculated from admission date time and discharge date time), hospital service at discharge, the discharge attending, discharge disposition of the patient, and the CMI, a marker of the severity of illness of the patient during that hospitalization. EPSI was also used to collect data on the admission type of all patients, either emergent, urgent, or routine, and the insurance status of the patient during that hospitalization.
Data on time of discharge were entered by the discharging nurse or unit assistant to reflect the time the patient left the hospital. Using these data, we defined a before‐noon discharge as one taking place between 8:00 am and 12:00 pm.
Statistical Analysis
Wilcoxon rank sum test and 2 statistics were used to compare baseline characteristics of hospitalizations of patients discharged before and after noon.
We used generalized linear models to assess the association of a discharge before noon on the LOS with gamma models. We accounted for clustering of discharge attendings using generalized estimating equations with exchangeable working correlation and robust standard errors. After the initial unadjusted analyses, covariates were included in the adjusted analysis if they were associated with an LOS at P < 0.05 or reasons of face validity. These variables are shown in Table 1. Because an effort to increase the discharges before noon was started in the 2014 academic year, we added an interaction term between the date of discharge and whether a discharge occurred before noon. The interaction term was included by dividing the study period into time periods corresponding to sequential 6‐month intervals. A new variable was defined by a categorical variable that indicated in which of these time periods a discharge occurred.
Discharged Before Noon | Discharged After Noon | P Value | |
---|---|---|---|
| |||
Median LOS (IQR) | 3.4 (2.25.9) | 3.7 (2.36.3) | <0.0005 |
Median CMI (IQR) | 1.8 (1.12.4) | 1.7 (1.12.5) | 0.006 |
Service type, N (%) | |||
Hospital medicine | 1,919 (29.6) | 11,290 (35.4) | |
Surgical services | 4,565 (70.4) | 20,591 (64.6) | <0.0005 |
Discharged before noon, N (%) | 6,484 (16.9) | 31,881 (83.1) | |
Discharged on weekend, N (%) | |||
Yes | 1,543 (23.8) | 7,411 (23.3) | |
No | 4,941 (76.2) | 24,470 (76.8) | 0.34 |
Discharge disposition, N (%) | |||
Home with home health | 748 (11.5) | 5,774 (18.1) | |
Home without home health | 3,997 (61.6) | 17,862 (56.0) | |
SNF | 837 (12.9) | 3,082 (9.7) | |
Other | 902 (13.9) | 5,163 (16.2) | <0.0005 |
6‐month interval, N (%) | |||
JulyDecember 2012 | 993 (15.3) | 5,596 (17.6) | |
JanuaryJune 2013 | 980 (15.1) | 5,721 (17.9) | |
JulyDecember 2013 | 1,088 (16.8) | 5,690 (17.9) | |
JanuaryJune 2014 | 1,288 (19.9) | 5,441 (17.1) | |
JulyDecember 2014 | 1,275 (19.7) | 5,656 (17.7) | |
JanuaryApril 2015 | 860 (13.3) | 3,777 (11.9) | <0.0005 |
Age category, N (%) | |||
1864 years | 4,177 (64.4) | 20,044 (62.9) | |
65+ years | 2,307 (35.6) | 11,837 (37.1) | 0.02 |
Male, N (%) | 3,274 (50.5) | 15,596 (48.9) | |
Female, N (%) | 3,210 (49.5) | 16,284 (51.1) | 0.06 |
Race, N (%) | |||
White or Caucasian | 4,133 (63.7) | 18,798 (59.0) | |
African American | 518 (8.0) | 3,020 (9.5) | |
Asian | 703 (10.8) | 4,052 (12.7) | |
Other | 1,130 (17.4) | 6,011 (18.9) | <0.0005 |
Ethnicity, N (%) | |||
Hispanic or Latino | 691 (10.7) | 3,713 (11.7) | |
Not Hispanic or Latino | 5,597 (86.3) | 27,209 (85.4) | |
Unknown/declined | 196 (3.0) | 959 (3.0) | 0.07 |
Admission type, N (%) | |||
Elective | 3,494 (53.9) | 13,881 (43.5) | |
Emergency | 2,047 (31.6) | 12,145 (38.1) | |
Urgent | 889 (13.7) | 5,459 (17.1) | |
Other | 54 (0.8) | 396 (1.2) | <0.0005 |
Payor class, N (%) | |||
Medicare | 2,648 (40.8) | 13,808 (43.3) | |
Medi‐Cal | 1,060 (16.4) | 5,913 (18.6) | |
Commercial | 2,633 (40.6) | 11,242 (35.3) | |
Other | 143 (2.2) | 918 (2.9) | <0.0005 |
We conducted a sensitivity analysis using propensity scores. The propensity score was based on demographic and clinical variables (as listed in Table 1) that exhibited P < 0.2 in bivariate analysis between the variable and being discharged before noon. We then used the propensity score as a covariate in a generalized linear model of the LOS with a gamma distribution and with generalized estimating equations as described above.
Finally, we performed prespecified secondary subset analyses of patients admitted emergently and nonemergently.
Statistical modeling and analysis was completed using Stata version 13 (StataCorp, College Station, TX).
RESULTS
Patient Demographics and Discharge Before Noon
Our study population comprised 27,983 patients for a total of 38,365 hospitalizations with a median LOS of 3.7 days. We observed 6484 discharges before noon (16.9%) and 31,881 discharges after noon (83.1%). The characteristics of the hospitalizations are shown in Table 1.
Patients who were discharged before noon tended to be younger, white, and discharged with a disposition to home without home health. The median CMI was slightly higher in discharges before noon (1.81, P = 0.006), and elective admissions were more likely than emergent to be discharged before noon (53.9% vs 31.6%, P < 0.0005).
Multivariable Analysis
A discharge before noon was associated with a 4.3% increase in LOS (adjusted odds ratio [OR]: 1.043, 95% confidence interval [CI]: 1.003‐1.086), adjusting for CMI, the service type, discharge on the weekend, discharge disposition, age, sex, ethnicity, race, urgency of admission, payor class, and a full interaction with the date of discharge (in 6‐month intervals). In preplanned subset analyses, the association between longer LOS and DCBN was more pronounced in patients admitted emergently (adjusted OR: 1.14, 95% CI: 1.033‐1.249) and less pronounced for patients not admitted emergently (adjusted OR: 1.03, 95% CI: 0.988‐1.074), although the latter did not meet statistical significance. In patients admitted emergently, this corresponds to approximately a 12‐hour increase in LOS. The interaction term of discharge date and DCBN was significant in the model. In further subset analyses, the association between longer LOS and DCBN was more pronounced in medicine patients (adjusted OR: 1.116, 95% CI: 1.014‐1.228) than in surgical patients (adjusted OR: 1.030, 95% CI: 0.989‐1.074), although the relationship in surgical patients did not meet statistical significance.
We also undertook sensitivity analyses utilizing propensity scores as a covariate in our base multivariable models. Results from these analyses did not differ from the base models and are not presented here. Results also did not differ when comparing discharges before and after the initiation of an attending only service.
DISCUSSION AND CONCLUSION
In our retrospective study of patients discharged from an academic medical center, discharge before noon was associated with a longer LOS, with the effect more pronounced in patients admitted emergently in the hospital. Our results suggest that efforts to discharge patients earlier in the day may have varying degrees of success depending on patient characteristics. Conceivably, elective admissions recover according to predictable plans, allowing for discharges earlier in the day. In contrast, patients discharged from emergent hospitalizations may have ongoing evolution of their care plan, making plans for discharging before noon more challenging.
Our results differ from a previous study,[3] which suggested that increasing the proportion of before‐noon discharges was associated with a fall in observed‐to‐expected LOS. However, observational studies of DCBN are challenging, because the association between early discharge and LOS is potentially bidirectional. One interpretation, for example, is that patients were kept longer in order to be discharged by noon the following day, which for the subgroups of patients admitted emergently corresponded to a roughly 12‐hour increase in LOS. However, it is also plausible that patients who stayed longer also had more time to plan for an early discharge. In either scenario, the ability of managers to utilize LOS as a key metric of throughput efforts may be flawed, and suggests that alternatives (eg, number of patients waiting for beds off unit) may be a more reasonable measure of throughput. Our results have several limitations. As in any observational study, our results are vulnerable to biases from unmeasured covariates that confound the analysis. We caution that a causal relationship between a discharge before noon and LOS cannot be determined from the nature of the study. Our results are also limited in that we were unable to adjust for day‐to‐day hospital capacity and other variables that affect LOS including caregiver and transportation availability, bed capacity at receiving care facilities, and patient consent to discharge. Finally, as a single‐site study, our findings may not be applicable to nonacademic settings.
In conclusion, our observational study discerned an association between discharging patients before noon and longer LOS. We believe our findings suggest a rationale for alternate approaches to measuring an early discharge program's effectiveness, namely, that the evaluation of the success of an early discharge initiative should consider multiple evaluation metrics including the effect on emergency department wait times, intensive care unit or postanesthesia transitions, and on patient reported experiences of care transitions.
Disclosures
Andrew Auerbach, MD, is supported by a K24 grant from the National Heart, Lung, and Blood Institute: K24HL098372. The authors report no conflicts of interest.
Slow hospital throughputthe process whereby a patient is admitted, placed in a room, and eventually dischargedcan worsen outcomes if admitted patients are boarded in emergency rooms or postanesthesia units.[1] One potential method to improve throughput is to discharge patients earlier in the day,[2] freeing up available beds and conceivably reducing hospital length of stay (LOS).
To quantify throughput, hospitals are beginning to measure the proportion of patients discharged before noon (DCBN). One study, looking at discharges on a single medical floor in an urban academic medical center, suggested that increasing the percentage of patients discharged by noon decreased observed‐to‐expected LOS in hospitalized medicine patients,[3] and a follow‐up study demonstrated that it was associated with admissions from the emergency department occurring earlier in the day.[4] However, these studies did not adjust for changes in case mix index (CMI) and other patient‐level characteristics that may also have affected these outcomes. Concerns persist that more efforts to discharge patients by noon could inadvertently increase LOS if staff chose to keep patients overnight for an early discharge the following day.
We undertook a retrospective analysis of data from patients discharged from a large academic medical center where an institution‐wide emphasis was placed on discharging more patients by noon. Using these data, we examined the association between discharges before noon and LOS in medical and surgical inpatients.
METHODS
Site and Subjects
Our study was based at the University of California, San Francisco (UCSF) Medical Center, a 400‐bed academic hospital located in San Francisco, California. We examined adult medical and surgical discharges from July 2012 through April 2015. Patients who stayed less than 24 hours or more than 20 days were excluded. Discharges from the hospital medicine service and the following surgical services were included in the analysis: cardiac surgery, colorectal surgery, cardiothoracic surgery, general surgery, gynecologic oncology, gynecology, neurosurgery, orthopedics, otolaryngology, head and neck surgery, plastic surgery, thoracic surgery, urology, and vascular surgery. No exclusions were made based on patient status (eg, observation vs inpatient). UCSF's institutional review board approved our study.
During the time of our study, discharges before noon time became an institutional priority. To this end, rates of DCBN were tracked using retrospective data, and various units undertook efforts such as informal afternoon meetings to prompt planning for the next morning's discharges. These efforts did not differentially affect medical or surgical units or emergent or nonemergent admissions, and no financial incentives or other changes in workflow were in place to increase DCBN rates.
Data Sources
We used the cost accounting system at UCSF (Enterprise Performance System Inc. [EPSI], Chicago, IL) to collect demographic information about each patient, including age, sex, primary race, and primary ethnicity. This system was also used to collect characteristics of each hospitalization including LOS (calculated from admission date time and discharge date time), hospital service at discharge, the discharge attending, discharge disposition of the patient, and the CMI, a marker of the severity of illness of the patient during that hospitalization. EPSI was also used to collect data on the admission type of all patients, either emergent, urgent, or routine, and the insurance status of the patient during that hospitalization.
Data on time of discharge were entered by the discharging nurse or unit assistant to reflect the time the patient left the hospital. Using these data, we defined a before‐noon discharge as one taking place between 8:00 am and 12:00 pm.
Statistical Analysis
Wilcoxon rank sum test and 2 statistics were used to compare baseline characteristics of hospitalizations of patients discharged before and after noon.
We used generalized linear models to assess the association of a discharge before noon on the LOS with gamma models. We accounted for clustering of discharge attendings using generalized estimating equations with exchangeable working correlation and robust standard errors. After the initial unadjusted analyses, covariates were included in the adjusted analysis if they were associated with an LOS at P < 0.05 or reasons of face validity. These variables are shown in Table 1. Because an effort to increase the discharges before noon was started in the 2014 academic year, we added an interaction term between the date of discharge and whether a discharge occurred before noon. The interaction term was included by dividing the study period into time periods corresponding to sequential 6‐month intervals. A new variable was defined by a categorical variable that indicated in which of these time periods a discharge occurred.
Discharged Before Noon | Discharged After Noon | P Value | |
---|---|---|---|
| |||
Median LOS (IQR) | 3.4 (2.25.9) | 3.7 (2.36.3) | <0.0005 |
Median CMI (IQR) | 1.8 (1.12.4) | 1.7 (1.12.5) | 0.006 |
Service type, N (%) | |||
Hospital medicine | 1,919 (29.6) | 11,290 (35.4) | |
Surgical services | 4,565 (70.4) | 20,591 (64.6) | <0.0005 |
Discharged before noon, N (%) | 6,484 (16.9) | 31,881 (83.1) | |
Discharged on weekend, N (%) | |||
Yes | 1,543 (23.8) | 7,411 (23.3) | |
No | 4,941 (76.2) | 24,470 (76.8) | 0.34 |
Discharge disposition, N (%) | |||
Home with home health | 748 (11.5) | 5,774 (18.1) | |
Home without home health | 3,997 (61.6) | 17,862 (56.0) | |
SNF | 837 (12.9) | 3,082 (9.7) | |
Other | 902 (13.9) | 5,163 (16.2) | <0.0005 |
6‐month interval, N (%) | |||
JulyDecember 2012 | 993 (15.3) | 5,596 (17.6) | |
JanuaryJune 2013 | 980 (15.1) | 5,721 (17.9) | |
JulyDecember 2013 | 1,088 (16.8) | 5,690 (17.9) | |
JanuaryJune 2014 | 1,288 (19.9) | 5,441 (17.1) | |
JulyDecember 2014 | 1,275 (19.7) | 5,656 (17.7) | |
JanuaryApril 2015 | 860 (13.3) | 3,777 (11.9) | <0.0005 |
Age category, N (%) | |||
1864 years | 4,177 (64.4) | 20,044 (62.9) | |
65+ years | 2,307 (35.6) | 11,837 (37.1) | 0.02 |
Male, N (%) | 3,274 (50.5) | 15,596 (48.9) | |
Female, N (%) | 3,210 (49.5) | 16,284 (51.1) | 0.06 |
Race, N (%) | |||
White or Caucasian | 4,133 (63.7) | 18,798 (59.0) | |
African American | 518 (8.0) | 3,020 (9.5) | |
Asian | 703 (10.8) | 4,052 (12.7) | |
Other | 1,130 (17.4) | 6,011 (18.9) | <0.0005 |
Ethnicity, N (%) | |||
Hispanic or Latino | 691 (10.7) | 3,713 (11.7) | |
Not Hispanic or Latino | 5,597 (86.3) | 27,209 (85.4) | |
Unknown/declined | 196 (3.0) | 959 (3.0) | 0.07 |
Admission type, N (%) | |||
Elective | 3,494 (53.9) | 13,881 (43.5) | |
Emergency | 2,047 (31.6) | 12,145 (38.1) | |
Urgent | 889 (13.7) | 5,459 (17.1) | |
Other | 54 (0.8) | 396 (1.2) | <0.0005 |
Payor class, N (%) | |||
Medicare | 2,648 (40.8) | 13,808 (43.3) | |
Medi‐Cal | 1,060 (16.4) | 5,913 (18.6) | |
Commercial | 2,633 (40.6) | 11,242 (35.3) | |
Other | 143 (2.2) | 918 (2.9) | <0.0005 |
We conducted a sensitivity analysis using propensity scores. The propensity score was based on demographic and clinical variables (as listed in Table 1) that exhibited P < 0.2 in bivariate analysis between the variable and being discharged before noon. We then used the propensity score as a covariate in a generalized linear model of the LOS with a gamma distribution and with generalized estimating equations as described above.
Finally, we performed prespecified secondary subset analyses of patients admitted emergently and nonemergently.
Statistical modeling and analysis was completed using Stata version 13 (StataCorp, College Station, TX).
RESULTS
Patient Demographics and Discharge Before Noon
Our study population comprised 27,983 patients for a total of 38,365 hospitalizations with a median LOS of 3.7 days. We observed 6484 discharges before noon (16.9%) and 31,881 discharges after noon (83.1%). The characteristics of the hospitalizations are shown in Table 1.
Patients who were discharged before noon tended to be younger, white, and discharged with a disposition to home without home health. The median CMI was slightly higher in discharges before noon (1.81, P = 0.006), and elective admissions were more likely than emergent to be discharged before noon (53.9% vs 31.6%, P < 0.0005).
Multivariable Analysis
A discharge before noon was associated with a 4.3% increase in LOS (adjusted odds ratio [OR]: 1.043, 95% confidence interval [CI]: 1.003‐1.086), adjusting for CMI, the service type, discharge on the weekend, discharge disposition, age, sex, ethnicity, race, urgency of admission, payor class, and a full interaction with the date of discharge (in 6‐month intervals). In preplanned subset analyses, the association between longer LOS and DCBN was more pronounced in patients admitted emergently (adjusted OR: 1.14, 95% CI: 1.033‐1.249) and less pronounced for patients not admitted emergently (adjusted OR: 1.03, 95% CI: 0.988‐1.074), although the latter did not meet statistical significance. In patients admitted emergently, this corresponds to approximately a 12‐hour increase in LOS. The interaction term of discharge date and DCBN was significant in the model. In further subset analyses, the association between longer LOS and DCBN was more pronounced in medicine patients (adjusted OR: 1.116, 95% CI: 1.014‐1.228) than in surgical patients (adjusted OR: 1.030, 95% CI: 0.989‐1.074), although the relationship in surgical patients did not meet statistical significance.
We also undertook sensitivity analyses utilizing propensity scores as a covariate in our base multivariable models. Results from these analyses did not differ from the base models and are not presented here. Results also did not differ when comparing discharges before and after the initiation of an attending only service.
DISCUSSION AND CONCLUSION
In our retrospective study of patients discharged from an academic medical center, discharge before noon was associated with a longer LOS, with the effect more pronounced in patients admitted emergently in the hospital. Our results suggest that efforts to discharge patients earlier in the day may have varying degrees of success depending on patient characteristics. Conceivably, elective admissions recover according to predictable plans, allowing for discharges earlier in the day. In contrast, patients discharged from emergent hospitalizations may have ongoing evolution of their care plan, making plans for discharging before noon more challenging.
Our results differ from a previous study,[3] which suggested that increasing the proportion of before‐noon discharges was associated with a fall in observed‐to‐expected LOS. However, observational studies of DCBN are challenging, because the association between early discharge and LOS is potentially bidirectional. One interpretation, for example, is that patients were kept longer in order to be discharged by noon the following day, which for the subgroups of patients admitted emergently corresponded to a roughly 12‐hour increase in LOS. However, it is also plausible that patients who stayed longer also had more time to plan for an early discharge. In either scenario, the ability of managers to utilize LOS as a key metric of throughput efforts may be flawed, and suggests that alternatives (eg, number of patients waiting for beds off unit) may be a more reasonable measure of throughput. Our results have several limitations. As in any observational study, our results are vulnerable to biases from unmeasured covariates that confound the analysis. We caution that a causal relationship between a discharge before noon and LOS cannot be determined from the nature of the study. Our results are also limited in that we were unable to adjust for day‐to‐day hospital capacity and other variables that affect LOS including caregiver and transportation availability, bed capacity at receiving care facilities, and patient consent to discharge. Finally, as a single‐site study, our findings may not be applicable to nonacademic settings.
In conclusion, our observational study discerned an association between discharging patients before noon and longer LOS. We believe our findings suggest a rationale for alternate approaches to measuring an early discharge program's effectiveness, namely, that the evaluation of the success of an early discharge initiative should consider multiple evaluation metrics including the effect on emergency department wait times, intensive care unit or postanesthesia transitions, and on patient reported experiences of care transitions.
Disclosures
Andrew Auerbach, MD, is supported by a K24 grant from the National Heart, Lung, and Blood Institute: K24HL098372. The authors report no conflicts of interest.
- The effect of emergency department crowding on clinically oriented outcomes. Acad Emerg Med. 2009;16(1):1–10. , , , et al.
- Centers for Medicare 2013.
- Discharge before noon: an achievable hospital goal. J Hosp Med. 2014;9(4):210–214. , , , et al.
- Discharge before noon: effect on throughput and sustainability. J Hosp Med. 2015;10(10):664–669. , , , et al.
- The effect of emergency department crowding on clinically oriented outcomes. Acad Emerg Med. 2009;16(1):1–10. , , , et al.
- Centers for Medicare 2013.
- Discharge before noon: an achievable hospital goal. J Hosp Med. 2014;9(4):210–214. , , , et al.
- Discharge before noon: effect on throughput and sustainability. J Hosp Med. 2015;10(10):664–669. , , , et al.
© 2015 Society of Hospital Medicine
Assessing Vascular Nursing Experience
Peripherally inserted central catheters (PICCs) are among the most prevalent of venous access devices in hospitalized patients.[1, 2] Although growing use of these devices reflects clinical advantages, such as a reduced risk of complications during insertion and durable venous access, use of PICCs is also likely related to the growth of vascular access nursing.[3, 4] A relatively new specialty, vascular access nurses obtain, maintain, and manage venous access in hospitalized patients.[4, 5] Depending on their scope of practice, these professionals are responsible not only for insertion of devices, such as peripheral intravenous catheters and PICCs, but also nontunneled central venous catheters and arterial catheters in some settings.[6]
Although a growing number of US hospitals have introduced vascular nursing teams,[7] little is known about the experience, practice, knowledge, and beliefs of vascular access nurses. This knowledge gap is relevant for hospitalists and hospital medicine as (1) vascular access nurses increasingly represent a key partner in the care of hospitalized patients; (2) the knowledge and practice of these individuals directly affects patient safety and clinical outcomes; and (3) understanding experience, practice, and beliefs of these specialists can help inform decision making and quality‐improvement efforts related to PICCs. As hospitalists increasingly order the placement of and care for patients with PICCs, they are also well suited to improve PICC practice.
Therefore, we conducted a survey of vascular access nurses employed by hospitals that participate in the Michigan Hospital Medicine Safety (HMS) Consortium, a Blue Cross Blue Shield of Michiganfunded collaborative quality initiative.[6] We aimed to understand experience, practice, knowledge, and beliefs related to PICC care and use.
METHODS
Study Setting and Participants
To quantify vascular nursing experience, practice, knowledge, and beliefs, we conducted a Web‐based survey of vascular nurses across 47 Michigan hospitals that participate in HMS. A statewide quality‐improvement initiative, HMS aims to prevent adverse events in hospitalized medical patients through the creation of a data registry and sharing of best practices. The setting and design of this multicenter initiative have been previously described.[8, 9] Although participation is voluntary, each hospital receives payment for participating in the consortium and for data collection. Because HMS has an ongoing initiative aimed at identifying and preventing PICC‐related complications, this study was particularly relevant for participating hospitals and nurses.
Each HMS site has a designated quality‐improvement lead, physician champion, and data abstractor. To coordinate distribution and dissemination of the survey, we contacted the quality‐improvement leads at each site and enquired whether their hospital employed vascular access nurses who placed PICCs. Because we were only interested in responses from vascular access nurses, HMS hospitals that did not have these providers or stated PICCs were placed by other specialists (eg, interventional radiology) were excluded. At eligible sites, we obtained the total number of vascular nurses employed so as to determine the number of eligible respondents. In this manner, a purposeful sample of vascular nurses at participating HMS hospitals was constituted.
Participation in the survey was solicited through hospital quality leads that either distributed an electronic survey link to vascular nurses at their facilities or sent us individual email addresses to contact them directly. A cover letter explaining the rationale and the purpose of the survey along with the survey link was then sent to respondents through either of these routes. The survey was administered at all HMS sites contemporaneously and kept open for a period of 5 weeks. During the 5‐week period, 2 e‐mail reminders were sent to encourage participation. As a token of appreciation, a $10 Amazon gift card was offered to those who took the survey.
Development and Validation of the Survey
We developed the survey instrument (which we call PICC1 as we hope to administer longitudinally to track changes over time) by first conducting a literature search to identify relevant evidence‐based guidelines and studies regarding vascular access nursing practices and experiences.[10, 11, 12, 13] In addition, we consulted and involved national and international leaders in vascular access nursing to ensure validity and representativeness of the questions posed. We were specifically interested in nursing background, hospital practices, types of PICCs used, use of various technologies, relationships with healthcare providers, and management of complications. To understand participant characteristics and quantify potential variation in responses, we collected basic participant data including demographics, years in practice, number of PICCs placed, leadership roles, and vascular access certification status. Based on clinical reasoning and existing studies,[14, 15] we hypothesized that responses regarding certain practices (ultrasound use, electrocardiography [ECG] guidance system use), management of complications, or perceptions regarding leadership might vary based on years of experience, number of PICCs placed, or certification status. We therefore examined these associations as prespecified subgroup analyses.
The initial survey instrument was pilot tested with vascular nurses outside of the sampling frame. Based on feedback from the pilot testers, the instrument was refined and edited to improve clarity of the questions. In addition, specific skip patterns and logic were programmed into the final survey to reduce respondent burden and allow participants to seamlessly bypass questions that were contingent on a prior response (eg, use of ECG to place PICCs would lead to a series of questions about ECG‐assisted placement only for those respondents who used the technology). This final version of the survey was tested by members of the study team (V.C., L.K., S.L.K.) and then posted to SurveyMonkey for dissemination.
Statistical Analysis
Descriptive statistics (percentage, n/N) were used to tabulate results. In accordance with our a priori hypothesis that variation to responses might be associated with respondent characteristics, responses to questions regarding insertion practice (eg, use of ultrasound, measurement of catheter:vein ratio, trimming of catheters) and approach to complications (eg, catheter occlusion, deep vein thrombosis [DVT] notification, and PICC removal in the setting of fever) were compared by respondent years in practice (dichotomized to <5 vs >5 years), volume of PICCs placed (<999 vs 1000), and certification status (yes/no). Bivariate comparisons were made using 2 or Fisher exact tests based on the number of responses in a cell as appropriate; 2‐sided with a P value <0.05 was considered statistically significant. All statistical analyses were conducted using SAS version 9.3 (SAS Institute, Cary, NC).
Ethical and Regulatory Oversight
Because our study sought to describe existing practice without collecting any individual or facility level identifiable information, the project received a Not Regulated status by the University of Michigan Medical School Institutional Review Board (HUM00088351).
RESULTS
Of 172 vascular nurses who received invitations, 140 completed the survey for a response rate of 81%. Respondents reported working in not‐for‐profit hospitals (36%), academic medical centers (29%), and for‐profit hospitals (21%). Although multiple providers (eg, interventional radiology staff and providers, physicians) placed PICCs, 95% of those surveyed reported that they placed the majority of the PICCs at their institutions. Although most respondents placed PICCs in adult patients (86%), a few also placed PICCs in pediatric populations (17%). Vascular nursing programs were largely housed in their own department, but some reported to general nursing or subspecialties such as interventional radiology, cardiology, and critical care. Most respondents indicated their facilities had written policies regarding standard insertion and care practices (87% and 95%, respectively), but only 30% had policies regarding the necessity or appropriateness of PICCs.
Experience among respondents was variable: approximately a third had placed PICCs for <5 years (28.6%), whereas 58% reported placing PICCs for 5 years Correspondingly, 26% reported having placed 100 to 500 PICCs, whereas 34% had placed 1000 or more PICCs. Only 23% of those surveyed held a dedicated vascular access certification, such as board certified in vascular access or certified registered nurse infusion, whereas 16% indicated that they served as the vascular access lead nurse for their facility. Following placement, 94% of respondents reported that their facilities tracked the number of PICCs inserted, but only 40% indicated that dwell times of devices were also recorded. Only 30% of nurses reported that their hospitals had a written policy to evaluate PICC necessity or appropriateness following placement (Table 1).
