-

chph
Main menu
CHEST Main Menu
Explore menu
CHEST Explore Menu
Proclivity ID
18829001
Unpublish
Specialty Focus
Pulmonology
Critical Care
Sleep Medicine
Cardiology
Cardiothoracic Surgery
Hospice & Palliative Medicine
Negative Keywords Excluded Elements
header[@id='header']
div[contains(@class, 'header__large-screen')]
div[contains(@class, 'read-next-article')]
div[contains(@class, 'main-prefix')]
div[contains(@class, 'nav-primary')]
nav[contains(@class, 'nav-primary')]
section[contains(@class, 'footer-nav-section-wrapper')]
footer[@id='footer']
section[contains(@class, 'nav-hidden')]
div[contains(@class, 'ce-card-content')]
nav[contains(@class, 'nav-ce-stack')]
div[contains(@class, 'view-medstat-quiz-listing-panes')]
div[contains(@class, 'pane-article-sidebar-latest-news')]
Altmetric
Article Authors "autobrand" affiliation
MDedge News
DSM Affiliated
Display in offset block
Disqus Exclude
Best Practices
CE/CME
Education Center
Medical Education Library
Enable Disqus
Display Author and Disclosure Link
Publication Type
News
Slot System
Featured Buckets
Disable Sticky Ads
Disable Ad Block Mitigation
Featured Buckets Admin
LayerRx Clinical Edge Id
784
Non-Overridden Topics
Show Ads on this Publication's Homepage
Consolidated Pub
Show Article Page Numbers on TOC
Expire Announcement Bar
Thu, 09/19/2024 - 15:01
Use larger logo size
Off
publication_blueconic_enabled
Off
Show More Destinations Menu
Disable Adhesion on Publication
On
Mobile Logo Image
Restore Menu Label on Mobile Navigation
Disable Facebook Pixel from Publication
Exclude this publication from publication selection on articles and quiz
Challenge Center
Disable Inline Native ads
survey writer start date
Thu, 09/19/2024 - 15:01
Mobile Logo Media

Trauma epidemiology and the organization of trauma care in the US

Article Type
Changed
Thu, 08/01/2024 - 10:43

Dr. John Agapian
CHEST
Dr. John Agapian

CHEST INFECTIONS AND DISASTER RESPONSE NETWORK

Disaster Response and Global Health Section

Patients who are injured do better when treated at trauma centers.

During CHEST 2023 in Honolulu last year, the Disaster Response and Global Health Section hosted a presentation to a packed audience highlighting the history of the trauma model system in America. Attendees learned about the emergence of trauma systems in the US and the survival advantages that trauma centers offer to patients who are injured.

Inception of the first “trauma manual” was during President Lincoln’s term to address the need for a process to care for patients who were injured during the Civil War. By the time World War II occurred, hospitals had established the idea of emergency departments, and the foundations for an emergency medical system (EMS) emerged on the world scene. Eventually, the Hill-Burton Act of 1946 required hospitals to have emergency departments by incentivizing them with federal funding.

The Committee on Trauma was founded in the 1950s as an adaptation of the American College of Surgeons’ 1922 Committee on Fractures. Then, in 1966, the National Highway Traffic Safety Administration rolled out a federal initiative for all states to develop EMS programs. Additionally, the National Academy of Sciences published Accidental Death and Disability: The Neglected Disease of Modern Society, paving the way for the Emergency Medical Services Systems Act of 1973, along with the Wedworth-Townsend Paramedic Act in California.

The concept of today’s trauma centers started in 1966 at Cook County Hospital in Chicago and Shock Trauma Center in Baltimore.

Publications
Topics
Sections

Dr. John Agapian
CHEST
Dr. John Agapian

CHEST INFECTIONS AND DISASTER RESPONSE NETWORK

Disaster Response and Global Health Section

Patients who are injured do better when treated at trauma centers.

During CHEST 2023 in Honolulu last year, the Disaster Response and Global Health Section hosted a presentation to a packed audience highlighting the history of the trauma model system in America. Attendees learned about the emergence of trauma systems in the US and the survival advantages that trauma centers offer to patients who are injured.

Inception of the first “trauma manual” was during President Lincoln’s term to address the need for a process to care for patients who were injured during the Civil War. By the time World War II occurred, hospitals had established the idea of emergency departments, and the foundations for an emergency medical system (EMS) emerged on the world scene. Eventually, the Hill-Burton Act of 1946 required hospitals to have emergency departments by incentivizing them with federal funding.

The Committee on Trauma was founded in the 1950s as an adaptation of the American College of Surgeons’ 1922 Committee on Fractures. Then, in 1966, the National Highway Traffic Safety Administration rolled out a federal initiative for all states to develop EMS programs. Additionally, the National Academy of Sciences published Accidental Death and Disability: The Neglected Disease of Modern Society, paving the way for the Emergency Medical Services Systems Act of 1973, along with the Wedworth-Townsend Paramedic Act in California.

The concept of today’s trauma centers started in 1966 at Cook County Hospital in Chicago and Shock Trauma Center in Baltimore.

Dr. John Agapian
CHEST
Dr. John Agapian

CHEST INFECTIONS AND DISASTER RESPONSE NETWORK

Disaster Response and Global Health Section

Patients who are injured do better when treated at trauma centers.

During CHEST 2023 in Honolulu last year, the Disaster Response and Global Health Section hosted a presentation to a packed audience highlighting the history of the trauma model system in America. Attendees learned about the emergence of trauma systems in the US and the survival advantages that trauma centers offer to patients who are injured.

Inception of the first “trauma manual” was during President Lincoln’s term to address the need for a process to care for patients who were injured during the Civil War. By the time World War II occurred, hospitals had established the idea of emergency departments, and the foundations for an emergency medical system (EMS) emerged on the world scene. Eventually, the Hill-Burton Act of 1946 required hospitals to have emergency departments by incentivizing them with federal funding.

The Committee on Trauma was founded in the 1950s as an adaptation of the American College of Surgeons’ 1922 Committee on Fractures. Then, in 1966, the National Highway Traffic Safety Administration rolled out a federal initiative for all states to develop EMS programs. Additionally, the National Academy of Sciences published Accidental Death and Disability: The Neglected Disease of Modern Society, paving the way for the Emergency Medical Services Systems Act of 1973, along with the Wedworth-Townsend Paramedic Act in California.

The concept of today’s trauma centers started in 1966 at Cook County Hospital in Chicago and Shock Trauma Center in Baltimore.

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

On 5 Ps: PSG, PM, PPG, PulseOx, and PAT

Article Type
Changed
Fri, 08/02/2024 - 08:58

Dr. Anisha Tauquir, Medical College of Wisconsin
CHEST
Dr. Anoosha Tauquir
  
Dr. Octavian C. Ioachimescu, Medical College of Wisconsin
CHEST
Dr. Octavian C. Ioachimescu

OSA is a very prevalent condition in the general population, but still many patients remain undiagnosed and untreated. Prolonged, untreated OSA is an independent risk factor for major cardiovascular morbidity and mortality. Therefore, timely diagnosis and treatment are required.

Polysomnography (PSG) remains to this day the gold standard for diagnosing sleep apnea. A standard PSG (type I) is performed in a sleep laboratory in the presence of specialized sleep technicians and utilizes EEG, electrooculogram (EOG), and electromyogram (EMG) to determine sleep stages, oronasal thermal and pressure transducer sensors to monitor airflow, respiratory inductance plethysmography to record respiratory effort, EMG for limb movements, pulse oximetry (PulseOx), ECG, and video or body sensor devices to confirm body position. Rising rates of sleep testing have created demand for an alternative to cumbersome, costly, and resource-intensive in-lab PSGs. As such, home sleep apnea testing (HSAT) has emerged as a simpler, more accessible, and cost-effective alternative diagnostic tool.

In 2007, the American Academy of Sleep Medicine (AASM) endorsed Portable Monitoring (PM) as an alternative to standard PSG, with the caveat that it should be used only in patients with a high pretest probability of sleep apnea, without respiratory or cardiovascular disorders and comorbid sleep disorders. All HSAT devices (type II-IV) are required to have a minimum of an oronasal thermal sensor/nasal pressure transducer, respiratory inductance plethysmography, and PulseOx. A major limitation of most HSAT devices is the lack of EEG, preventing detection of cortical arousals and wake time, forcing the use of total recording time as a surrogate for total sleep time.

Peripheral arterial tonometry (PAT)-based HSAT devices are unique in this respect, as their proprietary algorithms allow estimates of total sleep time by monitoring changes in peripheral vascular tone. Anyone who has seen a PAT-based HSAT may have noticed very different outputs from traditional HSATs.

PAT is based on the concept that airflow obstruction may lead to a surge in sympathetic tone, causing vasoconstriction and reduced blood volume in the peripheral vascular bed. A PAT-based device measures relative changes in blood volume and combines this information with actigraphy signals, PulseOx, and heart rate to diagnose the presence of respiratory events. Sleep apnea severity stratification is accomplished by the use of pAHI or pRDI (PAT-based apnea-hypopnea index and respiratory disturbance index, respectively).

PAT-based technology was first approved by the FDA in 2001 as a diagnostic tool for sleep apnea. The 2 best-known medical devices are WatchPAT® and NightOwl®, both of which have been FDA-approved and studied against PSG. To obtain an accurate and sustainable PAT signal, WatchPAT has a pneumo-optic finger probe designed to generate a uniform, subdiastolic pressure on the finger that minimizes venous blood pooling, prevents uncontrolled venous backflow, and effectively unloads the arterial wall tension without blocking digital arterial flow. NightOwl is a smaller device, with a single fingertip sensor that acquires actigraphy and PPG data to measure heart rate, Pulse Ox, and PAT.

The physiological basis of PAT relies on photoplethysmography (PPG), a noninvasive optical monitoring technique that generates a waveform, which ultimately correlates with the circulatory volume of the respective tissue.1 The PPG technology relies on the fact that when a specific tissue is exposed to light signal of a specific wavelength, its absorbance by tissue fluctuates with arterial pulsations. Pulse oximetry represents the most used application of PPG. Recent advances in PPG signal analysis have fueled its use in clinical and consumer sleep technologies and allowed new capabilities, including capturing heart rate and rhythm, pulse rate variability, arterial stiffness, and even—with somewhat less accuracy—energy expenditure, maximum O2 consumption, and blood pressure. Combining actigraphy monitoring with PPG technology took both consumer and medical-grade sleep technologies further, allowing the estimation of parameters such as sleep stages, sleep times, and respiratory events. With myriad new sleep trackers claiming to assess total sleep time, wake time, light or deep sleep, and even respiratory events, the obvious clinical question is centered on their comparative accuracy, as well against more traditional PM and the gold standard PSG.

Numerous studies have evaluated the efficacy of PAT-based devices for diagnosing sleep apnea, with variable findings. Many have shown good correlations between mean AHI and pAHI. Others have highlighted significant discrepancies in the measurements between PSG and PAT, questioning the reliability of PAT-based devices in the diagnosis and severity stratification of OSA. One meta-analysis of 14 studies showed a high degree of correlation between pAHI and AHI.2 Another study reported a concordance of 80% between PAT-based testing and consecutive PSG, with an increase to 86% at a higher AHI (>15/h).3 A subsequent meta-analysis showed that PAT was significantly less sensitive for diagnosing OSA than PSG, particularly for mild or moderate severity disease, emphasizing the need for further confirmation with PSG when faced with inconclusive or negative results.4 A large sleep clinic-based cohort study of 500 patients with OSA showed that WatchPAT devices misclassify OSA in a sizeable proportion of patients (30%-50%), leading to both over- and under-estimation of severity.5 Van Pee, et al, found that their PAT-based HSAT NightOwl performed better, using both the 3% and 4% hypopnea scoring rules and a novel near-border zone labeling.6

Some of the discordance in AHI between PAT and PSG appears to be related to age and sex. In our large sample comparing PAT to PSG, we found that using PAT-based data in concert with demographic (age, gender) and anthropometric (neck circumference, body mass index) variables improved the diagnostic accuracy of PAT-based testing.7 Another study concluded that manual scoring of WatchPAT automated results improved concordance with PSG, particularly in older participants and women. Several studies on WatchPAT recordings have demonstrated significant artifacts and inaccuracies in the PulseOx data. Although WatchPAT employs automated algorithms to remove erroneous data, a thorough visual inspection and manual correction of study data is still essential to derive accurate results.

Recent studies have found that PAT-based tests can also differentiate between central and obstructive respiratory events by using pulse signal upstroke variations caused by changes in intrathoracic pressure and respiratory/chest wall movement recorded by body position sensors, but large-scale studies are needed to confirm these findings. Korkalainen, et al, recently employed a deep-learning model to perform sleep staging on the PPG PulseOx signals from nearly 900 PSGs in patients with suspected OSA.8 The deep learning approach enabled the differentiation of sleep stages and accurate estimation of the total sleep time. Going forward, this could easily enhance the diagnostic yield of PM recordings and enable cost-efficient, long-term monitoring of sleep.

Although PAT-based home sleep tests have emerged as a simple and convenient option for the evaluation of sleep apnea, several studies have highlighted their limited sensitivity as a screening tool for mild and moderate cases of sleep apnea. Furthermore, the scope of these tests remains limited, rendering them rather unsuitable for assessment of more complex sleep disorders like narcolepsy or restless leg syndrome. Therefore, when OSA is suspected, the PAT-based sleep study is a good screening tool, but negative tests should not preclude further investigation. Where a high probability of sleep apnea exists but PAT-based testing shows no or mild OSA, an in-lab sleep study should be performed.


References

1. Ryals S, Chiang A, Schutte-Rodin S, et al. Photoplethysmography -- new applications for an old technology: a sleep technology review. J Clin Sleep Med. 2023;19(1):189-195.

2. Yalamanchali S, Farajian V, Hamilton C, Pott TR, Samuelson CG, Friedman M. Diagnosis of obstructive sleep apnea by peripheral arterial tonometry: meta-analysis. JAMA Otolaryngol Head Neck Surg. 2013;139(12):1343-1350.

3. Röcken J, Schumann DM, Herrmann MJ, et al. Peripheral arterial tonometry versus polysomnography in suspected obstructive sleep apnoea. Eur J Med Res. 2023;28(1):251.

4. Iftikhar IH, Finch CE, Shah AS, Augunstein CA, Ioachimescu OC. A meta-analysis of diagnostic test performance of peripheral arterial tonometry studies. J Clin Sleep Med. 2022;18(4):1093-1102.

5. Ioachimescu OC, Allam JS, Samarghandi A, et al. Performance of peripheral arterial tonometry-based testing for the diagnosis of obstructive sleep apnea in a large sleep clinic cohort. J Clin Sleep Med. 2020;16(10):1663-1674.

6. Van Pee B, Massie F, Vits S, et al. A multicentric validation study of a novel home sleep apnea test based on peripheral arterial tonometry. Sleep. 2022;45(5).

7. Ioachimescu OC, Dholakia SA, Venkateshiah SB, et al. Improving the performance of peripheral arterial tonometry-based testing for the diagnosis of obstructive sleep apnea. J Investig Med. 2020;68(8):1370-1378.

8. Korkalainen H, Aakko J, Duce B, et al. Deep learning enables sleep staging from photoplethysmogram for patients with suspected sleep apnea. Sleep. 2020;43(11).
 

