User login
Compassionate Communication Amid the COVID-19 Pandemic
The coronavirus disease of 2019 (COVID-19) pandemic is the health crisis of our generation and will inevitably leave a lasting mark on how we practice medicine.1,2 It has already rapidly changed the way we communicate with patients, families, and colleagues. From the explosion of virtual care—which has been accelerated by need and new reimbursement policies3—to the physical barriers created by personal protective equipment (PPE) and no-visitor policies, the landscape of caring for hospitalized patients has seismically shifted in a few short months. At its core, the practice of medicine is about human connection—a connection between healers and the sick—and should remain as such to provide compassionate care to patients and their loved ones.4,5 In this perspective, we discuss challenges arising from communication barriers in the time of COVID-19 and opportunities to overcome them by preserving human connection to deliver high-quality care (Table).
COMMUNICATION WITH PATIENTS
While critically important to prevent transmission of the COVID-19 pathogen (ie, SARS-CoV-2), physical distancing and PPE create myriad challenges to achieving effective communication between healthcare providers and patients. Telemedicine has been leveraged to allow distanced communication between patients with COVID-19 and their providers from separate rooms. For face-to-face conversations, physical barriers, including distance between individuals and the wearing of face masks, impose new types of hindrances to nonverbal and verbal communication.
Challenges
Nonverbal communication helps build the therapeutic alliance and influences patient adherence to care plans, satisfaction, trust, and clinical outcomes.6,7 Expressions of emotion and reciprocity of nonverbal communication serve as important foundations for physician-patient encounters.6 Face masks, a necessity to reduce transmission of SARS-CoV-2, lead to fewer facial cues and may impede the ability to express and recognize emotional cues for patients and providers. A study of over 1,000 patients randomized to mask-wearing and non–mask-wearing physicians revealed a significant and negative effect on patient perception of physician empathy in consultations performed by mask-wearing physicians.8 Additionally, simple handshakes that convey respect and appreciation are no longer practiced.
Verbal communication is also affected by measures designed to reduce infection. The face mask and face shield worn by clinicians caring for patients with respiratory illnesses like COVID-19 diminish the volume and clarity of the spoken word. This is particularly problematic for patients who have sensory disturbances like hearing impairment. Additionally, these patients may rely on lipreading to effectively understand others, a strategy lost once the face mask is donned.
Opportunities
Healthcare providers may respond to nonverbal communication impediments by explicitly shifting nonverbal to verbal communication. For instance, when delivering serious news, a physician might previously have “mirrored” the patient’s sadness through a light touch on the hand and facial expressions congruent with that emotion. With physical distancing and PPE, the physician may instead express empathy through verbal statements such as acknowledging, validating, and respecting the patient’s emotions; making supportive statements; or exploring the patient’s feelings. The physician may also thank the patient for providing their input for the conversation.
Physicians should introduce themselves at the start of every daily encounter with a patient since there may be few distinct features above the face mask to distinguish the numerous individuals on a healthcare team. Some medical teams have provided “facesheets” with photographs and information about each member in an effort to humanize the team and connect more genuinely with the patient. In some cases, this may be the only way for a patient to see their healthcare providers’ faces.
To address obstacles to effective verbal communication, physicians should inquire about patients’ possible sensory disturbances on admission and, if necessary, arrange for hearing aids or other assistive devices. When communicating, physicians should articulate, enunciate, and increase volume to overcome the physical barrier created by the face mask. They should speak slowly, use plain language without jargon, and intentionally pause to check for understanding using the teach-back method.9
COMMUNICATION WITH FAMILIES AND CAREGIVERS
Challenges
With the aim of mitigating SARS-CoV-2 transmission, most healthcare systems have implemented no-visitor policies for hospitalized patients. This often leads to feelings of isolation among patients and their families. Goals-of-care discussions for COVID-19 and other serious diagnoses such as cancer can become even more difficult because family members often cannot witness how ill patients have become and clinicians cannot easily communicate virtually with multiple family members simultaneously.