No.* | % | |
---|---|---|
| ||
Participant characteristics | ||
For how many years have you been inserting PICCs? | ||
<5 years | 40 | 28.6% |
5 years | 81 | 57.9% |
Missing | ||
In which of the following populations do you insert PICCs? | ||
Adult patients | 121 | 86.4% |
Pediatric patients | 24 | 17.1% |
Neonatal patients | 1 | 0.7% |
In which of the following locations do you place PICCs? (Select all that apply.) | ||
Adult medical ward | 115 | 82.1% |
General adult surgical ward | 110 | 78.6% |
General pediatric medical ward | 34 | 24.3% |
General pediatric surgical ward | 24 | 17.1% |
Adult intensive care unit | 114 | 81.4% |
Pediatric intensive care unit | 19 | 13.6% |
Neonatal intensive care unit | 3 | 2.1% |
Other intensive care unit | 59 | 42.1% |
Outpatient clinic or emergency department | 17 | 12.1% |
Other | 10 | 7.1% |
Approximately how many PICCs may you have placed in your career? | ||
099 | 15 | 10.7% |
100499 | 36 | 25.7% |
500999 | 23 | 16.4% |
1,000 | 47 | 33.6% |
Are you the vascular access lead nurse for your facility or organization? | ||
Yes | 22 | 15.7% |
No | 98 | 70.0% |
Do you currently hold a dedicated vascular access certification (BC‐VA, CRNI, etc.)? | ||
Yes | 32 | 22.9% |
No | 89 | 63.6% |
Facility characteristics | ||
Which of the following best describes your primary work location? | ||
Academic medical center | 41 | 29.3% |
For‐profit community‐based hospital or medical center | 30 | 21.4% |
Not‐for‐profit community‐based hospital or medical center | 50 | 35.7% |
Who inserts the most PICCs in your facility? | ||
Vascular access nurses | 133 | 95.0% |
Interventional radiology or other providers | 7 | 5.0% |
In which department is vascular access nursing located? | ||
Vascular nursing | 76 | 54.3% |
General nursing | 38 | 27.1% |
Interventional radiology | 15 | 10.7% |
Other | 11 | 7.9% |
Using your best guess, how many PICCs do you think your facility inserts each month? | ||
<25 | 5 | 3.6% |
2549 | 13 | 9.3% |
50100 | 39 | 27.9% |
>100 | 78 | 55.7% |
Unknown | 2 | 1.4% |
How many vascular access nurses are employed by your facility? | ||
<4 | 14 | 10.0% |
46 | 33 | 23.6% |
79 | 15 | 10.7% |
1015 | 25 | 17.9% |
>15 | 53 | 37.9% |
Does your facility track the number of PICCs placed? | ||
Yes | 132 | 94.3% |
No | 5 | 3.6% |
Unknown | 3 | 2.1% |
Does your facility track the duration or dwell time of PICCs? | ||
Yes | 56 | 40.0% |
No | 60 | 42.9% |
Unknown | 24 | 17.1% |
Does your facility have a written policy regarding standard PICC insertion practices? | ||
Yes | 122 | 87.1% |
No | 8 | 5.7% |
Unknown | 7 | 5.0% |
Does your facility have a written policy regarding standard PICC care and maintenance? | ||
Yes | 133 | 95.0% |
No | 3 | 2.1% |
Unknown | 1 | 0.7% |
Does your facility have a written process to review the necessity or appropriateness of a PICC? | ||
Yes | 42 | 30.0% |
No | 63 | 45.0% |
Unknown | 20 | 14.3% |
The most commonly reported indications for PICC placement included intravenous antibiotics at discharge, difficult venous access, and placement for chemotherapy in patients with cancer. Forty‐six percent of nurses indicated they had placed a PICC in a patient receiving some form of dialysis in the past several months; however, 91% of these respondents reported receiving approval from nephrology prior to placement in these patients. Although almost all nurses (91%) used ultrasound to find a suitable vein for PICC placement, a smaller percentage used ultrasound to estimate the catheter‐to‐vein ratio to prevent thrombosis (79%), and only a few (14%) documented this figure in the medical record. Three‐quarters of those surveyed (76%) indicated they used ECG‐based systems to position PICC tips at the cavoatrial junction to prevent thrombosis. Of those who used this technology, 36% still obtained chest x‐rays to verify the position of the PICC tip. According to 84% of respondents, flushing of PICCs was performed mainly by bedside nurses, whereas scheduled weekly dressing changes were most often performed by vascular access nurses (Table 2).
Question | No. | % |
---|---|---|
| ||
Do you use ultrasound to find a suitable vein prior to PICC insertion? | ||
Yes | 128 | 91.4% |
No | 0 | 0.0% |
Do you use ultrasound to estimate the catheter‐to‐vein ratio prior to PICC insertion? | ||
Yes | 110 | 78.6% |
No | 18 | 12.9% |
When using ultrasound, do you document the catheter‐to‐vein ratio in the PICC insertion note? | ||
Yes | 20 | 14.3% |
No | 89 | 63.6% |
Do you use ECG guidance‐assisted systems to place PICCs? | ||
Yes | 106 | 75.7% |
No | 21 | 15.0% |
If using ECG guidance, do you still routinely obtain a chest x‐ray to verify PICC tip position after placing the PICC using ECG guidance? | ||
Yes | 38 | 27.1% |
No | 68 | 48.6% |
Who is primarily responsible for administering and adhering to a flushing protocol after PICC insertion at your facility? | ||
Bedside nurses | 118 | 83.6% |
Patients | 1 | 0.7% |
Vascular access nurses | 8 | 5.7% |
Which of the following agents are most often used to flush PICCs? | ||
Both heparin and normal saline flushes | 61 | 43.6% |
Normal saline only | 63 | 45.0% |
Heparin only | 3 | 2.1% |
Who is responsible for scheduled weekly dressing changes for PICCs? | ||
Vascular access nurses | 110 | 78.6% |
Bedside nurses | 14 | 10.0% |
Other (eg, IR staff, ICU staff) | 3 | 2.1% |
In the past few months, have you placed a PICC in a patient who was receiving a form of dialysis (eg, peritoneal or hemodialysis)? | ||
Yes | 65 | 46.4% |
No | 64 | 45.7% |
If you have placed PICCs in patients on dialysis, do you discuss PICC placement or receive approval from nephrology prior to inserting the PICC? | ||
Yes | 59 | 90.8% |
No | 6 | 9.2% |
With respect to complications, catheter occlusion, migration, and DVT were reported as the 3 most prevalent adverse events. Interestingly, respondents did not report central lineassociated bloodstream infection (CLABSI) as a common complication. Additionally, 51% of those surveyed indicated that physicians unnecessarily removed PICCs when CLABSI was suspected but not confirmed. When managing catheter occlusion, 50% of respondents began with normal saline flushes but used tissue‐plasminogen activator if saline failed to resolve occlusion. Management of catheter migration varied based on degree of device movement: when the PICC had migrated <5 cm, most respondents (77%) indicated they would first obtain a chest x‐ray to determine the position of the PICC tip, with few (4%) performing catheter exchange. However, if the PICC had migrated more than 5 cm, a significantly greater proportion of respondents (21%) indicated they would perform a catheter exchange. With regard to managing DVT, most vascular nurses reported they notified nurses and physicians to continue using the PICC but recommended tests to confirm the diagnosis.
To better understand the experiences of vascular nurses, we asked for their perceptions regarding appropriateness of PICC use and relationships with bedside nurses, physicians, and leadership. Over a third of respondents (36%) felt that <5% of all PICCs may be inappropriate in their facility, whereas 1 in 5 indicated that 10% to 24% of PICCs placed in their facilities may be inappropriate or could have been avoided. Almost all (98%) of the nurses stated they were not empowered to remove idle or clinically unnecessary PICCs without physician authorization. Although 51% of nurses described the support received from hospital leadership as excellent, very good, or good, 43% described leadership support as either fair or poor. Conversely, relationships with bedside nurses and physicians were rated as being very good or good by nearly two‐thirds of those surveyed (64% and 65%, respectively) (Table 3).
Question | No. | % |
---|---|---|
| ||
Which of the following PICC‐related complications have you most frequently encountered in your practice? | ||
Catheter occlusion | 81 | 57.9% |
Catheter migration | 27 | 19.3% |
PICC‐associated DVT | 6 | 4.3% |
Catheter fracture or embolization | 3 | 2.1% |
Exit site infection | 3 | 2.1% |
Coiling or kinking after insertion | 2 | 1.4% |
If you suspect a patient has catheter occlusion, which of the following best describes your approach to resolving this problem? | ||
Begin with normal saline but use a tPA product if this fails to restore patency | 70 | 50.0% |
Use a tPA product (eg, Cathflo, Activase, or Retavase) to restore patency | 44 | 31.4% |
Begin with heparin‐based flushes but use a tPA product if this fails to restore | 7 | 5.0% |
Use only normal saline flushes to restore patency | 3 | 2.1% |
If you find a PICC that has migrated out or has been accidentally dislodged <5 cm in a patient without symptoms, and the device is still clinically needed, which of the following best describes your practice? | ||
Obtain a chest x‐ray to verify tip position | 108 | 77.1% |
Perform a complete catheter exchange over a guidewire if possible | 5 | 3.6% |
Notify/discuss next steps with physician | 5 | 3.6% |
Other | 6 | 4.3% |
If you find a PICC that has migrated out or has been accidentally dislodged >5 cm in a patient without symptoms, and the device is still clinically needed, which of the following best describes your practice? | ||
Obtain a chest x‐ray to verify tip position | 72 | 51.4% |
Perform a catheter exchange over a guidewire if possible | 30 | 21.4% |
Notify/discuss next steps with physician | 10 | 7.1% |
Other | 12 | 8.6% |
Which of the following best describes your first approach when you suspect a patient has PICC‐associated phlebitis? | ||
Discuss best course of action with physician or nurse | 79 | 56.4% |
Supportive measures (eg, warm compresses, analgesics, monitoring) | 25 | 17.9% |
Remove the PICC | 15 | 10.7% |
Other | 5 | 3.6% |
Which of the following best describes your first approach when you suspect a patient has a PICC‐related DVT? | ||
Notify caregivers to continue using PICC and consider tests such as ultrasound | 82 | 58.6% |
Notify bedside nurse and physician not to continue use of the PICC and consider tests such as ultrasound | 42 | 30.0% |
PICCs are often removed when physicians suspect, but have not yet confirmed, CLABSI. Considering your experiences, what percentage of PICCs may have been removed in this manner at your facility? | ||
<5% | 11 | 7.9% |
59% | 16 | 11.4% |
1024% | 24 | 17.1% |
25% | 71 | 50.7% |
Based on your experience, what percentage of PICCs do you think are inappropriate or could have been avoided at your facility? | ||
<5% | 51 | 36.4% |
59% | 25 | 17.9% |
1024% | 28 | 20.0% |
2550% | 13 | 9.3% |
>50% | 5 | 3.6% |
Are vascular access nurses empowered to remove PICCs that are idle or clinically unnecessary without physician authorization? | ||
Yes | 3 | 2.1% |
No | 122 | 87.1% |
How would you rank the overall support your vascular access service receives from hospital leadership? | ||
Excellent | 5 | 3.6% |
Very good | 32 | 22.9% |
Good | 40 | 28.6% |
Fair | 35 | 25.0% |
Poor | 25 | 17.9% |
How would you describe your relationship with physicians at your facility when it comes to communicating recommendations or management of PICCs? | ||
Very good | 28 | 20.0% |
Good | 63 | 45.0% |
Fair | 35 | 25.0% |
Poor | 7 | 5.0% |
Very poor | 4 | 2.9% |
How would you describe your relationship with bedside nurses at your facility when it comes to communicating recommendations or management of PICCs? | ||
Very good | 32 | 22.9% |
Good | 58 | 41.4% |
Fair | 38 | 27.1% |
Poor | 7 | 5.0% |
Very poor | 2 | 1.4% |
Variation in Responses Based on Years in Practice or Certification
We initially hypothesized that responses regarding practice (ultrasound use, ECG guidance system use), management of complications, or perceptions regarding leadership might vary based on years of experience, number of PICCs placed, or certification status. However, no statistically significant associations with these factors and individual responses were identified.
DISCUSSION
In this survey of 140 vascular access nurses in hospitals across Michigan, new insights regarding the experience, practice, knowledge, and beliefs of this group of providers were obtained. We found that vascular access nurses varied with respect to years in practice, volume of PICCs placed, and certification status, reflecting heterogeneity in this provider group. Variation in insertion techniques, such as use of ultrasound to examine catheter‐to‐vein ratio (a key way to prevent thrombosis) or newer ECG technology to position the PICC, was also noted. Although indications for PICC insertion appeared consistent with published literature, the frequency with which these devices were placed in patients receiving dialysis (reportedly with nephrology approval) was surprising given national calls to avoid such use.[16] Opportunities to improve hospital practices, such as tracking PICC dwell times and PICC necessity, as well as the potential need to better educate physicians on when to remove PICCs for suspected CLABSI, were also identified. Collectively, these data are highly relevant to hospitalists and health systems as they help to identify areas for quality improvement and inform clinical practice regarding the use of PICCs in hospitalized patients. As hospitalists increasingly order PICCs and manage complications associated with these devices, they are well suited to use these data so as to improve patient safety and clinical outcomes.
Venous access is the most common medical procedure performed in hospitalized medical patients. Although a number of devices including peripheral intravenous catheters, central venous catheters, and PICCs are used for this purpose, the growing use of PICCs to secure venous access has been documented in several studies.[17] Such growth, in part, undoubtedly reflects increasing availability of vascular access nurses. Traditionally placed by interventional radiologists, the creation of dedicated vascular nursing teams has resulted in these subspecialists now serving in more of a backup or trouble‐shooting role rather than that of primary operator.[4, 14] This paradigm shift is well illustrated in a recent survey of infection preventionists, where over 60% of respondents reported that they had a vascular nursing team in their facility.[7] The growth of these nursing‐led vascular access teams has produced not only high rates of insertion success and low rates of complications, but also greater cost‐effectiveness when compared to interventional radiologybased insertion.[18]
Nonetheless, our survey also identified a number of important concerns regarding PICC practices and vascular nursing providers. First, we found variation in areas such as insertion practices and management of complications. Such variability highlights the importance of both growing and disseminating the evidence base for consistent practice in vascular nursing. Through their close clinical affiliation with vascular nurses and shared interests in obtaining safe and appropriate venous access for patients, hospitalists are ideally poised to lead this effort. Second, similarities between vascular nurse opinions regarding appropriateness of PICCs and those of hospitalists from a prior survey were noted.[19] Namely, a substantial proportion of both vascular nurses and hospitalists felt that some PICCs were inappropriate and could be avoided. Third, although relationships between vascular access nurses and leadership were reported as being variable, the survey responses suggested relatively good interprovider relationships with bedside nurses and physicians. Such relationships likely reflect the close clinical ties that emerge from being in the trenches of patient care and suggest that interventions to improve care in partnership with these providers are highly viable.
Our study has some limitations. First, despite a high response rate, our study used a survey design and reports findings from a convenience sample of vascular access nurses in a single state. Thus, nonrespondent and selection biases remain threats to our conclusions. Additionally, some respondents did not complete all responses, perhaps due to nonapplicability to practice or other unknown reasons. The pattern of missingness observed, however, suggested that such responses were missing at random. Second, we surveyed vascular nurses in hospitals that are actively engaged in improving PICC practices; our findings may therefore not be representative of vascular nursing professionals as a whole and may instead reflect those of a highly motivated group of individuals. Relatedly, the underlying reasons for adoption of specific practices or techniques cannot be discerned from our study. Third, although we did not find differences based on years in practice or certification status, our sample size was relatively small and likely underpowered for these comparisons. Finally, our study sample consists of vascular nurses who are clustered within hospitals in which they are employed. Therefore, overlap between reported practices and those required by the facility are possible.
Despite these limitations, our study has important strengths. First, this is among the most comprehensive of surveys examining vascular nursing experience, practice, knowledge, and beliefs. The growing presence of these providers across US hospitals, coupled with limited insight regarding their clinical practices, highlight the importance and utility of these data. Second, we noted important differences in experience, practices, and interprovider relationships between vascular providers in this field. Although we are unable to ascertain the drivers or significance of such variation, hospitals and health systems focused on improving patient safety should consider quantifying and exploring these factors. Third, findings from our survey within Michigan suggest the need for similar, larger studies across the country. Partnerships with nursing organizations or larger professional groups that represent vascular nursing specialists may be helpful in this regard.
In conclusion, we found important similarities and differences in vascular nursing experience, practice, knowledge, and beliefs in Michigan. These data are useful as they help provide context regarding the constitution of these teams, current practices, and opportunities for improving care. Hospitalists seeking to improve patient safety may use these data to better inform vascular access practice in hospitalized patients.
Acknowledgements
The authors thank Claire Rickard, PhD, RN, Britt Meyer, RN, Peter Carr, PhD, and David Dempsey, RN for their assistance in developing the survey instrument used in this study.
Disclosures: This project was funded through an Investigator Initiated Research Grant from the Blue Cross Blue Shield of Michigan (BCBSM) Foundation (grant number 2140.II). The funding source played no role in study design, data acquisition, analysis, or reporting of the data. Support for the Hospital Medicine Safety (HMS) Consortium is provided by BCBSM and the Blue Care Network as part of the BCBSM Value Partnerships program. Although BCBSM and HMS work collaboratively, the opinions, beliefs, and viewpoints expressed by the authors do not necessarily reflect the opinions, beliefs, and viewpoints of BCBSM or any of its employees. This work was also supported with resources from the Veterans Affairs Ann Arbor Healthcare System. The views expressed in this article are those of the authors and do not necessarily reflect the position or policy of the Department of Veterans Affairs or the United States government.
- Peripherally inserted central venous catheters in the acute care setting: a safe alternative to high‐risk short‐term central venous catheters. Am J Infect Control. 2010;38(2):149–153. , , , et al.
- Risk of venous thromboembolism in hospitalized patients with peripherally inserted central catheters. J Hosp Med. 2009;4(7):417–422. , , , , .
- Central venous catheter placement by advanced practice nurses demonstrates low procedural complication and infection rates‐‐a report from 13 years of service. Crit Care Med. 2014;42(3):536–543. , , , , , .
- Developing an alternative workflow model for peripherally inserted central catheter placement. J Infus Nurs. 2012;34(1):34–42. .
- Facility wide benefits of radiology vascular access teams. Radiol Manage. 2010;32(1):28–32; quiz 33–34. , .
- Moving the needle forward: the imperative for collaboration in vascular access. J Infus Nurs. 2015;38(2):100–102. , .
- Use of designated PICC teams by U.S. hospitals: a survey‐based study [published online November 10, 2015]. J Patient Saf. doi: 10.1097/PTS.0000000000000246 , , , .
- The association between PICC use and venous thromboembolism in upper and lower extremities. American J Med. 2015;128(9):986–993.e1. , , , , .
- Hospital performance for pharmacologic venous thromboembolism prophylaxis and rate of venous thromboembolism: a cohort study. JAMA Intern Med. 2014;174(10):1577–1584. , , , et al.
- Risk of venous thromboembolism associated with peripherally inserted central catheters: a systematic review and meta‐analysis. Lancet. 2013;382(9889):311–325. , , , et al.
- Infusion Nurses Society. Infusion nursing standards of practice. J Infus Nurs. 2006;29(1 suppl):S1–S92.
- Guidelines for the prevention of intravascular catheter‐related infections. Am J Infect Control. 2011;39(4 suppl 1):S1–S34. , , , et al.
- International evidence‐based recommendations on ultrasound‐guided vascular access. Intensive Care Med. 2012;38(7):1105–1117. , , , et al.
- A single institution experience of seven hundred consecutively placed peripherally inserted central venous catheters. J Vasc Access. 2014;15(6):498–502. , , .
- Central venous access devices site care practices: an international survey of 34 countries [published online September 3, 2015]. J Vasc Access. doi: 10.5301/jva.5000450 , , .
- American Society of Nephrology. World's Leading Kidney Society Joins Effort to Reduce Unnecessary Medical Tests and Procedures. Available at: https://www.asn‐online.org/policy/choosingwisely/PressReleaseChoosingWisely.pdf. Accessed September 4, 2015.
- A survey of the current use of peripherally inserted central venous catheter (PICC) in Swedish oncology departments. Acta Oncol. 2013;52(6):1241–1242. , , , .
- Nurse‐led PICC insertion: is it cost effective? Br J Nurs. 2013;22(19):S9–S15. , .
- Hospitalist experiences, practice, opinions, and knowledge regarding peripherally inserted central catheters: a Michigan survey. J Hosp Med. 2013;8(6):309–314. , , , et al.
Peripherally inserted central catheters (PICCs) are among the most prevalent of venous access devices in hospitalized patients.[1, 2] Although growing use of these devices reflects clinical advantages, such as a reduced risk of complications during insertion and durable venous access, use of PICCs is also likely related to the growth of vascular access nursing.[3, 4] A relatively new specialty, vascular access nurses obtain, maintain, and manage venous access in hospitalized patients.[4, 5] Depending on their scope of practice, these professionals are responsible not only for insertion of devices, such as peripheral intravenous catheters and PICCs, but also nontunneled central venous catheters and arterial catheters in some settings.[6]
Although a growing number of US hospitals have introduced vascular nursing teams,[7] little is known about the experience, practice, knowledge, and beliefs of vascular access nurses. This knowledge gap is relevant for hospitalists and hospital medicine as (1) vascular access nurses increasingly represent a key partner in the care of hospitalized patients; (2) the knowledge and practice of these individuals directly affects patient safety and clinical outcomes; and (3) understanding experience, practice, and beliefs of these specialists can help inform decision making and quality‐improvement efforts related to PICCs. As hospitalists increasingly order the placement of and care for patients with PICCs, they are also well suited to improve PICC practice.
Therefore, we conducted a survey of vascular access nurses employed by hospitals that participate in the Michigan Hospital Medicine Safety (HMS) Consortium, a Blue Cross Blue Shield of Michiganfunded collaborative quality initiative.[6] We aimed to understand experience, practice, knowledge, and beliefs related to PICC care and use.
METHODS
Study Setting and Participants
To quantify vascular nursing experience, practice, knowledge, and beliefs, we conducted a Web‐based survey of vascular nurses across 47 Michigan hospitals that participate in HMS. A statewide quality‐improvement initiative, HMS aims to prevent adverse events in hospitalized medical patients through the creation of a data registry and sharing of best practices. The setting and design of this multicenter initiative have been previously described.[8, 9] Although participation is voluntary, each hospital receives payment for participating in the consortium and for data collection. Because HMS has an ongoing initiative aimed at identifying and preventing PICC‐related complications, this study was particularly relevant for participating hospitals and nurses.
Each HMS site has a designated quality‐improvement lead, physician champion, and data abstractor. To coordinate distribution and dissemination of the survey, we contacted the quality‐improvement leads at each site and enquired whether their hospital employed vascular access nurses who placed PICCs. Because we were only interested in responses from vascular access nurses, HMS hospitals that did not have these providers or stated PICCs were placed by other specialists (eg, interventional radiology) were excluded. At eligible sites, we obtained the total number of vascular nurses employed so as to determine the number of eligible respondents. In this manner, a purposeful sample of vascular nurses at participating HMS hospitals was constituted.
Participation in the survey was solicited through hospital quality leads that either distributed an electronic survey link to vascular nurses at their facilities or sent us individual email addresses to contact them directly. A cover letter explaining the rationale and the purpose of the survey along with the survey link was then sent to respondents through either of these routes. The survey was administered at all HMS sites contemporaneously and kept open for a period of 5 weeks. During the 5‐week period, 2 e‐mail reminders were sent to encourage participation. As a token of appreciation, a $10 Amazon gift card was offered to those who took the survey.
Development and Validation of the Survey
We developed the survey instrument (which we call PICC1 as we hope to administer longitudinally to track changes over time) by first conducting a literature search to identify relevant evidence‐based guidelines and studies regarding vascular access nursing practices and experiences.[10, 11, 12, 13] In addition, we consulted and involved national and international leaders in vascular access nursing to ensure validity and representativeness of the questions posed. We were specifically interested in nursing background, hospital practices, types of PICCs used, use of various technologies, relationships with healthcare providers, and management of complications. To understand participant characteristics and quantify potential variation in responses, we collected basic participant data including demographics, years in practice, number of PICCs placed, leadership roles, and vascular access certification status. Based on clinical reasoning and existing studies,[14, 15] we hypothesized that responses regarding certain practices (ultrasound use, electrocardiography [ECG] guidance system use), management of complications, or perceptions regarding leadership might vary based on years of experience, number of PICCs placed, or certification status. We therefore examined these associations as prespecified subgroup analyses.
The initial survey instrument was pilot tested with vascular nurses outside of the sampling frame. Based on feedback from the pilot testers, the instrument was refined and edited to improve clarity of the questions. In addition, specific skip patterns and logic were programmed into the final survey to reduce respondent burden and allow participants to seamlessly bypass questions that were contingent on a prior response (eg, use of ECG to place PICCs would lead to a series of questions about ECG‐assisted placement only for those respondents who used the technology). This final version of the survey was tested by members of the study team (V.C., L.K., S.L.K.) and then posted to SurveyMonkey for dissemination.
Statistical Analysis
Descriptive statistics (percentage, n/N) were used to tabulate results. In accordance with our a priori hypothesis that variation to responses might be associated with respondent characteristics, responses to questions regarding insertion practice (eg, use of ultrasound, measurement of catheter:vein ratio, trimming of catheters) and approach to complications (eg, catheter occlusion, deep vein thrombosis [DVT] notification, and PICC removal in the setting of fever) were compared by respondent years in practice (dichotomized to <5 vs >5 years), volume of PICCs placed (<999 vs 1000), and certification status (yes/no). Bivariate comparisons were made using 2 or Fisher exact tests based on the number of responses in a cell as appropriate; 2‐sided with a P value <0.05 was considered statistically significant. All statistical analyses were conducted using SAS version 9.3 (SAS Institute, Cary, NC).
Ethical and Regulatory Oversight
Because our study sought to describe existing practice without collecting any individual or facility level identifiable information, the project received a Not Regulated status by the University of Michigan Medical School Institutional Review Board (HUM00088351).
RESULTS
Of 172 vascular nurses who received invitations, 140 completed the survey for a response rate of 81%. Respondents reported working in not‐for‐profit hospitals (36%), academic medical centers (29%), and for‐profit hospitals (21%). Although multiple providers (eg, interventional radiology staff and providers, physicians) placed PICCs, 95% of those surveyed reported that they placed the majority of the PICCs at their institutions. Although most respondents placed PICCs in adult patients (86%), a few also placed PICCs in pediatric populations (17%). Vascular nursing programs were largely housed in their own department, but some reported to general nursing or subspecialties such as interventional radiology, cardiology, and critical care. Most respondents indicated their facilities had written policies regarding standard insertion and care practices (87% and 95%, respectively), but only 30% had policies regarding the necessity or appropriateness of PICCs.
Experience among respondents was variable: approximately a third had placed PICCs for <5 years (28.6%), whereas 58% reported placing PICCs for 5 years Correspondingly, 26% reported having placed 100 to 500 PICCs, whereas 34% had placed 1000 or more PICCs. Only 23% of those surveyed held a dedicated vascular access certification, such as board certified in vascular access or certified registered nurse infusion, whereas 16% indicated that they served as the vascular access lead nurse for their facility. Following placement, 94% of respondents reported that their facilities tracked the number of PICCs inserted, but only 40% indicated that dwell times of devices were also recorded. Only 30% of nurses reported that their hospitals had a written policy to evaluate PICC necessity or appropriateness following placement (Table 1).
No.* | % | |
---|---|---|
| ||
Participant characteristics | ||
For how many years have you been inserting PICCs? | ||
<5 years | 40 | 28.6% |
5 years | 81 | 57.9% |
Missing | ||
In which of the following populations do you insert PICCs? | ||
Adult patients | 121 | 86.4% |
Pediatric patients | 24 | 17.1% |
Neonatal patients | 1 | 0.7% |
In which of the following locations do you place PICCs? (Select all that apply.) | ||
Adult medical ward | 115 | 82.1% |
General adult surgical ward | 110 | 78.6% |
General pediatric medical ward | 34 | 24.3% |
General pediatric surgical ward | 24 | 17.1% |
Adult intensive care unit | 114 | 81.4% |
Pediatric intensive care unit | 19 | 13.6% |
Neonatal intensive care unit | 3 | 2.1% |
Other intensive care unit | 59 | 42.1% |
Outpatient clinic or emergency department | 17 | 12.1% |
Other | 10 | 7.1% |
Approximately how many PICCs may you have placed in your career? | ||
099 | 15 | 10.7% |
100499 | 36 | 25.7% |
500999 | 23 | 16.4% |
1,000 | 47 | 33.6% |
Are you the vascular access lead nurse for your facility or organization? | ||
Yes | 22 | 15.7% |
No | 98 | 70.0% |
Do you currently hold a dedicated vascular access certification (BC‐VA, CRNI, etc.)? | ||
Yes | 32 | 22.9% |
No | 89 | 63.6% |
Facility characteristics | ||
Which of the following best describes your primary work location? | ||
Academic medical center | 41 | 29.3% |
For‐profit community‐based hospital or medical center | 30 | 21.4% |
Not‐for‐profit community‐based hospital or medical center | 50 | 35.7% |
Who inserts the most PICCs in your facility? | ||
Vascular access nurses | 133 | 95.0% |
Interventional radiology or other providers | 7 | 5.0% |
In which department is vascular access nursing located? | ||
Vascular nursing | 76 | 54.3% |
General nursing | 38 | 27.1% |
Interventional radiology | 15 | 10.7% |
Other | 11 | 7.9% |
Using your best guess, how many PICCs do you think your facility inserts each month? | ||
<25 | 5 | 3.6% |
2549 | 13 | 9.3% |
50100 | 39 | 27.9% |
>100 | 78 | 55.7% |
Unknown | 2 | 1.4% |
How many vascular access nurses are employed by your facility? | ||
<4 | 14 | 10.0% |
46 | 33 | 23.6% |
79 | 15 | 10.7% |
1015 | 25 | 17.9% |
>15 | 53 | 37.9% |
Does your facility track the number of PICCs placed? | ||
Yes | 132 | 94.3% |
No | 5 | 3.6% |
Unknown | 3 | 2.1% |
Does your facility track the duration or dwell time of PICCs? | ||
Yes | 56 | 40.0% |
No | 60 | 42.9% |
Unknown | 24 | 17.1% |
Does your facility have a written policy regarding standard PICC insertion practices? | ||
Yes | 122 | 87.1% |
No | 8 | 5.7% |
Unknown | 7 | 5.0% |
Does your facility have a written policy regarding standard PICC care and maintenance? | ||
Yes | 133 | 95.0% |
No | 3 | 2.1% |
Unknown | 1 | 0.7% |
Does your facility have a written process to review the necessity or appropriateness of a PICC? | ||
Yes | 42 | 30.0% |
No | 63 | 45.0% |
Unknown | 20 | 14.3% |
The most commonly reported indications for PICC placement included intravenous antibiotics at discharge, difficult venous access, and placement for chemotherapy in patients with cancer. Forty‐six percent of nurses indicated they had placed a PICC in a patient receiving some form of dialysis in the past several months; however, 91% of these respondents reported receiving approval from nephrology prior to placement in these patients. Although almost all nurses (91%) used ultrasound to find a suitable vein for PICC placement, a smaller percentage used ultrasound to estimate the catheter‐to‐vein ratio to prevent thrombosis (79%), and only a few (14%) documented this figure in the medical record. Three‐quarters of those surveyed (76%) indicated they used ECG‐based systems to position PICC tips at the cavoatrial junction to prevent thrombosis. Of those who used this technology, 36% still obtained chest x‐rays to verify the position of the PICC tip. According to 84% of respondents, flushing of PICCs was performed mainly by bedside nurses, whereas scheduled weekly dressing changes were most often performed by vascular access nurses (Table 2).