Publications
Topics
Sections

Dr. Anisha Tauquir, Medical College of Wisconsin
CHEST
Dr. Anoosha Tauquir
  
Dr. Octavian C. Ioachimescu, Medical College of Wisconsin
CHEST
Dr. Octavian C. Ioachimescu

OSA is a very prevalent condition in the general population, but still many patients remain undiagnosed and untreated. Prolonged, untreated OSA is an independent risk factor for major cardiovascular morbidity and mortality. Therefore, timely diagnosis and treatment are required.

Polysomnography (PSG) remains to this day the gold standard for diagnosing sleep apnea. A standard PSG (type I) is performed in a sleep laboratory in the presence of specialized sleep technicians and utilizes EEG, electrooculogram (EOG), and electromyogram (EMG) to determine sleep stages, oronasal thermal and pressure transducer sensors to monitor airflow, respiratory inductance plethysmography to record respiratory effort, EMG for limb movements, pulse oximetry (PulseOx), ECG, and video or body sensor devices to confirm body position. Rising rates of sleep testing have created demand for an alternative to cumbersome, costly, and resource-intensive in-lab PSGs. As such, home sleep apnea testing (HSAT) has emerged as a simpler, more accessible, and cost-effective alternative diagnostic tool.

In 2007, the American Academy of Sleep Medicine (AASM) endorsed Portable Monitoring (PM) as an alternative to standard PSG, with the caveat that it should be used only in patients with a high pretest probability of sleep apnea, without respiratory or cardiovascular disorders and comorbid sleep disorders. All HSAT devices (type II-IV) are required to have a minimum of an oronasal thermal sensor/nasal pressure transducer, respiratory inductance plethysmography, and PulseOx. A major limitation of most HSAT devices is the lack of EEG, preventing detection of cortical arousals and wake time, forcing the use of total recording time as a surrogate for total sleep time.

Peripheral arterial tonometry (PAT)-based HSAT devices are unique in this respect, as their proprietary algorithms allow estimates of total sleep time by monitoring changes in peripheral vascular tone. Anyone who has seen a PAT-based HSAT may have noticed very different outputs from traditional HSATs.

PAT is based on the concept that airflow obstruction may lead to a surge in sympathetic tone, causing vasoconstriction and reduced blood volume in the peripheral vascular bed. A PAT-based device measures relative changes in blood volume and combines this information with actigraphy signals, PulseOx, and heart rate to diagnose the presence of respiratory events. Sleep apnea severity stratification is accomplished by the use of pAHI or pRDI (PAT-based apnea-hypopnea index and respiratory disturbance index, respectively).

PAT-based technology was first approved by the FDA in 2001 as a diagnostic tool for sleep apnea. The 2 best-known medical devices are WatchPAT® and NightOwl®, both of which have been FDA-approved and studied against PSG. To obtain an accurate and sustainable PAT signal, WatchPAT has a pneumo-optic finger probe designed to generate a uniform, subdiastolic pressure on the finger that minimizes venous blood pooling, prevents uncontrolled venous backflow, and effectively unloads the arterial wall tension without blocking digital arterial flow. NightOwl is a smaller device, with a single fingertip sensor that acquires actigraphy and PPG data to measure heart rate, Pulse Ox, and PAT.

The physiological basis of PAT relies on photoplethysmography (PPG), a noninvasive optical monitoring technique that generates a waveform, which ultimately correlates with the circulatory volume of the respective tissue.1 The PPG technology relies on the fact that when a specific tissue is exposed to light signal of a specific wavelength, its absorbance by tissue fluctuates with arterial pulsations. Pulse oximetry represents the most used application of PPG. Recent advances in PPG signal analysis have fueled its use in clinical and consumer sleep technologies and allowed new capabilities, including capturing heart rate and rhythm, pulse rate variability, arterial stiffness, and even—with somewhat less accuracy—energy expenditure, maximum O2 consumption, and blood pressure. Combining actigraphy monitoring with PPG technology took both consumer and medical-grade sleep technologies further, allowing the estimation of parameters such as sleep stages, sleep times, and respiratory events. With myriad new sleep trackers claiming to assess total sleep time, wake time, light or deep sleep, and even respiratory events, the obvious clinical question is centered on their comparative accuracy, as well against more traditional PM and the gold standard PSG.

Numerous studies have evaluated the efficacy of PAT-based devices for diagnosing sleep apnea, with variable findings. Many have shown good correlations between mean AHI and pAHI. Others have highlighted significant discrepancies in the measurements between PSG and PAT, questioning the reliability of PAT-based devices in the diagnosis and severity stratification of OSA. One meta-analysis of 14 studies showed a high degree of correlation between pAHI and AHI.2 Another study reported a concordance of 80% between PAT-based testing and consecutive PSG, with an increase to 86% at a higher AHI (>15/h).3 A subsequent meta-analysis showed that PAT was significantly less sensitive for diagnosing OSA than PSG, particularly for mild or moderate severity disease, emphasizing the need for further confirmation with PSG when faced with inconclusive or negative results.4 A large sleep clinic-based cohort study of 500 patients with OSA showed that WatchPAT devices misclassify OSA in a sizeable proportion of patients (30%-50%), leading to both over- and under-estimation of severity.5 Van Pee, et al, found that their PAT-based HSAT NightOwl performed better, using both the 3% and 4% hypopnea scoring rules and a novel near-border zone labeling.6

Some of the discordance in AHI between PAT and PSG appears to be related to age and sex. In our large sample comparing PAT to PSG, we found that using PAT-based data in concert with demographic (age, gender) and anthropometric (neck circumference, body mass index) variables improved the diagnostic accuracy of PAT-based testing.7 Another study concluded that manual scoring of WatchPAT automated results improved concordance with PSG, particularly in older participants and women. Several studies on WatchPAT recordings have demonstrated significant artifacts and inaccuracies in the PulseOx data. Although WatchPAT employs automated algorithms to remove erroneous data, a thorough visual inspection and manual correction of study data is still essential to derive accurate results.

Recent studies have found that PAT-based tests can also differentiate between central and obstructive respiratory events by using pulse signal upstroke variations caused by changes in intrathoracic pressure and respiratory/chest wall movement recorded by body position sensors, but large-scale studies are needed to confirm these findings. Korkalainen, et al, recently employed a deep-learning model to perform sleep staging on the PPG PulseOx signals from nearly 900 PSGs in patients with suspected OSA.8 The deep learning approach enabled the differentiation of sleep stages and accurate estimation of the total sleep time. Going forward, this could easily enhance the diagnostic yield of PM recordings and enable cost-efficient, long-term monitoring of sleep.

Although PAT-based home sleep tests have emerged as a simple and convenient option for the evaluation of sleep apnea, several studies have highlighted their limited sensitivity as a screening tool for mild and moderate cases of sleep apnea. Furthermore, the scope of these tests remains limited, rendering them rather unsuitable for assessment of more complex sleep disorders like narcolepsy or restless leg syndrome. Therefore, when OSA is suspected, the PAT-based sleep study is a good screening tool, but negative tests should not preclude further investigation. Where a high probability of sleep apnea exists but PAT-based testing shows no or mild OSA, an in-lab sleep study should be performed.


References

1. Ryals S, Chiang A, Schutte-Rodin S, et al. Photoplethysmography -- new applications for an old technology: a sleep technology review. J Clin Sleep Med. 2023;19(1):189-195.

2. Yalamanchali S, Farajian V, Hamilton C, Pott TR, Samuelson CG, Friedman M. Diagnosis of obstructive sleep apnea by peripheral arterial tonometry: meta-analysis. JAMA Otolaryngol Head Neck Surg. 2013;139(12):1343-1350.

3. Röcken J, Schumann DM, Herrmann MJ, et al. Peripheral arterial tonometry versus polysomnography in suspected obstructive sleep apnoea. Eur J Med Res. 2023;28(1):251.

4. Iftikhar IH, Finch CE, Shah AS, Augunstein CA, Ioachimescu OC. A meta-analysis of diagnostic test performance of peripheral arterial tonometry studies. J Clin Sleep Med. 2022;18(4):1093-1102.

5. Ioachimescu OC, Allam JS, Samarghandi A, et al. Performance of peripheral arterial tonometry-based testing for the diagnosis of obstructive sleep apnea in a large sleep clinic cohort. J Clin Sleep Med. 2020;16(10):1663-1674.

6. Van Pee B, Massie F, Vits S, et al. A multicentric validation study of a novel home sleep apnea test based on peripheral arterial tonometry. Sleep. 2022;45(5).

7. Ioachimescu OC, Dholakia SA, Venkateshiah SB, et al. Improving the performance of peripheral arterial tonometry-based testing for the diagnosis of obstructive sleep apnea. J Investig Med. 2020;68(8):1370-1378.

8. Korkalainen H, Aakko J, Duce B, et al. Deep learning enables sleep staging from photoplethysmogram for patients with suspected sleep apnea. Sleep. 2020;43(11).
 

Dr. Anisha Tauquir, Medical College of Wisconsin
CHEST
Dr. Anoosha Tauquir
  
Dr. Octavian C. Ioachimescu, Medical College of Wisconsin
CHEST
Dr. Octavian C. Ioachimescu

OSA is a very prevalent condition in the general population, but still many patients remain undiagnosed and untreated. Prolonged, untreated OSA is an independent risk factor for major cardiovascular morbidity and mortality. Therefore, timely diagnosis and treatment are required.

Polysomnography (PSG) remains to this day the gold standard for diagnosing sleep apnea. A standard PSG (type I) is performed in a sleep laboratory in the presence of specialized sleep technicians and utilizes EEG, electrooculogram (EOG), and electromyogram (EMG) to determine sleep stages, oronasal thermal and pressure transducer sensors to monitor airflow, respiratory inductance plethysmography to record respiratory effort, EMG for limb movements, pulse oximetry (PulseOx), ECG, and video or body sensor devices to confirm body position. Rising rates of sleep testing have created demand for an alternative to cumbersome, costly, and resource-intensive in-lab PSGs. As such, home sleep apnea testing (HSAT) has emerged as a simpler, more accessible, and cost-effective alternative diagnostic tool.

In 2007, the American Academy of Sleep Medicine (AASM) endorsed Portable Monitoring (PM) as an alternative to standard PSG, with the caveat that it should be used only in patients with a high pretest probability of sleep apnea, without respiratory or cardiovascular disorders and comorbid sleep disorders. All HSAT devices (type II-IV) are required to have a minimum of an oronasal thermal sensor/nasal pressure transducer, respiratory inductance plethysmography, and PulseOx. A major limitation of most HSAT devices is the lack of EEG, preventing detection of cortical arousals and wake time, forcing the use of total recording time as a surrogate for total sleep time.

Peripheral arterial tonometry (PAT)-based HSAT devices are unique in this respect, as their proprietary algorithms allow estimates of total sleep time by monitoring changes in peripheral vascular tone. Anyone who has seen a PAT-based HSAT may have noticed very different outputs from traditional HSATs.

PAT is based on the concept that airflow obstruction may lead to a surge in sympathetic tone, causing vasoconstriction and reduced blood volume in the peripheral vascular bed. A PAT-based device measures relative changes in blood volume and combines this information with actigraphy signals, PulseOx, and heart rate to diagnose the presence of respiratory events. Sleep apnea severity stratification is accomplished by the use of pAHI or pRDI (PAT-based apnea-hypopnea index and respiratory disturbance index, respectively).

PAT-based technology was first approved by the FDA in 2001 as a diagnostic tool for sleep apnea. The 2 best-known medical devices are WatchPAT® and NightOwl®, both of which have been FDA-approved and studied against PSG. To obtain an accurate and sustainable PAT signal, WatchPAT has a pneumo-optic finger probe designed to generate a uniform, subdiastolic pressure on the finger that minimizes venous blood pooling, prevents uncontrolled venous backflow, and effectively unloads the arterial wall tension without blocking digital arterial flow. NightOwl is a smaller device, with a single fingertip sensor that acquires actigraphy and PPG data to measure heart rate, Pulse Ox, and PAT.

The physiological basis of PAT relies on photoplethysmography (PPG), a noninvasive optical monitoring technique that generates a waveform, which ultimately correlates with the circulatory volume of the respective tissue.1 The PPG technology relies on the fact that when a specific tissue is exposed to light signal of a specific wavelength, its absorbance by tissue fluctuates with arterial pulsations. Pulse oximetry represents the most used application of PPG. Recent advances in PPG signal analysis have fueled its use in clinical and consumer sleep technologies and allowed new capabilities, including capturing heart rate and rhythm, pulse rate variability, arterial stiffness, and even—with somewhat less accuracy—energy expenditure, maximum O2 consumption, and blood pressure. Combining actigraphy monitoring with PPG technology took both consumer and medical-grade sleep technologies further, allowing the estimation of parameters such as sleep stages, sleep times, and respiratory events. With myriad new sleep trackers claiming to assess total sleep time, wake time, light or deep sleep, and even respiratory events, the obvious clinical question is centered on their comparative accuracy, as well against more traditional PM and the gold standard PSG.

Numerous studies have evaluated the efficacy of PAT-based devices for diagnosing sleep apnea, with variable findings. Many have shown good correlations between mean AHI and pAHI. Others have highlighted significant discrepancies in the measurements between PSG and PAT, questioning the reliability of PAT-based devices in the diagnosis and severity stratification of OSA. One meta-analysis of 14 studies showed a high degree of correlation between pAHI and AHI.2 Another study reported a concordance of 80% between PAT-based testing and consecutive PSG, with an increase to 86% at a higher AHI (>15/h).3 A subsequent meta-analysis showed that PAT was significantly less sensitive for diagnosing OSA than PSG, particularly for mild or moderate severity disease, emphasizing the need for further confirmation with PSG when faced with inconclusive or negative results.4 A large sleep clinic-based cohort study of 500 patients with OSA showed that WatchPAT devices misclassify OSA in a sizeable proportion of patients (30%-50%), leading to both over- and under-estimation of severity.5 Van Pee, et al, found that their PAT-based HSAT NightOwl performed better, using both the 3% and 4% hypopnea scoring rules and a novel near-border zone labeling.6

Some of the discordance in AHI between PAT and PSG appears to be related to age and sex. In our large sample comparing PAT to PSG, we found that using PAT-based data in concert with demographic (age, gender) and anthropometric (neck circumference, body mass index) variables improved the diagnostic accuracy of PAT-based testing.7 Another study concluded that manual scoring of WatchPAT automated results improved concordance with PSG, particularly in older participants and women. Several studies on WatchPAT recordings have demonstrated significant artifacts and inaccuracies in the PulseOx data. Although WatchPAT employs automated algorithms to remove erroneous data, a thorough visual inspection and manual correction of study data is still essential to derive accurate results.

Recent studies have found that PAT-based tests can also differentiate between central and obstructive respiratory events by using pulse signal upstroke variations caused by changes in intrathoracic pressure and respiratory/chest wall movement recorded by body position sensors, but large-scale studies are needed to confirm these findings. Korkalainen, et al, recently employed a deep-learning model to perform sleep staging on the PPG PulseOx signals from nearly 900 PSGs in patients with suspected OSA.8 The deep learning approach enabled the differentiation of sleep stages and accurate estimation of the total sleep time. Going forward, this could easily enhance the diagnostic yield of PM recordings and enable cost-efficient, long-term monitoring of sleep.

Although PAT-based home sleep tests have emerged as a simple and convenient option for the evaluation of sleep apnea, several studies have highlighted their limited sensitivity as a screening tool for mild and moderate cases of sleep apnea. Furthermore, the scope of these tests remains limited, rendering them rather unsuitable for assessment of more complex sleep disorders like narcolepsy or restless leg syndrome. Therefore, when OSA is suspected, the PAT-based sleep study is a good screening tool, but negative tests should not preclude further investigation. Where a high probability of sleep apnea exists but PAT-based testing shows no or mild OSA, an in-lab sleep study should be performed.