Lack of family at the bedside also makes critical activities, such as discharge planning and education, more vulnerable to poor coordination and medical errors.10 Patients who are continuing to recover from acute illness may be expected to learn the details of home infusion for intravenous antibiotics, tracheostomy care, or specialized nutritional feeds. Without caregiver support, the patient may be at risk for readmission or other untoward safety events.
Opportunities
Several strategies may be used to improve virtual communication with families. The healthcare team should identify one family point of contact (ideally with the durable power of attorney for healthcare) who will receive and disseminate to others information about the patient’s status. This reduces the potential for multiple telephone conversations. We have witnessed some remarkable family points of contact call many family members to relay medical updates and moderate discussion. Care teams may decide to call the family contact during rounds so that they may listen in on the conversation with the patient or call after rounds to provide succinct updates. Family meetings may benefit greatly if conducted through a video platform, when possible, particularly if significant interval events have occurred. Connection through video allows eye contact and recognition of other nonverbal cues, as well as allowing findings like diagnostic images to be shared.
Because of increased anxiety associated with isolation, we recommend that one member of the primary healthcare team conduct telephone updates to the family point of contact on at least a daily basis. This simple act reduces potential for disjointed or discrepant messages from the healthcare team.11 It also demonstrates the value of keeping those individuals most important to the patient informed and has been shown to increase satisfaction with care and perceived effectiveness of meeting informational needs.12
Regarding discharge planning, physicians should engage the patient and family/caregivers in developing a patient-centered plan as early in the hospital stay as possible. The adage “discharge planning starts at admission” has never been more relevant. The team should avoid assumptions about patient/family sophistication for understanding complex healthcare concepts. Rather, physicians should assess patients’ and caregivers’ health literacy at the beginning of a hospital stay by asking simple, validated questions in a nonjudgmental way.13,14 This valuable information then allows the team to tailor medical information and discharge education appropriately for both patients and caregivers.
COMMUNICATION WITHIN THE HEALTHCARE TEAM
Challenges
As a result of the COVID-19 pandemic, various members of the healthcare team may be working remotely, and therefore, team members may feel less connected with each other. This could lead to a loss of camaraderie and fellowship within the team, as well as depersonalization, one of the main facets of burnout.15 Even if colocalized in the same area, those wearing face masks may experience disconnection and depersonalization. In an anecdote at our medical center, one clinician did not know what her team members’ faces looked like until they removed their masks for a moment to have a snack just before the end of the rotation.
In addition, healthcare systems have witnessed an increase in the volume of electronic consultations in which faculty and house staff review the patient’s medical record and render medical decision-making and recommendations without physically examining or interviewing the patient at the bedside. The purpose of this is twofold: to reduce the risk of transmitting SARS-CoV-2 and to conserve PPE. Electronic consultations could threaten to reduce collaborative communication and teaching among primary and consulting teams, which may lead to greater misunderstanding, less-effective patient care, and decreased satisfaction within the healthcare team.
Opportunities
Now more than ever, physicians should purposefully engage in regular communication with the multidisciplinary healthcare team that includes nurses, pharmacists, social workers, and other critical members. Because many of these individuals may now be working remotely or not joining in-person rounds, several strategies are needed to ensure care coordination within the primary healthcare team. For example, all members should “huddle” at least once daily to review each patient’s care and progress in meeting discharge goals. Team members who are working remotely should be dialed into these huddles and included in coordinating the plan for the day. While in-person multidisciplinary rounds may be temporarily halted to allow for physical distancing of staff, physician leaders can still encourage regular check-ins and updates throughout the day with multidisciplinary team members by other means, such as discussions by phone or a secure instant messenger, if available.