Question | No. | % |
---|---|---|
| ||
Do you use ultrasound to find a suitable vein prior to PICC insertion? | ||
Yes | 128 | 91.4% |
No | 0 | 0.0% |
Do you use ultrasound to estimate the catheter‐to‐vein ratio prior to PICC insertion? | ||
Yes | 110 | 78.6% |
No | 18 | 12.9% |
When using ultrasound, do you document the catheter‐to‐vein ratio in the PICC insertion note? | ||
Yes | 20 | 14.3% |
No | 89 | 63.6% |
Do you use ECG guidance‐assisted systems to place PICCs? | ||
Yes | 106 | 75.7% |
No | 21 | 15.0% |
If using ECG guidance, do you still routinely obtain a chest x‐ray to verify PICC tip position after placing the PICC using ECG guidance? | ||
Yes | 38 | 27.1% |
No | 68 | 48.6% |
Who is primarily responsible for administering and adhering to a flushing protocol after PICC insertion at your facility? | ||
Bedside nurses | 118 | 83.6% |
Patients | 1 | 0.7% |
Vascular access nurses | 8 | 5.7% |
Which of the following agents are most often used to flush PICCs? | ||
Both heparin and normal saline flushes | 61 | 43.6% |
Normal saline only | 63 | 45.0% |
Heparin only | 3 | 2.1% |
Who is responsible for scheduled weekly dressing changes for PICCs? | ||
Vascular access nurses | 110 | 78.6% |
Bedside nurses | 14 | 10.0% |
Other (eg, IR staff, ICU staff) | 3 | 2.1% |
In the past few months, have you placed a PICC in a patient who was receiving a form of dialysis (eg, peritoneal or hemodialysis)? | ||
Yes | 65 | 46.4% |
No | 64 | 45.7% |
If you have placed PICCs in patients on dialysis, do you discuss PICC placement or receive approval from nephrology prior to inserting the PICC? | ||
Yes | 59 | 90.8% |
No | 6 | 9.2% |
With respect to complications, catheter occlusion, migration, and DVT were reported as the 3 most prevalent adverse events. Interestingly, respondents did not report central lineassociated bloodstream infection (CLABSI) as a common complication. Additionally, 51% of those surveyed indicated that physicians unnecessarily removed PICCs when CLABSI was suspected but not confirmed. When managing catheter occlusion, 50% of respondents began with normal saline flushes but used tissue‐plasminogen activator if saline failed to resolve occlusion. Management of catheter migration varied based on degree of device movement: when the PICC had migrated <5 cm, most respondents (77%) indicated they would first obtain a chest x‐ray to determine the position of the PICC tip, with few (4%) performing catheter exchange. However, if the PICC had migrated more than 5 cm, a significantly greater proportion of respondents (21%) indicated they would perform a catheter exchange. With regard to managing DVT, most vascular nurses reported they notified nurses and physicians to continue using the PICC but recommended tests to confirm the diagnosis.
To better understand the experiences of vascular nurses, we asked for their perceptions regarding appropriateness of PICC use and relationships with bedside nurses, physicians, and leadership. Over a third of respondents (36%) felt that <5% of all PICCs may be inappropriate in their facility, whereas 1 in 5 indicated that 10% to 24% of PICCs placed in their facilities may be inappropriate or could have been avoided. Almost all (98%) of the nurses stated they were not empowered to remove idle or clinically unnecessary PICCs without physician authorization. Although 51% of nurses described the support received from hospital leadership as excellent, very good, or good, 43% described leadership support as either fair or poor. Conversely, relationships with bedside nurses and physicians were rated as being very good or good by nearly two‐thirds of those surveyed (64% and 65%, respectively) (Table 3).
Question | No. | % |
---|---|---|
| ||
Which of the following PICC‐related complications have you most frequently encountered in your practice? | ||
Catheter occlusion | 81 | 57.9% |
Catheter migration | 27 | 19.3% |
PICC‐associated DVT | 6 | 4.3% |
Catheter fracture or embolization | 3 | 2.1% |
Exit site infection | 3 | 2.1% |
Coiling or kinking after insertion | 2 | 1.4% |
If you suspect a patient has catheter occlusion, which of the following best describes your approach to resolving this problem? | ||
Begin with normal saline but use a tPA product if this fails to restore patency | 70 | 50.0% |
Use a tPA product (eg, Cathflo, Activase, or Retavase) to restore patency | 44 | 31.4% |
Begin with heparin‐based flushes but use a tPA product if this fails to restore | 7 | 5.0% |
Use only normal saline flushes to restore patency | 3 | 2.1% |
If you find a PICC that has migrated out or has been accidentally dislodged <5 cm in a patient without symptoms, and the device is still clinically needed, which of the following best describes your practice? | ||
Obtain a chest x‐ray to verify tip position | 108 | 77.1% |
Perform a complete catheter exchange over a guidewire if possible | 5 | 3.6% |
Notify/discuss next steps with physician | 5 | 3.6% |
Other | 6 | 4.3% |
If you find a PICC that has migrated out or has been accidentally dislodged >5 cm in a patient without symptoms, and the device is still clinically needed, which of the following best describes your practice? | ||
Obtain a chest x‐ray to verify tip position | 72 | 51.4% |
Perform a catheter exchange over a guidewire if possible | 30 | 21.4% |
Notify/discuss next steps with physician | 10 | 7.1% |
Other | 12 | 8.6% |
Which of the following best describes your first approach when you suspect a patient has PICC‐associated phlebitis? | ||
Discuss best course of action with physician or nurse | 79 | 56.4% |
Supportive measures (eg, warm compresses, analgesics, monitoring) | 25 | 17.9% |
Remove the PICC | 15 | 10.7% |
Other | 5 | 3.6% |
Which of the following best describes your first approach when you suspect a patient has a PICC‐related DVT? | ||
Notify caregivers to continue using PICC and consider tests such as ultrasound | 82 | 58.6% |
Notify bedside nurse and physician not to continue use of the PICC and consider tests such as ultrasound | 42 | 30.0% |
PICCs are often removed when physicians suspect, but have not yet confirmed, CLABSI. Considering your experiences, what percentage of PICCs may have been removed in this manner at your facility? | ||
<5% | 11 | 7.9% |
59% | 16 | 11.4% |
1024% | 24 | 17.1% |
25% | 71 | 50.7% |
Based on your experience, what percentage of PICCs do you think are inappropriate or could have been avoided at your facility? | ||
<5% | 51 | 36.4% |
59% | 25 | 17.9% |
1024% | 28 | 20.0% |
2550% | 13 | 9.3% |
>50% | 5 | 3.6% |
Are vascular access nurses empowered to remove PICCs that are idle or clinically unnecessary without physician authorization? | ||
Yes | 3 | 2.1% |
No | 122 | 87.1% |
How would you rank the overall support your vascular access service receives from hospital leadership? | ||
Excellent | 5 | 3.6% |
Very good | 32 | 22.9% |
Good | 40 | 28.6% |
Fair | 35 | 25.0% |
Poor | 25 | 17.9% |
How would you describe your relationship with physicians at your facility when it comes to communicating recommendations or management of PICCs? | ||
Very good | 28 | 20.0% |
Good | 63 | 45.0% |
Fair | 35 | 25.0% |
Poor | 7 | 5.0% |
Very poor | 4 | 2.9% |
How would you describe your relationship with bedside nurses at your facility when it comes to communicating recommendations or management of PICCs? | ||
Very good | 32 | 22.9% |
Good | 58 | 41.4% |
Fair | 38 | 27.1% |
Poor | 7 | 5.0% |
Very poor | 2 | 1.4% |
Variation in Responses Based on Years in Practice or Certification
We initially hypothesized that responses regarding practice (ultrasound use, ECG guidance system use), management of complications, or perceptions regarding leadership might vary based on years of experience, number of PICCs placed, or certification status. However, no statistically significant associations with these factors and individual responses were identified.
DISCUSSION
In this survey of 140 vascular access nurses in hospitals across Michigan, new insights regarding the experience, practice, knowledge, and beliefs of this group of providers were obtained. We found that vascular access nurses varied with respect to years in practice, volume of PICCs placed, and certification status, reflecting heterogeneity in this provider group. Variation in insertion techniques, such as use of ultrasound to examine catheter‐to‐vein ratio (a key way to prevent thrombosis) or newer ECG technology to position the PICC, was also noted. Although indications for PICC insertion appeared consistent with published literature, the frequency with which these devices were placed in patients receiving dialysis (reportedly with nephrology approval) was surprising given national calls to avoid such use.[16] Opportunities to improve hospital practices, such as tracking PICC dwell times and PICC necessity, as well as the potential need to better educate physicians on when to remove PICCs for suspected CLABSI, were also identified. Collectively, these data are highly relevant to hospitalists and health systems as they help to identify areas for quality improvement and inform clinical practice regarding the use of PICCs in hospitalized patients. As hospitalists increasingly order PICCs and manage complications associated with these devices, they are well suited to use these data so as to improve patient safety and clinical outcomes.
Venous access is the most common medical procedure performed in hospitalized medical patients. Although a number of devices including peripheral intravenous catheters, central venous catheters, and PICCs are used for this purpose, the growing use of PICCs to secure venous access has been documented in several studies.[17] Such growth, in part, undoubtedly reflects increasing availability of vascular access nurses. Traditionally placed by interventional radiologists, the creation of dedicated vascular nursing teams has resulted in these subspecialists now serving in more of a backup or trouble‐shooting role rather than that of primary operator.[4, 14] This paradigm shift is well illustrated in a recent survey of infection preventionists, where over 60% of respondents reported that they had a vascular nursing team in their facility.[7] The growth of these nursing‐led vascular access teams has produced not only high rates of insertion success and low rates of complications, but also greater cost‐effectiveness when compared to interventional radiologybased insertion.[18]
Nonetheless, our survey also identified a number of important concerns regarding PICC practices and vascular nursing providers. First, we found variation in areas such as insertion practices and management of complications. Such variability highlights the importance of both growing and disseminating the evidence base for consistent practice in vascular nursing. Through their close clinical affiliation with vascular nurses and shared interests in obtaining safe and appropriate venous access for patients, hospitalists are ideally poised to lead this effort. Second, similarities between vascular nurse opinions regarding appropriateness of PICCs and those of hospitalists from a prior survey were noted.[19] Namely, a substantial proportion of both vascular nurses and hospitalists felt that some PICCs were inappropriate and could be avoided. Third, although relationships between vascular access nurses and leadership were reported as being variable, the survey responses suggested relatively good interprovider relationships with bedside nurses and physicians. Such relationships likely reflect the close clinical ties that emerge from being in the trenches of patient care and suggest that interventions to improve care in partnership with these providers are highly viable.
Our study has some limitations. First, despite a high response rate, our study used a survey design and reports findings from a convenience sample of vascular access nurses in a single state. Thus, nonrespondent and selection biases remain threats to our conclusions. Additionally, some respondents did not complete all responses, perhaps due to nonapplicability to practice or other unknown reasons. The pattern of missingness observed, however, suggested that such responses were missing at random. Second, we surveyed vascular nurses in hospitals that are actively engaged in improving PICC practices; our findings may therefore not be representative of vascular nursing professionals as a whole and may instead reflect those of a highly motivated group of individuals. Relatedly, the underlying reasons for adoption of specific practices or techniques cannot be discerned from our study. Third, although we did not find differences based on years in practice or certification status, our sample size was relatively small and likely underpowered for these comparisons. Finally, our study sample consists of vascular nurses who are clustered within hospitals in which they are employed. Therefore, overlap between reported practices and those required by the facility are possible.
Despite these limitations, our study has important strengths. First, this is among the most comprehensive of surveys examining vascular nursing experience, practice, knowledge, and beliefs. The growing presence of these providers across US hospitals, coupled with limited insight regarding their clinical practices, highlight the importance and utility of these data. Second, we noted important differences in experience, practices, and interprovider relationships between vascular providers in this field. Although we are unable to ascertain the drivers or significance of such variation, hospitals and health systems focused on improving patient safety should consider quantifying and exploring these factors. Third, findings from our survey within Michigan suggest the need for similar, larger studies across the country. Partnerships with nursing organizations or larger professional groups that represent vascular nursing specialists may be helpful in this regard.
In conclusion, we found important similarities and differences in vascular nursing experience, practice, knowledge, and beliefs in Michigan. These data are useful as they help provide context regarding the constitution of these teams, current practices, and opportunities for improving care. Hospitalists seeking to improve patient safety may use these data to better inform vascular access practice in hospitalized patients.
Acknowledgements
The authors thank Claire Rickard, PhD, RN, Britt Meyer, RN, Peter Carr, PhD, and David Dempsey, RN for their assistance in developing the survey instrument used in this study.
Disclosures: This project was funded through an Investigator Initiated Research Grant from the Blue Cross Blue Shield of Michigan (BCBSM) Foundation (grant number 2140.II). The funding source played no role in study design, data acquisition, analysis, or reporting of the data. Support for the Hospital Medicine Safety (HMS) Consortium is provided by BCBSM and the Blue Care Network as part of the BCBSM Value Partnerships program. Although BCBSM and HMS work collaboratively, the opinions, beliefs, and viewpoints expressed by the authors do not necessarily reflect the opinions, beliefs, and viewpoints of BCBSM or any of its employees. This work was also supported with resources from the Veterans Affairs Ann Arbor Healthcare System. The views expressed in this article are those of the authors and do not necessarily reflect the position or policy of the Department of Veterans Affairs or the United States government.
Peripherally inserted central catheters (PICCs) are among the most prevalent of venous access devices in hospitalized patients.[1, 2] Although growing use of these devices reflects clinical advantages, such as a reduced risk of complications during insertion and durable venous access, use of PICCs is also likely related to the growth of vascular access nursing.[3, 4] A relatively new specialty, vascular access nurses obtain, maintain, and manage venous access in hospitalized patients.[4, 5] Depending on their scope of practice, these professionals are responsible not only for insertion of devices, such as peripheral intravenous catheters and PICCs, but also nontunneled central venous catheters and arterial catheters in some settings.[6]
Although a growing number of US hospitals have introduced vascular nursing teams,[7] little is known about the experience, practice, knowledge, and beliefs of vascular access nurses. This knowledge gap is relevant for hospitalists and hospital medicine as (1) vascular access nurses increasingly represent a key partner in the care of hospitalized patients; (2) the knowledge and practice of these individuals directly affects patient safety and clinical outcomes; and (3) understanding experience, practice, and beliefs of these specialists can help inform decision making and quality‐improvement efforts related to PICCs. As hospitalists increasingly order the placement of and care for patients with PICCs, they are also well suited to improve PICC practice.
Therefore, we conducted a survey of vascular access nurses employed by hospitals that participate in the Michigan Hospital Medicine Safety (HMS) Consortium, a Blue Cross Blue Shield of Michiganfunded collaborative quality initiative.[6] We aimed to understand experience, practice, knowledge, and beliefs related to PICC care and use.
METHODS
Study Setting and Participants
To quantify vascular nursing experience, practice, knowledge, and beliefs, we conducted a Web‐based survey of vascular nurses across 47 Michigan hospitals that participate in HMS. A statewide quality‐improvement initiative, HMS aims to prevent adverse events in hospitalized medical patients through the creation of a data registry and sharing of best practices. The setting and design of this multicenter initiative have been previously described.[8, 9] Although participation is voluntary, each hospital receives payment for participating in the consortium and for data collection. Because HMS has an ongoing initiative aimed at identifying and preventing PICC‐related complications, this study was particularly relevant for participating hospitals and nurses.
Each HMS site has a designated quality‐improvement lead, physician champion, and data abstractor. To coordinate distribution and dissemination of the survey, we contacted the quality‐improvement leads at each site and enquired whether their hospital employed vascular access nurses who placed PICCs. Because we were only interested in responses from vascular access nurses, HMS hospitals that did not have these providers or stated PICCs were placed by other specialists (eg, interventional radiology) were excluded. At eligible sites, we obtained the total number of vascular nurses employed so as to determine the number of eligible respondents. In this manner, a purposeful sample of vascular nurses at participating HMS hospitals was constituted.
Participation in the survey was solicited through hospital quality leads that either distributed an electronic survey link to vascular nurses at their facilities or sent us individual email addresses to contact them directly. A cover letter explaining the rationale and the purpose of the survey along with the survey link was then sent to respondents through either of these routes. The survey was administered at all HMS sites contemporaneously and kept open for a period of 5 weeks. During the 5‐week period, 2 e‐mail reminders were sent to encourage participation. As a token of appreciation, a $10 Amazon gift card was offered to those who took the survey.
Development and Validation of the Survey
We developed the survey instrument (which we call PICC1 as we hope to administer longitudinally to track changes over time) by first conducting a literature search to identify relevant evidence‐based guidelines and studies regarding vascular access nursing practices and experiences.[10, 11, 12, 13] In addition, we consulted and involved national and international leaders in vascular access nursing to ensure validity and representativeness of the questions posed. We were specifically interested in nursing background, hospital practices, types of PICCs used, use of various technologies, relationships with healthcare providers, and management of complications. To understand participant characteristics and quantify potential variation in responses, we collected basic participant data including demographics, years in practice, number of PICCs placed, leadership roles, and vascular access certification status. Based on clinical reasoning and existing studies,[14, 15] we hypothesized that responses regarding certain practices (ultrasound use, electrocardiography [ECG] guidance system use), management of complications, or perceptions regarding leadership might vary based on years of experience, number of PICCs placed, or certification status. We therefore examined these associations as prespecified subgroup analyses.
The initial survey instrument was pilot tested with vascular nurses outside of the sampling frame. Based on feedback from the pilot testers, the instrument was refined and edited to improve clarity of the questions. In addition, specific skip patterns and logic were programmed into the final survey to reduce respondent burden and allow participants to seamlessly bypass questions that were contingent on a prior response (eg, use of ECG to place PICCs would lead to a series of questions about ECG‐assisted placement only for those respondents who used the technology). This final version of the survey was tested by members of the study team (V.C., L.K., S.L.K.) and then posted to SurveyMonkey for dissemination.
Statistical Analysis
Descriptive statistics (percentage, n/N) were used to tabulate results. In accordance with our a priori hypothesis that variation to responses might be associated with respondent characteristics, responses to questions regarding insertion practice (eg, use of ultrasound, measurement of catheter:vein ratio, trimming of catheters) and approach to complications (eg, catheter occlusion, deep vein thrombosis [DVT] notification, and PICC removal in the setting of fever) were compared by respondent years in practice (dichotomized to <5 vs >5 years), volume of PICCs placed (<999 vs 1000), and certification status (yes/no). Bivariate comparisons were made using 2 or Fisher exact tests based on the number of responses in a cell as appropriate; 2‐sided with a P value <0.05 was considered statistically significant. All statistical analyses were conducted using SAS version 9.3 (SAS Institute, Cary, NC).
Ethical and Regulatory Oversight
Because our study sought to describe existing practice without collecting any individual or facility level identifiable information, the project received a Not Regulated status by the University of Michigan Medical School Institutional Review Board (HUM00088351).
RESULTS
Of 172 vascular nurses who received invitations, 140 completed the survey for a response rate of 81%. Respondents reported working in not‐for‐profit hospitals (36%), academic medical centers (29%), and for‐profit hospitals (21%). Although multiple providers (eg, interventional radiology staff and providers, physicians) placed PICCs, 95% of those surveyed reported that they placed the majority of the PICCs at their institutions. Although most respondents placed PICCs in adult patients (86%), a few also placed PICCs in pediatric populations (17%). Vascular nursing programs were largely housed in their own department, but some reported to general nursing or subspecialties such as interventional radiology, cardiology, and critical care. Most respondents indicated their facilities had written policies regarding standard insertion and care practices (87% and 95%, respectively), but only 30% had policies regarding the necessity or appropriateness of PICCs.
Experience among respondents was variable: approximately a third had placed PICCs for <5 years (28.6%), whereas 58% reported placing PICCs for 5 years Correspondingly, 26% reported having placed 100 to 500 PICCs, whereas 34% had placed 1000 or more PICCs. Only 23% of those surveyed held a dedicated vascular access certification, such as board certified in vascular access or certified registered nurse infusion, whereas 16% indicated that they served as the vascular access lead nurse for their facility. Following placement, 94% of respondents reported that their facilities tracked the number of PICCs inserted, but only 40% indicated that dwell times of devices were also recorded. Only 30% of nurses reported that their hospitals had a written policy to evaluate PICC necessity or appropriateness following placement (Table 1).
No.* | % | |
---|---|---|
| ||
Participant characteristics | ||
For how many years have you been inserting PICCs? | ||
<5 years | 40 | 28.6% |
5 years | 81 | 57.9% |
Missing | ||
In which of the following populations do you insert PICCs? | ||
Adult patients | 121 | 86.4% |
Pediatric patients | 24 | 17.1% |
Neonatal patients | 1 | 0.7% |
In which of the following locations do you place PICCs? (Select all that apply.) | ||
Adult medical ward | 115 | 82.1% |
General adult surgical ward | 110 | 78.6% |
General pediatric medical ward | 34 | 24.3% |
General pediatric surgical ward | 24 | 17.1% |
Adult intensive care unit | 114 | 81.4% |
Pediatric intensive care unit | 19 | 13.6% |
Neonatal intensive care unit | 3 | 2.1% |
Other intensive care unit | 59 | 42.1% |
Outpatient clinic or emergency department | 17 | 12.1% |
Other | 10 | 7.1% |
Approximately how many PICCs may you have placed in your career? | ||
099 | 15 | 10.7% |
100499 | 36 | 25.7% |
500999 | 23 | 16.4% |
1,000 | 47 | 33.6% |
Are you the vascular access lead nurse for your facility or organization? | ||
Yes | 22 | 15.7% |
No | 98 | 70.0% |
Do you currently hold a dedicated vascular access certification (BC‐VA, CRNI, etc.)? | ||
Yes | 32 | 22.9% |
No | 89 | 63.6% |
Facility characteristics | ||
Which of the following best describes your primary work location? | ||
Academic medical center | 41 | 29.3% |
For‐profit community‐based hospital or medical center | 30 | 21.4% |
Not‐for‐profit community‐based hospital or medical center | 50 | 35.7% |
Who inserts the most PICCs in your facility? | ||
Vascular access nurses | 133 | 95.0% |
Interventional radiology or other providers | 7 | 5.0% |
In which department is vascular access nursing located? | ||
Vascular nursing | 76 | 54.3% |
General nursing | 38 | 27.1% |
Interventional radiology | 15 | 10.7% |
Other | 11 | 7.9% |
Using your best guess, how many PICCs do you think your facility inserts each month? | ||
<25 | 5 | 3.6% |
2549 | 13 | 9.3% |
50100 | 39 | 27.9% |
>100 | 78 | 55.7% |
Unknown | 2 | 1.4% |
How many vascular access nurses are employed by your facility? | ||
<4 | 14 | 10.0% |
46 | 33 | 23.6% |
79 | 15 | 10.7% |
1015 | 25 | 17.9% |
>15 | 53 | 37.9% |
Does your facility track the number of PICCs placed? | ||
Yes | 132 | 94.3% |
No | 5 | 3.6% |
Unknown | 3 | 2.1% |
Does your facility track the duration or dwell time of PICCs? | ||
Yes | 56 | 40.0% |
No | 60 | 42.9% |
Unknown | 24 | 17.1% |
Does your facility have a written policy regarding standard PICC insertion practices? | ||
Yes | 122 | 87.1% |
No | 8 | 5.7% |
Unknown | 7 | 5.0% |
Does your facility have a written policy regarding standard PICC care and maintenance? | ||
Yes | 133 | 95.0% |
No | 3 | 2.1% |
Unknown | 1 | 0.7% |
Does your facility have a written process to review the necessity or appropriateness of a PICC? | ||
Yes | 42 | 30.0% |
No | 63 | 45.0% |
Unknown | 20 | 14.3% |
The most commonly reported indications for PICC placement included intravenous antibiotics at discharge, difficult venous access, and placement for chemotherapy in patients with cancer. Forty‐six percent of nurses indicated they had placed a PICC in a patient receiving some form of dialysis in the past several months; however, 91% of these respondents reported receiving approval from nephrology prior to placement in these patients. Although almost all nurses (91%) used ultrasound to find a suitable vein for PICC placement, a smaller percentage used ultrasound to estimate the catheter‐to‐vein ratio to prevent thrombosis (79%), and only a few (14%) documented this figure in the medical record. Three‐quarters of those surveyed (76%) indicated they used ECG‐based systems to position PICC tips at the cavoatrial junction to prevent thrombosis. Of those who used this technology, 36% still obtained chest x‐rays to verify the position of the PICC tip. According to 84% of respondents, flushing of PICCs was performed mainly by bedside nurses, whereas scheduled weekly dressing changes were most often performed by vascular access nurses (Table 2).
Question | No. | % |
---|---|---|
| ||
Do you use ultrasound to find a suitable vein prior to PICC insertion? | ||
Yes | 128 | 91.4% |
No | 0 | 0.0% |
Do you use ultrasound to estimate the catheter‐to‐vein ratio prior to PICC insertion? | ||
Yes | 110 | 78.6% |
No | 18 | 12.9% |
When using ultrasound, do you document the catheter‐to‐vein ratio in the PICC insertion note? | ||
Yes | 20 | 14.3% |
No | 89 | 63.6% |
Do you use ECG guidance‐assisted systems to place PICCs? | ||
Yes | 106 | 75.7% |
No | 21 | 15.0% |
If using ECG guidance, do you still routinely obtain a chest x‐ray to verify PICC tip position after placing the PICC using ECG guidance? | ||
Yes | 38 | 27.1% |
No | 68 | 48.6% |
Who is primarily responsible for administering and adhering to a flushing protocol after PICC insertion at your facility? | ||
Bedside nurses | 118 | 83.6% |
Patients | 1 | 0.7% |
Vascular access nurses | 8 | 5.7% |
Which of the following agents are most often used to flush PICCs? | ||
Both heparin and normal saline flushes | 61 | 43.6% |
Normal saline only | 63 | 45.0% |
Heparin only | 3 | 2.1% |
Who is responsible for scheduled weekly dressing changes for PICCs? | ||
Vascular access nurses | 110 | 78.6% |
Bedside nurses | 14 | 10.0% |
Other (eg, IR staff, ICU staff) | 3 | 2.1% |
In the past few months, have you placed a PICC in a patient who was receiving a form of dialysis (eg, peritoneal or hemodialysis)? | ||
Yes | 65 | 46.4% |
No | 64 | 45.7% |
If you have placed PICCs in patients on dialysis, do you discuss PICC placement or receive approval from nephrology prior to inserting the PICC? | ||
Yes | 59 | 90.8% |
No | 6 | 9.2% |
With respect to complications, catheter occlusion, migration, and DVT were reported as the 3 most prevalent adverse events. Interestingly, respondents did not report central lineassociated bloodstream infection (CLABSI) as a common complication. Additionally, 51% of those surveyed indicated that physicians unnecessarily removed PICCs when CLABSI was suspected but not confirmed. When managing catheter occlusion, 50% of respondents began with normal saline flushes but used tissue‐plasminogen activator if saline failed to resolve occlusion. Management of catheter migration varied based on degree of device movement: when the PICC had migrated <5 cm, most respondents (77%) indicated they would first obtain a chest x‐ray to determine the position of the PICC tip, with few (4%) performing catheter exchange. However, if the PICC had migrated more than 5 cm, a significantly greater proportion of respondents (21%) indicated they would perform a catheter exchange. With regard to managing DVT, most vascular nurses reported they notified nurses and physicians to continue using the PICC but recommended tests to confirm the diagnosis.
To better understand the experiences of vascular nurses, we asked for their perceptions regarding appropriateness of PICC use and relationships with bedside nurses, physicians, and leadership. Over a third of respondents (36%) felt that <5% of all PICCs may be inappropriate in their facility, whereas 1 in 5 indicated that 10% to 24% of PICCs placed in their facilities may be inappropriate or could have been avoided. Almost all (98%) of the nurses stated they were not empowered to remove idle or clinically unnecessary PICCs without physician authorization. Although 51% of nurses described the support received from hospital leadership as excellent, very good, or good, 43% described leadership support as either fair or poor. Conversely, relationships with bedside nurses and physicians were rated as being very good or good by nearly two‐thirds of those surveyed (64% and 65%, respectively) (Table 3).