References

1. Ryals S, Chiang A, Schutte-Rodin S, et al. Photoplethysmography -- new applications for an old technology: a sleep technology review. J Clin Sleep Med. 2023;19(1):189-195.

2. Yalamanchali S, Farajian V, Hamilton C, Pott TR, Samuelson CG, Friedman M. Diagnosis of obstructive sleep apnea by peripheral arterial tonometry: meta-analysis. JAMA Otolaryngol Head Neck Surg. 2013;139(12):1343-1350.

3. Röcken J, Schumann DM, Herrmann MJ, et al. Peripheral arterial tonometry versus polysomnography in suspected obstructive sleep apnoea. Eur J Med Res. 2023;28(1):251.

4. Iftikhar IH, Finch CE, Shah AS, Augunstein CA, Ioachimescu OC. A meta-analysis of diagnostic test performance of peripheral arterial tonometry studies. J Clin Sleep Med. 2022;18(4):1093-1102.

5. Ioachimescu OC, Allam JS, Samarghandi A, et al. Performance of peripheral arterial tonometry-based testing for the diagnosis of obstructive sleep apnea in a large sleep clinic cohort. J Clin Sleep Med. 2020;16(10):1663-1674.

6. Van Pee B, Massie F, Vits S, et al. A multicentric validation study of a novel home sleep apnea test based on peripheral arterial tonometry. Sleep. 2022;45(5).

7. Ioachimescu OC, Dholakia SA, Venkateshiah SB, et al. Improving the performance of peripheral arterial tonometry-based testing for the diagnosis of obstructive sleep apnea. J Investig Med. 2020;68(8):1370-1378.

8. Korkalainen H, Aakko J, Duce B, et al. Deep learning enables sleep staging from photoplethysmogram for patients with suspected sleep apnea. Sleep. 2020;43(11).
 

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

On thoughtful selection of medications in the acute critical care setting

Article Type
Changed
Thu, 08/01/2024 - 10:41

Dr. Alexandra Wichmann
CHEST
Dr. Alexandra Wichmann

CRITICAL CARE NETWORK

Palliative and End-of-Life Care Section

As critical care medicine continues to advance understanding of ICU survivorship, thoughtful selection of medications in the acute setting can potentially mitigate long-term cognitive, physical, and affective effects.

As of yet, no significant studies have linked opioid use in critical care to new diagnoses of opioid use disorder, but the opioid epidemic has taught us that profligate use of opioids can have devastating effects despite best intentions. Continuous infusions of full agonist opioids for sedation remain an important tool in management of sedation. For acute pain, buprenorphine represents an attractive alternative for patients who are both intubated and nonintubated. It provides equal pain relief as full agonist opioids while causing less respiratory depression, less delirium, less nausea, less constipation, less euphoria, and less misuse potential. Its unique partial mu-opioid agonism is responsible for the improved nausea, constipation, and respiratory depression, while the kappa and delta receptor antagonisms are responsible for antidepressant effects as well as lessened opioid craving, sedation, and dysphoria. Given the variety of doses and routes for buprenorphine, palliative medicine consults can help navigate preventing precipitated withdrawal in patients who are opioid-tolerant and the variety of available dosing and routes.

It is a testament to the growth of critical care medicine that we now have the privilege and responsibility to account for long-term sequelae of our lifesaving interventions, rather than the old model of “prevent death at all costs.” Continued integration of high-quality symptom management into critical care offers an opportunity to better balance life-prolonging treatment and optimize quality of life.

References

1. Neale KJ, Weimer MB, Davis MP, et al. Top ten tips palliative care clinicians should know about buprenorphine. J Palliat Med. 2023;26(1):120-130. doi: 10.1089/jpm.2022.0399
 

Publications
Topics
Sections

Dr. Alexandra Wichmann
CHEST
Dr. Alexandra Wichmann

CRITICAL CARE NETWORK

Palliative and End-of-Life Care Section

As critical care medicine continues to advance understanding of ICU survivorship, thoughtful selection of medications in the acute setting can potentially mitigate long-term cognitive, physical, and affective effects.

As of yet, no significant studies have linked opioid use in critical care to new diagnoses of opioid use disorder, but the opioid epidemic has taught us that profligate use of opioids can have devastating effects despite best intentions. Continuous infusions of full agonist opioids for sedation remain an important tool in management of sedation. For acute pain, buprenorphine represents an attractive alternative for patients who are both intubated and nonintubated. It provides equal pain relief as full agonist opioids while causing less respiratory depression, less delirium, less nausea, less constipation, less euphoria, and less misuse potential. Its unique partial mu-opioid agonism is responsible for the improved nausea, constipation, and respiratory depression, while the kappa and delta receptor antagonisms are responsible for antidepressant effects as well as lessened opioid craving, sedation, and dysphoria. Given the variety of doses and routes for buprenorphine, palliative medicine consults can help navigate preventing precipitated withdrawal in patients who are opioid-tolerant and the variety of available dosing and routes.

It is a testament to the growth of critical care medicine that we now have the privilege and responsibility to account for long-term sequelae of our lifesaving interventions, rather than the old model of “prevent death at all costs.” Continued integration of high-quality symptom management into critical care offers an opportunity to better balance life-prolonging treatment and optimize quality of life.

References

1. Neale KJ, Weimer MB, Davis MP, et al. Top ten tips palliative care clinicians should know about buprenorphine. J Palliat Med. 2023;26(1):120-130. doi: 10.1089/jpm.2022.0399
 

Dr. Alexandra Wichmann
CHEST
Dr. Alexandra Wichmann

CRITICAL CARE NETWORK

Palliative and End-of-Life Care Section

As critical care medicine continues to advance understanding of ICU survivorship, thoughtful selection of medications in the acute setting can potentially mitigate long-term cognitive, physical, and affective effects.

As of yet, no significant studies have linked opioid use in critical care to new diagnoses of opioid use disorder, but the opioid epidemic has taught us that profligate use of opioids can have devastating effects despite best intentions. Continuous infusions of full agonist opioids for sedation remain an important tool in management of sedation. For acute pain, buprenorphine represents an attractive alternative for patients who are both intubated and nonintubated. It provides equal pain relief as full agonist opioids while causing less respiratory depression, less delirium, less nausea, less constipation, less euphoria, and less misuse potential. Its unique partial mu-opioid agonism is responsible for the improved nausea, constipation, and respiratory depression, while the kappa and delta receptor antagonisms are responsible for antidepressant effects as well as lessened opioid craving, sedation, and dysphoria. Given the variety of doses and routes for buprenorphine, palliative medicine consults can help navigate preventing precipitated withdrawal in patients who are opioid-tolerant and the variety of available dosing and routes.

It is a testament to the growth of critical care medicine that we now have the privilege and responsibility to account for long-term sequelae of our lifesaving interventions, rather than the old model of “prevent death at all costs.” Continued integration of high-quality symptom management into critical care offers an opportunity to better balance life-prolonging treatment and optimize quality of life.

References

1. Neale KJ, Weimer MB, Davis MP, et al. Top ten tips palliative care clinicians should know about buprenorphine. J Palliat Med. 2023;26(1):120-130. doi: 10.1089/jpm.2022.0399
 

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Atypical pulmonary cysts: Why to care

Article Type
Changed
Thu, 08/01/2024 - 10:24

Dr. Rage Geringer
CHEST
Dr. Rage Geringer

THORACIC ONCOLOGY AND CHEST PROCEDURES NETWORK

Lung Cancer Section

Since the American College of Radiology (ACR) updated its Lung CT Screening Reporting & Data System (Lung-RADS) to include atypical pulmonary cysts in 2022, there has been little discussion among chest physicians regarding the significance of pulmonary cysts and why these changes were made.

Lung-RADS 2022 defined atypical pulmonary cysts as single, unilocular cysts with a wall thickness greater than 2 mm or any multilocular cysts. These can be uniform, asymmetric, or have a focal nodularity. This change was prompted by data derived from multiple studies. First, a finding that 3.6% of lung cancers were associated with cysts at baseline.1 This was followed by a reanalysis of the NELSON trial’s missed cancers showing 22% of those overlooked during initial screening had findings of cystic disease, reaffirming the significance of atypical pulmonary cysts.2 Though the number is low, we now know 1.1% of all cancers present as an atypical cyst, with 4.7% of them being malignant.3
 

Based on these studies, cysts are a baseline Lung-RADS 4A—a finding that correlates to a higher risk and needs to be followed with a short-term CT scan in 3 months vs a PET. ACR does recommend reserving PET scans for wall thickness > 8 mm. If the repeat CT scan is stable, then the Lung-RADS designation is dropped to a 3 for follow-up.

References

1. Farooqi AO, Cham M, Zhang L, et al. Lung cancer associated with cystic airspaces. AJR Am J Roentgenol. 2012;199(4):781-786.

2. Scholten ET, Horeweg N, Koning HJ, et al. Computed tomographic characteristics of interval and post screen carcinomas in lung cancer screening. Eur Radiol. 2015;25(1):81-88.

3. Mascalchi M, Attinà D, Bertelli E, et al. Lung cancer associated with cystic airspaces. J Comput Assist Tomogr. 2015;39(1):102-108.

Publications
Topics
Sections

Dr. Rage Geringer
CHEST
Dr. Rage Geringer

THORACIC ONCOLOGY AND CHEST PROCEDURES NETWORK

Lung Cancer Section

Since the American College of Radiology (ACR) updated its Lung CT Screening Reporting & Data System (Lung-RADS) to include atypical pulmonary cysts in 2022, there has been little discussion among chest physicians regarding the significance of pulmonary cysts and why these changes were made.

Lung-RADS 2022 defined atypical pulmonary cysts as single, unilocular cysts with a wall thickness greater than 2 mm or any multilocular cysts. These can be uniform, asymmetric, or have a focal nodularity. This change was prompted by data derived from multiple studies. First, a finding that 3.6% of lung cancers were associated with cysts at baseline.1 This was followed by a reanalysis of the NELSON trial’s missed cancers showing 22% of those overlooked during initial screening had findings of cystic disease, reaffirming the significance of atypical pulmonary cysts.2 Though the number is low, we now know 1.1% of all cancers present as an atypical cyst, with 4.7% of them being malignant.3
 

Based on these studies, cysts are a baseline Lung-RADS 4A—a finding that correlates to a higher risk and needs to be followed with a short-term CT scan in 3 months vs a PET. ACR does recommend reserving PET scans for wall thickness > 8 mm. If the repeat CT scan is stable, then the Lung-RADS designation is dropped to a 3 for follow-up.

References

1. Farooqi AO, Cham M, Zhang L, et al. Lung cancer associated with cystic airspaces. AJR Am J Roentgenol. 2012;199(4):781-786.

2. Scholten ET, Horeweg N, Koning HJ, et al. Computed tomographic characteristics of interval and post screen carcinomas in lung cancer screening. Eur Radiol. 2015;25(1):81-88.

3. Mascalchi M, Attinà D, Bertelli E, et al. Lung cancer associated with cystic airspaces. J Comput Assist Tomogr. 2015;39(1):102-108.

Dr. Rage Geringer
CHEST
Dr. Rage Geringer

THORACIC ONCOLOGY AND CHEST PROCEDURES NETWORK

Lung Cancer Section

Since the American College of Radiology (ACR) updated its Lung CT Screening Reporting & Data System (Lung-RADS) to include atypical pulmonary cysts in 2022, there has been little discussion among chest physicians regarding the significance of pulmonary cysts and why these changes were made.

Lung-RADS 2022 defined atypical pulmonary cysts as single, unilocular cysts with a wall thickness greater than 2 mm or any multilocular cysts. These can be uniform, asymmetric, or have a focal nodularity. This change was prompted by data derived from multiple studies. First, a finding that 3.6% of lung cancers were associated with cysts at baseline.1 This was followed by a reanalysis of the NELSON trial’s missed cancers showing 22% of those overlooked during initial screening had findings of cystic disease, reaffirming the significance of atypical pulmonary cysts.2 Though the number is low, we now know 1.1% of all cancers present as an atypical cyst, with 4.7% of them being malignant.3
 

Based on these studies, cysts are a baseline Lung-RADS 4A—a finding that correlates to a higher risk and needs to be followed with a short-term CT scan in 3 months vs a PET. ACR does recommend reserving PET scans for wall thickness > 8 mm. If the repeat CT scan is stable, then the Lung-RADS designation is dropped to a 3 for follow-up.

References

1. Farooqi AO, Cham M, Zhang L, et al. Lung cancer associated with cystic airspaces. AJR Am J Roentgenol. 2012;199(4):781-786.

2. Scholten ET, Horeweg N, Koning HJ, et al. Computed tomographic characteristics of interval and post screen carcinomas in lung cancer screening. Eur Radiol. 2015;25(1):81-88.

3. Mascalchi M, Attinà D, Bertelli E, et al. Lung cancer associated with cystic airspaces. J Comput Assist Tomogr. 2015;39(1):102-108.

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Improving ILD diagnosis in primary care settings

Article Type
Changed
Thu, 08/01/2024 - 16:13

Interstitial lung diseases (ILDs), with their many ubiquitous symptoms, are often hard to diagnose in patients. That’s why Amirahwaty Abdullah, MBBS, and Kavitha Selvan, MD, see value in educating clinicians on how to identify and diagnose ILD.

CHEST
Dr. Amirahwaty Abdullah

Both Dr. Abdullah and Dr. Selvan received quality improvement grants from CHEST in October 2023 to do just that. Their projects put the ILD Clinician Toolkit into practice, created as a part of CHEST’s Bridging Specialties™: Timely Diagnosis for ILD initiative aimed at shortening the time to diagnosis.

CHEST
Dr. Kavitha Selvan


Recently, CHEST caught up with the two grant recipients to see how their projects were progressing.

Combating the prevalence of ILDs in West Virginia

Dr. Abdullah and Co-Principal Investigator, Haroon Ahmed, MD, are two of the staff members supporting West Virginia University ILD Clinic, where the ILD Clinician Toolkit is being utilized.

CHEST
Dr. Haroon Ahmed

“ILD is so prevalent here, so we thought it would be an excellent opportunity to do the quality improvement project here because we really do need the resources to improve the care of our [patients with ILD],” Dr. Abdullah said. “For the entire state of West Virginia, we’re the only ILD center.”

In West Virginia, many factors are at play making ILD prevalent, with a recent study showing that the state has the second highest rate of interstitial pulmonary fibrosis, Dr. Abdullah said. This is all due to the economy of the state, the rurality of the population, and the occupational hazards with common jobs like coal mining and farming.

“This topic about bridging the gap and early diagnosis really resonated with us because we see all these patients who end up seeing us after they’ve been on oxygen for years, when they can’t do anything else,” Dr. Ahmed said. “There was a big gap here, and we saw that every day in our clinical practice.”

Since implementing the ILD Clinician Toolkit into their program, the two have started providing these resources to primary care physicians in an effort to help expedite their workups when they see patients with common ILD symptoms. This was done through grand rounds and educational conferences for those practicing in family medicine, internal medicine, and pediatric medicine. And more education is planned for the future.

They have also created working relationships with these departments and have encouraged them to send patients to the ILD Clinic, so patients don’t have to be referred to multiple different physicians.

The next step for the project is to implement telemedicine capabilities, which will allow the team to roll out the patient questionnaire from the toolkit. The questionnaire helps physicians gather a detailed history about their patients, including their past and current medications, surgeries, occupational and environmental exposures, and known comorbidities.