Another strategy to improve care coordination is to engage consulting teams in direct patient/family communication at critical junctures. For example, when a patient’s renal failure has gotten severe enough that dialysis is a consideration, the primary team may ask the nephrology consult service to participate in a joint telephone discussion with the family about risks, benefits, and alternatives to renal replacement therapy. Additionally, our palliative care consult service volunteered to be automatically consulted for all COVID-19 patients in the intensive care unit and high-risk COVID-19 patients on the acute care wards because of the disease’s high potential morbidity and mortality. Their roles included proactively confirming the patient’s surrogate decision maker, reviewing the patient’s decision-making capacity, eliciting specific goals of care and life-sustaining treatment preferences, and establishing relationships with the family. They also conducted daily huddles with the respective teams, another approach that fostered high-quality, collaborative care.
CONCLUSION
The COVID-19 pandemic has forced us to change the approaches we usually employ to interact with patients and their loved ones, as well as healthcare team members, but it has not changed the heart of medicine, which is to heal. Here we provide tangible and discrete strategies to achieve this goal through clear and compassionate communication, including shifting nonverbal to verbal communication with patients, speaking at least daily to one family point of contact, ensuring early and tailored discharge planning, emphasizing continued close care coordination among the multidisciplinary team, and thoughtfully engaging consultants in patient/family communication. We hope this guidance will assist us in striving to cultivate connection with our patients, their loved ones, and each other, just as we have always sought to do. With these strategies in mind, coupled with a continued focus on patient- and family-centered care for hospitalized patients, no amount of distance or PPE will diminish the power of human connection.
Acknowledgments
The authors wish to thank their colleagues—the physicians, nurses, respiratory therapists, clerks, custodial staff, security, and administrative professionals, to name a few—of the VA Ann Arbor Healthcare System for their collaboration, dedication, and grace in this time of crisis. The authors are indebted to the patients and their loved ones for putting their trust in their team, for teaching team members, and for providing the privilege of being a part of their lives.
Disclosures
The authors reported having nothing to disclose.
1. Ross JE. Resident response during pandemic: this is our time [online first]. Ann Intern Med. 2020. https://doi.org/10.7326/M20-1240
2. Berwick DM. Choices for the “new normal” [online first]. JAMA. 2020. https://doi.org/10.1001/jama.2020.6949.
3. Centers for Medicare & Medicaid Services. President Trump expands telehealth benefits for Medicare beneficiaries during COVID-19 outbreak. CMS.gov. Mar 17, 2020. https://www.cms.gov/newsroom/press-releases/president-trump-expands-telehealth-benefits-medicare-beneficiaries-during-covid-19-outbreak. Accessed May 09, 2020.
4. Zulman DM, Haverfield MC, Shaw JG, et al. Practices to foster physician presence and connection with patients in the clinical encounter. JAMA. 2020;323(1):70‐81. https://doi.org/10.1001/jama.2019.19003.
5. Haverfield MC, Tierney A, Schwartz R, et al. Can patient-provider interpersonal interventions achieve the quadruple aim of healthcare? a systematic review [online first]. J Gen Intern Med. 2020. https://doi.org/10.1007/s11606-019-05525-2.
6. Roter DL, Frankel RM, Hall JA, Sluyter D. The expression of emotion through nonverbal behavior in medical visits: mechanisms and outcomes. J Gen Intern Med. 2006;21(Suppl 1):S28-S34. https://doi.org/10.1111/j.1525-1497.2006.00306.x.
7. Mast MS. On the importance of nonverbal communication in the physician-patient interaction. Patient Educ Couns. 2007;67(3):315-318. https://doi.org/10.1016/j.pec.2007.03.005.
8. Wong CK, Yip BH, Mercer S, et al. Effect of facemasks on empathy and relational continuity: a randomised controlled trial in primary care. BMC Fam Pract. 2013;14:200. https://doi.org/10.1186/1471-2296-14-200.
9. Talevski J, Wong Shee A, Rasmussen B, Kemp G, Beauchamp A. Teach-back: a systematic review of implementation and impacts. PLoS One. 2020;15(4):e0231350. https://doi.org/10.1371/journal.pone.0231350.