Question | No. | % |
---|---|---|
| ||
Which of the following PICC‐related complications have you most frequently encountered in your practice? | ||
Catheter occlusion | 81 | 57.9% |
Catheter migration | 27 | 19.3% |
PICC‐associated DVT | 6 | 4.3% |
Catheter fracture or embolization | 3 | 2.1% |
Exit site infection | 3 | 2.1% |
Coiling or kinking after insertion | 2 | 1.4% |
If you suspect a patient has catheter occlusion, which of the following best describes your approach to resolving this problem? | ||
Begin with normal saline but use a tPA product if this fails to restore patency | 70 | 50.0% |
Use a tPA product (eg, Cathflo, Activase, or Retavase) to restore patency | 44 | 31.4% |
Begin with heparin‐based flushes but use a tPA product if this fails to restore | 7 | 5.0% |
Use only normal saline flushes to restore patency | 3 | 2.1% |
If you find a PICC that has migrated out or has been accidentally dislodged <5 cm in a patient without symptoms, and the device is still clinically needed, which of the following best describes your practice? | ||
Obtain a chest x‐ray to verify tip position | 108 | 77.1% |
Perform a complete catheter exchange over a guidewire if possible | 5 | 3.6% |
Notify/discuss next steps with physician | 5 | 3.6% |
Other | 6 | 4.3% |
If you find a PICC that has migrated out or has been accidentally dislodged >5 cm in a patient without symptoms, and the device is still clinically needed, which of the following best describes your practice? | ||
Obtain a chest x‐ray to verify tip position | 72 | 51.4% |
Perform a catheter exchange over a guidewire if possible | 30 | 21.4% |
Notify/discuss next steps with physician | 10 | 7.1% |
Other | 12 | 8.6% |
Which of the following best describes your first approach when you suspect a patient has PICC‐associated phlebitis? | ||
Discuss best course of action with physician or nurse | 79 | 56.4% |
Supportive measures (eg, warm compresses, analgesics, monitoring) | 25 | 17.9% |
Remove the PICC | 15 | 10.7% |
Other | 5 | 3.6% |
Which of the following best describes your first approach when you suspect a patient has a PICC‐related DVT? | ||
Notify caregivers to continue using PICC and consider tests such as ultrasound | 82 | 58.6% |
Notify bedside nurse and physician not to continue use of the PICC and consider tests such as ultrasound | 42 | 30.0% |
PICCs are often removed when physicians suspect, but have not yet confirmed, CLABSI. Considering your experiences, what percentage of PICCs may have been removed in this manner at your facility? | ||
<5% | 11 | 7.9% |
59% | 16 | 11.4% |
1024% | 24 | 17.1% |
25% | 71 | 50.7% |
Based on your experience, what percentage of PICCs do you think are inappropriate or could have been avoided at your facility? | ||
<5% | 51 | 36.4% |
59% | 25 | 17.9% |
1024% | 28 | 20.0% |
2550% | 13 | 9.3% |
>50% | 5 | 3.6% |
Are vascular access nurses empowered to remove PICCs that are idle or clinically unnecessary without physician authorization? | ||
Yes | 3 | 2.1% |
No | 122 | 87.1% |
How would you rank the overall support your vascular access service receives from hospital leadership? | ||
Excellent | 5 | 3.6% |
Very good | 32 | 22.9% |
Good | 40 | 28.6% |
Fair | 35 | 25.0% |
Poor | 25 | 17.9% |
How would you describe your relationship with physicians at your facility when it comes to communicating recommendations or management of PICCs? | ||
Very good | 28 | 20.0% |
Good | 63 | 45.0% |
Fair | 35 | 25.0% |
Poor | 7 | 5.0% |
Very poor | 4 | 2.9% |
How would you describe your relationship with bedside nurses at your facility when it comes to communicating recommendations or management of PICCs? | ||
Very good | 32 | 22.9% |
Good | 58 | 41.4% |
Fair | 38 | 27.1% |
Poor | 7 | 5.0% |
Very poor | 2 | 1.4% |
Variation in Responses Based on Years in Practice or Certification
We initially hypothesized that responses regarding practice (ultrasound use, ECG guidance system use), management of complications, or perceptions regarding leadership might vary based on years of experience, number of PICCs placed, or certification status. However, no statistically significant associations with these factors and individual responses were identified.
DISCUSSION
In this survey of 140 vascular access nurses in hospitals across Michigan, new insights regarding the experience, practice, knowledge, and beliefs of this group of providers were obtained. We found that vascular access nurses varied with respect to years in practice, volume of PICCs placed, and certification status, reflecting heterogeneity in this provider group. Variation in insertion techniques, such as use of ultrasound to examine catheter‐to‐vein ratio (a key way to prevent thrombosis) or newer ECG technology to position the PICC, was also noted. Although indications for PICC insertion appeared consistent with published literature, the frequency with which these devices were placed in patients receiving dialysis (reportedly with nephrology approval) was surprising given national calls to avoid such use.[16] Opportunities to improve hospital practices, such as tracking PICC dwell times and PICC necessity, as well as the potential need to better educate physicians on when to remove PICCs for suspected CLABSI, were also identified. Collectively, these data are highly relevant to hospitalists and health systems as they help to identify areas for quality improvement and inform clinical practice regarding the use of PICCs in hospitalized patients. As hospitalists increasingly order PICCs and manage complications associated with these devices, they are well suited to use these data so as to improve patient safety and clinical outcomes.
Venous access is the most common medical procedure performed in hospitalized medical patients. Although a number of devices including peripheral intravenous catheters, central venous catheters, and PICCs are used for this purpose, the growing use of PICCs to secure venous access has been documented in several studies.[17] Such growth, in part, undoubtedly reflects increasing availability of vascular access nurses. Traditionally placed by interventional radiologists, the creation of dedicated vascular nursing teams has resulted in these subspecialists now serving in more of a backup or trouble‐shooting role rather than that of primary operator.[4, 14] This paradigm shift is well illustrated in a recent survey of infection preventionists, where over 60% of respondents reported that they had a vascular nursing team in their facility.[7] The growth of these nursing‐led vascular access teams has produced not only high rates of insertion success and low rates of complications, but also greater cost‐effectiveness when compared to interventional radiologybased insertion.[18]
Nonetheless, our survey also identified a number of important concerns regarding PICC practices and vascular nursing providers. First, we found variation in areas such as insertion practices and management of complications. Such variability highlights the importance of both growing and disseminating the evidence base for consistent practice in vascular nursing. Through their close clinical affiliation with vascular nurses and shared interests in obtaining safe and appropriate venous access for patients, hospitalists are ideally poised to lead this effort. Second, similarities between vascular nurse opinions regarding appropriateness of PICCs and those of hospitalists from a prior survey were noted.[19] Namely, a substantial proportion of both vascular nurses and hospitalists felt that some PICCs were inappropriate and could be avoided. Third, although relationships between vascular access nurses and leadership were reported as being variable, the survey responses suggested relatively good interprovider relationships with bedside nurses and physicians. Such relationships likely reflect the close clinical ties that emerge from being in the trenches of patient care and suggest that interventions to improve care in partnership with these providers are highly viable.
Our study has some limitations. First, despite a high response rate, our study used a survey design and reports findings from a convenience sample of vascular access nurses in a single state. Thus, nonrespondent and selection biases remain threats to our conclusions. Additionally, some respondents did not complete all responses, perhaps due to nonapplicability to practice or other unknown reasons. The pattern of missingness observed, however, suggested that such responses were missing at random. Second, we surveyed vascular nurses in hospitals that are actively engaged in improving PICC practices; our findings may therefore not be representative of vascular nursing professionals as a whole and may instead reflect those of a highly motivated group of individuals. Relatedly, the underlying reasons for adoption of specific practices or techniques cannot be discerned from our study. Third, although we did not find differences based on years in practice or certification status, our sample size was relatively small and likely underpowered for these comparisons. Finally, our study sample consists of vascular nurses who are clustered within hospitals in which they are employed. Therefore, overlap between reported practices and those required by the facility are possible.
Despite these limitations, our study has important strengths. First, this is among the most comprehensive of surveys examining vascular nursing experience, practice, knowledge, and beliefs. The growing presence of these providers across US hospitals, coupled with limited insight regarding their clinical practices, highlight the importance and utility of these data. Second, we noted important differences in experience, practices, and interprovider relationships between vascular providers in this field. Although we are unable to ascertain the drivers or significance of such variation, hospitals and health systems focused on improving patient safety should consider quantifying and exploring these factors. Third, findings from our survey within Michigan suggest the need for similar, larger studies across the country. Partnerships with nursing organizations or larger professional groups that represent vascular nursing specialists may be helpful in this regard.
In conclusion, we found important similarities and differences in vascular nursing experience, practice, knowledge, and beliefs in Michigan. These data are useful as they help provide context regarding the constitution of these teams, current practices, and opportunities for improving care. Hospitalists seeking to improve patient safety may use these data to better inform vascular access practice in hospitalized patients.
Acknowledgements
The authors thank Claire Rickard, PhD, RN, Britt Meyer, RN, Peter Carr, PhD, and David Dempsey, RN for their assistance in developing the survey instrument used in this study.
Disclosures: This project was funded through an Investigator Initiated Research Grant from the Blue Cross Blue Shield of Michigan (BCBSM) Foundation (grant number 2140.II). The funding source played no role in study design, data acquisition, analysis, or reporting of the data. Support for the Hospital Medicine Safety (HMS) Consortium is provided by BCBSM and the Blue Care Network as part of the BCBSM Value Partnerships program. Although BCBSM and HMS work collaboratively, the opinions, beliefs, and viewpoints expressed by the authors do not necessarily reflect the opinions, beliefs, and viewpoints of BCBSM or any of its employees. This work was also supported with resources from the Veterans Affairs Ann Arbor Healthcare System. The views expressed in this article are those of the authors and do not necessarily reflect the position or policy of the Department of Veterans Affairs or the United States government.
- Peripherally inserted central venous catheters in the acute care setting: a safe alternative to high‐risk short‐term central venous catheters. Am J Infect Control. 2010;38(2):149–153. , , , et al.
- Risk of venous thromboembolism in hospitalized patients with peripherally inserted central catheters. J Hosp Med. 2009;4(7):417–422. , , , , .
- Central venous catheter placement by advanced practice nurses demonstrates low procedural complication and infection rates‐‐a report from 13 years of service. Crit Care Med. 2014;42(3):536–543. , , , , , .
- Developing an alternative workflow model for peripherally inserted central catheter placement. J Infus Nurs. 2012;34(1):34–42. .
- Facility wide benefits of radiology vascular access teams. Radiol Manage. 2010;32(1):28–32; quiz 33–34. , .
- Moving the needle forward: the imperative for collaboration in vascular access. J Infus Nurs. 2015;38(2):100–102. , .
- Use of designated PICC teams by U.S. hospitals: a survey‐based study [published online November 10, 2015]. J Patient Saf. doi: 10.1097/PTS.0000000000000246 , , , .
- The association between PICC use and venous thromboembolism in upper and lower extremities. American J Med. 2015;128(9):986–993.e1. , , , , .
- Hospital performance for pharmacologic venous thromboembolism prophylaxis and rate of venous thromboembolism: a cohort study. JAMA Intern Med. 2014;174(10):1577–1584. , , , et al.
- Risk of venous thromboembolism associated with peripherally inserted central catheters: a systematic review and meta‐analysis. Lancet. 2013;382(9889):311–325. , , , et al.
- Infusion Nurses Society. Infusion nursing standards of practice. J Infus Nurs. 2006;29(1 suppl):S1–S92.
- Guidelines for the prevention of intravascular catheter‐related infections. Am J Infect Control. 2011;39(4 suppl 1):S1–S34. , , , et al.
- International evidence‐based recommendations on ultrasound‐guided vascular access. Intensive Care Med. 2012;38(7):1105–1117. , , , et al.
- A single institution experience of seven hundred consecutively placed peripherally inserted central venous catheters. J Vasc Access. 2014;15(6):498–502. , , .
- Central venous access devices site care practices: an international survey of 34 countries [published online September 3, 2015]. J Vasc Access. doi: 10.5301/jva.5000450 , , .
- American Society of Nephrology. World's Leading Kidney Society Joins Effort to Reduce Unnecessary Medical Tests and Procedures. Available at: https://www.asn‐online.org/policy/choosingwisely/PressReleaseChoosingWisely.pdf. Accessed September 4, 2015.
- A survey of the current use of peripherally inserted central venous catheter (PICC) in Swedish oncology departments. Acta Oncol. 2013;52(6):1241–1242. , , , .
- Nurse‐led PICC insertion: is it cost effective? Br J Nurs. 2013;22(19):S9–S15. , .
- Hospitalist experiences, practice, opinions, and knowledge regarding peripherally inserted central catheters: a Michigan survey. J Hosp Med. 2013;8(6):309–314. , , , et al.
- Peripherally inserted central venous catheters in the acute care setting: a safe alternative to high‐risk short‐term central venous catheters. Am J Infect Control. 2010;38(2):149–153. , , , et al.
- Risk of venous thromboembolism in hospitalized patients with peripherally inserted central catheters. J Hosp Med. 2009;4(7):417–422. , , , , .
- Central venous catheter placement by advanced practice nurses demonstrates low procedural complication and infection rates‐‐a report from 13 years of service. Crit Care Med. 2014;42(3):536–543. , , , , , .
- Developing an alternative workflow model for peripherally inserted central catheter placement. J Infus Nurs. 2012;34(1):34–42. .
- Facility wide benefits of radiology vascular access teams. Radiol Manage. 2010;32(1):28–32; quiz 33–34. , .
- Moving the needle forward: the imperative for collaboration in vascular access. J Infus Nurs. 2015;38(2):100–102. , .
- Use of designated PICC teams by U.S. hospitals: a survey‐based study [published online November 10, 2015]. J Patient Saf. doi: 10.1097/PTS.0000000000000246 , , , .
- The association between PICC use and venous thromboembolism in upper and lower extremities. American J Med. 2015;128(9):986–993.e1. , , , , .
- Hospital performance for pharmacologic venous thromboembolism prophylaxis and rate of venous thromboembolism: a cohort study. JAMA Intern Med. 2014;174(10):1577–1584. , , , et al.
- Risk of venous thromboembolism associated with peripherally inserted central catheters: a systematic review and meta‐analysis. Lancet. 2013;382(9889):311–325. , , , et al.
- Infusion Nurses Society. Infusion nursing standards of practice. J Infus Nurs. 2006;29(1 suppl):S1–S92.
- Guidelines for the prevention of intravascular catheter‐related infections. Am J Infect Control. 2011;39(4 suppl 1):S1–S34. , , , et al.
- International evidence‐based recommendations on ultrasound‐guided vascular access. Intensive Care Med. 2012;38(7):1105–1117. , , , et al.
- A single institution experience of seven hundred consecutively placed peripherally inserted central venous catheters. J Vasc Access. 2014;15(6):498–502. , , .
- Central venous access devices site care practices: an international survey of 34 countries [published online September 3, 2015]. J Vasc Access. doi: 10.5301/jva.5000450 , , .
- American Society of Nephrology. World's Leading Kidney Society Joins Effort to Reduce Unnecessary Medical Tests and Procedures. Available at: https://www.asn‐online.org/policy/choosingwisely/PressReleaseChoosingWisely.pdf. Accessed September 4, 2015.
- A survey of the current use of peripherally inserted central venous catheter (PICC) in Swedish oncology departments. Acta Oncol. 2013;52(6):1241–1242. , , , .
- Nurse‐led PICC insertion: is it cost effective? Br J Nurs. 2013;22(19):S9–S15. , .
- Hospitalist experiences, practice, opinions, and knowledge regarding peripherally inserted central catheters: a Michigan survey. J Hosp Med. 2013;8(6):309–314. , , , et al.
© 2015 Society of Hospital Medicine
USPSTF urges extra step before treating hypertension
Screening for and treating high blood pressure (HBP) to prevent cardiovascular and renal disease is a tried-and-true preventive intervention that is supported by strong evidence. And not surprisingly, when the US Preventive Services Task Force (USPSTF) recently updated its 2007 recommendation for blood pressure screening for adults, it once again gave an A recommendation for those ages 18 years and older. What is noteworthy, however, is that this update concentrates on the accuracy of blood pressure measurement methods and optimal frequency of screening.1
The most significant modification of past recommendations is that HBP found with office measurement of blood pressure (OMBP) should be confirmed with either ambulatory blood pressure monitoring (ABPM) or home blood pressure monitoring (HBPM) before starting treatment. (For its recommendation, the USPSTF used the HBP definition from the Seventh Report of the Joint National Committee on Prevention, Detection, Evaluation, and Treatment of High Blood Pressure [TABLE 1].2,3)
Ensuring accurate blood-pressure measurements. More than 30% of adults in the United States have HBP, with prevalence increasing with age (TABLE 2).2 Only about half of this population has HBP under control.4 This modifiable condition contributes to more than 360,000 deaths annually.2 However, while treatment of true HBP results in substantial benefits, it is important not to over-diagnose HBP and over-treat it.
Studies have shown that 15% to 30% of individuals diagnosed with HBP in a clinical setting will have blood pressure in the normal range when measurements are taken outside of the doctor’s office.1 This discrepancy can be due to measurement error, regression to the mean, recent caffeine ingestion by the patient, or isolated clinical hypertension wherein the stress and anxiety caused by clinic visits elevates blood pressure transiently.
With this in mind, the USPSTF recommends that OMBP-detected HBP be confirmed with either ABPM or HBPM. Of these 2 follow-up methods, ABPM is supported by stronger evidence and is preferred. The USPSTF includes HBPM as an alternative because ABPM equipment may not always be available—or affordable—and using the equipment may present logistical challenges.
Starting off on the right foot
Screening for HBP in a clinical setting is more accurate if conducted according to recommended procedures: use an appropriately sized cuff; take the measurement at least 5 minutes after the patient’s arrival while he or she is seated with legs uncrossed and the cuffed arm is at the level of the heart; and record the mean of 2 separate measurements. There appears to be no real difference in the accuracy of automated vs manual sphygmomanometers.
Optimal frequency of screening varies. While the USPSTF found little evidence to support any particular overall screening frequency, it recommends annual screening for those who are 40 years of age or older and those ages 18 to 39 who are obese or overweight, are African American, or who have high-normal blood pressure (TABLE 3).1 Screening every 3 to 5 years is recommended for individuals not in these categories.
Initial steps in treating HBP. The Task Force also commented on which medications to use when initiating HBP treatment (after lifestyle and dietary interventions). Non-African Americans should receive a thiazide diuretic, calcium channel blocker, angiotensin-converting enzyme inhibitor, or angiotensin-receptor blocker. African Americans should begin treatment with a thiazide diuretic or calcium channel blocker. These recommendations appear to have been adopted from the Eighth Joint National Committee, since the accompanying evidence report for the USPSTF’s update did not address this issue.5
Don't forget patient support
Patient support is key. As of June 2015, the Community Preventive Services Task Force (CPSTF) recommends self-measured blood pressure monitoring combined with additional support as a means of improving blood pressure control in those with HBP.4
Supportive measures include things such as patient counseling on medications and health behavior changes (eg, diet and exercise); education on HBP and blood pressure self-management; and use of secure electronic or Web-based tools such as text or e-mail reminders to measure blood pressure, show up for appointments, or communicate blood pressure readings to healthcare providers. Patients who participate in home self-measurement of blood pressure with additional support lower their systolic blood pressure, on average, 1.4 mm Hg more than those who do not.4
Remaining questions
The new USPSTF recommendation leaves several issues unaddressed. For one thing, the Affordable Care Act mandates that commercial health insurance plans provide services with an A or B Task Force recommendation to patients with no copayments. So does the new HBP recommendation mean payers have to make ABPM and HBPM available to patients at no charge?
There are other questions, too. If HBP detected by OMBP is not confirmed when ABPM is performed, should ABPM be repeated, and if so, at what interval? What is the role of emerging technologies that use devices other than arm cuffs to measure blood pressure?
Despite these uncertainties, the new USPSTF and CPSTF recommendations refine the longstanding in-office–only approach to diagnosing and monitoring HBP and advocate newer technologies that could help improve diagnostic accuracy, avoid over-diagnosis and over-treatment, and improve patient adherence to treatment goals.
1. US Preventive Services Task Force. High blood pressure in adults: screening. Available at: http://www.uspreventiveservicestaskforce.org/Page/Document/UpdateSummaryFinal/high-blood-pressure-in-adults-screening. Accessed November 24, 2015.
2. Piper MA, Evans CV, Burda BU, et al. Screening for high blood pressure in adults: a systematic evidence review for the U.S. Preventive Services Task Force. Available at: http://www.ncbi.nlm.nih.gov/books/NBK269495/. Accessed November 24, 2015.
3. US Department of Health and Human Services. The seventh report of the Joint National Committee on Prevention, Detection, Evaluation, and Treatment of High Blood Pressure. Available at: http://www.nhlbi.nih.gov/files/docs/guidelines/jnc7full.pdf. Accessed November 24, 2015.
4. Community Preventive Services Task Force. Cardiovascular disease prevention and control: self-measured blood pressure monitoring interventions for improved blood pressure control — when combined with additional support. Available at: http://www.thecommunityguide.org/cvd/SMBP-additional.html. Accessed November 24, 2015.
5. James PA, Oparil S, Carter BL, et al. 2014 evidence-based guideline for the management of high blood pressure in adults: report from the panel members appointed to the Eighth Joint National Committee (JNC8). JAMA. 2014;311:507-520.
Screening for and treating high blood pressure (HBP) to prevent cardiovascular and renal disease is a tried-and-true preventive intervention that is supported by strong evidence. And not surprisingly, when the US Preventive Services Task Force (USPSTF) recently updated its 2007 recommendation for blood pressure screening for adults, it once again gave an A recommendation for those ages 18 years and older. What is noteworthy, however, is that this update concentrates on the accuracy of blood pressure measurement methods and optimal frequency of screening.1
The most significant modification of past recommendations is that HBP found with office measurement of blood pressure (OMBP) should be confirmed with either ambulatory blood pressure monitoring (ABPM) or home blood pressure monitoring (HBPM) before starting treatment. (For its recommendation, the USPSTF used the HBP definition from the Seventh Report of the Joint National Committee on Prevention, Detection, Evaluation, and Treatment of High Blood Pressure [TABLE 1].2,3)
Ensuring accurate blood-pressure measurements. More than 30% of adults in the United States have HBP, with prevalence increasing with age (TABLE 2).2 Only about half of this population has HBP under control.4 This modifiable condition contributes to more than 360,000 deaths annually.2 However, while treatment of true HBP results in substantial benefits, it is important not to over-diagnose HBP and over-treat it.
Studies have shown that 15% to 30% of individuals diagnosed with HBP in a clinical setting will have blood pressure in the normal range when measurements are taken outside of the doctor’s office.1 This discrepancy can be due to measurement error, regression to the mean, recent caffeine ingestion by the patient, or isolated clinical hypertension wherein the stress and anxiety caused by clinic visits elevates blood pressure transiently.
With this in mind, the USPSTF recommends that OMBP-detected HBP be confirmed with either ABPM or HBPM. Of these 2 follow-up methods, ABPM is supported by stronger evidence and is preferred. The USPSTF includes HBPM as an alternative because ABPM equipment may not always be available—or affordable—and using the equipment may present logistical challenges.
Starting off on the right foot
Screening for HBP in a clinical setting is more accurate if conducted according to recommended procedures: use an appropriately sized cuff; take the measurement at least 5 minutes after the patient’s arrival while he or she is seated with legs uncrossed and the cuffed arm is at the level of the heart; and record the mean of 2 separate measurements. There appears to be no real difference in the accuracy of automated vs manual sphygmomanometers.
Optimal frequency of screening varies. While the USPSTF found little evidence to support any particular overall screening frequency, it recommends annual screening for those who are 40 years of age or older and those ages 18 to 39 who are obese or overweight, are African American, or who have high-normal blood pressure (TABLE 3).1 Screening every 3 to 5 years is recommended for individuals not in these categories.
Initial steps in treating HBP. The Task Force also commented on which medications to use when initiating HBP treatment (after lifestyle and dietary interventions). Non-African Americans should receive a thiazide diuretic, calcium channel blocker, angiotensin-converting enzyme inhibitor, or angiotensin-receptor blocker. African Americans should begin treatment with a thiazide diuretic or calcium channel blocker. These recommendations appear to have been adopted from the Eighth Joint National Committee, since the accompanying evidence report for the USPSTF’s update did not address this issue.5
Don't forget patient support
Patient support is key. As of June 2015, the Community Preventive Services Task Force (CPSTF) recommends self-measured blood pressure monitoring combined with additional support as a means of improving blood pressure control in those with HBP.4
Supportive measures include things such as patient counseling on medications and health behavior changes (eg, diet and exercise); education on HBP and blood pressure self-management; and use of secure electronic or Web-based tools such as text or e-mail reminders to measure blood pressure, show up for appointments, or communicate blood pressure readings to healthcare providers. Patients who participate in home self-measurement of blood pressure with additional support lower their systolic blood pressure, on average, 1.4 mm Hg more than those who do not.4
Remaining questions
The new USPSTF recommendation leaves several issues unaddressed. For one thing, the Affordable Care Act mandates that commercial health insurance plans provide services with an A or B Task Force recommendation to patients with no copayments. So does the new HBP recommendation mean payers have to make ABPM and HBPM available to patients at no charge?
There are other questions, too. If HBP detected by OMBP is not confirmed when ABPM is performed, should ABPM be repeated, and if so, at what interval? What is the role of emerging technologies that use devices other than arm cuffs to measure blood pressure?
Despite these uncertainties, the new USPSTF and CPSTF recommendations refine the longstanding in-office–only approach to diagnosing and monitoring HBP and advocate newer technologies that could help improve diagnostic accuracy, avoid over-diagnosis and over-treatment, and improve patient adherence to treatment goals.
Screening for and treating high blood pressure (HBP) to prevent cardiovascular and renal disease is a tried-and-true preventive intervention that is supported by strong evidence. And not surprisingly, when the US Preventive Services Task Force (USPSTF) recently updated its 2007 recommendation for blood pressure screening for adults, it once again gave an A recommendation for those ages 18 years and older. What is noteworthy, however, is that this update concentrates on the accuracy of blood pressure measurement methods and optimal frequency of screening.1
The most significant modification of past recommendations is that HBP found with office measurement of blood pressure (OMBP) should be confirmed with either ambulatory blood pressure monitoring (ABPM) or home blood pressure monitoring (HBPM) before starting treatment. (For its recommendation, the USPSTF used the HBP definition from the Seventh Report of the Joint National Committee on Prevention, Detection, Evaluation, and Treatment of High Blood Pressure [TABLE 1].2,3)
Ensuring accurate blood-pressure measurements. More than 30% of adults in the United States have HBP, with prevalence increasing with age (TABLE 2).2 Only about half of this population has HBP under control.4 This modifiable condition contributes to more than 360,000 deaths annually.2 However, while treatment of true HBP results in substantial benefits, it is important not to over-diagnose HBP and over-treat it.
Studies have shown that 15% to 30% of individuals diagnosed with HBP in a clinical setting will have blood pressure in the normal range when measurements are taken outside of the doctor’s office.1 This discrepancy can be due to measurement error, regression to the mean, recent caffeine ingestion by the patient, or isolated clinical hypertension wherein the stress and anxiety caused by clinic visits elevates blood pressure transiently.
With this in mind, the USPSTF recommends that OMBP-detected HBP be confirmed with either ABPM or HBPM. Of these 2 follow-up methods, ABPM is supported by stronger evidence and is preferred. The USPSTF includes HBPM as an alternative because ABPM equipment may not always be available—or affordable—and using the equipment may present logistical challenges.
Starting off on the right foot
Screening for HBP in a clinical setting is more accurate if conducted according to recommended procedures: use an appropriately sized cuff; take the measurement at least 5 minutes after the patient’s arrival while he or she is seated with legs uncrossed and the cuffed arm is at the level of the heart; and record the mean of 2 separate measurements. There appears to be no real difference in the accuracy of automated vs manual sphygmomanometers.
Optimal frequency of screening varies. While the USPSTF found little evidence to support any particular overall screening frequency, it recommends annual screening for those who are 40 years of age or older and those ages 18 to 39 who are obese or overweight, are African American, or who have high-normal blood pressure (TABLE 3).1 Screening every 3 to 5 years is recommended for individuals not in these categories.
Initial steps in treating HBP. The Task Force also commented on which medications to use when initiating HBP treatment (after lifestyle and dietary interventions). Non-African Americans should receive a thiazide diuretic, calcium channel blocker, angiotensin-converting enzyme inhibitor, or angiotensin-receptor blocker. African Americans should begin treatment with a thiazide diuretic or calcium channel blocker. These recommendations appear to have been adopted from the Eighth Joint National Committee, since the accompanying evidence report for the USPSTF’s update did not address this issue.5
Don't forget patient support
Patient support is key. As of June 2015, the Community Preventive Services Task Force (CPSTF) recommends self-measured blood pressure monitoring combined with additional support as a means of improving blood pressure control in those with HBP.4
Supportive measures include things such as patient counseling on medications and health behavior changes (eg, diet and exercise); education on HBP and blood pressure self-management; and use of secure electronic or Web-based tools such as text or e-mail reminders to measure blood pressure, show up for appointments, or communicate blood pressure readings to healthcare providers. Patients who participate in home self-measurement of blood pressure with additional support lower their systolic blood pressure, on average, 1.4 mm Hg more than those who do not.4
Remaining questions
The new USPSTF recommendation leaves several issues unaddressed. For one thing, the Affordable Care Act mandates that commercial health insurance plans provide services with an A or B Task Force recommendation to patients with no copayments. So does the new HBP recommendation mean payers have to make ABPM and HBPM available to patients at no charge?
There are other questions, too. If HBP detected by OMBP is not confirmed when ABPM is performed, should ABPM be repeated, and if so, at what interval? What is the role of emerging technologies that use devices other than arm cuffs to measure blood pressure?
Despite these uncertainties, the new USPSTF and CPSTF recommendations refine the longstanding in-office–only approach to diagnosing and monitoring HBP and advocate newer technologies that could help improve diagnostic accuracy, avoid over-diagnosis and over-treatment, and improve patient adherence to treatment goals.
1. US Preventive Services Task Force. High blood pressure in adults: screening. Available at: http://www.uspreventiveservicestaskforce.org/Page/Document/UpdateSummaryFinal/high-blood-pressure-in-adults-screening. Accessed November 24, 2015.
2. Piper MA, Evans CV, Burda BU, et al. Screening for high blood pressure in adults: a systematic evidence review for the U.S. Preventive Services Task Force. Available at: http://www.ncbi.nlm.nih.gov/books/NBK269495/. Accessed November 24, 2015.