“We definitely want to reach out via telemedicine to patients because, at this point in time, some of our patients travel 3, 4 hours one way just to come see us. So, if we can make it more accessible, we will,” Dr. Abdullah said.

Their plan is to provide iPads that are equipped with the questionnaire and other toolkit resources to the providers. Through this method, the team will be able to see how often the questionnaire is used.

“We are very thankful for this grant because I do think we are saving lives,” Dr. Abdullah said. “Any little thing that we can do to improve the outcomes of these patients who have a rare but difficult-to-treat disease—it’s crucial. This gives us the ability to reach out and help patients who are out of our physical bounds.”
 

 

 

Diagnosing ILD among underrepresented minority populations in Chicago

Dr. Kavitha Selvan is conducting her quality improvement research project at the University of Chicago, alongside her team.

“The community we serve in South Chicago houses a significant number of underrepresented [minority populations],” she said.

“We know that Black patients with ILD experience higher rates of hospitalization, compared with White patients,” she said. “And they are hospitalized, require lung transplants, and die at a younger age too.”

In clinical practice, Dr. Selvan has noticed a trend of patients being referred for evaluation of possible ILD later and later. In some cases, several years go by before the appropriate work for ILD is initiated, and that valuable time lost leads to irreversible loss of lung function.

Dr. Selvan’s plan is to partner with primary care providers who are seeing these patients first in order to expedite the work-up and referral of ILD when appropriate and, ultimately, reduce the amount of time that passes between symptom onset and definitive diagnosis.

“To me, this grant and the resources it provided represented an important opportunity to improve outcomes in the high-risk patients we care for through early disease recognition and treatment,” she said.

Dr. Selvan’s project began with educating members of the Primary Care Group at UChicago on risk factors and exam findings that may suggest ILD in patients coming to clinic with nonspecific respiratory complaints. Then they had to equip providers with the ILD Clinician Toolkit and reach patients by handing out the patient questionnaire in primary care clinic.

The next step for this project is going to be dissecting the answers to a postintervention survey that was sent out to understand the practices and comfort of primary care providers in evaluating suspected ILD and the utility of the additional resources created by Dr. Selvan’s team.

“My hope is that we can utilize this partnership with the Primary Care Group to provide the education that’s needed both on the patient side and the provider side, like knowing not to ignore respiratory symptoms, knowing which patients warrant ILD-specific testing, and knowing what the appropriate tests are. In doing so, we can get patients into ILD clinic earlier, confirm their diagnosis, and get them initiated on the appropriate therapies sooner,” she said.

Dr. Selvan credits the quality improvement grant as being fundamental in her success as a fellow and early faculty at UChicago Medicine and believes that this project has created a culture of awareness that wouldn’t have been possible without funding.

“I truly believe that early detection and risk factor modification is the most critical aspect of interstitial lung disease care, and the mechanism for how we’re going to actually improve patient outcomes in our community,” Dr. Selvan said. “Building the infrastructure to accomplish that requires time, resources, and support from institutions and philanthropic foundations that believe in that mission too."

Opportunities like this are made possible by generous contributions from our donors. Make a gift to CHEST and select “Clinical Research” to help aid future research into interstitial lung disease and much more.

Publications
Topics
Sections

Interstitial lung diseases (ILDs), with their many ubiquitous symptoms, are often hard to diagnose in patients. That’s why Amirahwaty Abdullah, MBBS, and Kavitha Selvan, MD, see value in educating clinicians on how to identify and diagnose ILD.

CHEST
Dr. Amirahwaty Abdullah

Both Dr. Abdullah and Dr. Selvan received quality improvement grants from CHEST in October 2023 to do just that. Their projects put the ILD Clinician Toolkit into practice, created as a part of CHEST’s Bridging Specialties™: Timely Diagnosis for ILD initiative aimed at shortening the time to diagnosis.

CHEST
Dr. Kavitha Selvan


Recently, CHEST caught up with the two grant recipients to see how their projects were progressing.

Combating the prevalence of ILDs in West Virginia

Dr. Abdullah and Co-Principal Investigator, Haroon Ahmed, MD, are two of the staff members supporting West Virginia University ILD Clinic, where the ILD Clinician Toolkit is being utilized.

CHEST
Dr. Haroon Ahmed

“ILD is so prevalent here, so we thought it would be an excellent opportunity to do the quality improvement project here because we really do need the resources to improve the care of our [patients with ILD],” Dr. Abdullah said. “For the entire state of West Virginia, we’re the only ILD center.”

In West Virginia, many factors are at play making ILD prevalent, with a recent study showing that the state has the second highest rate of interstitial pulmonary fibrosis, Dr. Abdullah said. This is all due to the economy of the state, the rurality of the population, and the occupational hazards with common jobs like coal mining and farming.

“This topic about bridging the gap and early diagnosis really resonated with us because we see all these patients who end up seeing us after they’ve been on oxygen for years, when they can’t do anything else,” Dr. Ahmed said. “There was a big gap here, and we saw that every day in our clinical practice.”

Since implementing the ILD Clinician Toolkit into their program, the two have started providing these resources to primary care physicians in an effort to help expedite their workups when they see patients with common ILD symptoms. This was done through grand rounds and educational conferences for those practicing in family medicine, internal medicine, and pediatric medicine. And more education is planned for the future.

They have also created working relationships with these departments and have encouraged them to send patients to the ILD Clinic, so patients don’t have to be referred to multiple different physicians.

The next step for the project is to implement telemedicine capabilities, which will allow the team to roll out the patient questionnaire from the toolkit. The questionnaire helps physicians gather a detailed history about their patients, including their past and current medications, surgeries, occupational and environmental exposures, and known comorbidities.

“We definitely want to reach out via telemedicine to patients because, at this point in time, some of our patients travel 3, 4 hours one way just to come see us. So, if we can make it more accessible, we will,” Dr. Abdullah said.

Their plan is to provide iPads that are equipped with the questionnaire and other toolkit resources to the providers. Through this method, the team will be able to see how often the questionnaire is used.

“We are very thankful for this grant because I do think we are saving lives,” Dr. Abdullah said. “Any little thing that we can do to improve the outcomes of these patients who have a rare but difficult-to-treat disease—it’s crucial. This gives us the ability to reach out and help patients who are out of our physical bounds.”
 

 

 

Diagnosing ILD among underrepresented minority populations in Chicago

Dr. Kavitha Selvan is conducting her quality improvement research project at the University of Chicago, alongside her team.

“The community we serve in South Chicago houses a significant number of underrepresented [minority populations],” she said.

“We know that Black patients with ILD experience higher rates of hospitalization, compared with White patients,” she said. “And they are hospitalized, require lung transplants, and die at a younger age too.”

In clinical practice, Dr. Selvan has noticed a trend of patients being referred for evaluation of possible ILD later and later. In some cases, several years go by before the appropriate work for ILD is initiated, and that valuable time lost leads to irreversible loss of lung function.

Dr. Selvan’s plan is to partner with primary care providers who are seeing these patients first in order to expedite the work-up and referral of ILD when appropriate and, ultimately, reduce the amount of time that passes between symptom onset and definitive diagnosis.

“To me, this grant and the resources it provided represented an important opportunity to improve outcomes in the high-risk patients we care for through early disease recognition and treatment,” she said.

Dr. Selvan’s project began with educating members of the Primary Care Group at UChicago on risk factors and exam findings that may suggest ILD in patients coming to clinic with nonspecific respiratory complaints. Then they had to equip providers with the ILD Clinician Toolkit and reach patients by handing out the patient questionnaire in primary care clinic.

The next step for this project is going to be dissecting the answers to a postintervention survey that was sent out to understand the practices and comfort of primary care providers in evaluating suspected ILD and the utility of the additional resources created by Dr. Selvan’s team.

“My hope is that we can utilize this partnership with the Primary Care Group to provide the education that’s needed both on the patient side and the provider side, like knowing not to ignore respiratory symptoms, knowing which patients warrant ILD-specific testing, and knowing what the appropriate tests are. In doing so, we can get patients into ILD clinic earlier, confirm their diagnosis, and get them initiated on the appropriate therapies sooner,” she said.

Dr. Selvan credits the quality improvement grant as being fundamental in her success as a fellow and early faculty at UChicago Medicine and believes that this project has created a culture of awareness that wouldn’t have been possible without funding.

“I truly believe that early detection and risk factor modification is the most critical aspect of interstitial lung disease care, and the mechanism for how we’re going to actually improve patient outcomes in our community,” Dr. Selvan said. “Building the infrastructure to accomplish that requires time, resources, and support from institutions and philanthropic foundations that believe in that mission too."

Opportunities like this are made possible by generous contributions from our donors. Make a gift to CHEST and select “Clinical Research” to help aid future research into interstitial lung disease and much more.

Interstitial lung diseases (ILDs), with their many ubiquitous symptoms, are often hard to diagnose in patients. That’s why Amirahwaty Abdullah, MBBS, and Kavitha Selvan, MD, see value in educating clinicians on how to identify and diagnose ILD.

CHEST
Dr. Amirahwaty Abdullah

Both Dr. Abdullah and Dr. Selvan received quality improvement grants from CHEST in October 2023 to do just that. Their projects put the ILD Clinician Toolkit into practice, created as a part of CHEST’s Bridging Specialties™: Timely Diagnosis for ILD initiative aimed at shortening the time to diagnosis.

CHEST
Dr. Kavitha Selvan


Recently, CHEST caught up with the two grant recipients to see how their projects were progressing.

Combating the prevalence of ILDs in West Virginia

Dr. Abdullah and Co-Principal Investigator, Haroon Ahmed, MD, are two of the staff members supporting West Virginia University ILD Clinic, where the ILD Clinician Toolkit is being utilized.

CHEST
Dr. Haroon Ahmed

“ILD is so prevalent here, so we thought it would be an excellent opportunity to do the quality improvement project here because we really do need the resources to improve the care of our [patients with ILD],” Dr. Abdullah said. “For the entire state of West Virginia, we’re the only ILD center.”

In West Virginia, many factors are at play making ILD prevalent, with a recent study showing that the state has the second highest rate of interstitial pulmonary fibrosis, Dr. Abdullah said. This is all due to the economy of the state, the rurality of the population, and the occupational hazards with common jobs like coal mining and farming.

“This topic about bridging the gap and early diagnosis really resonated with us because we see all these patients who end up seeing us after they’ve been on oxygen for years, when they can’t do anything else,” Dr. Ahmed said. “There was a big gap here, and we saw that every day in our clinical practice.”

Since implementing the ILD Clinician Toolkit into their program, the two have started providing these resources to primary care physicians in an effort to help expedite their workups when they see patients with common ILD symptoms. This was done through grand rounds and educational conferences for those practicing in family medicine, internal medicine, and pediatric medicine. And more education is planned for the future.

They have also created working relationships with these departments and have encouraged them to send patients to the ILD Clinic, so patients don’t have to be referred to multiple different physicians.

The next step for the project is to implement telemedicine capabilities, which will allow the team to roll out the patient questionnaire from the toolkit. The questionnaire helps physicians gather a detailed history about their patients, including their past and current medications, surgeries, occupational and environmental exposures, and known comorbidities.

“We definitely want to reach out via telemedicine to patients because, at this point in time, some of our patients travel 3, 4 hours one way just to come see us. So, if we can make it more accessible, we will,” Dr. Abdullah said.

Their plan is to provide iPads that are equipped with the questionnaire and other toolkit resources to the providers. Through this method, the team will be able to see how often the questionnaire is used.

“We are very thankful for this grant because I do think we are saving lives,” Dr. Abdullah said. “Any little thing that we can do to improve the outcomes of these patients who have a rare but difficult-to-treat disease—it’s crucial. This gives us the ability to reach out and help patients who are out of our physical bounds.”
 

 

 

Diagnosing ILD among underrepresented minority populations in Chicago

Dr. Kavitha Selvan is conducting her quality improvement research project at the University of Chicago, alongside her team.

“The community we serve in South Chicago houses a significant number of underrepresented [minority populations],” she said.

“We know that Black patients with ILD experience higher rates of hospitalization, compared with White patients,” she said. “And they are hospitalized, require lung transplants, and die at a younger age too.”

In clinical practice, Dr. Selvan has noticed a trend of patients being referred for evaluation of possible ILD later and later. In some cases, several years go by before the appropriate work for ILD is initiated, and that valuable time lost leads to irreversible loss of lung function.

Dr. Selvan’s plan is to partner with primary care providers who are seeing these patients first in order to expedite the work-up and referral of ILD when appropriate and, ultimately, reduce the amount of time that passes between symptom onset and definitive diagnosis.

“To me, this grant and the resources it provided represented an important opportunity to improve outcomes in the high-risk patients we care for through early disease recognition and treatment,” she said.

Dr. Selvan’s project began with educating members of the Primary Care Group at UChicago on risk factors and exam findings that may suggest ILD in patients coming to clinic with nonspecific respiratory complaints. Then they had to equip providers with the ILD Clinician Toolkit and reach patients by handing out the patient questionnaire in primary care clinic.

The next step for this project is going to be dissecting the answers to a postintervention survey that was sent out to understand the practices and comfort of primary care providers in evaluating suspected ILD and the utility of the additional resources created by Dr. Selvan’s team.

“My hope is that we can utilize this partnership with the Primary Care Group to provide the education that’s needed both on the patient side and the provider side, like knowing not to ignore respiratory symptoms, knowing which patients warrant ILD-specific testing, and knowing what the appropriate tests are. In doing so, we can get patients into ILD clinic earlier, confirm their diagnosis, and get them initiated on the appropriate therapies sooner,” she said.

Dr. Selvan credits the quality improvement grant as being fundamental in her success as a fellow and early faculty at UChicago Medicine and believes that this project has created a culture of awareness that wouldn’t have been possible without funding.

“I truly believe that early detection and risk factor modification is the most critical aspect of interstitial lung disease care, and the mechanism for how we’re going to actually improve patient outcomes in our community,” Dr. Selvan said. “Building the infrastructure to accomplish that requires time, resources, and support from institutions and philanthropic foundations that believe in that mission too."

Opportunities like this are made possible by generous contributions from our donors. Make a gift to CHEST and select “Clinical Research” to help aid future research into interstitial lung disease and much more.

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Top reads from the CHEST journal portfolio

Article Type
Changed
Thu, 08/01/2024 - 12:03
Display Headline
Top reads from the CHEST journal portfolio

Studying endotypes in sleep apnea, diagnostic testing in ILD, and inhaled corticosteroid use in children

 

Journal CHEST®

Differences in Physiologic Endotypes Between Nonpositional and Positional OSA: Results From the Shanghai Sleep Health Study Cohort

By Xiaoting Wang, MM, and colleagues

This study from the Shanghai Sleep Health Study cohort enhances our understanding of physiologic endotypes in sleep apnea—nonpositional OSA (NPOSA) and positional OSA (POSA). The study found that nonanatomic traits play a more significant role in POSA severity. Patients with POSA exhibited lower arousal thresholds and greater ventilatory compensation, while anatomical abnormalities were more prominent in NPOSA. This research underscores the need for personalized treatment strategies based on specific endotypes, especially with the increasing emergence of alternatives to CPAP therapy, such as mandibular advancement therapy and hypoglossal nerve stimulation. Patients with obesity with NPOSA were found to have elevated loop gain, which is known to be impacted by weight loss. This is particularly relevant in light of recent studies showing that new GLP-1 agonists can dramatically reduce sleep apnea severity. Future research should focus on refining these personalized approaches and integrating alternative treatments to provide a more individualized approach to managing sleep apnea.