10. Kripalani S, Jackson AT, Schnipper JL, Coleman EA. Promoting effective transitions of care at hospital discharge: a review of key issues for hospitalists. J Hosp Med. 2007;2(5):314-323. https://doi.org/10.1002/jhm.228.
11. Ahrens T, Yancey V, Kollef M. Improving family communications at the end of life: implications for length of stay in the intensive care unit and resource use. Am J Crit Care. 2003;12(4):317-324.
12. Medland JJ, Ferrans CE. Effectiveness of a structured communication program for family members of patients in an ICU. Am J Crit Care. 1998;7(1):24-29.
13. Chew LD, Bradley KA, Boyko EJ. Brief questions to identify patients with inadequate health literacy. Fam Med. 2004;36(8):588-594.
14. Wallace LS, Rogers ES, Roskos SE, Holiday DB, Weiss BD. Brief report: screening items to identify patients with limited health literacy skills. J Gen Intern Med. 2006;21:874-877. https://doi.org/10.1111/j.1525-1497.2006.00532.x.
15. West CP, Dyrbye LN, Shanafelt TD. Physician burnout: contributors, consequences and solutions. J Intern Med. 2018;283(6):516‐529. https://doi.org/10.1111/joim.12752.
The coronavirus disease of 2019 (COVID-19) pandemic is the health crisis of our generation and will inevitably leave a lasting mark on how we practice medicine.1,2 It has already rapidly changed the way we communicate with patients, families, and colleagues. From the explosion of virtual care—which has been accelerated by need and new reimbursement policies3—to the physical barriers created by personal protective equipment (PPE) and no-visitor policies, the landscape of caring for hospitalized patients has seismically shifted in a few short months. At its core, the practice of medicine is about human connection—a connection between healers and the sick—and should remain as such to provide compassionate care to patients and their loved ones.4,5 In this perspective, we discuss challenges arising from communication barriers in the time of COVID-19 and opportunities to overcome them by preserving human connection to deliver high-quality care (Table).
COMMUNICATION WITH PATIENTS
While critically important to prevent transmission of the COVID-19 pathogen (ie, SARS-CoV-2), physical distancing and PPE create myriad challenges to achieving effective communication between healthcare providers and patients. Telemedicine has been leveraged to allow distanced communication between patients with COVID-19 and their providers from separate rooms. For face-to-face conversations, physical barriers, including distance between individuals and the wearing of face masks, impose new types of hindrances to nonverbal and verbal communication.
Challenges
Nonverbal communication helps build the therapeutic alliance and influences patient adherence to care plans, satisfaction, trust, and clinical outcomes.6,7 Expressions of emotion and reciprocity of nonverbal communication serve as important foundations for physician-patient encounters.6 Face masks, a necessity to reduce transmission of SARS-CoV-2, lead to fewer facial cues and may impede the ability to express and recognize emotional cues for patients and providers. A study of over 1,000 patients randomized to mask-wearing and non–mask-wearing physicians revealed a significant and negative effect on patient perception of physician empathy in consultations performed by mask-wearing physicians.8 Additionally, simple handshakes that convey respect and appreciation are no longer practiced.
Verbal communication is also affected by measures designed to reduce infection. The face mask and face shield worn by clinicians caring for patients with respiratory illnesses like COVID-19 diminish the volume and clarity of the spoken word. This is particularly problematic for patients who have sensory disturbances like hearing impairment. Additionally, these patients may rely on lipreading to effectively understand others, a strategy lost once the face mask is donned.
Opportunities
Healthcare providers may respond to nonverbal communication impediments by explicitly shifting nonverbal to verbal communication. For instance, when delivering serious news, a physician might previously have “mirrored” the patient’s sadness through a light touch on the hand and facial expressions congruent with that emotion. With physical distancing and PPE, the physician may instead express empathy through verbal statements such as acknowledging, validating, and respecting the patient’s emotions; making supportive statements; or exploring the patient’s feelings. The physician may also thank the patient for providing their input for the conversation.