3. US Department of Health and Human Services. The seventh report of the Joint National Committee on Prevention, Detection, Evaluation, and Treatment of High Blood Pressure. Available at: http://www.nhlbi.nih.gov/files/docs/guidelines/jnc7full.pdf. Accessed November 24, 2015.
4. Community Preventive Services Task Force. Cardiovascular disease prevention and control: self-measured blood pressure monitoring interventions for improved blood pressure control — when combined with additional support. Available at: http://www.thecommunityguide.org/cvd/SMBP-additional.html. Accessed November 24, 2015.
5. James PA, Oparil S, Carter BL, et al. 2014 evidence-based guideline for the management of high blood pressure in adults: report from the panel members appointed to the Eighth Joint National Committee (JNC8). JAMA. 2014;311:507-520.
1. US Preventive Services Task Force. High blood pressure in adults: screening. Available at: http://www.uspreventiveservicestaskforce.org/Page/Document/UpdateSummaryFinal/high-blood-pressure-in-adults-screening. Accessed November 24, 2015.
2. Piper MA, Evans CV, Burda BU, et al. Screening for high blood pressure in adults: a systematic evidence review for the U.S. Preventive Services Task Force. Available at: http://www.ncbi.nlm.nih.gov/books/NBK269495/. Accessed November 24, 2015.
3. US Department of Health and Human Services. The seventh report of the Joint National Committee on Prevention, Detection, Evaluation, and Treatment of High Blood Pressure. Available at: http://www.nhlbi.nih.gov/files/docs/guidelines/jnc7full.pdf. Accessed November 24, 2015.
4. Community Preventive Services Task Force. Cardiovascular disease prevention and control: self-measured blood pressure monitoring interventions for improved blood pressure control — when combined with additional support. Available at: http://www.thecommunityguide.org/cvd/SMBP-additional.html. Accessed November 24, 2015.
5. James PA, Oparil S, Carter BL, et al. 2014 evidence-based guideline for the management of high blood pressure in adults: report from the panel members appointed to the Eighth Joint National Committee (JNC8). JAMA. 2014;311:507-520.
What Should Hospitalists Know about Surgical Tubes and Drains?
Case
A 45-year-old woman was admitted with choledocholithiasis. Two days prior, following endoscopic retrograde cholangiopancreatography (ERCP), she had gone to the OR for cholecystectomy. The procedure was completed laparoscopically, though the surgeon reported a difficult dissection. The surgeon left a Blake drain in the gallbladder fossa, which initially contained punch-colored fluid. Today, there is bilious fluid in the drain.
Overview
Surgical drains are used to monitor for postoperative leaks or abscesses, to collect normal physiologic fluid, or to minimize dead space. A hospitalist caring for surgical patients may be the first provider to note when something changes in the color or volume of surgical drains. Table 1 lists various types of drains with their indications for use.
Surgical Tubes and Drains
Chest tubes. Chest tubes are placed in the pleural space to evacuate air or fluid. They can be as thin as 20 French or as thick as 40 French (for adults). Chest tubes are typically placed between the fourth and fifth intercostal spaces in the anterior axillary or mid-axillary line; however, the location may vary according to the indication for placement. The tubes can be straight or angled.
The tubes are connected to a collecting system with a three-way chamber. The water chamber holds a column of water, which prevents air from being sucked into the pleural space with inhalation. The suction chamber can be attached to continuous wall suction to remove air or fluid, or it can be placed on “water seal” with no active suction mechanism. The third chamber is the collection chamber for fluid drainage.
Indications for a chest tube include pneumothorax, hemothorax, or a persistent or large pleural effusion. Pneumothorax and hemothorax usually require immediate chest tube placement. Chest tubes are also commonly placed at the end of thoracic surgeries to allow for appropriate re-expansion of the lung tissue.
A chest X-ray should be obtained after any chest tube insertion to ensure appropriate placement. Chest tubes are equipped with a radiopaque line along the longitudinal axis, which should be visible on X-ray. Respiratory variation in the fluid in the collecting tube, called “tidling,” should also be seen in a correctly placed chest tube, and should be monitored at the bedside to reassure continued appropriate location. The interventional radiologist or surgeon who placed the tube should determine the subsequent frequency of serial chest X-rays required to monitor the location of the chest tube.
If the patient has a pneumothorax, air bubbles will be visible in the water chamber; called an air leak, these are often more apparent when the patient coughs. The chest tube should initially be set to continuous suction at -20 mmHg to evacuate the air. Once the air leak has stopped, the chest tube should be placed on water seal to confirm resolution of the pneumothorax (water seal mimics normal physiology). If, after the transition from suction to water seal, resumption of the air leak is noted, it may indicate recurrence of the patient’s pneumothorax. A stat chest X-ray should be obtained, and the chest tube should be placed back on continuous suction. In general, a chest X-ray should be obtained any time the chest tube is changed from suction to water seal or vice versa.
If the patient experiences ongoing or worsening pain, fever, or inadequate drainage, a chest computed tomographic (CT) scan may be warranted to identify inappropriate positioning or other complications, such as occlusion or effusion of the tube. Blood or other debris might clog chest tubes; the surgical team may be able to evacuate the tube with suction tubing at the bedside. If unsuccessful, the tube may need to be removed and reinserted.
The team that placed the tube should help the hospitalist determine the timing of the chest tube removal. If the patient has a pleural effusion, the chest tube can usually be removed when the output is less than 100-200 mL per day and the lung is expanded. The tube should usually be taken off suction and placed on water seal to rule out pneumothorax prior to tube removal.
Penrose drains. Penrose drains are often used to drain fluid or to keep a space open for drainage. Surgeons may use sutures to anchor Penrose drains to skin. Common indications include:
- Ventral hernia repair;
- Debridement of infected pancreatitis; and
- Drainage of superficial abscess cavities.
Penrose drains are simple, flexible tubes that are open at both ends; in contrast to closed drains, they permit ingress as well as egress, facilitating colonization.
Closed suction drains. Closed suction drains with a plastic bulb attachment (i.e., Jackson-Pratt, Blake, Hemovac) are used to collect fluid from a postoperative cavity. Common indications include:
- Post-mastectomy to drain subcutaneous fluid;
- Abdominal surgery;
- Plastic surgery to prevent seroma formation and promote tissue apposition;
- Cholecystectomy if there is concern for damage to ducts of Luschka or other source of bile leak;
- Inadvertent postoperative leakage following a difficult rectal anastomosis; and
- Post-pancreatic surgery.
The quality and quantity of fluid drained should always be carefully noted and recorded. Changes in the fluid can imply development of bleed, leak, or other complications. The surgical team should be contacted immediately if changes are noted.
Typically, closed suction drains will be left in place until the drainage is less than 20 mL per day. These drains can be left in for weeks if necessary and will often be removed during the patient’s scheduled surgical follow-up. Rare complications include erosion into surrounding tissues and inadvertent suturing of the drain in place, such that reexploration is required to remove it. If a closed suction drain becomes occluded, contact the team that placed the drain for further recommendations on adjustment, replacement, or removal.
Nasogastric and duodenal tubes. Nasogastric tubes (NGTs) are often used in the nonoperative management of small bowel obstruction or ileus. They should be placed in the most dependent portion of the gastric lumen and confirmed by chest or abdominal X-ray. NGTs are sump pumps and have a double lumen, which includes an air port to assure flow. The air port should be patent for optimal functioning. The tube may be connected to continuous wall suction or intermittent suction, set to low (less than 60 mmHg) to avoid mucosal avulsion.
NGT output should decrease during the resolution of obstruction or ileus, and symptoms of nausea, vomiting, and abdominal distention should concomitantly improve. Persistently high output in a patient with other indicators of bowel function (flatus, for example) may suggest postpyloric placement (and placement should be checked by X-ray). The timing of NGT removal depends on resumption of bowel function.
Gastrostomy and jejunostomy tubes. Gastrostomy tubes are most commonly used for feeding but may also be used for decompression of functional or anatomic gastric outlet obstruction. They are indicated when patients need prolonged enteral access, such as those with prolonged mechanical ventilation or head and neck pathology that prohibits oral feeding. They are also rarely used for gastropexy to tack an atonic or patulous stomach to the abdominal wall or to prevent recurrence of paraesophageal hernias. These tubes can be placed percutaneously by interventional radiologists, endoscopically by surgeons and gastroenterologists, or laparoscopically or laparotomally by surgeons. This last option is often reserved for patients with difficult anatomy or those who are having laparotomy for another reason.
Because of the stomach’s generous lumen, gastrostomy tubes rarely clog. In the event that they do get clogged, carbonated liquids, meat tenderizer, or enzymes can help dissolve the obstruction. If a gastrostomy tube is left to drain, the patient may experience significant fluid and electrolyte losses, so these need to be carefully monitored.
Jejunostomy tubes are used exclusively for feeding and are usually placed 10-20 cm distal to the ligament of Treitz. These tubes are indicated in patients who require distal feedings due to gastric dysfunction or in those who have undergone a surgery in which a proximal anastomosis requires time to heal. These tubes are more apt to clog and can be more difficult to manage because the lumen of the small bowel is smaller than the stomach. Some prefer not to put pills down the tube to mitigate this risk. Routine flushes with water or saline (30 mL every four to six hours) are also helpful in mitigating the risk of clogging. In the event that they do get clogged, they may be treated like gastrostomy tubes, using carbonated liquids, meat tenderizer, or enzymes to help dissolve the obstruction.
Percutaneous tube sites should be examined frequently for signs of infection. Though gastrostomy and jejunostomy tubes are typically well secured intraabdominally, they can become dislodged. If a gastrostomy or jejunostomy tube has been in place for more than two weeks, it can easily be replaced at the bedside with a tube of comparable caliber by a member of the surgical team or by an experienced hospitalist. If the tube has been in place less than two weeks, it requires replacement with radiographic guidance, as the risk of creating a false lumen is high. Over time, tubes can become loose and fall out. If they need replacement, the preceding guidelines apply.
Back to the Case
A potential major complication of cholecystectomy is severance of the common bile duct, which necessitates significant further surgery. Less severe complications include injuries to the cholecystohepatic ducts (otherwise known as the ducts of Luschka), which can result in leakage of bile into the peritoneal cavity. A bile leak can lead to abscess and systemic infection if left undrained.
Surgeons who are concerned for such a complication intraoperatively may opt to leave a closed suction drain in the gallbladder fossa, such as a Blake drain, for monitoring and subsequent drainage. The drain will remain in place at least until the patient’s diet has been advanced fully, because digestion promotes the secretion of bile and may elucidate a leak. Bilious fluid in the Blake drain is suspicious for a leak.
The surgeon should be notified, and imaging should be obtained to find the nature of the injury to the biliary tree (CT scan with IV contrast, hepatobiliary iminodiacetic acid scan, or endoscopic retrograde cholangiopancreatography). If injury to major biliary structures (the cystic duct stump, the hepatic ducts, or the common bile duct) is diagnosed, a stent may be placed in order to restore ductile continuity.
Minor leaks, with damage to the cystic duct stump, hepatic ducts, and common bile duct ruled out, more often resolve on their own over time, and thus the patient’s closed suction drain will be left in place until biliary drainage ceases, without further initial intervention.
Bottom Line
Surgical tubes and drains have several placement indications. Alterations in quality and quantity of output can indicate changes in clinical status, and hospitalists should be able to handle initial troubleshooting. TH
Dr. Columbus is a general surgery resident at Brigham and Women’s Hospital in Boston. Dr. Havens is an instructor for the department of surgery at Brigham and Women’s Hospital. Dr. Peetz is an instructor for the department of surgery at University Hospital Case Medical Center in Cleveland.
Case
A 45-year-old woman was admitted with choledocholithiasis. Two days prior, following endoscopic retrograde cholangiopancreatography (ERCP), she had gone to the OR for cholecystectomy. The procedure was completed laparoscopically, though the surgeon reported a difficult dissection. The surgeon left a Blake drain in the gallbladder fossa, which initially contained punch-colored fluid. Today, there is bilious fluid in the drain.
Overview
Surgical drains are used to monitor for postoperative leaks or abscesses, to collect normal physiologic fluid, or to minimize dead space. A hospitalist caring for surgical patients may be the first provider to note when something changes in the color or volume of surgical drains. Table 1 lists various types of drains with their indications for use.
Surgical Tubes and Drains
Chest tubes. Chest tubes are placed in the pleural space to evacuate air or fluid. They can be as thin as 20 French or as thick as 40 French (for adults). Chest tubes are typically placed between the fourth and fifth intercostal spaces in the anterior axillary or mid-axillary line; however, the location may vary according to the indication for placement. The tubes can be straight or angled.
The tubes are connected to a collecting system with a three-way chamber. The water chamber holds a column of water, which prevents air from being sucked into the pleural space with inhalation. The suction chamber can be attached to continuous wall suction to remove air or fluid, or it can be placed on “water seal” with no active suction mechanism. The third chamber is the collection chamber for fluid drainage.
Indications for a chest tube include pneumothorax, hemothorax, or a persistent or large pleural effusion. Pneumothorax and hemothorax usually require immediate chest tube placement. Chest tubes are also commonly placed at the end of thoracic surgeries to allow for appropriate re-expansion of the lung tissue.
A chest X-ray should be obtained after any chest tube insertion to ensure appropriate placement. Chest tubes are equipped with a radiopaque line along the longitudinal axis, which should be visible on X-ray. Respiratory variation in the fluid in the collecting tube, called “tidling,” should also be seen in a correctly placed chest tube, and should be monitored at the bedside to reassure continued appropriate location. The interventional radiologist or surgeon who placed the tube should determine the subsequent frequency of serial chest X-rays required to monitor the location of the chest tube.
If the patient has a pneumothorax, air bubbles will be visible in the water chamber; called an air leak, these are often more apparent when the patient coughs. The chest tube should initially be set to continuous suction at -20 mmHg to evacuate the air. Once the air leak has stopped, the chest tube should be placed on water seal to confirm resolution of the pneumothorax (water seal mimics normal physiology). If, after the transition from suction to water seal, resumption of the air leak is noted, it may indicate recurrence of the patient’s pneumothorax. A stat chest X-ray should be obtained, and the chest tube should be placed back on continuous suction. In general, a chest X-ray should be obtained any time the chest tube is changed from suction to water seal or vice versa.
If the patient experiences ongoing or worsening pain, fever, or inadequate drainage, a chest computed tomographic (CT) scan may be warranted to identify inappropriate positioning or other complications, such as occlusion or effusion of the tube. Blood or other debris might clog chest tubes; the surgical team may be able to evacuate the tube with suction tubing at the bedside. If unsuccessful, the tube may need to be removed and reinserted.
The team that placed the tube should help the hospitalist determine the timing of the chest tube removal. If the patient has a pleural effusion, the chest tube can usually be removed when the output is less than 100-200 mL per day and the lung is expanded. The tube should usually be taken off suction and placed on water seal to rule out pneumothorax prior to tube removal.
Penrose drains. Penrose drains are often used to drain fluid or to keep a space open for drainage. Surgeons may use sutures to anchor Penrose drains to skin. Common indications include:
- Ventral hernia repair;
- Debridement of infected pancreatitis; and
- Drainage of superficial abscess cavities.
Penrose drains are simple, flexible tubes that are open at both ends; in contrast to closed drains, they permit ingress as well as egress, facilitating colonization.
Closed suction drains. Closed suction drains with a plastic bulb attachment (i.e., Jackson-Pratt, Blake, Hemovac) are used to collect fluid from a postoperative cavity. Common indications include:
- Post-mastectomy to drain subcutaneous fluid;
- Abdominal surgery;
- Plastic surgery to prevent seroma formation and promote tissue apposition;
- Cholecystectomy if there is concern for damage to ducts of Luschka or other source of bile leak;
- Inadvertent postoperative leakage following a difficult rectal anastomosis; and
- Post-pancreatic surgery.
The quality and quantity of fluid drained should always be carefully noted and recorded. Changes in the fluid can imply development of bleed, leak, or other complications. The surgical team should be contacted immediately if changes are noted.
Typically, closed suction drains will be left in place until the drainage is less than 20 mL per day. These drains can be left in for weeks if necessary and will often be removed during the patient’s scheduled surgical follow-up. Rare complications include erosion into surrounding tissues and inadvertent suturing of the drain in place, such that reexploration is required to remove it. If a closed suction drain becomes occluded, contact the team that placed the drain for further recommendations on adjustment, replacement, or removal.
Nasogastric and duodenal tubes. Nasogastric tubes (NGTs) are often used in the nonoperative management of small bowel obstruction or ileus. They should be placed in the most dependent portion of the gastric lumen and confirmed by chest or abdominal X-ray. NGTs are sump pumps and have a double lumen, which includes an air port to assure flow. The air port should be patent for optimal functioning. The tube may be connected to continuous wall suction or intermittent suction, set to low (less than 60 mmHg) to avoid mucosal avulsion.
NGT output should decrease during the resolution of obstruction or ileus, and symptoms of nausea, vomiting, and abdominal distention should concomitantly improve. Persistently high output in a patient with other indicators of bowel function (flatus, for example) may suggest postpyloric placement (and placement should be checked by X-ray). The timing of NGT removal depends on resumption of bowel function.
Gastrostomy and jejunostomy tubes. Gastrostomy tubes are most commonly used for feeding but may also be used for decompression of functional or anatomic gastric outlet obstruction. They are indicated when patients need prolonged enteral access, such as those with prolonged mechanical ventilation or head and neck pathology that prohibits oral feeding. They are also rarely used for gastropexy to tack an atonic or patulous stomach to the abdominal wall or to prevent recurrence of paraesophageal hernias. These tubes can be placed percutaneously by interventional radiologists, endoscopically by surgeons and gastroenterologists, or laparoscopically or laparotomally by surgeons. This last option is often reserved for patients with difficult anatomy or those who are having laparotomy for another reason.
Because of the stomach’s generous lumen, gastrostomy tubes rarely clog. In the event that they do get clogged, carbonated liquids, meat tenderizer, or enzymes can help dissolve the obstruction. If a gastrostomy tube is left to drain, the patient may experience significant fluid and electrolyte losses, so these need to be carefully monitored.
Jejunostomy tubes are used exclusively for feeding and are usually placed 10-20 cm distal to the ligament of Treitz. These tubes are indicated in patients who require distal feedings due to gastric dysfunction or in those who have undergone a surgery in which a proximal anastomosis requires time to heal. These tubes are more apt to clog and can be more difficult to manage because the lumen of the small bowel is smaller than the stomach. Some prefer not to put pills down the tube to mitigate this risk. Routine flushes with water or saline (30 mL every four to six hours) are also helpful in mitigating the risk of clogging. In the event that they do get clogged, they may be treated like gastrostomy tubes, using carbonated liquids, meat tenderizer, or enzymes to help dissolve the obstruction.
Percutaneous tube sites should be examined frequently for signs of infection. Though gastrostomy and jejunostomy tubes are typically well secured intraabdominally, they can become dislodged. If a gastrostomy or jejunostomy tube has been in place for more than two weeks, it can easily be replaced at the bedside with a tube of comparable caliber by a member of the surgical team or by an experienced hospitalist. If the tube has been in place less than two weeks, it requires replacement with radiographic guidance, as the risk of creating a false lumen is high. Over time, tubes can become loose and fall out. If they need replacement, the preceding guidelines apply.
Back to the Case
A potential major complication of cholecystectomy is severance of the common bile duct, which necessitates significant further surgery. Less severe complications include injuries to the cholecystohepatic ducts (otherwise known as the ducts of Luschka), which can result in leakage of bile into the peritoneal cavity. A bile leak can lead to abscess and systemic infection if left undrained.
Surgeons who are concerned for such a complication intraoperatively may opt to leave a closed suction drain in the gallbladder fossa, such as a Blake drain, for monitoring and subsequent drainage. The drain will remain in place at least until the patient’s diet has been advanced fully, because digestion promotes the secretion of bile and may elucidate a leak. Bilious fluid in the Blake drain is suspicious for a leak.
The surgeon should be notified, and imaging should be obtained to find the nature of the injury to the biliary tree (CT scan with IV contrast, hepatobiliary iminodiacetic acid scan, or endoscopic retrograde cholangiopancreatography). If injury to major biliary structures (the cystic duct stump, the hepatic ducts, or the common bile duct) is diagnosed, a stent may be placed in order to restore ductile continuity.
Minor leaks, with damage to the cystic duct stump, hepatic ducts, and common bile duct ruled out, more often resolve on their own over time, and thus the patient’s closed suction drain will be left in place until biliary drainage ceases, without further initial intervention.
Bottom Line
Surgical tubes and drains have several placement indications. Alterations in quality and quantity of output can indicate changes in clinical status, and hospitalists should be able to handle initial troubleshooting. TH
Dr. Columbus is a general surgery resident at Brigham and Women’s Hospital in Boston. Dr. Havens is an instructor for the department of surgery at Brigham and Women’s Hospital. Dr. Peetz is an instructor for the department of surgery at University Hospital Case Medical Center in Cleveland.
Case
A 45-year-old woman was admitted with choledocholithiasis. Two days prior, following endoscopic retrograde cholangiopancreatography (ERCP), she had gone to the OR for cholecystectomy. The procedure was completed laparoscopically, though the surgeon reported a difficult dissection. The surgeon left a Blake drain in the gallbladder fossa, which initially contained punch-colored fluid. Today, there is bilious fluid in the drain.
Overview
Surgical drains are used to monitor for postoperative leaks or abscesses, to collect normal physiologic fluid, or to minimize dead space. A hospitalist caring for surgical patients may be the first provider to note when something changes in the color or volume of surgical drains. Table 1 lists various types of drains with their indications for use.
Surgical Tubes and Drains
Chest tubes. Chest tubes are placed in the pleural space to evacuate air or fluid. They can be as thin as 20 French or as thick as 40 French (for adults). Chest tubes are typically placed between the fourth and fifth intercostal spaces in the anterior axillary or mid-axillary line; however, the location may vary according to the indication for placement. The tubes can be straight or angled.
The tubes are connected to a collecting system with a three-way chamber. The water chamber holds a column of water, which prevents air from being sucked into the pleural space with inhalation. The suction chamber can be attached to continuous wall suction to remove air or fluid, or it can be placed on “water seal” with no active suction mechanism. The third chamber is the collection chamber for fluid drainage.
Indications for a chest tube include pneumothorax, hemothorax, or a persistent or large pleural effusion. Pneumothorax and hemothorax usually require immediate chest tube placement. Chest tubes are also commonly placed at the end of thoracic surgeries to allow for appropriate re-expansion of the lung tissue.
A chest X-ray should be obtained after any chest tube insertion to ensure appropriate placement. Chest tubes are equipped with a radiopaque line along the longitudinal axis, which should be visible on X-ray. Respiratory variation in the fluid in the collecting tube, called “tidling,” should also be seen in a correctly placed chest tube, and should be monitored at the bedside to reassure continued appropriate location. The interventional radiologist or surgeon who placed the tube should determine the subsequent frequency of serial chest X-rays required to monitor the location of the chest tube.
If the patient has a pneumothorax, air bubbles will be visible in the water chamber; called an air leak, these are often more apparent when the patient coughs. The chest tube should initially be set to continuous suction at -20 mmHg to evacuate the air. Once the air leak has stopped, the chest tube should be placed on water seal to confirm resolution of the pneumothorax (water seal mimics normal physiology). If, after the transition from suction to water seal, resumption of the air leak is noted, it may indicate recurrence of the patient’s pneumothorax. A stat chest X-ray should be obtained, and the chest tube should be placed back on continuous suction. In general, a chest X-ray should be obtained any time the chest tube is changed from suction to water seal or vice versa.
If the patient experiences ongoing or worsening pain, fever, or inadequate drainage, a chest computed tomographic (CT) scan may be warranted to identify inappropriate positioning or other complications, such as occlusion or effusion of the tube. Blood or other debris might clog chest tubes; the surgical team may be able to evacuate the tube with suction tubing at the bedside. If unsuccessful, the tube may need to be removed and reinserted.
The team that placed the tube should help the hospitalist determine the timing of the chest tube removal. If the patient has a pleural effusion, the chest tube can usually be removed when the output is less than 100-200 mL per day and the lung is expanded. The tube should usually be taken off suction and placed on water seal to rule out pneumothorax prior to tube removal.
Penrose drains. Penrose drains are often used to drain fluid or to keep a space open for drainage. Surgeons may use sutures to anchor Penrose drains to skin. Common indications include:
- Ventral hernia repair;
- Debridement of infected pancreatitis; and
- Drainage of superficial abscess cavities.
Penrose drains are simple, flexible tubes that are open at both ends; in contrast to closed drains, they permit ingress as well as egress, facilitating colonization.
Closed suction drains. Closed suction drains with a plastic bulb attachment (i.e., Jackson-Pratt, Blake, Hemovac) are used to collect fluid from a postoperative cavity. Common indications include:
- Post-mastectomy to drain subcutaneous fluid;
- Abdominal surgery;
- Plastic surgery to prevent seroma formation and promote tissue apposition;
- Cholecystectomy if there is concern for damage to ducts of Luschka or other source of bile leak;
- Inadvertent postoperative leakage following a difficult rectal anastomosis; and
- Post-pancreatic surgery.
The quality and quantity of fluid drained should always be carefully noted and recorded. Changes in the fluid can imply development of bleed, leak, or other complications. The surgical team should be contacted immediately if changes are noted.
Typically, closed suction drains will be left in place until the drainage is less than 20 mL per day. These drains can be left in for weeks if necessary and will often be removed during the patient’s scheduled surgical follow-up. Rare complications include erosion into surrounding tissues and inadvertent suturing of the drain in place, such that reexploration is required to remove it. If a closed suction drain becomes occluded, contact the team that placed the drain for further recommendations on adjustment, replacement, or removal.
Nasogastric and duodenal tubes. Nasogastric tubes (NGTs) are often used in the nonoperative management of small bowel obstruction or ileus. They should be placed in the most dependent portion of the gastric lumen and confirmed by chest or abdominal X-ray. NGTs are sump pumps and have a double lumen, which includes an air port to assure flow. The air port should be patent for optimal functioning. The tube may be connected to continuous wall suction or intermittent suction, set to low (less than 60 mmHg) to avoid mucosal avulsion.
NGT output should decrease during the resolution of obstruction or ileus, and symptoms of nausea, vomiting, and abdominal distention should concomitantly improve. Persistently high output in a patient with other indicators of bowel function (flatus, for example) may suggest postpyloric placement (and placement should be checked by X-ray). The timing of NGT removal depends on resumption of bowel function.
Gastrostomy and jejunostomy tubes. Gastrostomy tubes are most commonly used for feeding but may also be used for decompression of functional or anatomic gastric outlet obstruction. They are indicated when patients need prolonged enteral access, such as those with prolonged mechanical ventilation or head and neck pathology that prohibits oral feeding. They are also rarely used for gastropexy to tack an atonic or patulous stomach to the abdominal wall or to prevent recurrence of paraesophageal hernias. These tubes can be placed percutaneously by interventional radiologists, endoscopically by surgeons and gastroenterologists, or laparoscopically or laparotomally by surgeons. This last option is often reserved for patients with difficult anatomy or those who are having laparotomy for another reason.
Because of the stomach’s generous lumen, gastrostomy tubes rarely clog. In the event that they do get clogged, carbonated liquids, meat tenderizer, or enzymes can help dissolve the obstruction. If a gastrostomy tube is left to drain, the patient may experience significant fluid and electrolyte losses, so these need to be carefully monitored.
Jejunostomy tubes are used exclusively for feeding and are usually placed 10-20 cm distal to the ligament of Treitz. These tubes are indicated in patients who require distal feedings due to gastric dysfunction or in those who have undergone a surgery in which a proximal anastomosis requires time to heal. These tubes are more apt to clog and can be more difficult to manage because the lumen of the small bowel is smaller than the stomach. Some prefer not to put pills down the tube to mitigate this risk. Routine flushes with water or saline (30 mL every four to six hours) are also helpful in mitigating the risk of clogging. In the event that they do get clogged, they may be treated like gastrostomy tubes, using carbonated liquids, meat tenderizer, or enzymes to help dissolve the obstruction.
Percutaneous tube sites should be examined frequently for signs of infection. Though gastrostomy and jejunostomy tubes are typically well secured intraabdominally, they can become dislodged. If a gastrostomy or jejunostomy tube has been in place for more than two weeks, it can easily be replaced at the bedside with a tube of comparable caliber by a member of the surgical team or by an experienced hospitalist. If the tube has been in place less than two weeks, it requires replacement with radiographic guidance, as the risk of creating a false lumen is high. Over time, tubes can become loose and fall out. If they need replacement, the preceding guidelines apply.
Back to the Case
A potential major complication of cholecystectomy is severance of the common bile duct, which necessitates significant further surgery. Less severe complications include injuries to the cholecystohepatic ducts (otherwise known as the ducts of Luschka), which can result in leakage of bile into the peritoneal cavity. A bile leak can lead to abscess and systemic infection if left undrained.
Surgeons who are concerned for such a complication intraoperatively may opt to leave a closed suction drain in the gallbladder fossa, such as a Blake drain, for monitoring and subsequent drainage. The drain will remain in place at least until the patient’s diet has been advanced fully, because digestion promotes the secretion of bile and may elucidate a leak. Bilious fluid in the Blake drain is suspicious for a leak.
The surgeon should be notified, and imaging should be obtained to find the nature of the injury to the biliary tree (CT scan with IV contrast, hepatobiliary iminodiacetic acid scan, or endoscopic retrograde cholangiopancreatography). If injury to major biliary structures (the cystic duct stump, the hepatic ducts, or the common bile duct) is diagnosed, a stent may be placed in order to restore ductile continuity.
Minor leaks, with damage to the cystic duct stump, hepatic ducts, and common bile duct ruled out, more often resolve on their own over time, and thus the patient’s closed suction drain will be left in place until biliary drainage ceases, without further initial intervention.