Dr. Shyamsunder Subramanian, Sutter Gould Medical Foundation, Tracy, Calif.
CHEST
Dr. Shyam Subramanian

– Commentary by Shyam Subramanian, MD, FCCP, Member of the CHEST Physician Editorial Board
 

CHEST® Pulmonary

Employment of the Envisia Genomic Classifier in Conjunction With Cryobiopsy in Patients With Undiagnosed Interstitial Lung Disease

By Fayez Kheir, MD, and colleagues

The novel mRNA genomic array, Envisia Genomic Classifier (EGC), used in this study has been shown to be highly specific for a histopathologic diagnosis of usual interstitial pneumonia (UIP). A diagnosis of UIP carries significant prognostic import as patients exhibit a higher risk for progressive fibrosis and death compared with other interstitial lung diseases (ILDs). In this retrospective study, the combination of bronchoscopic lung cryobiopsy and EGC testing was associated with a significant change in clinical management of those living with an indeterminate ILD. Participants were more likely to be started on antifibrotic therapy following this intervention. This study emphasizes the need for prospective trials on the diagnostic yield of molecular phenotyping with bronchoscopic lung biopsy compared with the traditional surgical approach for the undifferentiated ILD.

CHEST
Dr. Michael Marll

– Commentary by Michael Marll, MD, Member of the CHEST Physician Editorial Board


CHEST® Critical Care

Inhaled Corticosteroids Use Before Hospitalization May Be Protective in Children With Direct Lung Injury

By Elizabeth Landzberg, MD, and colleagues

Respiratory illness is the most common reason for both hospitalization and ICU admission in children. Studies suggest inhaled corticosteroids (ICSs) may protect adults with direct lung injury (DLI) from developing respiratory failure. However, there is a paucity of literature on this therapeutic intervention within the pediatric population. This retrospective, large, single-center study identified children seeking treatment at the emergency department (ED) with DLI before hospitalization. ICS use before hospitalization was associated with one-half the odds of escalation to intubation and noninvasive respiratory support (NRS) in pediatric patients seeking treatment in the ED with DLI. The protective effect was greatest in patients with a history of asthma. According to the authors, this is the first study to assess ICS exposure before hospitalization and progression to respiratory failure in all-cause pediatric DLI. It highlights the need for future studies to evaluate whether a role exists for early prophylactic ICS use in pediatric DLI without status asthmaticus, stratified by history of asthma.

Dr. Anne C. Coates, pediatric pulmonology, Portland, Me.
CHEST
Dr. Anne C. Coates

– Commentary by Anne C. Coates, MD, FCCP, Member of the CHEST Physician Editorial Board

Publications
Topics
Sections

Studying endotypes in sleep apnea, diagnostic testing in ILD, and inhaled corticosteroid use in children

Studying endotypes in sleep apnea, diagnostic testing in ILD, and inhaled corticosteroid use in children

 

Journal CHEST®

Differences in Physiologic Endotypes Between Nonpositional and Positional OSA: Results From the Shanghai Sleep Health Study Cohort

By Xiaoting Wang, MM, and colleagues

This study from the Shanghai Sleep Health Study cohort enhances our understanding of physiologic endotypes in sleep apnea—nonpositional OSA (NPOSA) and positional OSA (POSA). The study found that nonanatomic traits play a more significant role in POSA severity. Patients with POSA exhibited lower arousal thresholds and greater ventilatory compensation, while anatomical abnormalities were more prominent in NPOSA. This research underscores the need for personalized treatment strategies based on specific endotypes, especially with the increasing emergence of alternatives to CPAP therapy, such as mandibular advancement therapy and hypoglossal nerve stimulation. Patients with obesity with NPOSA were found to have elevated loop gain, which is known to be impacted by weight loss. This is particularly relevant in light of recent studies showing that new GLP-1 agonists can dramatically reduce sleep apnea severity. Future research should focus on refining these personalized approaches and integrating alternative treatments to provide a more individualized approach to managing sleep apnea.

Dr. Shyamsunder Subramanian, Sutter Gould Medical Foundation, Tracy, Calif.
CHEST
Dr. Shyam Subramanian

– Commentary by Shyam Subramanian, MD, FCCP, Member of the CHEST Physician Editorial Board
 

CHEST® Pulmonary

Employment of the Envisia Genomic Classifier in Conjunction With Cryobiopsy in Patients With Undiagnosed Interstitial Lung Disease

By Fayez Kheir, MD, and colleagues

The novel mRNA genomic array, Envisia Genomic Classifier (EGC), used in this study has been shown to be highly specific for a histopathologic diagnosis of usual interstitial pneumonia (UIP). A diagnosis of UIP carries significant prognostic import as patients exhibit a higher risk for progressive fibrosis and death compared with other interstitial lung diseases (ILDs). In this retrospective study, the combination of bronchoscopic lung cryobiopsy and EGC testing was associated with a significant change in clinical management of those living with an indeterminate ILD. Participants were more likely to be started on antifibrotic therapy following this intervention. This study emphasizes the need for prospective trials on the diagnostic yield of molecular phenotyping with bronchoscopic lung biopsy compared with the traditional surgical approach for the undifferentiated ILD.

CHEST
Dr. Michael Marll

– Commentary by Michael Marll, MD, Member of the CHEST Physician Editorial Board


CHEST® Critical Care

Inhaled Corticosteroids Use Before Hospitalization May Be Protective in Children With Direct Lung Injury

By Elizabeth Landzberg, MD, and colleagues

Respiratory illness is the most common reason for both hospitalization and ICU admission in children. Studies suggest inhaled corticosteroids (ICSs) may protect adults with direct lung injury (DLI) from developing respiratory failure. However, there is a paucity of literature on this therapeutic intervention within the pediatric population. This retrospective, large, single-center study identified children seeking treatment at the emergency department (ED) with DLI before hospitalization. ICS use before hospitalization was associated with one-half the odds of escalation to intubation and noninvasive respiratory support (NRS) in pediatric patients seeking treatment in the ED with DLI. The protective effect was greatest in patients with a history of asthma. According to the authors, this is the first study to assess ICS exposure before hospitalization and progression to respiratory failure in all-cause pediatric DLI. It highlights the need for future studies to evaluate whether a role exists for early prophylactic ICS use in pediatric DLI without status asthmaticus, stratified by history of asthma.

Dr. Anne C. Coates, pediatric pulmonology, Portland, Me.
CHEST
Dr. Anne C. Coates

– Commentary by Anne C. Coates, MD, FCCP, Member of the CHEST Physician Editorial Board

 

Journal CHEST®

Differences in Physiologic Endotypes Between Nonpositional and Positional OSA: Results From the Shanghai Sleep Health Study Cohort

By Xiaoting Wang, MM, and colleagues

This study from the Shanghai Sleep Health Study cohort enhances our understanding of physiologic endotypes in sleep apnea—nonpositional OSA (NPOSA) and positional OSA (POSA). The study found that nonanatomic traits play a more significant role in POSA severity. Patients with POSA exhibited lower arousal thresholds and greater ventilatory compensation, while anatomical abnormalities were more prominent in NPOSA. This research underscores the need for personalized treatment strategies based on specific endotypes, especially with the increasing emergence of alternatives to CPAP therapy, such as mandibular advancement therapy and hypoglossal nerve stimulation. Patients with obesity with NPOSA were found to have elevated loop gain, which is known to be impacted by weight loss. This is particularly relevant in light of recent studies showing that new GLP-1 agonists can dramatically reduce sleep apnea severity. Future research should focus on refining these personalized approaches and integrating alternative treatments to provide a more individualized approach to managing sleep apnea.

Dr. Shyamsunder Subramanian, Sutter Gould Medical Foundation, Tracy, Calif.
CHEST
Dr. Shyam Subramanian

– Commentary by Shyam Subramanian, MD, FCCP, Member of the CHEST Physician Editorial Board
 

CHEST® Pulmonary

Employment of the Envisia Genomic Classifier in Conjunction With Cryobiopsy in Patients With Undiagnosed Interstitial Lung Disease

By Fayez Kheir, MD, and colleagues

The novel mRNA genomic array, Envisia Genomic Classifier (EGC), used in this study has been shown to be highly specific for a histopathologic diagnosis of usual interstitial pneumonia (UIP). A diagnosis of UIP carries significant prognostic import as patients exhibit a higher risk for progressive fibrosis and death compared with other interstitial lung diseases (ILDs). In this retrospective study, the combination of bronchoscopic lung cryobiopsy and EGC testing was associated with a significant change in clinical management of those living with an indeterminate ILD. Participants were more likely to be started on antifibrotic therapy following this intervention. This study emphasizes the need for prospective trials on the diagnostic yield of molecular phenotyping with bronchoscopic lung biopsy compared with the traditional surgical approach for the undifferentiated ILD.

CHEST
Dr. Michael Marll

– Commentary by Michael Marll, MD, Member of the CHEST Physician Editorial Board


CHEST® Critical Care

Inhaled Corticosteroids Use Before Hospitalization May Be Protective in Children With Direct Lung Injury

By Elizabeth Landzberg, MD, and colleagues

Respiratory illness is the most common reason for both hospitalization and ICU admission in children. Studies suggest inhaled corticosteroids (ICSs) may protect adults with direct lung injury (DLI) from developing respiratory failure. However, there is a paucity of literature on this therapeutic intervention within the pediatric population. This retrospective, large, single-center study identified children seeking treatment at the emergency department (ED) with DLI before hospitalization. ICS use before hospitalization was associated with one-half the odds of escalation to intubation and noninvasive respiratory support (NRS) in pediatric patients seeking treatment in the ED with DLI. The protective effect was greatest in patients with a history of asthma. According to the authors, this is the first study to assess ICS exposure before hospitalization and progression to respiratory failure in all-cause pediatric DLI. It highlights the need for future studies to evaluate whether a role exists for early prophylactic ICS use in pediatric DLI without status asthmaticus, stratified by history of asthma.

Dr. Anne C. Coates, pediatric pulmonology, Portland, Me.
CHEST
Dr. Anne C. Coates

– Commentary by Anne C. Coates, MD, FCCP, Member of the CHEST Physician Editorial Board

Publications
Publications
Topics
Article Type
Display Headline
Top reads from the CHEST journal portfolio
Display Headline
Top reads from the CHEST journal portfolio
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

APPs and POCUS: Overcoming credentialing challenges

Article Type
Changed
Thu, 08/01/2024 - 13:37

Rhea Votipka, NP
CHEST
Rhea Votipka

APP INTERSECTION

Advanced practice providers (APPs) play an integral role in the care and management of patients both in the ICU and across the spectrum of health care. Due to reduced residency hours and the coming physician shortage, APPs are playing, and will continue to play, a greater role in the care and management of critically ill patients.

Point of care ultrasound (POCUS) is a key diagnostic modality that promotes rapid diagnosis, shortens time to key interventions, and allows for interval reassessment. However, the benefit relies heavily on operator skill to obtain good quality images, interpret this within the context of the patient, and know what interventions, if any, to make. It is imperative that APPs, given their role within critical care, become not only familiar with POCUS but demonstrate proficiency in practice.

Before this can become standard, there are a number of challenges to overcome. Educational institutions are behind on integrating POCUS into the APP curriculum.1 Additionally, APPs have few opportunities to develop these skills as residencies and fellowships are not required prior to entering the workforce. Instead, for a majority of APPs, POCUS training relies on nonstandardized on-the-job training or an (typically expensive) off-site training, for which they may not receive reimbursement. Other barriers include cultural practices that limit POCUS use by APPs; inability to upload and document images obtained; lack of willing, skilled mentors with time to provide feedback and assess for clinical competence; and even accessibility of ultrasound machines.2

Despite these challenges, the clinical benefits of utilizing ultrasound in practice necessitates overcoming these barriers. Institutionally, when APPs perform POCUS, it can lead to more rapid diagnosis and triage of patients, provide billing opportunities, and may reduce overall costs of care. From an APP perspective, having formalized training and credentialing can lead to more consistent POCUS assessment between providers, comprehensive patient care, security in one’s skills and knowledge base, and a tangible way of communicating one’s credibility between institutions.

While some institutions have overcome these barriers and APPs are trained and credentialed in POCUS, this is not always the norm. For this to come to fruition, we need educators to incorporate this into curriculum, colleagues who are willing to mentor learners, cultural changes to allow APPs to develop and utilize necessary skills for practice, clear structure for obtaining and maintaining competency, physicians to champion these efforts, and institutions to credential skilled APPs. Until then, it is important that we as APPs seek to facilitate these institutional changes and continue to set and maintain high quality standards for ourselves.


CHEST 2024 is for APPs too

With more than 300 educational sessions on the CHEST 2024 program, it can be overwhelming to figure out which ones you should put on your schedule. That’s why CHEST asked Danielle McCamey, DNP, CRNP, ACNP-BC, FCCP, and LaDonna Brown, DNP, CRNA, of DNPs of Color (DOC), as well as Corinne Young, MSN, FNP-C, FCCP, of the Association of Pulmonary Advanced Practice Providers (APAPP), for their recommendations on sessions that advanced practice providers shouldn’t miss. Visit chestnet.org/annual-meeting-apps for all of their recommendations.


References

1. Rudy S, Widmar B, Wilbeck J. Ultrasound education for nurse practitioner students: strategies for curricular integration. J Nurse Pract. 2024;20(6):104986.

2. Resnyk J, Weichold A. Barriers to learning and performing point-of-care ultrasound (POCUS): An integrative review. J Prof Nursing. 2024;54:54-62.




 

Publications
Topics
Sections

Rhea Votipka, NP
CHEST
Rhea Votipka

APP INTERSECTION

Advanced practice providers (APPs) play an integral role in the care and management of patients both in the ICU and across the spectrum of health care. Due to reduced residency hours and the coming physician shortage, APPs are playing, and will continue to play, a greater role in the care and management of critically ill patients.

Point of care ultrasound (POCUS) is a key diagnostic modality that promotes rapid diagnosis, shortens time to key interventions, and allows for interval reassessment. However, the benefit relies heavily on operator skill to obtain good quality images, interpret this within the context of the patient, and know what interventions, if any, to make. It is imperative that APPs, given their role within critical care, become not only familiar with POCUS but demonstrate proficiency in practice.

Before this can become standard, there are a number of challenges to overcome. Educational institutions are behind on integrating POCUS into the APP curriculum.1 Additionally, APPs have few opportunities to develop these skills as residencies and fellowships are not required prior to entering the workforce. Instead, for a majority of APPs, POCUS training relies on nonstandardized on-the-job training or an (typically expensive) off-site training, for which they may not receive reimbursement. Other barriers include cultural practices that limit POCUS use by APPs; inability to upload and document images obtained; lack of willing, skilled mentors with time to provide feedback and assess for clinical competence; and even accessibility of ultrasound machines.2

Despite these challenges, the clinical benefits of utilizing ultrasound in practice necessitates overcoming these barriers. Institutionally, when APPs perform POCUS, it can lead to more rapid diagnosis and triage of patients, provide billing opportunities, and may reduce overall costs of care. From an APP perspective, having formalized training and credentialing can lead to more consistent POCUS assessment between providers, comprehensive patient care, security in one’s skills and knowledge base, and a tangible way of communicating one’s credibility between institutions.

While some institutions have overcome these barriers and APPs are trained and credentialed in POCUS, this is not always the norm. For this to come to fruition, we need educators to incorporate this into curriculum, colleagues who are willing to mentor learners, cultural changes to allow APPs to develop and utilize necessary skills for practice, clear structure for obtaining and maintaining competency, physicians to champion these efforts, and institutions to credential skilled APPs. Until then, it is important that we as APPs seek to facilitate these institutional changes and continue to set and maintain high quality standards for ourselves.