Physicians should introduce themselves at the start of every daily encounter with a patient since there may be few distinct features above the face mask to distinguish the numerous individuals on a healthcare team. Some medical teams have provided “facesheets” with photographs and information about each member in an effort to humanize the team and connect more genuinely with the patient. In some cases, this may be the only way for a patient to see their healthcare providers’ faces.
To address obstacles to effective verbal communication, physicians should inquire about patients’ possible sensory disturbances on admission and, if necessary, arrange for hearing aids or other assistive devices. When communicating, physicians should articulate, enunciate, and increase volume to overcome the physical barrier created by the face mask. They should speak slowly, use plain language without jargon, and intentionally pause to check for understanding using the teach-back method.9
COMMUNICATION WITH FAMILIES AND CAREGIVERS
Challenges
With the aim of mitigating SARS-CoV-2 transmission, most healthcare systems have implemented no-visitor policies for hospitalized patients. This often leads to feelings of isolation among patients and their families. Goals-of-care discussions for COVID-19 and other serious diagnoses such as cancer can become even more difficult because family members often cannot witness how ill patients have become and clinicians cannot easily communicate virtually with multiple family members simultaneously.
Lack of family at the bedside also makes critical activities, such as discharge planning and education, more vulnerable to poor coordination and medical errors.10 Patients who are continuing to recover from acute illness may be expected to learn the details of home infusion for intravenous antibiotics, tracheostomy care, or specialized nutritional feeds. Without caregiver support, the patient may be at risk for readmission or other untoward safety events.
Opportunities
Several strategies may be used to improve virtual communication with families. The healthcare team should identify one family point of contact (ideally with the durable power of attorney for healthcare) who will receive and disseminate to others information about the patient’s status. This reduces the potential for multiple telephone conversations. We have witnessed some remarkable family points of contact call many family members to relay medical updates and moderate discussion. Care teams may decide to call the family contact during rounds so that they may listen in on the conversation with the patient or call after rounds to provide succinct updates. Family meetings may benefit greatly if conducted through a video platform, when possible, particularly if significant interval events have occurred. Connection through video allows eye contact and recognition of other nonverbal cues, as well as allowing findings like diagnostic images to be shared.
Because of increased anxiety associated with isolation, we recommend that one member of the primary healthcare team conduct telephone updates to the family point of contact on at least a daily basis. This simple act reduces potential for disjointed or discrepant messages from the healthcare team.11 It also demonstrates the value of keeping those individuals most important to the patient informed and has been shown to increase satisfaction with care and perceived effectiveness of meeting informational needs.12
Regarding discharge planning, physicians should engage the patient and family/caregivers in developing a patient-centered plan as early in the hospital stay as possible. The adage “discharge planning starts at admission” has never been more relevant. The team should avoid assumptions about patient/family sophistication for understanding complex healthcare concepts. Rather, physicians should assess patients’ and caregivers’ health literacy at the beginning of a hospital stay by asking simple, validated questions in a nonjudgmental way.13,14 This valuable information then allows the team to tailor medical information and discharge education appropriately for both patients and caregivers.
COMMUNICATION WITHIN THE HEALTHCARE TEAM
Challenges
As a result of the COVID-19 pandemic, various members of the healthcare team may be working remotely, and therefore, team members may feel less connected with each other. This could lead to a loss of camaraderie and fellowship within the team, as well as depersonalization, one of the main facets of burnout.15 Even if colocalized in the same area, those wearing face masks may experience disconnection and depersonalization. In an anecdote at our medical center, one clinician did not know what her team members’ faces looked like until they removed their masks for a moment to have a snack just before the end of the rotation.
In addition, healthcare systems have witnessed an increase in the volume of electronic consultations in which faculty and house staff review the patient’s medical record and render medical decision-making and recommendations without physically examining or interviewing the patient at the bedside. The purpose of this is twofold: to reduce the risk of transmitting SARS-CoV-2 and to conserve PPE. Electronic consultations could threaten to reduce collaborative communication and teaching among primary and consulting teams, which may lead to greater misunderstanding, less-effective patient care, and decreased satisfaction within the healthcare team.