Bottom Line
Surgical tubes and drains have several placement indications. Alterations in quality and quantity of output can indicate changes in clinical status, and hospitalists should be able to handle initial troubleshooting. TH
Dr. Columbus is a general surgery resident at Brigham and Women’s Hospital in Boston. Dr. Havens is an instructor for the department of surgery at Brigham and Women’s Hospital. Dr. Peetz is an instructor for the department of surgery at University Hospital Case Medical Center in Cleveland.
Heavily pretreated myeloma responds to pembrolizumab combo
ORLANDO – The one-two punch of combining the programmed cell death-1 (PD-1) inhibitor pembrolizumab with the immunomodulatory drug lenalidomide and low-dose dexamethasone produced responses in 76% of 17 heavily pretreated patients with relapsed or refractory multiple myeloma in the KEYNOTE-023 study.
This included four very good partial responses (24%) and nine partial responses (53%).
In nine lenalidomide-refractory patients, the overall response rate was 56%, including two very good partial responses (22%) and three partial responses (33%).
The efficacy results are preliminary, but support the continued development of pembrolizumab (Keytruda) in patients with multiple myeloma, Dr. Jesús San Miguel of Clinica Universidad de Navarra, Pamplona, Spain, said at the annual meeting of the American Society of Hematology.
He closed his presentation with two illustrative cases highlighting a rapid response lasting more than a year and a half in a 49-year-old man with myeloma triple-refractory to autologous stem cell transplant (ASCT), lenalidomide (Revlimid), and dexamethasone.
The second case involved a patient with double-refractory myeloma and extramedullary disease who achieved a stringent complete response after two cycles of fourth-line pembrolizumab that was associated with a “striking” reduction in lesion volume on computed tomography scans, he said.
The median duration of response among the 17 evaluable patients was 9.7 months.
The median time to first response was 1.2 months (range 1.0 months to 6.5 months). But some patients require more time and, interestingly, the quality of the response was upgraded in 11% with continued treatment, Dr. San Miguel said.
The rationale for combining PD-1 inhibitors with immunomodulatory drugs (IMiD) lies in recent research showing that lenalidomide reduces PD-ligand 1 and PD-1 expression on multiple myeloma cells as well as on T and myeloid-derived suppressor cells, he explained. In addition, lenalidomide enhances checkpoint blockade–induced effector cytokine production in multiple myeloma bone marrow and induces cytotoxicity against myeloma cells.
“Lenalidomide will increase the number of T cells and the T-cell activation and anti-PD-1 will release the brake in order to allow these activated T cells to interact with the tumor,” Dr. San Miguel said.
Patients enrolled in KEYSTONE-023 were heavily pretreated, with 26% previously exposed to pomalidomide, 76% refractory to lenalidomide, 80% refractory to their last line of therapy, and 86% having undergone prior ASCT. Half the patients were double, triple, or quadruple refractory, he noted.
The study (Abstract 505) was designed to identify the maximum tolerated dose (MTD) of pembrolizumab and to assess its safety and tolerability when given with lenalidomide and dexamethasone in patients with multiple myeloma failing at least two prior lines of therapy including a proteasome inhibitor and an IMiD. Their median age was 62 years; 64% were male.
In the dose-determination stage, three of six patients treated with pembrolizumab 2 mg/kg plus lenalidomide 25 mg and dexamethasone 40 mg experienced dose-limiting toxicities that resolved without treatment discontinuation.
After dose adjustments, a “flat dose” of pembrolizumab 200 mg given every other week in a rapid 30-minute intravenous infusion without premedication with lenalidomide 25 mg on days 1-21 and dexamethasone 40 mg weekly did not cause any dose-limiting toxicities and was identified as the final MTD, Dr. San Miguel said.
The regimen is to continue for 24 months or until tumor progression or excessive side effects and was carried forward into the dose-expansion stage in 33 additional patients with a median follow-up of 48 days.
Among all 50 patients evaluable for safety, 72% experienced at least one treatment-related adverse event of any grade and 46% (23/50 patients) had grade 3/4 adverse events including neutropenia (22%), thrombocytopenia and anemia (8% each), hyperglycemia (6%), and fatigue, muscle spasms, and diarrhea (2% each).
The adverse events were consistent with the individual drug safety profiles, but “the incidence may be underestimated due to the limited drug exposure,” Dr. San Miguel cautioned.
Immune-mediated adverse events included two cases each of hyper- and hypothyroidism, one case of thyroiditis, and one grade 2 adrenal insufficiency. No cases of colitis or pneumonitis were reported. No dose modifications or treatment discontinuations were required to mange the immune-related side effects, he said. No treatment-related deaths occurred.
In a second study reported during the same oral myeloma session, pneumonitis cropped up in 10% of heavily pretreated patients with relapsed multiple myeloma receiving a slightly different regimen of pembrolizumab plus the IMiD pomalidomide (Pomalyst) and dexamethasone. The overall response rate in the phase II study was 60% among 27 evaluable patients and 55% in those double-refractory to IMiDs and proteasome inhibitors.
ORLANDO – The one-two punch of combining the programmed cell death-1 (PD-1) inhibitor pembrolizumab with the immunomodulatory drug lenalidomide and low-dose dexamethasone produced responses in 76% of 17 heavily pretreated patients with relapsed or refractory multiple myeloma in the KEYNOTE-023 study.
This included four very good partial responses (24%) and nine partial responses (53%).
In nine lenalidomide-refractory patients, the overall response rate was 56%, including two very good partial responses (22%) and three partial responses (33%).
The efficacy results are preliminary, but support the continued development of pembrolizumab (Keytruda) in patients with multiple myeloma, Dr. Jesús San Miguel of Clinica Universidad de Navarra, Pamplona, Spain, said at the annual meeting of the American Society of Hematology.
He closed his presentation with two illustrative cases highlighting a rapid response lasting more than a year and a half in a 49-year-old man with myeloma triple-refractory to autologous stem cell transplant (ASCT), lenalidomide (Revlimid), and dexamethasone.
The second case involved a patient with double-refractory myeloma and extramedullary disease who achieved a stringent complete response after two cycles of fourth-line pembrolizumab that was associated with a “striking” reduction in lesion volume on computed tomography scans, he said.
The median duration of response among the 17 evaluable patients was 9.7 months.
The median time to first response was 1.2 months (range 1.0 months to 6.5 months). But some patients require more time and, interestingly, the quality of the response was upgraded in 11% with continued treatment, Dr. San Miguel said.
The rationale for combining PD-1 inhibitors with immunomodulatory drugs (IMiD) lies in recent research showing that lenalidomide reduces PD-ligand 1 and PD-1 expression on multiple myeloma cells as well as on T and myeloid-derived suppressor cells, he explained. In addition, lenalidomide enhances checkpoint blockade–induced effector cytokine production in multiple myeloma bone marrow and induces cytotoxicity against myeloma cells.
“Lenalidomide will increase the number of T cells and the T-cell activation and anti-PD-1 will release the brake in order to allow these activated T cells to interact with the tumor,” Dr. San Miguel said.
Patients enrolled in KEYSTONE-023 were heavily pretreated, with 26% previously exposed to pomalidomide, 76% refractory to lenalidomide, 80% refractory to their last line of therapy, and 86% having undergone prior ASCT. Half the patients were double, triple, or quadruple refractory, he noted.
The study (Abstract 505) was designed to identify the maximum tolerated dose (MTD) of pembrolizumab and to assess its safety and tolerability when given with lenalidomide and dexamethasone in patients with multiple myeloma failing at least two prior lines of therapy including a proteasome inhibitor and an IMiD. Their median age was 62 years; 64% were male.
In the dose-determination stage, three of six patients treated with pembrolizumab 2 mg/kg plus lenalidomide 25 mg and dexamethasone 40 mg experienced dose-limiting toxicities that resolved without treatment discontinuation.
After dose adjustments, a “flat dose” of pembrolizumab 200 mg given every other week in a rapid 30-minute intravenous infusion without premedication with lenalidomide 25 mg on days 1-21 and dexamethasone 40 mg weekly did not cause any dose-limiting toxicities and was identified as the final MTD, Dr. San Miguel said.
The regimen is to continue for 24 months or until tumor progression or excessive side effects and was carried forward into the dose-expansion stage in 33 additional patients with a median follow-up of 48 days.
Among all 50 patients evaluable for safety, 72% experienced at least one treatment-related adverse event of any grade and 46% (23/50 patients) had grade 3/4 adverse events including neutropenia (22%), thrombocytopenia and anemia (8% each), hyperglycemia (6%), and fatigue, muscle spasms, and diarrhea (2% each).
The adverse events were consistent with the individual drug safety profiles, but “the incidence may be underestimated due to the limited drug exposure,” Dr. San Miguel cautioned.
Immune-mediated adverse events included two cases each of hyper- and hypothyroidism, one case of thyroiditis, and one grade 2 adrenal insufficiency. No cases of colitis or pneumonitis were reported. No dose modifications or treatment discontinuations were required to mange the immune-related side effects, he said. No treatment-related deaths occurred.
In a second study reported during the same oral myeloma session, pneumonitis cropped up in 10% of heavily pretreated patients with relapsed multiple myeloma receiving a slightly different regimen of pembrolizumab plus the IMiD pomalidomide (Pomalyst) and dexamethasone. The overall response rate in the phase II study was 60% among 27 evaluable patients and 55% in those double-refractory to IMiDs and proteasome inhibitors.
ORLANDO – The one-two punch of combining the programmed cell death-1 (PD-1) inhibitor pembrolizumab with the immunomodulatory drug lenalidomide and low-dose dexamethasone produced responses in 76% of 17 heavily pretreated patients with relapsed or refractory multiple myeloma in the KEYNOTE-023 study.
This included four very good partial responses (24%) and nine partial responses (53%).
In nine lenalidomide-refractory patients, the overall response rate was 56%, including two very good partial responses (22%) and three partial responses (33%).
The efficacy results are preliminary, but support the continued development of pembrolizumab (Keytruda) in patients with multiple myeloma, Dr. Jesús San Miguel of Clinica Universidad de Navarra, Pamplona, Spain, said at the annual meeting of the American Society of Hematology.
He closed his presentation with two illustrative cases highlighting a rapid response lasting more than a year and a half in a 49-year-old man with myeloma triple-refractory to autologous stem cell transplant (ASCT), lenalidomide (Revlimid), and dexamethasone.
The second case involved a patient with double-refractory myeloma and extramedullary disease who achieved a stringent complete response after two cycles of fourth-line pembrolizumab that was associated with a “striking” reduction in lesion volume on computed tomography scans, he said.
The median duration of response among the 17 evaluable patients was 9.7 months.
The median time to first response was 1.2 months (range 1.0 months to 6.5 months). But some patients require more time and, interestingly, the quality of the response was upgraded in 11% with continued treatment, Dr. San Miguel said.
The rationale for combining PD-1 inhibitors with immunomodulatory drugs (IMiD) lies in recent research showing that lenalidomide reduces PD-ligand 1 and PD-1 expression on multiple myeloma cells as well as on T and myeloid-derived suppressor cells, he explained. In addition, lenalidomide enhances checkpoint blockade–induced effector cytokine production in multiple myeloma bone marrow and induces cytotoxicity against myeloma cells.
“Lenalidomide will increase the number of T cells and the T-cell activation and anti-PD-1 will release the brake in order to allow these activated T cells to interact with the tumor,” Dr. San Miguel said.
Patients enrolled in KEYSTONE-023 were heavily pretreated, with 26% previously exposed to pomalidomide, 76% refractory to lenalidomide, 80% refractory to their last line of therapy, and 86% having undergone prior ASCT. Half the patients were double, triple, or quadruple refractory, he noted.
The study (Abstract 505) was designed to identify the maximum tolerated dose (MTD) of pembrolizumab and to assess its safety and tolerability when given with lenalidomide and dexamethasone in patients with multiple myeloma failing at least two prior lines of therapy including a proteasome inhibitor and an IMiD. Their median age was 62 years; 64% were male.
In the dose-determination stage, three of six patients treated with pembrolizumab 2 mg/kg plus lenalidomide 25 mg and dexamethasone 40 mg experienced dose-limiting toxicities that resolved without treatment discontinuation.
After dose adjustments, a “flat dose” of pembrolizumab 200 mg given every other week in a rapid 30-minute intravenous infusion without premedication with lenalidomide 25 mg on days 1-21 and dexamethasone 40 mg weekly did not cause any dose-limiting toxicities and was identified as the final MTD, Dr. San Miguel said.
The regimen is to continue for 24 months or until tumor progression or excessive side effects and was carried forward into the dose-expansion stage in 33 additional patients with a median follow-up of 48 days.
Among all 50 patients evaluable for safety, 72% experienced at least one treatment-related adverse event of any grade and 46% (23/50 patients) had grade 3/4 adverse events including neutropenia (22%), thrombocytopenia and anemia (8% each), hyperglycemia (6%), and fatigue, muscle spasms, and diarrhea (2% each).
The adverse events were consistent with the individual drug safety profiles, but “the incidence may be underestimated due to the limited drug exposure,” Dr. San Miguel cautioned.
Immune-mediated adverse events included two cases each of hyper- and hypothyroidism, one case of thyroiditis, and one grade 2 adrenal insufficiency. No cases of colitis or pneumonitis were reported. No dose modifications or treatment discontinuations were required to mange the immune-related side effects, he said. No treatment-related deaths occurred.
In a second study reported during the same oral myeloma session, pneumonitis cropped up in 10% of heavily pretreated patients with relapsed multiple myeloma receiving a slightly different regimen of pembrolizumab plus the IMiD pomalidomide (Pomalyst) and dexamethasone. The overall response rate in the phase II study was 60% among 27 evaluable patients and 55% in those double-refractory to IMiDs and proteasome inhibitors.
AT ASH 2015
Key clinical point: Initial results show promising activity for pembrolizumab in combination with lenalidomide and low-dose dexamethasone in heavily pretreated relapsed or refractory multiple myeloma.
Major finding: The overall response rate was 76% (13/17 patients).
Data source: Phase I study in 50 patients with relapsed or refractory multiple myeloma.
Disclosures: The study was supported by Merck. Dr. San Miguel reported consulting for Merck and relationships with Millennium, Janssen, Celgene, Novartis, Onyx, Bristol-Myers Squibb, and Sanofi.
The palliative path: Talking with elderly patients facing emergency surgery
An expert panel has developed a communication framework to improve treatment of older, seriously ill patients who have surgical emergencies, which has been published online in Annals of Surgery.
A substantial portion of older patients who undergo emergency surgeries already have serious life-limiting illnesses such as cardiopulmonary disease, renal failure, liver failure, dementia, severe neurological impairment, or malignancy. The advisory panel based its work on the premise that surgery in these circumstances can lead to significant further morbidity, health care utilization, functional decline, prolonged hospital stay or institutionalization, and death, with attendant physical discomfort and psychological distress at the end of these patients’ lives.
Surgeons consulted in the emergency setting for these patients are hampered by patients unable to communicate well because they are in extremis, by surrogates who are unprepared for their role, and by time constraints, lack of familiarity with the patient, poor understanding of the illness by patients and families, prognostic uncertainty, and inadequate advance care planning. In addition, “many surgeons lack skills to engage in conversations about end-of-life care, or are too unfamiliar with palliative options to discuss them well,” or feel obligated to maintain postoperative life support despite the patient’s wishes, said Dr. Zara Cooper, of Ariadne Labs and the Center for Surgery and Public Health at Brigham and Women’s Hospital, both in Boston, and her associates.
To address these issues and assist surgeons in caring for such patients, an expert panel of 23 national leaders in acute care surgery, general surgery, surgical oncology, palliative medicine, critical care, emergency medicine, anesthesia, and health care innovation was convened at Harvard Medical School, Boston.
The focus of the panel’s recommendations was a structured communications framework prototype to facilitate shared decision-making in these difficult circumstances.
Among the panel’s recommendations for surgeons were the following priorities:
• Review the medical record and consult the treatment team to fully understand the patient’s current condition, comorbidities, expected illness trajectory, and preferences for end-of-life care.
• Assess functional performance as part of the routine history and physical to fully understand the patient’s fitness for surgery.
• Formulate a prognosis regarding the patient’s overall health both with and without surgery.
The panel offered a set of principles and specific elements for the meeting with the patient and family:
• The surgeon should begin by introducing himself or herself; according to reports in the literature, physicians fail to do this approximately half of the time.
• Pay attention to nonverbal communication, such as eye contact and physical contact, as this is critical to building rapport. Immediately address pain, anxiety, and other indicators of distress, to maximize the patients’ and the families’ engagement in subsequent medical discussions. “Although adequate analgesia may render a patient unable to make their own decisions, surrogates are more likely to make appropriate decisions when they feel their loved one is comfortable,” the panel noted.
• Allow pauses and silences to occur. Let the patient and the family process information and their own emotions.
• Elicit the patients’ or the surrogates’ understanding of the illness and their views of the patients’ likely trajectory, correcting any inaccuracies. This substantially influences their decisions regarding the aggressiveness of subsequent treatments.
• Inform the patient and family of the life-threatening nature of the patient’s acute condition and its potential impact on the rest of his or her life, including the possibility of prolonged life support, ICU stay, burdensome treatment, and loss of independence. Use accepted techniques for breaking bad news, and check to be sure the patient understands what was conveyed.
• At this point, the surgeon should synthesize and summarize the information from the patient, the family, and the medical record, then pause to give them time to process the information and to assess their emotional state. It is helpful to label and respond to the patient’s emotions at this juncture, and to build empathy with statements such as “I know this is difficult news, and I wish it were different.”
• Describe the benefits, burdens, and range of likely outcomes if surgery is undertaken and if it is not. The surgeon should use nonmedical language to describe symptoms, and should convey his or her expectations regarding length of hospitalization, need for and duration of life support, burdensome symptoms, discharge to an institution, and functional recovery.
• Surgeons should be able to communicate palliative options possible either in combination with surgery or instead of surgery. Palliative care can aid in managing advanced symptoms, providing psychosocial support for patients and caregivers, facilitating interdisciplinary communication, and facilitating medical decisions and care transitions.
• Avoid describing surgical procedures as “doing everything” and palliative care as “doing nothing.” This can make patients and families “feel abandoned, fearful, isolated, and angry, and fails to encompass palliative care’s practices of proactive communication, aggressive symptom management, and timely emotional support to alleviate suffering and affirm quality of life,” the panel said.
• Surgeons should explicitly support the patients’ medical decisions, whether or not they choose surgery.
The panel also cited a few factors that would assist surgeons in following these recommendations. First, surgeons must recognize the importance of communicating well with seriously ill older patients and acknowledge that this is a crucial clinical skill for them to cultivate. They must also recognize that palliative care is vital to delivering high-quality surgical care. Surgeons should consider discharging patients to hospice, which can improve pain and symptom management, improve patient and family satisfaction with care, and avoid unwanted hospitalization or cardiopulmonary resuscitation.
“There are a number of major barriers to introducing palliative care in these situations. One is an education problem - the perception on the part of patients and clinicians, and surgeons in particular, that palliative care is only limited to end-of-life care, which it is not. It is a misperception of what palliative care means in this equation - that palliative care and hospice are the same thing, which they absolutely are not,”said Dr. Cooper in an interview.
”The definition of palliative care has evolved over the past decade and the focus of palliative care is on quality of life and alleviating symptoms. End-of-life palliative care is part of that, and as patients get closer to the end of life, symptom management and quality of life become more focal than life-prolonging treatment... But for patients with chronic and serious illness, there has to be a role for palliative care because we know that when patients feel better, they tend to live longer. And when patients feel their emotional concerns and physical needs are being addressed, they tend to do better. Patients families have improved satisfaction when their loved one receives palliative care,” she noted.”
However, the number of palliative providers is completely inadequate to meet the needs of the number of seriously ill patients, she said. And a lot of hospital-based palliative care is by necessity limited to end-of-life care because of a lack of palliative resources.
Dr. Atul Gawande, a coauthor of the panel recommendations, wrote a best-selling book, Being Mortal (New York: Metropolitan Books, 2014) addressing the shortcomings and potential remaking of medical care in the context of age-related frailty, grave illness, and death. Dr. Cooper noted that there is a growing sentiment among the general public that they want to have their quality of life addressed in the type of medical care they receive. She said that Dr. Gawande’s book tapped into the perception of a lack of recognition of personhood of seriously ill patients.
“We often focus on diagnosis and we don’t have the ‘bandwidth’ to focus on the person carrying that diagnosis, and our patients and focus on the person carrying that diagnosis, but our patients and their families are demanding different types of care. So, ultimately, the patients will be the ones to push us to do better for them.”
The next steps to further developing a widely used and validated communication framework would be to create educational opportunities for clinicians to develop clinical skills in communication with seriously ill patients and palliative care, and to study the impact of these initiatives on improving outcomes most relevant to older patient. This work was supported by the Ariadne Labs, a Joint Center for Health System Innovation at Brigham and Women’s Hospital. Dr. Cooper and her associates reported having no relevant financial disclosures.
An expert panel has developed a communication framework to improve treatment of older, seriously ill patients who have surgical emergencies, which has been published online in Annals of Surgery.
A substantial portion of older patients who undergo emergency surgeries already have serious life-limiting illnesses such as cardiopulmonary disease, renal failure, liver failure, dementia, severe neurological impairment, or malignancy. The advisory panel based its work on the premise that surgery in these circumstances can lead to significant further morbidity, health care utilization, functional decline, prolonged hospital stay or institutionalization, and death, with attendant physical discomfort and psychological distress at the end of these patients’ lives.
Surgeons consulted in the emergency setting for these patients are hampered by patients unable to communicate well because they are in extremis, by surrogates who are unprepared for their role, and by time constraints, lack of familiarity with the patient, poor understanding of the illness by patients and families, prognostic uncertainty, and inadequate advance care planning. In addition, “many surgeons lack skills to engage in conversations about end-of-life care, or are too unfamiliar with palliative options to discuss them well,” or feel obligated to maintain postoperative life support despite the patient’s wishes, said Dr. Zara Cooper, of Ariadne Labs and the Center for Surgery and Public Health at Brigham and Women’s Hospital, both in Boston, and her associates.
To address these issues and assist surgeons in caring for such patients, an expert panel of 23 national leaders in acute care surgery, general surgery, surgical oncology, palliative medicine, critical care, emergency medicine, anesthesia, and health care innovation was convened at Harvard Medical School, Boston.
The focus of the panel’s recommendations was a structured communications framework prototype to facilitate shared decision-making in these difficult circumstances.
Among the panel’s recommendations for surgeons were the following priorities:
• Review the medical record and consult the treatment team to fully understand the patient’s current condition, comorbidities, expected illness trajectory, and preferences for end-of-life care.
• Assess functional performance as part of the routine history and physical to fully understand the patient’s fitness for surgery.
• Formulate a prognosis regarding the patient’s overall health both with and without surgery.
The panel offered a set of principles and specific elements for the meeting with the patient and family:
• The surgeon should begin by introducing himself or herself; according to reports in the literature, physicians fail to do this approximately half of the time.
• Pay attention to nonverbal communication, such as eye contact and physical contact, as this is critical to building rapport. Immediately address pain, anxiety, and other indicators of distress, to maximize the patients’ and the families’ engagement in subsequent medical discussions. “Although adequate analgesia may render a patient unable to make their own decisions, surrogates are more likely to make appropriate decisions when they feel their loved one is comfortable,” the panel noted.
• Allow pauses and silences to occur. Let the patient and the family process information and their own emotions.
• Elicit the patients’ or the surrogates’ understanding of the illness and their views of the patients’ likely trajectory, correcting any inaccuracies. This substantially influences their decisions regarding the aggressiveness of subsequent treatments.
• Inform the patient and family of the life-threatening nature of the patient’s acute condition and its potential impact on the rest of his or her life, including the possibility of prolonged life support, ICU stay, burdensome treatment, and loss of independence. Use accepted techniques for breaking bad news, and check to be sure the patient understands what was conveyed.
• At this point, the surgeon should synthesize and summarize the information from the patient, the family, and the medical record, then pause to give them time to process the information and to assess their emotional state. It is helpful to label and respond to the patient’s emotions at this juncture, and to build empathy with statements such as “I know this is difficult news, and I wish it were different.”
• Describe the benefits, burdens, and range of likely outcomes if surgery is undertaken and if it is not. The surgeon should use nonmedical language to describe symptoms, and should convey his or her expectations regarding length of hospitalization, need for and duration of life support, burdensome symptoms, discharge to an institution, and functional recovery.
• Surgeons should be able to communicate palliative options possible either in combination with surgery or instead of surgery. Palliative care can aid in managing advanced symptoms, providing psychosocial support for patients and caregivers, facilitating interdisciplinary communication, and facilitating medical decisions and care transitions.
• Avoid describing surgical procedures as “doing everything” and palliative care as “doing nothing.” This can make patients and families “feel abandoned, fearful, isolated, and angry, and fails to encompass palliative care’s practices of proactive communication, aggressive symptom management, and timely emotional support to alleviate suffering and affirm quality of life,” the panel said.
• Surgeons should explicitly support the patients’ medical decisions, whether or not they choose surgery.
The panel also cited a few factors that would assist surgeons in following these recommendations. First, surgeons must recognize the importance of communicating well with seriously ill older patients and acknowledge that this is a crucial clinical skill for them to cultivate. They must also recognize that palliative care is vital to delivering high-quality surgical care. Surgeons should consider discharging patients to hospice, which can improve pain and symptom management, improve patient and family satisfaction with care, and avoid unwanted hospitalization or cardiopulmonary resuscitation.
“There are a number of major barriers to introducing palliative care in these situations. One is an education problem - the perception on the part of patients and clinicians, and surgeons in particular, that palliative care is only limited to end-of-life care, which it is not. It is a misperception of what palliative care means in this equation - that palliative care and hospice are the same thing, which they absolutely are not,”said Dr. Cooper in an interview.
”The definition of palliative care has evolved over the past decade and the focus of palliative care is on quality of life and alleviating symptoms. End-of-life palliative care is part of that, and as patients get closer to the end of life, symptom management and quality of life become more focal than life-prolonging treatment... But for patients with chronic and serious illness, there has to be a role for palliative care because we know that when patients feel better, they tend to live longer. And when patients feel their emotional concerns and physical needs are being addressed, they tend to do better. Patients families have improved satisfaction when their loved one receives palliative care,” she noted.”
However, the number of palliative providers is completely inadequate to meet the needs of the number of seriously ill patients, she said. And a lot of hospital-based palliative care is by necessity limited to end-of-life care because of a lack of palliative resources.
Dr. Atul Gawande, a coauthor of the panel recommendations, wrote a best-selling book, Being Mortal (New York: Metropolitan Books, 2014) addressing the shortcomings and potential remaking of medical care in the context of age-related frailty, grave illness, and death. Dr. Cooper noted that there is a growing sentiment among the general public that they want to have their quality of life addressed in the type of medical care they receive. She said that Dr. Gawande’s book tapped into the perception of a lack of recognition of personhood of seriously ill patients.
“We often focus on diagnosis and we don’t have the ‘bandwidth’ to focus on the person carrying that diagnosis, and our patients and focus on the person carrying that diagnosis, but our patients and their families are demanding different types of care. So, ultimately, the patients will be the ones to push us to do better for them.”
The next steps to further developing a widely used and validated communication framework would be to create educational opportunities for clinicians to develop clinical skills in communication with seriously ill patients and palliative care, and to study the impact of these initiatives on improving outcomes most relevant to older patient. This work was supported by the Ariadne Labs, a Joint Center for Health System Innovation at Brigham and Women’s Hospital. Dr. Cooper and her associates reported having no relevant financial disclosures.
An expert panel has developed a communication framework to improve treatment of older, seriously ill patients who have surgical emergencies, which has been published online in Annals of Surgery.
A substantial portion of older patients who undergo emergency surgeries already have serious life-limiting illnesses such as cardiopulmonary disease, renal failure, liver failure, dementia, severe neurological impairment, or malignancy. The advisory panel based its work on the premise that surgery in these circumstances can lead to significant further morbidity, health care utilization, functional decline, prolonged hospital stay or institutionalization, and death, with attendant physical discomfort and psychological distress at the end of these patients’ lives.
Surgeons consulted in the emergency setting for these patients are hampered by patients unable to communicate well because they are in extremis, by surrogates who are unprepared for their role, and by time constraints, lack of familiarity with the patient, poor understanding of the illness by patients and families, prognostic uncertainty, and inadequate advance care planning. In addition, “many surgeons lack skills to engage in conversations about end-of-life care, or are too unfamiliar with palliative options to discuss them well,” or feel obligated to maintain postoperative life support despite the patient’s wishes, said Dr. Zara Cooper, of Ariadne Labs and the Center for Surgery and Public Health at Brigham and Women’s Hospital, both in Boston, and her associates.
To address these issues and assist surgeons in caring for such patients, an expert panel of 23 national leaders in acute care surgery, general surgery, surgical oncology, palliative medicine, critical care, emergency medicine, anesthesia, and health care innovation was convened at Harvard Medical School, Boston.
The focus of the panel’s recommendations was a structured communications framework prototype to facilitate shared decision-making in these difficult circumstances.
Among the panel’s recommendations for surgeons were the following priorities:
• Review the medical record and consult the treatment team to fully understand the patient’s current condition, comorbidities, expected illness trajectory, and preferences for end-of-life care.
• Assess functional performance as part of the routine history and physical to fully understand the patient’s fitness for surgery.
• Formulate a prognosis regarding the patient’s overall health both with and without surgery.
The panel offered a set of principles and specific elements for the meeting with the patient and family:
• The surgeon should begin by introducing himself or herself; according to reports in the literature, physicians fail to do this approximately half of the time.
• Pay attention to nonverbal communication, such as eye contact and physical contact, as this is critical to building rapport. Immediately address pain, anxiety, and other indicators of distress, to maximize the patients’ and the families’ engagement in subsequent medical discussions. “Although adequate analgesia may render a patient unable to make their own decisions, surrogates are more likely to make appropriate decisions when they feel their loved one is comfortable,” the panel noted.