CHEST 2024 is for APPs too

With more than 300 educational sessions on the CHEST 2024 program, it can be overwhelming to figure out which ones you should put on your schedule. That’s why CHEST asked Danielle McCamey, DNP, CRNP, ACNP-BC, FCCP, and LaDonna Brown, DNP, CRNA, of DNPs of Color (DOC), as well as Corinne Young, MSN, FNP-C, FCCP, of the Association of Pulmonary Advanced Practice Providers (APAPP), for their recommendations on sessions that advanced practice providers shouldn’t miss. Visit chestnet.org/annual-meeting-apps for all of their recommendations.


References

1. Rudy S, Widmar B, Wilbeck J. Ultrasound education for nurse practitioner students: strategies for curricular integration. J Nurse Pract. 2024;20(6):104986.

2. Resnyk J, Weichold A. Barriers to learning and performing point-of-care ultrasound (POCUS): An integrative review. J Prof Nursing. 2024;54:54-62.




 

Rhea Votipka, NP
CHEST
Rhea Votipka

APP INTERSECTION

Advanced practice providers (APPs) play an integral role in the care and management of patients both in the ICU and across the spectrum of health care. Due to reduced residency hours and the coming physician shortage, APPs are playing, and will continue to play, a greater role in the care and management of critically ill patients.

Point of care ultrasound (POCUS) is a key diagnostic modality that promotes rapid diagnosis, shortens time to key interventions, and allows for interval reassessment. However, the benefit relies heavily on operator skill to obtain good quality images, interpret this within the context of the patient, and know what interventions, if any, to make. It is imperative that APPs, given their role within critical care, become not only familiar with POCUS but demonstrate proficiency in practice.

Before this can become standard, there are a number of challenges to overcome. Educational institutions are behind on integrating POCUS into the APP curriculum.1 Additionally, APPs have few opportunities to develop these skills as residencies and fellowships are not required prior to entering the workforce. Instead, for a majority of APPs, POCUS training relies on nonstandardized on-the-job training or an (typically expensive) off-site training, for which they may not receive reimbursement. Other barriers include cultural practices that limit POCUS use by APPs; inability to upload and document images obtained; lack of willing, skilled mentors with time to provide feedback and assess for clinical competence; and even accessibility of ultrasound machines.2

Despite these challenges, the clinical benefits of utilizing ultrasound in practice necessitates overcoming these barriers. Institutionally, when APPs perform POCUS, it can lead to more rapid diagnosis and triage of patients, provide billing opportunities, and may reduce overall costs of care. From an APP perspective, having formalized training and credentialing can lead to more consistent POCUS assessment between providers, comprehensive patient care, security in one’s skills and knowledge base, and a tangible way of communicating one’s credibility between institutions.

While some institutions have overcome these barriers and APPs are trained and credentialed in POCUS, this is not always the norm. For this to come to fruition, we need educators to incorporate this into curriculum, colleagues who are willing to mentor learners, cultural changes to allow APPs to develop and utilize necessary skills for practice, clear structure for obtaining and maintaining competency, physicians to champion these efforts, and institutions to credential skilled APPs. Until then, it is important that we as APPs seek to facilitate these institutional changes and continue to set and maintain high quality standards for ourselves.


CHEST 2024 is for APPs too

With more than 300 educational sessions on the CHEST 2024 program, it can be overwhelming to figure out which ones you should put on your schedule. That’s why CHEST asked Danielle McCamey, DNP, CRNP, ACNP-BC, FCCP, and LaDonna Brown, DNP, CRNA, of DNPs of Color (DOC), as well as Corinne Young, MSN, FNP-C, FCCP, of the Association of Pulmonary Advanced Practice Providers (APAPP), for their recommendations on sessions that advanced practice providers shouldn’t miss. Visit chestnet.org/annual-meeting-apps for all of their recommendations.


References

1. Rudy S, Widmar B, Wilbeck J. Ultrasound education for nurse practitioner students: strategies for curricular integration. J Nurse Pract. 2024;20(6):104986.

2. Resnyk J, Weichold A. Barriers to learning and performing point-of-care ultrasound (POCUS): An integrative review. J Prof Nursing. 2024;54:54-62.




 

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

In memoriam: Dr. James E. Dalen

Article Type
Changed
Thu, 08/01/2024 - 10:09

Dr. James E. Dalen
CHEST
Dr. James E. Dalen

CHEST was recently informed of the death of former CHEST President, James E. Dalen, MD, MPH, Master FCCP. He served as CHEST President from 1985 to 1986.

Dr. Dalen was a graduate of Washington State University. (His undergraduate education was temporarily interrupted when he served as a Corpsman in the Navy and Marines.) He held a master’s degree from the University of Michigan, an MD from the University of Washington, an MPH from Harvard University, and an honorary Doctor of Science degree from the University of Massachusetts. He was internationally recognized as a medical visionary and esteemed cardiologist.

Dr. Dalen’s academic career spanned 3 medical schools. During the time he spent at each, he achieved great things and nurtured great ideas that helped transform medicine. From 1967 to 1975, he was on the faculty of Harvard Medical School (and the Peter Bent Brigham Hospital). From 1975 to 1988, he was a faculty member at the University of Massachusetts Medical School where he served as Chairman of Cardiovascular Medicine from 1975 to 1977 and as Chairman of Medicine from 1977 to 1988. From 1986 to 1987, he served as Interim Chancellor of the University of Massachusetts at the Worcester Campus. Dr. Dalen left Massachusetts in 1988 to become the Dean of the University of Arizona College of Medicine.

During his tenure as Dean and Vice-President at the University of Arizona, Dr. Dalen oversaw the establishment of the Zuckerman College of Public Health, the Arizona Center for Integrative Medicine, and the Arizona Telemedicine Program. Successful fundraising under his leadership led to the establishment of new research facilities, including the Children’s Research Center, the Sarver Heart Center, the Arizona Arthritis Center, and a major expansion of the Arizona Cancer Center. While at the University of Arizona College of Medicine, Dr. Dalen recognized the importance of integrative medicine and the need to promote integrative medicine in the academic environment. With the active participation of Dr. Andrew Weil, this led to the creation of the Academic Consortium for Integrative Medicine and Health and the Arizona Center for Integrative Medicine.

Dr. Dalen received many teaching awards. In 1987, he received the Distinguished Public Service Award from the University of Massachusetts. In 1988, he was named the University of Washington Distinguished Medical Alumnus of the Year and received the Alumni Achievement Award from Washington State University. In 2000, he received the College Medal from CHEST and was named a Master Fellow. In 2010, he was awarded the Harvard School of Public Health’s highest honor for its alumni: the 2010 Alumni Award of Merit. In 2012, he was named a Master Fellow of the American College of Physicians and was awarded an honorary Doctor of Science degree by the University of Massachusetts in 2013. In 2015, he received the Bravewell Distinguished Service Award from the Academic Consortium for Integrative Medicine and Health for his role as one of the founders of the consortium and as one of the founders of the Arizona Center for Integrative Medicine at the University of Arizona.

In his spare time, he enjoyed reading, gardening, sailing, art, and music and was a lifelong student with a passion for the pursuit of knowledge. Dr. Dalen enjoyed watching basketball, particularly the Arizona Wildcats. His favorite place to spend time was by the ocean in Coronado, California. Dr. Dalen was an advocate for public health, social justice, and health care reform throughout his life and career. He mentored thousands of medical students and championed access to health care for populations who were underserved. He touched many lives as a leading physician and caring friend.

Dr. Dalen is survived by his devoted wife, Priscilla M. Dalen (Dunton); loving children, James E. Dalen, Jr, and Angela M. Snodgrass (Dalen); daughter-in-law, Suzan M. Dalen; son-in-law, Daniel N. Snodgrass; and many step-grandchildren. We remember our colleague and extend our sincere condolences.

Publications
Topics
Sections

Dr. James E. Dalen
CHEST
Dr. James E. Dalen

CHEST was recently informed of the death of former CHEST President, James E. Dalen, MD, MPH, Master FCCP. He served as CHEST President from 1985 to 1986.

Dr. Dalen was a graduate of Washington State University. (His undergraduate education was temporarily interrupted when he served as a Corpsman in the Navy and Marines.) He held a master’s degree from the University of Michigan, an MD from the University of Washington, an MPH from Harvard University, and an honorary Doctor of Science degree from the University of Massachusetts. He was internationally recognized as a medical visionary and esteemed cardiologist.

Dr. Dalen’s academic career spanned 3 medical schools. During the time he spent at each, he achieved great things and nurtured great ideas that helped transform medicine. From 1967 to 1975, he was on the faculty of Harvard Medical School (and the Peter Bent Brigham Hospital). From 1975 to 1988, he was a faculty member at the University of Massachusetts Medical School where he served as Chairman of Cardiovascular Medicine from 1975 to 1977 and as Chairman of Medicine from 1977 to 1988. From 1986 to 1987, he served as Interim Chancellor of the University of Massachusetts at the Worcester Campus. Dr. Dalen left Massachusetts in 1988 to become the Dean of the University of Arizona College of Medicine.

During his tenure as Dean and Vice-President at the University of Arizona, Dr. Dalen oversaw the establishment of the Zuckerman College of Public Health, the Arizona Center for Integrative Medicine, and the Arizona Telemedicine Program. Successful fundraising under his leadership led to the establishment of new research facilities, including the Children’s Research Center, the Sarver Heart Center, the Arizona Arthritis Center, and a major expansion of the Arizona Cancer Center. While at the University of Arizona College of Medicine, Dr. Dalen recognized the importance of integrative medicine and the need to promote integrative medicine in the academic environment. With the active participation of Dr. Andrew Weil, this led to the creation of the Academic Consortium for Integrative Medicine and Health and the Arizona Center for Integrative Medicine.

Dr. Dalen received many teaching awards. In 1987, he received the Distinguished Public Service Award from the University of Massachusetts. In 1988, he was named the University of Washington Distinguished Medical Alumnus of the Year and received the Alumni Achievement Award from Washington State University. In 2000, he received the College Medal from CHEST and was named a Master Fellow. In 2010, he was awarded the Harvard School of Public Health’s highest honor for its alumni: the 2010 Alumni Award of Merit. In 2012, he was named a Master Fellow of the American College of Physicians and was awarded an honorary Doctor of Science degree by the University of Massachusetts in 2013. In 2015, he received the Bravewell Distinguished Service Award from the Academic Consortium for Integrative Medicine and Health for his role as one of the founders of the consortium and as one of the founders of the Arizona Center for Integrative Medicine at the University of Arizona.

In his spare time, he enjoyed reading, gardening, sailing, art, and music and was a lifelong student with a passion for the pursuit of knowledge. Dr. Dalen enjoyed watching basketball, particularly the Arizona Wildcats. His favorite place to spend time was by the ocean in Coronado, California. Dr. Dalen was an advocate for public health, social justice, and health care reform throughout his life and career. He mentored thousands of medical students and championed access to health care for populations who were underserved. He touched many lives as a leading physician and caring friend.

Dr. Dalen is survived by his devoted wife, Priscilla M. Dalen (Dunton); loving children, James E. Dalen, Jr, and Angela M. Snodgrass (Dalen); daughter-in-law, Suzan M. Dalen; son-in-law, Daniel N. Snodgrass; and many step-grandchildren. We remember our colleague and extend our sincere condolences.

Dr. James E. Dalen
CHEST
Dr. James E. Dalen

CHEST was recently informed of the death of former CHEST President, James E. Dalen, MD, MPH, Master FCCP. He served as CHEST President from 1985 to 1986.

Dr. Dalen was a graduate of Washington State University. (His undergraduate education was temporarily interrupted when he served as a Corpsman in the Navy and Marines.) He held a master’s degree from the University of Michigan, an MD from the University of Washington, an MPH from Harvard University, and an honorary Doctor of Science degree from the University of Massachusetts. He was internationally recognized as a medical visionary and esteemed cardiologist.

Dr. Dalen’s academic career spanned 3 medical schools. During the time he spent at each, he achieved great things and nurtured great ideas that helped transform medicine. From 1967 to 1975, he was on the faculty of Harvard Medical School (and the Peter Bent Brigham Hospital). From 1975 to 1988, he was a faculty member at the University of Massachusetts Medical School where he served as Chairman of Cardiovascular Medicine from 1975 to 1977 and as Chairman of Medicine from 1977 to 1988. From 1986 to 1987, he served as Interim Chancellor of the University of Massachusetts at the Worcester Campus. Dr. Dalen left Massachusetts in 1988 to become the Dean of the University of Arizona College of Medicine.

During his tenure as Dean and Vice-President at the University of Arizona, Dr. Dalen oversaw the establishment of the Zuckerman College of Public Health, the Arizona Center for Integrative Medicine, and the Arizona Telemedicine Program. Successful fundraising under his leadership led to the establishment of new research facilities, including the Children’s Research Center, the Sarver Heart Center, the Arizona Arthritis Center, and a major expansion of the Arizona Cancer Center. While at the University of Arizona College of Medicine, Dr. Dalen recognized the importance of integrative medicine and the need to promote integrative medicine in the academic environment. With the active participation of Dr. Andrew Weil, this led to the creation of the Academic Consortium for Integrative Medicine and Health and the Arizona Center for Integrative Medicine.

Dr. Dalen received many teaching awards. In 1987, he received the Distinguished Public Service Award from the University of Massachusetts. In 1988, he was named the University of Washington Distinguished Medical Alumnus of the Year and received the Alumni Achievement Award from Washington State University. In 2000, he received the College Medal from CHEST and was named a Master Fellow. In 2010, he was awarded the Harvard School of Public Health’s highest honor for its alumni: the 2010 Alumni Award of Merit. In 2012, he was named a Master Fellow of the American College of Physicians and was awarded an honorary Doctor of Science degree by the University of Massachusetts in 2013. In 2015, he received the Bravewell Distinguished Service Award from the Academic Consortium for Integrative Medicine and Health for his role as one of the founders of the consortium and as one of the founders of the Arizona Center for Integrative Medicine at the University of Arizona.

In his spare time, he enjoyed reading, gardening, sailing, art, and music and was a lifelong student with a passion for the pursuit of knowledge. Dr. Dalen enjoyed watching basketball, particularly the Arizona Wildcats. His favorite place to spend time was by the ocean in Coronado, California. Dr. Dalen was an advocate for public health, social justice, and health care reform throughout his life and career. He mentored thousands of medical students and championed access to health care for populations who were underserved. He touched many lives as a leading physician and caring friend.

Dr. Dalen is survived by his devoted wife, Priscilla M. Dalen (Dunton); loving children, James E. Dalen, Jr, and Angela M. Snodgrass (Dalen); daughter-in-law, Suzan M. Dalen; son-in-law, Daniel N. Snodgrass; and many step-grandchildren. We remember our colleague and extend our sincere condolences.

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Will Hospital-at-Home Go Mainstream?

Article Type
Changed
Wed, 07/31/2024 - 12:26

Jordan Stohler, a 42-year-old nurse in Knoxville, Tennessee, was readmitted to Fort Sanders Medical Center in June 2023 with sepsis after a double mastectomy. 

She spent 5 days in the hospital after surgery to clear up the infection. Then she was offered a choice: She could either stay in the hospital while she received IV antibiotics, or she could go home and have the antibiotics given to her there under the Advanced Care at Home program of Covenant Health, the nine-hospital system to which Fort Sanders belongs.

She opted to go home, where she knew she’d be more comfortable and would be close to her beloved dog. In the end, she was very glad she did. 