Opportunities
Now more than ever, physicians should purposefully engage in regular communication with the multidisciplinary healthcare team that includes nurses, pharmacists, social workers, and other critical members. Because many of these individuals may now be working remotely or not joining in-person rounds, several strategies are needed to ensure care coordination within the primary healthcare team. For example, all members should “huddle” at least once daily to review each patient’s care and progress in meeting discharge goals. Team members who are working remotely should be dialed into these huddles and included in coordinating the plan for the day. While in-person multidisciplinary rounds may be temporarily halted to allow for physical distancing of staff, physician leaders can still encourage regular check-ins and updates throughout the day with multidisciplinary team members by other means, such as discussions by phone or a secure instant messenger, if available.
Another strategy to improve care coordination is to engage consulting teams in direct patient/family communication at critical junctures. For example, when a patient’s renal failure has gotten severe enough that dialysis is a consideration, the primary team may ask the nephrology consult service to participate in a joint telephone discussion with the family about risks, benefits, and alternatives to renal replacement therapy. Additionally, our palliative care consult service volunteered to be automatically consulted for all COVID-19 patients in the intensive care unit and high-risk COVID-19 patients on the acute care wards because of the disease’s high potential morbidity and mortality. Their roles included proactively confirming the patient’s surrogate decision maker, reviewing the patient’s decision-making capacity, eliciting specific goals of care and life-sustaining treatment preferences, and establishing relationships with the family. They also conducted daily huddles with the respective teams, another approach that fostered high-quality, collaborative care.
CONCLUSION
The COVID-19 pandemic has forced us to change the approaches we usually employ to interact with patients and their loved ones, as well as healthcare team members, but it has not changed the heart of medicine, which is to heal. Here we provide tangible and discrete strategies to achieve this goal through clear and compassionate communication, including shifting nonverbal to verbal communication with patients, speaking at least daily to one family point of contact, ensuring early and tailored discharge planning, emphasizing continued close care coordination among the multidisciplinary team, and thoughtfully engaging consultants in patient/family communication. We hope this guidance will assist us in striving to cultivate connection with our patients, their loved ones, and each other, just as we have always sought to do. With these strategies in mind, coupled with a continued focus on patient- and family-centered care for hospitalized patients, no amount of distance or PPE will diminish the power of human connection.
Acknowledgments
The authors wish to thank their colleagues—the physicians, nurses, respiratory therapists, clerks, custodial staff, security, and administrative professionals, to name a few—of the VA Ann Arbor Healthcare System for their collaboration, dedication, and grace in this time of crisis. The authors are indebted to the patients and their loved ones for putting their trust in their team, for teaching team members, and for providing the privilege of being a part of their lives.
Disclosures
The authors reported having nothing to disclose.
The coronavirus disease of 2019 (COVID-19) pandemic is the health crisis of our generation and will inevitably leave a lasting mark on how we practice medicine.1,2 It has already rapidly changed the way we communicate with patients, families, and colleagues. From the explosion of virtual care—which has been accelerated by need and new reimbursement policies3—to the physical barriers created by personal protective equipment (PPE) and no-visitor policies, the landscape of caring for hospitalized patients has seismically shifted in a few short months. At its core, the practice of medicine is about human connection—a connection between healers and the sick—and should remain as such to provide compassionate care to patients and their loved ones.4,5 In this perspective, we discuss challenges arising from communication barriers in the time of COVID-19 and opportunities to overcome them by preserving human connection to deliver high-quality care (Table).
COMMUNICATION WITH PATIENTS
While critically important to prevent transmission of the COVID-19 pathogen (ie, SARS-CoV-2), physical distancing and PPE create myriad challenges to achieving effective communication between healthcare providers and patients. Telemedicine has been leveraged to allow distanced communication between patients with COVID-19 and their providers from separate rooms. For face-to-face conversations, physical barriers, including distance between individuals and the wearing of face masks, impose new types of hindrances to nonverbal and verbal communication.