• Allow pauses and silences to occur. Let the patient and the family process information and their own emotions.
• Elicit the patients’ or the surrogates’ understanding of the illness and their views of the patients’ likely trajectory, correcting any inaccuracies. This substantially influences their decisions regarding the aggressiveness of subsequent treatments.
• Inform the patient and family of the life-threatening nature of the patient’s acute condition and its potential impact on the rest of his or her life, including the possibility of prolonged life support, ICU stay, burdensome treatment, and loss of independence. Use accepted techniques for breaking bad news, and check to be sure the patient understands what was conveyed.
• At this point, the surgeon should synthesize and summarize the information from the patient, the family, and the medical record, then pause to give them time to process the information and to assess their emotional state. It is helpful to label and respond to the patient’s emotions at this juncture, and to build empathy with statements such as “I know this is difficult news, and I wish it were different.”
• Describe the benefits, burdens, and range of likely outcomes if surgery is undertaken and if it is not. The surgeon should use nonmedical language to describe symptoms, and should convey his or her expectations regarding length of hospitalization, need for and duration of life support, burdensome symptoms, discharge to an institution, and functional recovery.
• Surgeons should be able to communicate palliative options possible either in combination with surgery or instead of surgery. Palliative care can aid in managing advanced symptoms, providing psychosocial support for patients and caregivers, facilitating interdisciplinary communication, and facilitating medical decisions and care transitions.
• Avoid describing surgical procedures as “doing everything” and palliative care as “doing nothing.” This can make patients and families “feel abandoned, fearful, isolated, and angry, and fails to encompass palliative care’s practices of proactive communication, aggressive symptom management, and timely emotional support to alleviate suffering and affirm quality of life,” the panel said.
• Surgeons should explicitly support the patients’ medical decisions, whether or not they choose surgery.
The panel also cited a few factors that would assist surgeons in following these recommendations. First, surgeons must recognize the importance of communicating well with seriously ill older patients and acknowledge that this is a crucial clinical skill for them to cultivate. They must also recognize that palliative care is vital to delivering high-quality surgical care. Surgeons should consider discharging patients to hospice, which can improve pain and symptom management, improve patient and family satisfaction with care, and avoid unwanted hospitalization or cardiopulmonary resuscitation.
“There are a number of major barriers to introducing palliative care in these situations. One is an education problem - the perception on the part of patients and clinicians, and surgeons in particular, that palliative care is only limited to end-of-life care, which it is not. It is a misperception of what palliative care means in this equation - that palliative care and hospice are the same thing, which they absolutely are not,”said Dr. Cooper in an interview.
”The definition of palliative care has evolved over the past decade and the focus of palliative care is on quality of life and alleviating symptoms. End-of-life palliative care is part of that, and as patients get closer to the end of life, symptom management and quality of life become more focal than life-prolonging treatment... But for patients with chronic and serious illness, there has to be a role for palliative care because we know that when patients feel better, they tend to live longer. And when patients feel their emotional concerns and physical needs are being addressed, they tend to do better. Patients families have improved satisfaction when their loved one receives palliative care,” she noted.”
However, the number of palliative providers is completely inadequate to meet the needs of the number of seriously ill patients, she said. And a lot of hospital-based palliative care is by necessity limited to end-of-life care because of a lack of palliative resources.
Dr. Atul Gawande, a coauthor of the panel recommendations, wrote a best-selling book, Being Mortal (New York: Metropolitan Books, 2014) addressing the shortcomings and potential remaking of medical care in the context of age-related frailty, grave illness, and death. Dr. Cooper noted that there is a growing sentiment among the general public that they want to have their quality of life addressed in the type of medical care they receive. She said that Dr. Gawande’s book tapped into the perception of a lack of recognition of personhood of seriously ill patients.
“We often focus on diagnosis and we don’t have the ‘bandwidth’ to focus on the person carrying that diagnosis, and our patients and focus on the person carrying that diagnosis, but our patients and their families are demanding different types of care. So, ultimately, the patients will be the ones to push us to do better for them.”
The next steps to further developing a widely used and validated communication framework would be to create educational opportunities for clinicians to develop clinical skills in communication with seriously ill patients and palliative care, and to study the impact of these initiatives on improving outcomes most relevant to older patient. This work was supported by the Ariadne Labs, a Joint Center for Health System Innovation at Brigham and Women’s Hospital. Dr. Cooper and her associates reported having no relevant financial disclosures.
FROM ANNALS OF SURGERY
Private Insurers to Reap Bulk of Spending on Hospitalized Patient Care
Spending on care of hospitalized patients is expected to pass $1 trillion in 2015, a new high. Thomas Selden, PhD, of the Agency for Healthcare Research and Quality recently asked where that money is likely to go. The answer: private insurers.
Dr. Selden and his colleagues report in Health Affairs this month that in 2012, private insurers’ payment rates for inpatient hospital stays were approximately 75% greater than Medicare’s payment rates, a sharp increase from the differential of approximately 10% percent during the period of 1996 to 2001. “We need to understand who’s paying what,” Dr. Selden says. “It’s the first step to a better understanding of public policy.”
The report found that “the predicted percentage difference between the rates of private insurers and those of Medicare has increased substantially over time.” In 1996, private insurers paid 106.1% of Medicare payment rates, a payment rate difference of 6.1% (95% CI: -3.2, 15.5). The difference climbed to 64.1% (95% CI: 48.3, 80.0) in 2011 and 75.3% (95% CI: 52.0, 98.6) in 2012. Medicaid payment rates averaged approximately 90% of Medicare payment rates throughout the study period.
Dr. Selden is hopeful that stakeholders will use the data his team collected to determine the impetus for the widening gap. He also plans to research whether payment differences affect quality metrics.
“Anytime you’re talking about a trillion dollars, it’s really important when a payment difference opens up of this magnitude,” he adds. “The difference is real … what the policy implications are is for the policy makers to decide.” TH
Visit our website for more information on healthcare payment models.
Spending on care of hospitalized patients is expected to pass $1 trillion in 2015, a new high. Thomas Selden, PhD, of the Agency for Healthcare Research and Quality recently asked where that money is likely to go. The answer: private insurers.
Dr. Selden and his colleagues report in Health Affairs this month that in 2012, private insurers’ payment rates for inpatient hospital stays were approximately 75% greater than Medicare’s payment rates, a sharp increase from the differential of approximately 10% percent during the period of 1996 to 2001. “We need to understand who’s paying what,” Dr. Selden says. “It’s the first step to a better understanding of public policy.”
The report found that “the predicted percentage difference between the rates of private insurers and those of Medicare has increased substantially over time.” In 1996, private insurers paid 106.1% of Medicare payment rates, a payment rate difference of 6.1% (95% CI: -3.2, 15.5). The difference climbed to 64.1% (95% CI: 48.3, 80.0) in 2011 and 75.3% (95% CI: 52.0, 98.6) in 2012. Medicaid payment rates averaged approximately 90% of Medicare payment rates throughout the study period.
Dr. Selden is hopeful that stakeholders will use the data his team collected to determine the impetus for the widening gap. He also plans to research whether payment differences affect quality metrics.
“Anytime you’re talking about a trillion dollars, it’s really important when a payment difference opens up of this magnitude,” he adds. “The difference is real … what the policy implications are is for the policy makers to decide.” TH
Visit our website for more information on healthcare payment models.
Spending on care of hospitalized patients is expected to pass $1 trillion in 2015, a new high. Thomas Selden, PhD, of the Agency for Healthcare Research and Quality recently asked where that money is likely to go. The answer: private insurers.
Dr. Selden and his colleagues report in Health Affairs this month that in 2012, private insurers’ payment rates for inpatient hospital stays were approximately 75% greater than Medicare’s payment rates, a sharp increase from the differential of approximately 10% percent during the period of 1996 to 2001. “We need to understand who’s paying what,” Dr. Selden says. “It’s the first step to a better understanding of public policy.”
The report found that “the predicted percentage difference between the rates of private insurers and those of Medicare has increased substantially over time.” In 1996, private insurers paid 106.1% of Medicare payment rates, a payment rate difference of 6.1% (95% CI: -3.2, 15.5). The difference climbed to 64.1% (95% CI: 48.3, 80.0) in 2011 and 75.3% (95% CI: 52.0, 98.6) in 2012. Medicaid payment rates averaged approximately 90% of Medicare payment rates throughout the study period.
Dr. Selden is hopeful that stakeholders will use the data his team collected to determine the impetus for the widening gap. He also plans to research whether payment differences affect quality metrics.
“Anytime you’re talking about a trillion dollars, it’s really important when a payment difference opens up of this magnitude,” he adds. “The difference is real … what the policy implications are is for the policy makers to decide.” TH
Visit our website for more information on healthcare payment models.
New and Noteworthy Information—January 2016
Suicide attempts and recurrent suicide attempts are associated with subsequent epilepsy, suggesting a common underlying biology, according to a study published online ahead of print December 9 in JAMA Psychiatry. The population-based retrospective cohort study in the United Kingdom included patients with incident epilepsy and control patients without a history of epilepsy. For 14,059 patients who later had an onset of epilepsy, versus 56,184 control patients, the risk for a first suicide attempt during the time period before the case patients received a diagnosis of epilepsy was increased 2.9-fold. For 278 case patients who later had an onset of epilepsy, versus 434 control patients, the risk for a recurrent suicide attempt up to and including the day that epilepsy was diagnosed was increased 1.8-fold.
Asthma is associated with an increased risk of new-onset chronic migraine one year later among individuals with episodic migraine, and the highest risk is among people with the greatest number of respiratory symptoms, according to a study published online ahead of print November 19 in Headache. Using the European Community Respiratory Health Survey, researchers defined asthma as a binary variable based on an empirical cut score and developed a Respiratory Symptom Severity Score based on the number of positive responses. This study included 4,446 individuals with episodic migraine in 2008, of whom 17% had asthma. In 2009, new-onset chronic migraine developed in 2.9% of the 2008 episodic migraine cohort, including 5.4% of the asthma subgroup and 2.5% of the non-asthma subgroup.
Vagus nerve stimulation (VNS) paired with rehabilitation is feasible and safe, according to a study published online ahead of print December 8 in Stroke. Twenty-one participants with ischemic stroke more than six months earlier and moderate to severe upper-limb impairment were randomized to VNS plus rehabilitation or rehabilitation alone. Rehabilitation consisted of three two-hour sessions per week for six weeks. There were no serious adverse device effects. One patient had transient vocal cord palsy and dysphagia after implantation. Five patients had minor adverse device effects, including nausea and taste disturbance on the evening of therapy. In the intention-to-treat analysis, the change in Fugl-Meyer Assessment-Upper Extremity scores was not significantly different between groups. In the per-protocol analysis, researchers found a significant difference in change in Fugl-Meyer Assessment-Upper Extremity score between groups.
The Chikungunya virus is a significant cause of CNS disease, according to a study published online ahead of print November 25 in Neurology. During the La Réunion outbreak between September 2005 and June 2006, 57 patients were diagnosed with Chikungunya virus-associated CNS disease, including 24 with Chikungunya virus-associated encephalitis (which corresponded to a cumulative incidence rate of 8.6 per 100,000 people). Patients with encephalitis were observed at both extremes of age categories. The cumulative incidence rates per 100,000 persons were 187 and 37 in patients younger than 1 and patients older than 65, respectively. The case-fatality rate of Chikungunya virus-associated encephalitis was 16.6%, and the proportion of children discharged with persistent disabilities was estimated at between 30% and 45%. Beyond the neonatal period, the clinical presentation and outcomes were less severe in infants than in adults.
Acute stroke is preventable to some extent in most patients, according to a study published online ahead of print December 7 in JAMA Neurology. Researchers evaluated the medical records of 274 consecutive patients discharged with a diagnosis of ischemic stroke between December 2, 2010, and June 11, 2012. Mean patient age was 67.2. Seventy-one patients (25.9%) had scores of 4 or greater on a 10-point scale, indicating that the stroke was highly preventable. Severity of stroke was not related to preventability of stroke. However, 29.6% of patients whose stroke was highly preventable were treated with IV or intra-arterial acute stroke therapy. These treatments were provided for 19.4% of patients with scores of 0, and 14% of patients with scores of 1 to 3.
Alpha-blocker therapy is associated with an increased risk of ischemic stroke during the early initiation period, especially among patients who are not taking other antihypertensive agents, according to a study published online ahead of print December 7 in Canadian Medical Association Journal. Researchers identified 7,502 men ages 50 and older as of 2007 who were incident users of alpha-blockers and who had a diagnosis of ischemic stroke during the study period, which lasted from 2007 to 2009. Investigators examined the incidence of stroke during risk periods before and after alpha-blocker prescription. Compared with the risk in the unexposed period, the risk of ischemic stroke was increased within 21 days after alpha-blocker initiation among all patients in the study population and among patients without concomitant prescriptions.
In patients with mild Alzheimer’s disease, moderate alcohol consumption (ie, two to three units per day) is associated with a significantly lower mortality over a period of 36 months, according to a study published December 11 in BMJ Open. Investigators examined data collected as part of the Danish Alzheimer’s Intervention Study (DAISY). Information about current daily alcohol consumption was obtained from 321 study participants. In all, 8% abstained from drinking alcohol, 71% drank alcohol occasionally, 17% had two to three units per day, and 4% had more than three units per day. Mortality was not significantly different in abstinent patients or in patients with an alcohol consumption of more than three units per day, compared with patients drinking one or less than one unit per day.
Stress is a potentially remediable risk factor for amnestic mild cognitive impairment (aMCI), according to a study published online ahead of print December 10 in Alzheimer Disease & Associated Disorders. The Perceived Stress Scale (PSS) was administered annually in the Einstein Aging Study to participants age 70 and older who were free of aMCI and dementia at baseline PSS administration and who had at least one subsequent annual follow-up. Cox hazard models were used to examine time to aMCI onset, adjusting for covariates. High levels of perceived stress were associated with a 30% greater risk of incident aMCI, independent of covariates. Overall, understanding the effect that perceived stress has on cognition may lead to intervention strategies that prevent the onset of aMCI and Alzheimer’s-related dementia, said the investigators.
Heptachlor epoxide, a pesticide, is associated with higher risk for signs of Parkinson’s disease, according to a study published online ahead of print December 9 in Neurology. For the study, 449 Japanese-American men with an average age of 54 were followed for more than 30 years and until death in the Honolulu-Asia Aging Study. Tests determined whether participants had lost brain cells in the substantia nigra. In 116 brains, researchers also measured the amount of heptachlor epoxide residue, which was present at high levels in Hawaii’s milk in the early 1980s. Nonsmokers who drank more than two cups of milk per day had 40% fewer brain cells in the substantia nigra than people who drank less than two cups of milk per day.
An in vivo florbetapir PET study confirms previous postmortem evidence showing an association between Alzheimer’s disease pathology and gait speed, and provides additional evidence on potential regional effects of brain β-amyloid on motor function, according to data published online ahead of print December 7 in Neurology. Cross-sectional associations between brain β-amyloid, as measured with [18F]florbetapir PET, and gait speed were examined in 128 elderly participants. Researchers found a significant association between β-amyloid in the posterior and anterior putamen, occipital cortex, precuneus, and anterior cingulate and slow gait speed. A multivariate model emphasized the posterior putamen and the precuneus. The β-amyloid burden explained as much as 9% of the variance in gait speed and significantly improved regression models that contained demographic variables, BMI, and APOE status.
Blast-related injury and loss of consciousness are common in traumatic brain injury (TBI) that is sustained while in the military, according to a study published online ahead of print December 15 in Radiology. Study participants were military service members or dependents recruited between August 2009 and August 2014. There were 834 participants with a history of TBI and 42 participants in a control group without TBI. MRIs were performed at 3 T, primarily with three-dimensional volume imaging at voxels smaller than 1 mm3. In all, 84.2% of participants reported one or more blast-related incidents, and 63.0% reported loss of consciousness at the time of injury. White matter T2-weighted hyperintense areas were the most common pathologic finding and were observed in 51.8% of TBI participants.
Researchers have created a transgenic mouse models of familial amyotrophic lateral sclerosis (ALS), according to research published in the December 2 issue of Neuron. To investigate the pathologic role of C9ORF72 in ALS and frontotemporal dementia (FTD), researchers generated a line of mice carrying a bacterial artificial chromosome containing exons one to six of the human C9ORF72 gene with approximately 500 repeats of the GGGGCC motif. The mice showed no overt behavioral phenotype, but recapitulated distinctive histopathologic features of C9ORF72 ALS/FTD, including sense and antisense intranuclear RNA foci and poly(glycine-proline) dipeptide repeat proteins. Using an artificial microRNA that targets human C9ORF72 in cultures of primary cortical neurons from the C9BAC mice, investigators attenuated expression of the C9BAC transgene and the poly(GP) dipeptides.
Oxidative stress may underlie most of the migraine triggers encountered in clinical practice, according to a study published online ahead of print December 7 in Headache. Investigators searched the literature for studies of common migraine triggers published between 1990 and 2014. The reference lists of the resulting articles were examined for further relevant studies. In all cases except pericranial pain, common migraine triggers are capable of generating oxidative stress. Mechanisms include a high rate of energy production by the mitochondria, toxicity or altered membrane properties of the mitochondria, calcium overload and excitotoxicity, neuroinflammation and activation of microglia, and activation of neuronal nicotinamide adenine dinucleotide phosphate oxidase. For some triggers, oxidants also arise as a byproduct of monoamine oxidase or cytochrome P450 processing, or from uncoupling of nitric oxide synthase.
—Kimberly Williams
Suicide attempts and recurrent suicide attempts are associated with subsequent epilepsy, suggesting a common underlying biology, according to a study published online ahead of print December 9 in JAMA Psychiatry. The population-based retrospective cohort study in the United Kingdom included patients with incident epilepsy and control patients without a history of epilepsy. For 14,059 patients who later had an onset of epilepsy, versus 56,184 control patients, the risk for a first suicide attempt during the time period before the case patients received a diagnosis of epilepsy was increased 2.9-fold. For 278 case patients who later had an onset of epilepsy, versus 434 control patients, the risk for a recurrent suicide attempt up to and including the day that epilepsy was diagnosed was increased 1.8-fold.
Asthma is associated with an increased risk of new-onset chronic migraine one year later among individuals with episodic migraine, and the highest risk is among people with the greatest number of respiratory symptoms, according to a study published online ahead of print November 19 in Headache. Using the European Community Respiratory Health Survey, researchers defined asthma as a binary variable based on an empirical cut score and developed a Respiratory Symptom Severity Score based on the number of positive responses. This study included 4,446 individuals with episodic migraine in 2008, of whom 17% had asthma. In 2009, new-onset chronic migraine developed in 2.9% of the 2008 episodic migraine cohort, including 5.4% of the asthma subgroup and 2.5% of the non-asthma subgroup.
Vagus nerve stimulation (VNS) paired with rehabilitation is feasible and safe, according to a study published online ahead of print December 8 in Stroke. Twenty-one participants with ischemic stroke more than six months earlier and moderate to severe upper-limb impairment were randomized to VNS plus rehabilitation or rehabilitation alone. Rehabilitation consisted of three two-hour sessions per week for six weeks. There were no serious adverse device effects. One patient had transient vocal cord palsy and dysphagia after implantation. Five patients had minor adverse device effects, including nausea and taste disturbance on the evening of therapy. In the intention-to-treat analysis, the change in Fugl-Meyer Assessment-Upper Extremity scores was not significantly different between groups. In the per-protocol analysis, researchers found a significant difference in change in Fugl-Meyer Assessment-Upper Extremity score between groups.
The Chikungunya virus is a significant cause of CNS disease, according to a study published online ahead of print November 25 in Neurology. During the La Réunion outbreak between September 2005 and June 2006, 57 patients were diagnosed with Chikungunya virus-associated CNS disease, including 24 with Chikungunya virus-associated encephalitis (which corresponded to a cumulative incidence rate of 8.6 per 100,000 people). Patients with encephalitis were observed at both extremes of age categories. The cumulative incidence rates per 100,000 persons were 187 and 37 in patients younger than 1 and patients older than 65, respectively. The case-fatality rate of Chikungunya virus-associated encephalitis was 16.6%, and the proportion of children discharged with persistent disabilities was estimated at between 30% and 45%. Beyond the neonatal period, the clinical presentation and outcomes were less severe in infants than in adults.
Acute stroke is preventable to some extent in most patients, according to a study published online ahead of print December 7 in JAMA Neurology. Researchers evaluated the medical records of 274 consecutive patients discharged with a diagnosis of ischemic stroke between December 2, 2010, and June 11, 2012. Mean patient age was 67.2. Seventy-one patients (25.9%) had scores of 4 or greater on a 10-point scale, indicating that the stroke was highly preventable. Severity of stroke was not related to preventability of stroke. However, 29.6% of patients whose stroke was highly preventable were treated with IV or intra-arterial acute stroke therapy. These treatments were provided for 19.4% of patients with scores of 0, and 14% of patients with scores of 1 to 3.
Alpha-blocker therapy is associated with an increased risk of ischemic stroke during the early initiation period, especially among patients who are not taking other antihypertensive agents, according to a study published online ahead of print December 7 in Canadian Medical Association Journal. Researchers identified 7,502 men ages 50 and older as of 2007 who were incident users of alpha-blockers and who had a diagnosis of ischemic stroke during the study period, which lasted from 2007 to 2009. Investigators examined the incidence of stroke during risk periods before and after alpha-blocker prescription. Compared with the risk in the unexposed period, the risk of ischemic stroke was increased within 21 days after alpha-blocker initiation among all patients in the study population and among patients without concomitant prescriptions.
In patients with mild Alzheimer’s disease, moderate alcohol consumption (ie, two to three units per day) is associated with a significantly lower mortality over a period of 36 months, according to a study published December 11 in BMJ Open. Investigators examined data collected as part of the Danish Alzheimer’s Intervention Study (DAISY). Information about current daily alcohol consumption was obtained from 321 study participants. In all, 8% abstained from drinking alcohol, 71% drank alcohol occasionally, 17% had two to three units per day, and 4% had more than three units per day. Mortality was not significantly different in abstinent patients or in patients with an alcohol consumption of more than three units per day, compared with patients drinking one or less than one unit per day.
Stress is a potentially remediable risk factor for amnestic mild cognitive impairment (aMCI), according to a study published online ahead of print December 10 in Alzheimer Disease & Associated Disorders. The Perceived Stress Scale (PSS) was administered annually in the Einstein Aging Study to participants age 70 and older who were free of aMCI and dementia at baseline PSS administration and who had at least one subsequent annual follow-up. Cox hazard models were used to examine time to aMCI onset, adjusting for covariates. High levels of perceived stress were associated with a 30% greater risk of incident aMCI, independent of covariates. Overall, understanding the effect that perceived stress has on cognition may lead to intervention strategies that prevent the onset of aMCI and Alzheimer’s-related dementia, said the investigators.
Heptachlor epoxide, a pesticide, is associated with higher risk for signs of Parkinson’s disease, according to a study published online ahead of print December 9 in Neurology. For the study, 449 Japanese-American men with an average age of 54 were followed for more than 30 years and until death in the Honolulu-Asia Aging Study. Tests determined whether participants had lost brain cells in the substantia nigra. In 116 brains, researchers also measured the amount of heptachlor epoxide residue, which was present at high levels in Hawaii’s milk in the early 1980s. Nonsmokers who drank more than two cups of milk per day had 40% fewer brain cells in the substantia nigra than people who drank less than two cups of milk per day.
An in vivo florbetapir PET study confirms previous postmortem evidence showing an association between Alzheimer’s disease pathology and gait speed, and provides additional evidence on potential regional effects of brain β-amyloid on motor function, according to data published online ahead of print December 7 in Neurology. Cross-sectional associations between brain β-amyloid, as measured with [18F]florbetapir PET, and gait speed were examined in 128 elderly participants. Researchers found a significant association between β-amyloid in the posterior and anterior putamen, occipital cortex, precuneus, and anterior cingulate and slow gait speed. A multivariate model emphasized the posterior putamen and the precuneus. The β-amyloid burden explained as much as 9% of the variance in gait speed and significantly improved regression models that contained demographic variables, BMI, and APOE status.
Blast-related injury and loss of consciousness are common in traumatic brain injury (TBI) that is sustained while in the military, according to a study published online ahead of print December 15 in Radiology. Study participants were military service members or dependents recruited between August 2009 and August 2014. There were 834 participants with a history of TBI and 42 participants in a control group without TBI. MRIs were performed at 3 T, primarily with three-dimensional volume imaging at voxels smaller than 1 mm3. In all, 84.2% of participants reported one or more blast-related incidents, and 63.0% reported loss of consciousness at the time of injury. White matter T2-weighted hyperintense areas were the most common pathologic finding and were observed in 51.8% of TBI participants.
Researchers have created a transgenic mouse models of familial amyotrophic lateral sclerosis (ALS), according to research published in the December 2 issue of Neuron. To investigate the pathologic role of C9ORF72 in ALS and frontotemporal dementia (FTD), researchers generated a line of mice carrying a bacterial artificial chromosome containing exons one to six of the human C9ORF72 gene with approximately 500 repeats of the GGGGCC motif. The mice showed no overt behavioral phenotype, but recapitulated distinctive histopathologic features of C9ORF72 ALS/FTD, including sense and antisense intranuclear RNA foci and poly(glycine-proline) dipeptide repeat proteins. Using an artificial microRNA that targets human C9ORF72 in cultures of primary cortical neurons from the C9BAC mice, investigators attenuated expression of the C9BAC transgene and the poly(GP) dipeptides.
Oxidative stress may underlie most of the migraine triggers encountered in clinical practice, according to a study published online ahead of print December 7 in Headache. Investigators searched the literature for studies of common migraine triggers published between 1990 and 2014. The reference lists of the resulting articles were examined for further relevant studies. In all cases except pericranial pain, common migraine triggers are capable of generating oxidative stress. Mechanisms include a high rate of energy production by the mitochondria, toxicity or altered membrane properties of the mitochondria, calcium overload and excitotoxicity, neuroinflammation and activation of microglia, and activation of neuronal nicotinamide adenine dinucleotide phosphate oxidase. For some triggers, oxidants also arise as a byproduct of monoamine oxidase or cytochrome P450 processing, or from uncoupling of nitric oxide synthase.
—Kimberly Williams
Suicide attempts and recurrent suicide attempts are associated with subsequent epilepsy, suggesting a common underlying biology, according to a study published online ahead of print December 9 in JAMA Psychiatry. The population-based retrospective cohort study in the United Kingdom included patients with incident epilepsy and control patients without a history of epilepsy. For 14,059 patients who later had an onset of epilepsy, versus 56,184 control patients, the risk for a first suicide attempt during the time period before the case patients received a diagnosis of epilepsy was increased 2.9-fold. For 278 case patients who later had an onset of epilepsy, versus 434 control patients, the risk for a recurrent suicide attempt up to and including the day that epilepsy was diagnosed was increased 1.8-fold.
Asthma is associated with an increased risk of new-onset chronic migraine one year later among individuals with episodic migraine, and the highest risk is among people with the greatest number of respiratory symptoms, according to a study published online ahead of print November 19 in Headache. Using the European Community Respiratory Health Survey, researchers defined asthma as a binary variable based on an empirical cut score and developed a Respiratory Symptom Severity Score based on the number of positive responses. This study included 4,446 individuals with episodic migraine in 2008, of whom 17% had asthma. In 2009, new-onset chronic migraine developed in 2.9% of the 2008 episodic migraine cohort, including 5.4% of the asthma subgroup and 2.5% of the non-asthma subgroup.
Vagus nerve stimulation (VNS) paired with rehabilitation is feasible and safe, according to a study published online ahead of print December 8 in Stroke. Twenty-one participants with ischemic stroke more than six months earlier and moderate to severe upper-limb impairment were randomized to VNS plus rehabilitation or rehabilitation alone. Rehabilitation consisted of three two-hour sessions per week for six weeks. There were no serious adverse device effects. One patient had transient vocal cord palsy and dysphagia after implantation. Five patients had minor adverse device effects, including nausea and taste disturbance on the evening of therapy. In the intention-to-treat analysis, the change in Fugl-Meyer Assessment-Upper Extremity scores was not significantly different between groups. In the per-protocol analysis, researchers found a significant difference in change in Fugl-Meyer Assessment-Upper Extremity score between groups.
The Chikungunya virus is a significant cause of CNS disease, according to a study published online ahead of print November 25 in Neurology. During the La Réunion outbreak between September 2005 and June 2006, 57 patients were diagnosed with Chikungunya virus-associated CNS disease, including 24 with Chikungunya virus-associated encephalitis (which corresponded to a cumulative incidence rate of 8.6 per 100,000 people). Patients with encephalitis were observed at both extremes of age categories. The cumulative incidence rates per 100,000 persons were 187 and 37 in patients younger than 1 and patients older than 65, respectively. The case-fatality rate of Chikungunya virus-associated encephalitis was 16.6%, and the proportion of children discharged with persistent disabilities was estimated at between 30% and 45%. Beyond the neonatal period, the clinical presentation and outcomes were less severe in infants than in adults.
Acute stroke is preventable to some extent in most patients, according to a study published online ahead of print December 7 in JAMA Neurology. Researchers evaluated the medical records of 274 consecutive patients discharged with a diagnosis of ischemic stroke between December 2, 2010, and June 11, 2012. Mean patient age was 67.2. Seventy-one patients (25.9%) had scores of 4 or greater on a 10-point scale, indicating that the stroke was highly preventable. Severity of stroke was not related to preventability of stroke. However, 29.6% of patients whose stroke was highly preventable were treated with IV or intra-arterial acute stroke therapy. These treatments were provided for 19.4% of patients with scores of 0, and 14% of patients with scores of 1 to 3.