“I received great care in the hospital, but to be allowed to be in the comfort of your own home, to be around my dog, who I think is therapeutic, to be able to cook my own meals, and to have the same one-on-one nursing care that I would have gotten in the hospital was great,” Ms. Stohler said. “

Being cared for at home helped her heal, she said. “I probably would have gotten a little stir crazy if I’d stayed in the hospital any longer. I received excellent care at home.”

Covenant’s Advanced Care at Home program is an example of the hospital-at-home trend that has been growing rapidly since Medicare began reimbursing hospitals for this approach during the COVID pandemic. Currently, 322 hospitals in 37 states have Medicare waivers for these kinds of programs, although not all of them are currently functioning.

A recent survey published in JAMA found that nearly half of consumers would accept hospital-at-home, and more than a third were neutral on it. Only 17% said they’d rather be cared for in a brick-and-mortar hospital. 

The findings of the JAMA survey confirm those of earlier studies, said Bruce Leff, MD, a professor at Johns Hopkins Medical School in Baltimore, who has researched hospital-at-home since the 1990s. Like the new study, those trials found that the results had no relationship to individual traits, such as socioeconomic status, medical conditions, age, gender, or race. 

Whether a person felt comfortable with the idea of hospital-at-home boiled down “to a preference for receiving care at home or in the hospital,” he said. Some people distrust hospitals, and others feel insecure about receiving care at home, even if it is provided by qualified health care professionals.
 

How Patients Are Selected 

While the details of hospital-at-home vary from program to program, the basic scenario is that patients who need certain kinds of acute care can be sent home from hospitals, emergency departments, or clinics to receive that care at home. Among the kinds of conditions that make stable patients eligible are heart failure, COPD, pneumonia, cellulitis, and COVID-19, said John Busigin, MD, a hospitalist and medical director of Covenant Advanced Care at Home. 

When a patient is admitted to hospital-at-home, the hospital will send along whatever equipment and medications that person needs. In some cases, this may include a hospital bed, although Ms. Stohler used her own. An IV line was put into her arm, and the IV stand was placed next to the bed. 

Ms. Stohler received a computer tablet that she used to communicate with doctors and nurses in Covenant’s “command center” in Knoxville. She also wore a watch with a button she could push in case of an emergency. And she had a telephone line that went directly to her medical team, in case she had an issue and the tablet didn’t work.

Twice a day, or as needed, specially trained paramedics came to Ms. Stohler’s home. They checked on the IV line, changed the IV bag, performed tests, and uploaded vital signs from monitoring equipment to Ms. Stohler’s tablet so it could be transmitted to the command center. A physician assistant came in on the second and fourth days of her weeklong stay in the program, and she saw a hospitalist remotely every day.

While some hospital-at-home programs have registered nurses visit patients at home, RNs are in short supply. To fill this gap, Covenant’s program uses community paramedics who have been in the field for at least 5 years, doing everything from intubating patients and placing them on ventilators to providing advanced cardiac life support, Dr. Busigin said. To get certified as community paramedics, they go through a 3-month training program.

Shortly after Ms. Stohler went into hospital-at-home, she had another crisis. Excess fluid had built up in her body because of all the IV fluids she’d received in the hospital while fighting the sepsis. As a result, she became short of breath. If she had been discharged to home rather than hospital-at-home, she said, she would have had to go to the emergency room. Instead, she sent out a distress call. One of the paramedics rushed to her house and gave her an IV diuretic medication, which helped her urinate to get rid of the excess fluid.

A small number of the estimated 300 people who have gone through the program had to be admitted to the hospital, Dr. Busigin said. Nationally, he said, about 5%-10% are admitted. But readmissions among the patients in the Covenant program have been 25% lower than for patients who received conventional hospital care and had the same conditions as those in hospital-at-home.

Studies have shown that these programs not only reduce readmissions, but also cost less, on average, and create a better patient experience than traditional hospital care does. And, according to the JAMA survey, most consumers like the idea. Fifty-six percent of people who took the survey agreed with the statement that people recover faster at home than in the hospital. Fifty-nine percent agreed they’d feel safe being treated at home, and 49% said they’d be more comfortable if treated at home. 

The 1134 people who took the survey were also asked about their comfort level with providing various kinds of care to their loved ones during a hospital-at-home episode. The results varied with the type of task: For example, 82% of the respondents agreed or strongly agreed they could manage a patient’s medications, while just 41% said they’d be willing to change a feeding tube. Smaller percentages were willing to change an IV bag or a catheter or do wound care.

However, hospital-at-home programs don’t allow caregivers to take part in clinical care, which is prohibited by Medicare waivers and state licensing regulations. None of the 22 health systems that use the hospital-at-home services of Medically Home, including Covenant, ask caregivers to do anything along this line, said Pippa Shulman, DO, medical director of the company, which provides equipment, technology, and protocols for hospital programs

The only exception at Covenant, Dr. Busigin said, is that the hospital may train family members to do wound care when a patient is discharged from the hospital to Advanced Care at Home. They may also prepare meals for their loved ones, although the program provides balanced meals to patients if they want them. Ms. Stohler had some of these meals, which just had to be heated up, for the first few days of hospital-at-home, and later her relatives brought meals to her house.
 

 

 

Challenges for the Future

The number of Medicare hospital-at-home waivers has nearly doubled since 2021. A year earlier, when Medicare began reimbursing hospitals for acute care at home to help them cope with the overflow of COVID patients, there were only about 15-20 programs in the United States, said Dr. Leff of Johns Hopkins.

A big reason for the lack of use before the pandemic, Dr. Leff said, is that there was no payment system for hospitals that offered hospital-at-home. Now, they can get paid by Medicare and 10 state Medicaid programs, and a number of private payers are also coming on board. Ms. Stohler’s private insurer covered her hospital-at-home stay, and Dr. Busigin said several plans that contract with Covenant will pay for it.

Dr. Leff said he’s cautiously optimistic Congress will extend the Medicare waiver program, which is scheduled to end in December, for another 5 years. A couple of key House committees have signed off on a bill to do that, he said, and a Congressional Budget Office report found that the program did not cost Medicare more money. 

But even if the waiver is renewed, some health systems may find it tough to deliver the service. The current version of this model depends a lot on technology, because telemedicine is used and reliable communication is needed for patients in hospital-at-home. That’s why many of the hospitals hire outside vendors like Medically Home to provide the infrastructure they need.

Medically Home manages the tablets given to patients and all connection and networking services, including internet and cellphone connections. It also provides technical services in the command centers that hospitals set up for the doctors and nurses who provide care remotely. 

And the firm figures out how to deliver the standard care for each condition in each hospital-at-home. “We need to make sure that the patient is going to get what they need in the time frame it needs to be delivered in, and that it’s safe and effective for the patient,” Dr. Shulman said. “So we’ve developed logistical protocols for a multitude of disease states that allow us to provide high-acuity care in the home to a variety of complex patients.”

The health care workers use the hospital electronic health record for hospital-at-home patients, and vital signs uploaded from patient tablets flow directly into the electronic health record, she said.
 

Rural Areas Need Help

The use of hospital-at-home in rural areas holds a lot of promise, Dr. Leff said. 

“A lot of rural hospitals have been closing, and hospital-at-home could be a mechanism to create hospital-level care where facilities have closed down. It’s easier to do this in urban areas, but it can be done in rural environments as well.”

Rami Karjian, CEO of Medically Home, agreed. The firm services hospital-at-home programs in rural areas of Oklahoma and California, using cellphones and paramedics in areas that lack broadband connections and nurses, he pointed out. 

“Hospital-at-home can’t just be available to people who live in big cities,” he said. “The access problems in health care are pervasive, and this is part of how we solve access problems in rural areas.”
 

A version of this article first appeared on WebMD.com.

Publications
Topics
Sections

Jordan Stohler, a 42-year-old nurse in Knoxville, Tennessee, was readmitted to Fort Sanders Medical Center in June 2023 with sepsis after a double mastectomy. 

She spent 5 days in the hospital after surgery to clear up the infection. Then she was offered a choice: She could either stay in the hospital while she received IV antibiotics, or she could go home and have the antibiotics given to her there under the Advanced Care at Home program of Covenant Health, the nine-hospital system to which Fort Sanders belongs.

She opted to go home, where she knew she’d be more comfortable and would be close to her beloved dog. In the end, she was very glad she did. 

“I received great care in the hospital, but to be allowed to be in the comfort of your own home, to be around my dog, who I think is therapeutic, to be able to cook my own meals, and to have the same one-on-one nursing care that I would have gotten in the hospital was great,” Ms. Stohler said. “

Being cared for at home helped her heal, she said. “I probably would have gotten a little stir crazy if I’d stayed in the hospital any longer. I received excellent care at home.”

Covenant’s Advanced Care at Home program is an example of the hospital-at-home trend that has been growing rapidly since Medicare began reimbursing hospitals for this approach during the COVID pandemic. Currently, 322 hospitals in 37 states have Medicare waivers for these kinds of programs, although not all of them are currently functioning.

A recent survey published in JAMA found that nearly half of consumers would accept hospital-at-home, and more than a third were neutral on it. Only 17% said they’d rather be cared for in a brick-and-mortar hospital. 

The findings of the JAMA survey confirm those of earlier studies, said Bruce Leff, MD, a professor at Johns Hopkins Medical School in Baltimore, who has researched hospital-at-home since the 1990s. Like the new study, those trials found that the results had no relationship to individual traits, such as socioeconomic status, medical conditions, age, gender, or race. 

Whether a person felt comfortable with the idea of hospital-at-home boiled down “to a preference for receiving care at home or in the hospital,” he said. Some people distrust hospitals, and others feel insecure about receiving care at home, even if it is provided by qualified health care professionals.
 

How Patients Are Selected 

While the details of hospital-at-home vary from program to program, the basic scenario is that patients who need certain kinds of acute care can be sent home from hospitals, emergency departments, or clinics to receive that care at home. Among the kinds of conditions that make stable patients eligible are heart failure, COPD, pneumonia, cellulitis, and COVID-19, said John Busigin, MD, a hospitalist and medical director of Covenant Advanced Care at Home. 

When a patient is admitted to hospital-at-home, the hospital will send along whatever equipment and medications that person needs. In some cases, this may include a hospital bed, although Ms. Stohler used her own. An IV line was put into her arm, and the IV stand was placed next to the bed. 

Ms. Stohler received a computer tablet that she used to communicate with doctors and nurses in Covenant’s “command center” in Knoxville. She also wore a watch with a button she could push in case of an emergency. And she had a telephone line that went directly to her medical team, in case she had an issue and the tablet didn’t work.

Twice a day, or as needed, specially trained paramedics came to Ms. Stohler’s home. They checked on the IV line, changed the IV bag, performed tests, and uploaded vital signs from monitoring equipment to Ms. Stohler’s tablet so it could be transmitted to the command center. A physician assistant came in on the second and fourth days of her weeklong stay in the program, and she saw a hospitalist remotely every day.

While some hospital-at-home programs have registered nurses visit patients at home, RNs are in short supply. To fill this gap, Covenant’s program uses community paramedics who have been in the field for at least 5 years, doing everything from intubating patients and placing them on ventilators to providing advanced cardiac life support, Dr. Busigin said. To get certified as community paramedics, they go through a 3-month training program.

Shortly after Ms. Stohler went into hospital-at-home, she had another crisis. Excess fluid had built up in her body because of all the IV fluids she’d received in the hospital while fighting the sepsis. As a result, she became short of breath. If she had been discharged to home rather than hospital-at-home, she said, she would have had to go to the emergency room. Instead, she sent out a distress call. One of the paramedics rushed to her house and gave her an IV diuretic medication, which helped her urinate to get rid of the excess fluid.

A small number of the estimated 300 people who have gone through the program had to be admitted to the hospital, Dr. Busigin said. Nationally, he said, about 5%-10% are admitted. But readmissions among the patients in the Covenant program have been 25% lower than for patients who received conventional hospital care and had the same conditions as those in hospital-at-home.

Studies have shown that these programs not only reduce readmissions, but also cost less, on average, and create a better patient experience than traditional hospital care does. And, according to the JAMA survey, most consumers like the idea. Fifty-six percent of people who took the survey agreed with the statement that people recover faster at home than in the hospital. Fifty-nine percent agreed they’d feel safe being treated at home, and 49% said they’d be more comfortable if treated at home. 

The 1134 people who took the survey were also asked about their comfort level with providing various kinds of care to their loved ones during a hospital-at-home episode. The results varied with the type of task: For example, 82% of the respondents agreed or strongly agreed they could manage a patient’s medications, while just 41% said they’d be willing to change a feeding tube. Smaller percentages were willing to change an IV bag or a catheter or do wound care.

However, hospital-at-home programs don’t allow caregivers to take part in clinical care, which is prohibited by Medicare waivers and state licensing regulations. None of the 22 health systems that use the hospital-at-home services of Medically Home, including Covenant, ask caregivers to do anything along this line, said Pippa Shulman, DO, medical director of the company, which provides equipment, technology, and protocols for hospital programs

The only exception at Covenant, Dr. Busigin said, is that the hospital may train family members to do wound care when a patient is discharged from the hospital to Advanced Care at Home. They may also prepare meals for their loved ones, although the program provides balanced meals to patients if they want them. Ms. Stohler had some of these meals, which just had to be heated up, for the first few days of hospital-at-home, and later her relatives brought meals to her house.
 

 

 

Challenges for the Future

The number of Medicare hospital-at-home waivers has nearly doubled since 2021. A year earlier, when Medicare began reimbursing hospitals for acute care at home to help them cope with the overflow of COVID patients, there were only about 15-20 programs in the United States, said Dr. Leff of Johns Hopkins.

A big reason for the lack of use before the pandemic, Dr. Leff said, is that there was no payment system for hospitals that offered hospital-at-home. Now, they can get paid by Medicare and 10 state Medicaid programs, and a number of private payers are also coming on board. Ms. Stohler’s private insurer covered her hospital-at-home stay, and Dr. Busigin said several plans that contract with Covenant will pay for it.

Dr. Leff said he’s cautiously optimistic Congress will extend the Medicare waiver program, which is scheduled to end in December, for another 5 years. A couple of key House committees have signed off on a bill to do that, he said, and a Congressional Budget Office report found that the program did not cost Medicare more money. 

But even if the waiver is renewed, some health systems may find it tough to deliver the service. The current version of this model depends a lot on technology, because telemedicine is used and reliable communication is needed for patients in hospital-at-home. That’s why many of the hospitals hire outside vendors like Medically Home to provide the infrastructure they need.

Medically Home manages the tablets given to patients and all connection and networking services, including internet and cellphone connections. It also provides technical services in the command centers that hospitals set up for the doctors and nurses who provide care remotely. 

And the firm figures out how to deliver the standard care for each condition in each hospital-at-home. “We need to make sure that the patient is going to get what they need in the time frame it needs to be delivered in, and that it’s safe and effective for the patient,” Dr. Shulman said. “So we’ve developed logistical protocols for a multitude of disease states that allow us to provide high-acuity care in the home to a variety of complex patients.”

The health care workers use the hospital electronic health record for hospital-at-home patients, and vital signs uploaded from patient tablets flow directly into the electronic health record, she said.
 

Rural Areas Need Help

The use of hospital-at-home in rural areas holds a lot of promise, Dr. Leff said. 

“A lot of rural hospitals have been closing, and hospital-at-home could be a mechanism to create hospital-level care where facilities have closed down. It’s easier to do this in urban areas, but it can be done in rural environments as well.”

Rami Karjian, CEO of Medically Home, agreed. The firm services hospital-at-home programs in rural areas of Oklahoma and California, using cellphones and paramedics in areas that lack broadband connections and nurses, he pointed out. 

“Hospital-at-home can’t just be available to people who live in big cities,” he said. “The access problems in health care are pervasive, and this is part of how we solve access problems in rural areas.”
 

A version of this article first appeared on WebMD.com.

Jordan Stohler, a 42-year-old nurse in Knoxville, Tennessee, was readmitted to Fort Sanders Medical Center in June 2023 with sepsis after a double mastectomy. 

She spent 5 days in the hospital after surgery to clear up the infection. Then she was offered a choice: She could either stay in the hospital while she received IV antibiotics, or she could go home and have the antibiotics given to her there under the Advanced Care at Home program of Covenant Health, the nine-hospital system to which Fort Sanders belongs.

She opted to go home, where she knew she’d be more comfortable and would be close to her beloved dog. In the end, she was very glad she did. 

“I received great care in the hospital, but to be allowed to be in the comfort of your own home, to be around my dog, who I think is therapeutic, to be able to cook my own meals, and to have the same one-on-one nursing care that I would have gotten in the hospital was great,” Ms. Stohler said. “

Being cared for at home helped her heal, she said. “I probably would have gotten a little stir crazy if I’d stayed in the hospital any longer. I received excellent care at home.”

Covenant’s Advanced Care at Home program is an example of the hospital-at-home trend that has been growing rapidly since Medicare began reimbursing hospitals for this approach during the COVID pandemic. Currently, 322 hospitals in 37 states have Medicare waivers for these kinds of programs, although not all of them are currently functioning.

A recent survey published in JAMA found that nearly half of consumers would accept hospital-at-home, and more than a third were neutral on it. Only 17% said they’d rather be cared for in a brick-and-mortar hospital. 

The findings of the JAMA survey confirm those of earlier studies, said Bruce Leff, MD, a professor at Johns Hopkins Medical School in Baltimore, who has researched hospital-at-home since the 1990s. Like the new study, those trials found that the results had no relationship to individual traits, such as socioeconomic status, medical conditions, age, gender, or race. 

Whether a person felt comfortable with the idea of hospital-at-home boiled down “to a preference for receiving care at home or in the hospital,” he said. Some people distrust hospitals, and others feel insecure about receiving care at home, even if it is provided by qualified health care professionals.
 

How Patients Are Selected 

While the details of hospital-at-home vary from program to program, the basic scenario is that patients who need certain kinds of acute care can be sent home from hospitals, emergency departments, or clinics to receive that care at home. Among the kinds of conditions that make stable patients eligible are heart failure, COPD, pneumonia, cellulitis, and COVID-19, said John Busigin, MD, a hospitalist and medical director of Covenant Advanced Care at Home. 

When a patient is admitted to hospital-at-home, the hospital will send along whatever equipment and medications that person needs. In some cases, this may include a hospital bed, although Ms. Stohler used her own. An IV line was put into her arm, and the IV stand was placed next to the bed. 

Ms. Stohler received a computer tablet that she used to communicate with doctors and nurses in Covenant’s “command center” in Knoxville. She also wore a watch with a button she could push in case of an emergency. And she had a telephone line that went directly to her medical team, in case she had an issue and the tablet didn’t work.

Twice a day, or as needed, specially trained paramedics came to Ms. Stohler’s home. They checked on the IV line, changed the IV bag, performed tests, and uploaded vital signs from monitoring equipment to Ms. Stohler’s tablet so it could be transmitted to the command center. A physician assistant came in on the second and fourth days of her weeklong stay in the program, and she saw a hospitalist remotely every day.

While some hospital-at-home programs have registered nurses visit patients at home, RNs are in short supply. To fill this gap, Covenant’s program uses community paramedics who have been in the field for at least 5 years, doing everything from intubating patients and placing them on ventilators to providing advanced cardiac life support, Dr. Busigin said. To get certified as community paramedics, they go through a 3-month training program.

Shortly after Ms. Stohler went into hospital-at-home, she had another crisis. Excess fluid had built up in her body because of all the IV fluids she’d received in the hospital while fighting the sepsis. As a result, she became short of breath. If she had been discharged to home rather than hospital-at-home, she said, she would have had to go to the emergency room. Instead, she sent out a distress call. One of the paramedics rushed to her house and gave her an IV diuretic medication, which helped her urinate to get rid of the excess fluid.

A small number of the estimated 300 people who have gone through the program had to be admitted to the hospital, Dr. Busigin said. Nationally, he said, about 5%-10% are admitted. But readmissions among the patients in the Covenant program have been 25% lower than for patients who received conventional hospital care and had the same conditions as those in hospital-at-home.

Studies have shown that these programs not only reduce readmissions, but also cost less, on average, and create a better patient experience than traditional hospital care does. And, according to the JAMA survey, most consumers like the idea. Fifty-six percent of people who took the survey agreed with the statement that people recover faster at home than in the hospital. Fifty-nine percent agreed they’d feel safe being treated at home, and 49% said they’d be more comfortable if treated at home. 

The 1134 people who took the survey were also asked about their comfort level with providing various kinds of care to their loved ones during a hospital-at-home episode. The results varied with the type of task: For example, 82% of the respondents agreed or strongly agreed they could manage a patient’s medications, while just 41% said they’d be willing to change a feeding tube. Smaller percentages were willing to change an IV bag or a catheter or do wound care.

However, hospital-at-home programs don’t allow caregivers to take part in clinical care, which is prohibited by Medicare waivers and state licensing regulations. None of the 22 health systems that use the hospital-at-home services of Medically Home, including Covenant, ask caregivers to do anything along this line, said Pippa Shulman, DO, medical director of the company, which provides equipment, technology, and protocols for hospital programs

The only exception at Covenant, Dr. Busigin said, is that the hospital may train family members to do wound care when a patient is discharged from the hospital to Advanced Care at Home. They may also prepare meals for their loved ones, although the program provides balanced meals to patients if they want them. Ms. Stohler had some of these meals, which just had to be heated up, for the first few days of hospital-at-home, and later her relatives brought meals to her house.
 

 

 

Challenges for the Future

The number of Medicare hospital-at-home waivers has nearly doubled since 2021. A year earlier, when Medicare began reimbursing hospitals for acute care at home to help them cope with the overflow of COVID patients, there were only about 15-20 programs in the United States, said Dr. Leff of Johns Hopkins.

A big reason for the lack of use before the pandemic, Dr. Leff said, is that there was no payment system for hospitals that offered hospital-at-home. Now, they can get paid by Medicare and 10 state Medicaid programs, and a number of private payers are also coming on board. Ms. Stohler’s private insurer covered her hospital-at-home stay, and Dr. Busigin said several plans that contract with Covenant will pay for it.

Dr. Leff said he’s cautiously optimistic Congress will extend the Medicare waiver program, which is scheduled to end in December, for another 5 years. A couple of key House committees have signed off on a bill to do that, he said, and a Congressional Budget Office report found that the program did not cost Medicare more money. 

But even if the waiver is renewed, some health systems may find it tough to deliver the service. The current version of this model depends a lot on technology, because telemedicine is used and reliable communication is needed for patients in hospital-at-home. That’s why many of the hospitals hire outside vendors like Medically Home to provide the infrastructure they need.

Medically Home manages the tablets given to patients and all connection and networking services, including internet and cellphone connections. It also provides technical services in the command centers that hospitals set up for the doctors and nurses who provide care remotely. 

And the firm figures out how to deliver the standard care for each condition in each hospital-at-home. “We need to make sure that the patient is going to get what they need in the time frame it needs to be delivered in, and that it’s safe and effective for the patient,” Dr. Shulman said. “So we’ve developed logistical protocols for a multitude of disease states that allow us to provide high-acuity care in the home to a variety of complex patients.”

The health care workers use the hospital electronic health record for hospital-at-home patients, and vital signs uploaded from patient tablets flow directly into the electronic health record, she said.
 

Rural Areas Need Help

The use of hospital-at-home in rural areas holds a lot of promise, Dr. Leff said. 

“A lot of rural hospitals have been closing, and hospital-at-home could be a mechanism to create hospital-level care where facilities have closed down. It’s easier to do this in urban areas, but it can be done in rural environments as well.”

Rami Karjian, CEO of Medically Home, agreed. The firm services hospital-at-home programs in rural areas of Oklahoma and California, using cellphones and paramedics in areas that lack broadband connections and nurses, he pointed out. 

“Hospital-at-home can’t just be available to people who live in big cities,” he said. “The access problems in health care are pervasive, and this is part of how we solve access problems in rural areas.”
 

A version of this article first appeared on WebMD.com.

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Shortage of Blood Bottles Could Disrupt Care

Article Type
Changed
Tue, 07/30/2024 - 16:10

Hospitals and laboratories across the United States are grappling with a shortage of Becton Dickinson BACTEC blood culture bottles that threatens to extend at least until September.

In a health advisory, the Centers for Disease Control and Prevention (CDC) warned that the critical shortage could lead to “delays in diagnosis, misdiagnosis, or other challenges” in the management of patients with infectious diseases.

Most blood cultures in the United States are performed using continuous-monitoring blood culture systems; the Becton Dickinson system is used in about half of all US laboratories and is only compatible with the brand’s BACTEC blood culture media bottles.

Healthcare providers, laboratories, healthcare facility administrators, and state, tribal, local, and territorial health departments affected by the shortage “should immediately begin to assess their situations and develop plans and options to mitigate the potential impact,” according to the health advisory.
 

What to Do

To reduce the impact of the shortage, facilities are urged to:

  • Determine the type of blood culture bottles they have
  • Optimize the use of blood cultures at their facility
  • Take steps to prevent blood culture contamination
  • Ensure that the appropriate volume of blood is collected for culture
  • Assess alternate options for blood cultures
  • Work with a nearby facility or send samples to another laboratory

Health departments are advised to contact hospitals and laboratories in their jurisdictions to determine whether the shortage will affect them. Health departments are also encouraged to educate others on the supply shortage, optimal use of blood cultures, and mechanisms for reporting supply chain shortages or interruptions to the Food and Drug Administration (FDA), as well as to help with communication between laboratories and facilities willing to assist others in need.

To further assist affected providers, the CDC, in collaboration with the Infectious Diseases Society of America, hosted a webinar with speakers from Johns Hopkins University, Massachusetts General Hospital, and Vanderbilt University, who shared what their institutions are doing to cope with the shortage and protect patients.
 

Why It Happened

In June, Becton Dickinson warned its customers that they may experience “intermittent delays” in the supply of some BACTEC blood culture media over the coming months because of reduced availability of plastic bottles from its supplier.

In a July 22 update, the company said the supplier issues were “more complex” than originally communicated and it is taking steps to “resolve this challenge as quickly as possible.”

In July, the FDA published a letter to healthcare providers acknowledging the supply disruptions and recommended strategies to preserve the supply for patients at highest risk.

Becton Dickinson has promised an update by September to this “dynamic and evolving situation.”

A version of this article appeared on Medscape.com.

Publications
Topics
Sections

Hospitals and laboratories across the United States are grappling with a shortage of Becton Dickinson BACTEC blood culture bottles that threatens to extend at least until September.

In a health advisory, the Centers for Disease Control and Prevention (CDC) warned that the critical shortage could lead to “delays in diagnosis, misdiagnosis, or other challenges” in the management of patients with infectious diseases.

Most blood cultures in the United States are performed using continuous-monitoring blood culture systems; the Becton Dickinson system is used in about half of all US laboratories and is only compatible with the brand’s BACTEC blood culture media bottles.

Healthcare providers, laboratories, healthcare facility administrators, and state, tribal, local, and territorial health departments affected by the shortage “should immediately begin to assess their situations and develop plans and options to mitigate the potential impact,” according to the health advisory.
 

What to Do

To reduce the impact of the shortage, facilities are urged to:

  • Determine the type of blood culture bottles they have
  • Optimize the use of blood cultures at their facility
  • Take steps to prevent blood culture contamination
  • Ensure that the appropriate volume of blood is collected for culture
  • Assess alternate options for blood cultures
  • Work with a nearby facility or send samples to another laboratory

Health departments are advised to contact hospitals and laboratories in their jurisdictions to determine whether the shortage will affect them. Health departments are also encouraged to educate others on the supply shortage, optimal use of blood cultures, and mechanisms for reporting supply chain shortages or interruptions to the Food and Drug Administration (FDA), as well as to help with communication between laboratories and facilities willing to assist others in need.

To further assist affected providers, the CDC, in collaboration with the Infectious Diseases Society of America, hosted a webinar with speakers from Johns Hopkins University, Massachusetts General Hospital, and Vanderbilt University, who shared what their institutions are doing to cope with the shortage and protect patients.
 

Why It Happened

In June, Becton Dickinson warned its customers that they may experience “intermittent delays” in the supply of some BACTEC blood culture media over the coming months because of reduced availability of plastic bottles from its supplier.

In a July 22 update, the company said the supplier issues were “more complex” than originally communicated and it is taking steps to “resolve this challenge as quickly as possible.”

In July, the FDA published a letter to healthcare providers acknowledging the supply disruptions and recommended strategies to preserve the supply for patients at highest risk.

Becton Dickinson has promised an update by September to this “dynamic and evolving situation.”

A version of this article appeared on Medscape.com.

Hospitals and laboratories across the United States are grappling with a shortage of Becton Dickinson BACTEC blood culture bottles that threatens to extend at least until September.

In a health advisory, the Centers for Disease Control and Prevention (CDC) warned that the critical shortage could lead to “delays in diagnosis, misdiagnosis, or other challenges” in the management of patients with infectious diseases.

Most blood cultures in the United States are performed using continuous-monitoring blood culture systems; the Becton Dickinson system is used in about half of all US laboratories and is only compatible with the brand’s BACTEC blood culture media bottles.

Healthcare providers, laboratories, healthcare facility administrators, and state, tribal, local, and territorial health departments affected by the shortage “should immediately begin to assess their situations and develop plans and options to mitigate the potential impact,” according to the health advisory.
 

What to Do

To reduce the impact of the shortage, facilities are urged to:

  • Determine the type of blood culture bottles they have
  • Optimize the use of blood cultures at their facility
  • Take steps to prevent blood culture contamination
  • Ensure that the appropriate volume of blood is collected for culture
  • Assess alternate options for blood cultures
  • Work with a nearby facility or send samples to another laboratory

Health departments are advised to contact hospitals and laboratories in their jurisdictions to determine whether the shortage will affect them. Health departments are also encouraged to educate others on the supply shortage, optimal use of blood cultures, and mechanisms for reporting supply chain shortages or interruptions to the Food and Drug Administration (FDA), as well as to help with communication between laboratories and facilities willing to assist others in need.

To further assist affected providers, the CDC, in collaboration with the Infectious Diseases Society of America, hosted a webinar with speakers from Johns Hopkins University, Massachusetts General Hospital, and Vanderbilt University, who shared what their institutions are doing to cope with the shortage and protect patients.
 

Why It Happened

In June, Becton Dickinson warned its customers that they may experience “intermittent delays” in the supply of some BACTEC blood culture media over the coming months because of reduced availability of plastic bottles from its supplier.

In a July 22 update, the company said the supplier issues were “more complex” than originally communicated and it is taking steps to “resolve this challenge as quickly as possible.”

In July, the FDA published a letter to healthcare providers acknowledging the supply disruptions and recommended strategies to preserve the supply for patients at highest risk.

Becton Dickinson has promised an update by September to this “dynamic and evolving situation.”

A version of this article appeared on Medscape.com.

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article