Alpha-blocker therapy is associated with an increased risk of ischemic stroke during the early initiation period, especially among patients who are not taking other antihypertensive agents, according to a study published online ahead of print December 7 in Canadian Medical Association Journal. Researchers identified 7,502 men ages 50 and older as of 2007 who were incident users of alpha-blockers and who had a diagnosis of ischemic stroke during the study period, which lasted from 2007 to 2009. Investigators examined the incidence of stroke during risk periods before and after alpha-blocker prescription. Compared with the risk in the unexposed period, the risk of ischemic stroke was increased within 21 days after alpha-blocker initiation among all patients in the study population and among patients without concomitant prescriptions.
In patients with mild Alzheimer’s disease, moderate alcohol consumption (ie, two to three units per day) is associated with a significantly lower mortality over a period of 36 months, according to a study published December 11 in BMJ Open. Investigators examined data collected as part of the Danish Alzheimer’s Intervention Study (DAISY). Information about current daily alcohol consumption was obtained from 321 study participants. In all, 8% abstained from drinking alcohol, 71% drank alcohol occasionally, 17% had two to three units per day, and 4% had more than three units per day. Mortality was not significantly different in abstinent patients or in patients with an alcohol consumption of more than three units per day, compared with patients drinking one or less than one unit per day.
Stress is a potentially remediable risk factor for amnestic mild cognitive impairment (aMCI), according to a study published online ahead of print December 10 in Alzheimer Disease & Associated Disorders. The Perceived Stress Scale (PSS) was administered annually in the Einstein Aging Study to participants age 70 and older who were free of aMCI and dementia at baseline PSS administration and who had at least one subsequent annual follow-up. Cox hazard models were used to examine time to aMCI onset, adjusting for covariates. High levels of perceived stress were associated with a 30% greater risk of incident aMCI, independent of covariates. Overall, understanding the effect that perceived stress has on cognition may lead to intervention strategies that prevent the onset of aMCI and Alzheimer’s-related dementia, said the investigators.
Heptachlor epoxide, a pesticide, is associated with higher risk for signs of Parkinson’s disease, according to a study published online ahead of print December 9 in Neurology. For the study, 449 Japanese-American men with an average age of 54 were followed for more than 30 years and until death in the Honolulu-Asia Aging Study. Tests determined whether participants had lost brain cells in the substantia nigra. In 116 brains, researchers also measured the amount of heptachlor epoxide residue, which was present at high levels in Hawaii’s milk in the early 1980s. Nonsmokers who drank more than two cups of milk per day had 40% fewer brain cells in the substantia nigra than people who drank less than two cups of milk per day.
An in vivo florbetapir PET study confirms previous postmortem evidence showing an association between Alzheimer’s disease pathology and gait speed, and provides additional evidence on potential regional effects of brain β-amyloid on motor function, according to data published online ahead of print December 7 in Neurology. Cross-sectional associations between brain β-amyloid, as measured with [18F]florbetapir PET, and gait speed were examined in 128 elderly participants. Researchers found a significant association between β-amyloid in the posterior and anterior putamen, occipital cortex, precuneus, and anterior cingulate and slow gait speed. A multivariate model emphasized the posterior putamen and the precuneus. The β-amyloid burden explained as much as 9% of the variance in gait speed and significantly improved regression models that contained demographic variables, BMI, and APOE status.
Blast-related injury and loss of consciousness are common in traumatic brain injury (TBI) that is sustained while in the military, according to a study published online ahead of print December 15 in Radiology. Study participants were military service members or dependents recruited between August 2009 and August 2014. There were 834 participants with a history of TBI and 42 participants in a control group without TBI. MRIs were performed at 3 T, primarily with three-dimensional volume imaging at voxels smaller than 1 mm3. In all, 84.2% of participants reported one or more blast-related incidents, and 63.0% reported loss of consciousness at the time of injury. White matter T2-weighted hyperintense areas were the most common pathologic finding and were observed in 51.8% of TBI participants.
Researchers have created a transgenic mouse models of familial amyotrophic lateral sclerosis (ALS), according to research published in the December 2 issue of Neuron. To investigate the pathologic role of C9ORF72 in ALS and frontotemporal dementia (FTD), researchers generated a line of mice carrying a bacterial artificial chromosome containing exons one to six of the human C9ORF72 gene with approximately 500 repeats of the GGGGCC motif. The mice showed no overt behavioral phenotype, but recapitulated distinctive histopathologic features of C9ORF72 ALS/FTD, including sense and antisense intranuclear RNA foci and poly(glycine-proline) dipeptide repeat proteins. Using an artificial microRNA that targets human C9ORF72 in cultures of primary cortical neurons from the C9BAC mice, investigators attenuated expression of the C9BAC transgene and the poly(GP) dipeptides.
Oxidative stress may underlie most of the migraine triggers encountered in clinical practice, according to a study published online ahead of print December 7 in Headache. Investigators searched the literature for studies of common migraine triggers published between 1990 and 2014. The reference lists of the resulting articles were examined for further relevant studies. In all cases except pericranial pain, common migraine triggers are capable of generating oxidative stress. Mechanisms include a high rate of energy production by the mitochondria, toxicity or altered membrane properties of the mitochondria, calcium overload and excitotoxicity, neuroinflammation and activation of microglia, and activation of neuronal nicotinamide adenine dinucleotide phosphate oxidase. For some triggers, oxidants also arise as a byproduct of monoamine oxidase or cytochrome P450 processing, or from uncoupling of nitric oxide synthase.
—Kimberly Williams
91% who overdose on opioids continue to receive opioid prescriptions
Almost all patients who had nonfatal overdoses while taking long-term opioids for noncancer pain continued to receive opioid prescriptions, usually from the same physicians, in a nationwide cohort study published online Dec. 28 in Annals of Internal Medicine.
Clinical guidelines specify that adverse events related to the misuse of opioids are clear indications to discontinue long-term opioid therapy. But patterns of prescribing after opioid overdoses are not monitored. To examine prescribing trends following nonfatal opioid overdoses, researchers analyzed information in a database of inpatient, outpatient, and pharmacy claims from a large U.S. health insurer covering all 50 states.
They focused on 2,848 insured adults enrolled in 2000-2012 who received hospital or ED treatment for a prescription opioid overdose and were followed in the database for a median of 15 months. The prescribed drugs included codeine, dihydrocodeine, meperidine, morphine, oxycodone, hydrocodone, hydromorphone, fentanyl, oxymorphone, propoxyphene, methadone, tramadol, and levorphanol, said Dr. Marc R. Larochelle of Boston Medical Center and his associates.
A total of 2,597 of these patients (91%) continued to receive opioid prescriptions after their overdose. The primary prescriber was the same person before and after the overdose in 1,198 cases (61%). Two hundred twelve of these patients (7%) had another opioid overdose during follow-up. The likelihood of a second overdose was much higher for patients taking the highest doses of opioids (100 mg or more morphine-equivalent dosage per day), with hazard ratios of 1.13 for patients taking low doses of opioids, 1.89 for those taking mid-range doses, and 2.57 for those taking high doses.
“We could not determine the reason for the treatment patterns after the overdose; however, some prescribers may have been unaware that the opioid overdose had occurred” because there are no procedures in place to ensure provider notification in such cases. Newly introduced prescription monitoring programs may facilitate such communication, but a more rigorous approach would mandate that all overdoses be reported to public health departments, which would then notify providers and pharmacies, and perhaps secure patient referral to substance abuse treatment programs, the investigators said (Ann Intern Med. 2015 Dec 28. doi: 10.7326/M15-0038). It is possible that some overdoses stemmed from therapeutic error rather than opioid misuse, and that providers felt the risk-benefit ratio justified continued opioid treatment. But it also is likely that many providers simply did not have the knowledge and skills to identify and treat opioid misuse, they added.
“Simply eliminating opioid prescribing for patients who had an overdose is not sufficient. … because some [patients] may turn to diverted or illicit opioids. Rather, efforts to identify and treat substance use disorders in these patients are needed,” Dr. Larochelle and his associates said.
Overall, the study findings indicate that nonfatal overdoses provide a meaningful opportunity to improve the safety of opioid prescribing, but that most prescribers at present are missing this opportunity.
It’s tempting to attribute these astonishing findings to poor medical care, bad medical decisions, or sloppy prescribing, but the problem extends well beyond individual prescribers’ practices. These prescribing behaviors take place within a context in which substantial, even deadly, mistakes are inevitable.
Clinicians must be notified when their patients overdose and must know how to act on that notification. They must be taught to recognize pain and addiction as chronic diseases that require team approaches. They must learn how to taper opioid dosages appropriately, how to use buprenorphine, and what other resources in their communities are reliable. Health systems must give physicians the tools and the time they need to identify and coordinate care for affected patients, and would do well to connect overdose patients directly to addiction services at hospital discharge.
This approach would turn an opioid overdose from a devastating event into an opportunity for hope.
Dr. Jessica Gregg is at Central City Concern, a nonprofit agency serving adults and families impacted by homelessness, poverty, and addiction in Portland, Ore. She reported having no relevant financial conflicts of interest. Dr. Gregg made these remarks in an editorial accompanying Dr. Larochelle’s report (Ann Intern Med. 2015 Dec 28. doi: 10.7326/M152687).
It’s tempting to attribute these astonishing findings to poor medical care, bad medical decisions, or sloppy prescribing, but the problem extends well beyond individual prescribers’ practices. These prescribing behaviors take place within a context in which substantial, even deadly, mistakes are inevitable.
Clinicians must be notified when their patients overdose and must know how to act on that notification. They must be taught to recognize pain and addiction as chronic diseases that require team approaches. They must learn how to taper opioid dosages appropriately, how to use buprenorphine, and what other resources in their communities are reliable. Health systems must give physicians the tools and the time they need to identify and coordinate care for affected patients, and would do well to connect overdose patients directly to addiction services at hospital discharge.
This approach would turn an opioid overdose from a devastating event into an opportunity for hope.
Dr. Jessica Gregg is at Central City Concern, a nonprofit agency serving adults and families impacted by homelessness, poverty, and addiction in Portland, Ore. She reported having no relevant financial conflicts of interest. Dr. Gregg made these remarks in an editorial accompanying Dr. Larochelle’s report (Ann Intern Med. 2015 Dec 28. doi: 10.7326/M152687).
It’s tempting to attribute these astonishing findings to poor medical care, bad medical decisions, or sloppy prescribing, but the problem extends well beyond individual prescribers’ practices. These prescribing behaviors take place within a context in which substantial, even deadly, mistakes are inevitable.
Clinicians must be notified when their patients overdose and must know how to act on that notification. They must be taught to recognize pain and addiction as chronic diseases that require team approaches. They must learn how to taper opioid dosages appropriately, how to use buprenorphine, and what other resources in their communities are reliable. Health systems must give physicians the tools and the time they need to identify and coordinate care for affected patients, and would do well to connect overdose patients directly to addiction services at hospital discharge.
This approach would turn an opioid overdose from a devastating event into an opportunity for hope.
Dr. Jessica Gregg is at Central City Concern, a nonprofit agency serving adults and families impacted by homelessness, poverty, and addiction in Portland, Ore. She reported having no relevant financial conflicts of interest. Dr. Gregg made these remarks in an editorial accompanying Dr. Larochelle’s report (Ann Intern Med. 2015 Dec 28. doi: 10.7326/M152687).
Almost all patients who had nonfatal overdoses while taking long-term opioids for noncancer pain continued to receive opioid prescriptions, usually from the same physicians, in a nationwide cohort study published online Dec. 28 in Annals of Internal Medicine.
Clinical guidelines specify that adverse events related to the misuse of opioids are clear indications to discontinue long-term opioid therapy. But patterns of prescribing after opioid overdoses are not monitored. To examine prescribing trends following nonfatal opioid overdoses, researchers analyzed information in a database of inpatient, outpatient, and pharmacy claims from a large U.S. health insurer covering all 50 states.
They focused on 2,848 insured adults enrolled in 2000-2012 who received hospital or ED treatment for a prescription opioid overdose and were followed in the database for a median of 15 months. The prescribed drugs included codeine, dihydrocodeine, meperidine, morphine, oxycodone, hydrocodone, hydromorphone, fentanyl, oxymorphone, propoxyphene, methadone, tramadol, and levorphanol, said Dr. Marc R. Larochelle of Boston Medical Center and his associates.
A total of 2,597 of these patients (91%) continued to receive opioid prescriptions after their overdose. The primary prescriber was the same person before and after the overdose in 1,198 cases (61%). Two hundred twelve of these patients (7%) had another opioid overdose during follow-up. The likelihood of a second overdose was much higher for patients taking the highest doses of opioids (100 mg or more morphine-equivalent dosage per day), with hazard ratios of 1.13 for patients taking low doses of opioids, 1.89 for those taking mid-range doses, and 2.57 for those taking high doses.
“We could not determine the reason for the treatment patterns after the overdose; however, some prescribers may have been unaware that the opioid overdose had occurred” because there are no procedures in place to ensure provider notification in such cases. Newly introduced prescription monitoring programs may facilitate such communication, but a more rigorous approach would mandate that all overdoses be reported to public health departments, which would then notify providers and pharmacies, and perhaps secure patient referral to substance abuse treatment programs, the investigators said (Ann Intern Med. 2015 Dec 28. doi: 10.7326/M15-0038). It is possible that some overdoses stemmed from therapeutic error rather than opioid misuse, and that providers felt the risk-benefit ratio justified continued opioid treatment. But it also is likely that many providers simply did not have the knowledge and skills to identify and treat opioid misuse, they added.
“Simply eliminating opioid prescribing for patients who had an overdose is not sufficient. … because some [patients] may turn to diverted or illicit opioids. Rather, efforts to identify and treat substance use disorders in these patients are needed,” Dr. Larochelle and his associates said.
Overall, the study findings indicate that nonfatal overdoses provide a meaningful opportunity to improve the safety of opioid prescribing, but that most prescribers at present are missing this opportunity.
Almost all patients who had nonfatal overdoses while taking long-term opioids for noncancer pain continued to receive opioid prescriptions, usually from the same physicians, in a nationwide cohort study published online Dec. 28 in Annals of Internal Medicine.
Clinical guidelines specify that adverse events related to the misuse of opioids are clear indications to discontinue long-term opioid therapy. But patterns of prescribing after opioid overdoses are not monitored. To examine prescribing trends following nonfatal opioid overdoses, researchers analyzed information in a database of inpatient, outpatient, and pharmacy claims from a large U.S. health insurer covering all 50 states.
They focused on 2,848 insured adults enrolled in 2000-2012 who received hospital or ED treatment for a prescription opioid overdose and were followed in the database for a median of 15 months. The prescribed drugs included codeine, dihydrocodeine, meperidine, morphine, oxycodone, hydrocodone, hydromorphone, fentanyl, oxymorphone, propoxyphene, methadone, tramadol, and levorphanol, said Dr. Marc R. Larochelle of Boston Medical Center and his associates.
A total of 2,597 of these patients (91%) continued to receive opioid prescriptions after their overdose. The primary prescriber was the same person before and after the overdose in 1,198 cases (61%). Two hundred twelve of these patients (7%) had another opioid overdose during follow-up. The likelihood of a second overdose was much higher for patients taking the highest doses of opioids (100 mg or more morphine-equivalent dosage per day), with hazard ratios of 1.13 for patients taking low doses of opioids, 1.89 for those taking mid-range doses, and 2.57 for those taking high doses.
“We could not determine the reason for the treatment patterns after the overdose; however, some prescribers may have been unaware that the opioid overdose had occurred” because there are no procedures in place to ensure provider notification in such cases. Newly introduced prescription monitoring programs may facilitate such communication, but a more rigorous approach would mandate that all overdoses be reported to public health departments, which would then notify providers and pharmacies, and perhaps secure patient referral to substance abuse treatment programs, the investigators said (Ann Intern Med. 2015 Dec 28. doi: 10.7326/M15-0038). It is possible that some overdoses stemmed from therapeutic error rather than opioid misuse, and that providers felt the risk-benefit ratio justified continued opioid treatment. But it also is likely that many providers simply did not have the knowledge and skills to identify and treat opioid misuse, they added.
“Simply eliminating opioid prescribing for patients who had an overdose is not sufficient. … because some [patients] may turn to diverted or illicit opioids. Rather, efforts to identify and treat substance use disorders in these patients are needed,” Dr. Larochelle and his associates said.
Overall, the study findings indicate that nonfatal overdoses provide a meaningful opportunity to improve the safety of opioid prescribing, but that most prescribers at present are missing this opportunity.
FROM ANNALS OF INTERNAL MEDICINE
Key clinical point: Almost all patients treated at EDs for nonfatal opioid overdose continue to receive opioid prescriptions.
Major finding: 2,597 patients (91%) continued to receive opioid prescriptions after their overdose; the primary prescriber was the same person before and after the overdose in 1,198 cases (61%).
Data source: A retrospective cohort study involving 2,848 adults taking opioids for noncancer pain who overdosed and were followed for up to 2 years.
Disclosures: This study was funded by the U.S. Health Resources and Services Administration, which had no role in the design or conduct of the study. Dr. Larochelle reported also receiving support from the Ryoichi Sasakawa Fellowship Fund and Harvard Pilgrim Health Care Institute. His associates reported having no relevant financial disclosures.
My mental health hopes for 2016
The world has become a very complicated place. I suppose it always was, but today’s complexities are new and different and constantly changing with technology. It’s easy to think of ways our professional lives could be simpler and the treatment of our patients more attentive. Reflections, predictions, and all things “Happy New Year” tend to come as lists, and I don’t want to break with tradition. A list of my mental health hopes for 2016 follows:
1. I hope … that as our legislators rethink the Affordable Care Act (Obamacare), they don’t throw out the baby with the bath water, and the gains that were made remain in place. Among the good that’s been done has been the ability to keep adult children on a family policy to age 26, the elimination of “preexisting conditions” as exclusions from health insurance coverage, and the overall expansion of who is able to get some form of health insurance.
2. I hope … psychiatric treatment comes to be about so much more than medications, and that psychiatry resumes the priority of listening to patients on so many different levels. People are about more than checklists of symptoms and side effects, and their problems occur within the context of their lives. This is often more than a psychiatrist can piece together when seeing four or five patients an hour. In addition, there has been the added burden of attending to screens, data collection that is irrelevant to treatment, and paperwork burdens. I’d like to see the end of “meaningful use,” irrelevant maintenance of certification exams, and demands to use electronic medical records that divert psychiatrist time and attention without improving patient care. Technology should facilitate excellent care, not detract from it. And in my continued hope for our Internet-based world, I’ll wish for efficiency and innovation in how we use technology to learn, to communicate with one another, and to offer care to our patients, without compromising patient privacy.
3. I hope … discussions about involuntary treatment come to be about reducing the suffering of our patients, and not about preventing mass murders. It’s an expectation psychiatry simply cannot meet.
4. I hope … our legislators will come to understand that “mental illness” does not cause gun violence, and that a better predictor of violent behavior is a past history of violence, substance abuse, anger, and impulse control problems, and not the presence of a particular diagnosis or the catchall category of mental illness.
5. I hope ... we stop political discussions calling for an end to stigma while simultaneously stigmatizing those with psychiatric disorders.
6. I hope … that insurers and pharmacy benefit managers are required to limit preauthorization requirements to the most expensive and controversial forms of care. They must be required to standardize, simplify, and streamline any preauthorization procedures, and to be held to a level of accountability for the care that is denied.
7. I hope … that Medicare and Medicaid become user friendly entities that are easy to navigate and welcoming to psychiatrists so that our patients with limited incomes have access to treatment.
8. I hope … we come to appreciate the need for “housing-first” options for people who live and sleep on our streets. In a civilized society, this is a disgrace, and it benefits no one. It shouldn’t matter whether people with nowhere else to sleep do so because they are mentally ill, addicted, or simply impoverished: We need to provide better housing options. It is the humane thing to do, and it is more cost effective than paying for the correctional and medical care that result from homelessness. For those who are homeless because of untreated mental illness, it is so much easier to take medications when they have a shelf to put the bottles on.
9. I hope … for an understanding that our world is not black and white: We don’t neatly divide into those who are mentally ill and those who are not any more than we neatly divide into those who are good guys and those who are bad guys. People are complex, and mentally healthy people can behave in very disordered ways given the wrong set of circumstances. And, quite obviously, we all have a bit of both the good guy and the bad guy in us, and to believe otherwise is to be naive.
10. I hope … for both mental health and all of medicine, that all our treatments become available to all of our patients. It doesn’t matter how wonderful new treatments are if they are available only to those patients who are wealthy enough to pay for their own care, or who have Cadillac insurance policies that will reimburse for a given therapy.
Convention (or at least David Letterman) would suggest that I stop at 10 hopes. I never was one to follow arbitrary rules, so please allow me to express more mental health wishes for 2016.
11. I hope … we find some way of addressing the shortage of psychiatrists and services. We need to make it easier for patients to link with psychiatrists they find to be helpful. Our current system is difficult to negotiate for those who are healthy and have resources. Linking to care is nearly impossible for someone with limited resources who is psychotic or severely depressed.
12. And finally, I hope ... for better treatments for our patients, ones that cure mental illnesses without causing side effects, adverse reactions, or trade-offs that sometime make the cures worse than the diseases.
Thank you for bearing with me. I’m sure you have your own mental health hopes for the coming year, and perhaps some of our wishes might even come true. Wishing you a Healthy and Happy New Year!
Dr. Miller is a coauthor of “Shrink Rap: Three Psychiatrists Explain Their Work” (Baltimore: Johns Hopkins University Press, 2011).
The world has become a very complicated place. I suppose it always was, but today’s complexities are new and different and constantly changing with technology. It’s easy to think of ways our professional lives could be simpler and the treatment of our patients more attentive. Reflections, predictions, and all things “Happy New Year” tend to come as lists, and I don’t want to break with tradition. A list of my mental health hopes for 2016 follows:
1. I hope … that as our legislators rethink the Affordable Care Act (Obamacare), they don’t throw out the baby with the bath water, and the gains that were made remain in place. Among the good that’s been done has been the ability to keep adult children on a family policy to age 26, the elimination of “preexisting conditions” as exclusions from health insurance coverage, and the overall expansion of who is able to get some form of health insurance.
2. I hope … psychiatric treatment comes to be about so much more than medications, and that psychiatry resumes the priority of listening to patients on so many different levels. People are about more than checklists of symptoms and side effects, and their problems occur within the context of their lives. This is often more than a psychiatrist can piece together when seeing four or five patients an hour. In addition, there has been the added burden of attending to screens, data collection that is irrelevant to treatment, and paperwork burdens. I’d like to see the end of “meaningful use,” irrelevant maintenance of certification exams, and demands to use electronic medical records that divert psychiatrist time and attention without improving patient care. Technology should facilitate excellent care, not detract from it. And in my continued hope for our Internet-based world, I’ll wish for efficiency and innovation in how we use technology to learn, to communicate with one another, and to offer care to our patients, without compromising patient privacy.
3. I hope … discussions about involuntary treatment come to be about reducing the suffering of our patients, and not about preventing mass murders. It’s an expectation psychiatry simply cannot meet.
4. I hope … our legislators will come to understand that “mental illness” does not cause gun violence, and that a better predictor of violent behavior is a past history of violence, substance abuse, anger, and impulse control problems, and not the presence of a particular diagnosis or the catchall category of mental illness.
5. I hope ... we stop political discussions calling for an end to stigma while simultaneously stigmatizing those with psychiatric disorders.
6. I hope … that insurers and pharmacy benefit managers are required to limit preauthorization requirements to the most expensive and controversial forms of care. They must be required to standardize, simplify, and streamline any preauthorization procedures, and to be held to a level of accountability for the care that is denied.
7. I hope … that Medicare and Medicaid become user friendly entities that are easy to navigate and welcoming to psychiatrists so that our patients with limited incomes have access to treatment.
8. I hope … we come to appreciate the need for “housing-first” options for people who live and sleep on our streets. In a civilized society, this is a disgrace, and it benefits no one. It shouldn’t matter whether people with nowhere else to sleep do so because they are mentally ill, addicted, or simply impoverished: We need to provide better housing options. It is the humane thing to do, and it is more cost effective than paying for the correctional and medical care that result from homelessness. For those who are homeless because of untreated mental illness, it is so much easier to take medications when they have a shelf to put the bottles on.
9. I hope … for an understanding that our world is not black and white: We don’t neatly divide into those who are mentally ill and those who are not any more than we neatly divide into those who are good guys and those who are bad guys. People are complex, and mentally healthy people can behave in very disordered ways given the wrong set of circumstances. And, quite obviously, we all have a bit of both the good guy and the bad guy in us, and to believe otherwise is to be naive.
10. I hope … for both mental health and all of medicine, that all our treatments become available to all of our patients. It doesn’t matter how wonderful new treatments are if they are available only to those patients who are wealthy enough to pay for their own care, or who have Cadillac insurance policies that will reimburse for a given therapy.
Convention (or at least David Letterman) would suggest that I stop at 10 hopes. I never was one to follow arbitrary rules, so please allow me to express more mental health wishes for 2016.
11. I hope … we find some way of addressing the shortage of psychiatrists and services. We need to make it easier for patients to link with psychiatrists they find to be helpful. Our current system is difficult to negotiate for those who are healthy and have resources. Linking to care is nearly impossible for someone with limited resources who is psychotic or severely depressed.
12. And finally, I hope ... for better treatments for our patients, ones that cure mental illnesses without causing side effects, adverse reactions, or trade-offs that sometime make the cures worse than the diseases.
Thank you for bearing with me. I’m sure you have your own mental health hopes for the coming year, and perhaps some of our wishes might even come true. Wishing you a Healthy and Happy New Year!
Dr. Miller is a coauthor of “Shrink Rap: Three Psychiatrists Explain Their Work” (Baltimore: Johns Hopkins University Press, 2011).
The world has become a very complicated place. I suppose it always was, but today’s complexities are new and different and constantly changing with technology. It’s easy to think of ways our professional lives could be simpler and the treatment of our patients more attentive. Reflections, predictions, and all things “Happy New Year” tend to come as lists, and I don’t want to break with tradition. A list of my mental health hopes for 2016 follows:
1. I hope … that as our legislators rethink the Affordable Care Act (Obamacare), they don’t throw out the baby with the bath water, and the gains that were made remain in place. Among the good that’s been done has been the ability to keep adult children on a family policy to age 26, the elimination of “preexisting conditions” as exclusions from health insurance coverage, and the overall expansion of who is able to get some form of health insurance.
2. I hope … psychiatric treatment comes to be about so much more than medications, and that psychiatry resumes the priority of listening to patients on so many different levels. People are about more than checklists of symptoms and side effects, and their problems occur within the context of their lives. This is often more than a psychiatrist can piece together when seeing four or five patients an hour. In addition, there has been the added burden of attending to screens, data collection that is irrelevant to treatment, and paperwork burdens. I’d like to see the end of “meaningful use,” irrelevant maintenance of certification exams, and demands to use electronic medical records that divert psychiatrist time and attention without improving patient care. Technology should facilitate excellent care, not detract from it. And in my continued hope for our Internet-based world, I’ll wish for efficiency and innovation in how we use technology to learn, to communicate with one another, and to offer care to our patients, without compromising patient privacy.
3. I hope … discussions about involuntary treatment come to be about reducing the suffering of our patients, and not about preventing mass murders. It’s an expectation psychiatry simply cannot meet.
4. I hope … our legislators will come to understand that “mental illness” does not cause gun violence, and that a better predictor of violent behavior is a past history of violence, substance abuse, anger, and impulse control problems, and not the presence of a particular diagnosis or the catchall category of mental illness.
5. I hope ... we stop political discussions calling for an end to stigma while simultaneously stigmatizing those with psychiatric disorders.
6. I hope … that insurers and pharmacy benefit managers are required to limit preauthorization requirements to the most expensive and controversial forms of care. They must be required to standardize, simplify, and streamline any preauthorization procedures, and to be held to a level of accountability for the care that is denied.
7. I hope … that Medicare and Medicaid become user friendly entities that are easy to navigate and welcoming to psychiatrists so that our patients with limited incomes have access to treatment.
8. I hope … we come to appreciate the need for “housing-first” options for people who live and sleep on our streets. In a civilized society, this is a disgrace, and it benefits no one. It shouldn’t matter whether people with nowhere else to sleep do so because they are mentally ill, addicted, or simply impoverished: We need to provide better housing options. It is the humane thing to do, and it is more cost effective than paying for the correctional and medical care that result from homelessness. For those who are homeless because of untreated mental illness, it is so much easier to take medications when they have a shelf to put the bottles on.
9. I hope … for an understanding that our world is not black and white: We don’t neatly divide into those who are mentally ill and those who are not any more than we neatly divide into those who are good guys and those who are bad guys. People are complex, and mentally healthy people can behave in very disordered ways given the wrong set of circumstances. And, quite obviously, we all have a bit of both the good guy and the bad guy in us, and to believe otherwise is to be naive.
10. I hope … for both mental health and all of medicine, that all our treatments become available to all of our patients. It doesn’t matter how wonderful new treatments are if they are available only to those patients who are wealthy enough to pay for their own care, or who have Cadillac insurance policies that will reimburse for a given therapy.
Convention (or at least David Letterman) would suggest that I stop at 10 hopes. I never was one to follow arbitrary rules, so please allow me to express more mental health wishes for 2016.
11. I hope … we find some way of addressing the shortage of psychiatrists and services. We need to make it easier for patients to link with psychiatrists they find to be helpful. Our current system is difficult to negotiate for those who are healthy and have resources. Linking to care is nearly impossible for someone with limited resources who is psychotic or severely depressed.
12. And finally, I hope ... for better treatments for our patients, ones that cure mental illnesses without causing side effects, adverse reactions, or trade-offs that sometime make the cures worse than the diseases.
Thank you for bearing with me. I’m sure you have your own mental health hopes for the coming year, and perhaps some of our wishes might even come true. Wishing you a Healthy and Happy New Year!
Dr. Miller is a coauthor of “Shrink Rap: Three Psychiatrists Explain Their Work” (Baltimore: Johns Hopkins University Press, 2011).