Challenges
Nonverbal communication helps build the therapeutic alliance and influences patient adherence to care plans, satisfaction, trust, and clinical outcomes.6,7 Expressions of emotion and reciprocity of nonverbal communication serve as important foundations for physician-patient encounters.6 Face masks, a necessity to reduce transmission of SARS-CoV-2, lead to fewer facial cues and may impede the ability to express and recognize emotional cues for patients and providers. A study of over 1,000 patients randomized to mask-wearing and non–mask-wearing physicians revealed a significant and negative effect on patient perception of physician empathy in consultations performed by mask-wearing physicians.8 Additionally, simple handshakes that convey respect and appreciation are no longer practiced.
Verbal communication is also affected by measures designed to reduce infection. The face mask and face shield worn by clinicians caring for patients with respiratory illnesses like COVID-19 diminish the volume and clarity of the spoken word. This is particularly problematic for patients who have sensory disturbances like hearing impairment. Additionally, these patients may rely on lipreading to effectively understand others, a strategy lost once the face mask is donned.
Opportunities
Healthcare providers may respond to nonverbal communication impediments by explicitly shifting nonverbal to verbal communication. For instance, when delivering serious news, a physician might previously have “mirrored” the patient’s sadness through a light touch on the hand and facial expressions congruent with that emotion. With physical distancing and PPE, the physician may instead express empathy through verbal statements such as acknowledging, validating, and respecting the patient’s emotions; making supportive statements; or exploring the patient’s feelings. The physician may also thank the patient for providing their input for the conversation.
Physicians should introduce themselves at the start of every daily encounter with a patient since there may be few distinct features above the face mask to distinguish the numerous individuals on a healthcare team. Some medical teams have provided “facesheets” with photographs and information about each member in an effort to humanize the team and connect more genuinely with the patient. In some cases, this may be the only way for a patient to see their healthcare providers’ faces.
To address obstacles to effective verbal communication, physicians should inquire about patients’ possible sensory disturbances on admission and, if necessary, arrange for hearing aids or other assistive devices. When communicating, physicians should articulate, enunciate, and increase volume to overcome the physical barrier created by the face mask. They should speak slowly, use plain language without jargon, and intentionally pause to check for understanding using the teach-back method.9
COMMUNICATION WITH FAMILIES AND CAREGIVERS
Challenges
With the aim of mitigating SARS-CoV-2 transmission, most healthcare systems have implemented no-visitor policies for hospitalized patients. This often leads to feelings of isolation among patients and their families. Goals-of-care discussions for COVID-19 and other serious diagnoses such as cancer can become even more difficult because family members often cannot witness how ill patients have become and clinicians cannot easily communicate virtually with multiple family members simultaneously.
Lack of family at the bedside also makes critical activities, such as discharge planning and education, more vulnerable to poor coordination and medical errors.10 Patients who are continuing to recover from acute illness may be expected to learn the details of home infusion for intravenous antibiotics, tracheostomy care, or specialized nutritional feeds. Without caregiver support, the patient may be at risk for readmission or other untoward safety events.
Opportunities
Several strategies may be used to improve virtual communication with families. The healthcare team should identify one family point of contact (ideally with the durable power of attorney for healthcare) who will receive and disseminate to others information about the patient’s status. This reduces the potential for multiple telephone conversations. We have witnessed some remarkable family points of contact call many family members to relay medical updates and moderate discussion. Care teams may decide to call the family contact during rounds so that they may listen in on the conversation with the patient or call after rounds to provide succinct updates. Family meetings may benefit greatly if conducted through a video platform, when possible, particularly if significant interval events have occurred. Connection through video allows eye contact and recognition of other nonverbal cues, as well as allowing findings like diagnostic images to be shared.
Because of increased anxiety associated with isolation, we recommend that one member of the primary healthcare team conduct telephone updates to the family point of contact on at least a daily basis. This simple act reduces potential for disjointed or discrepant messages from the healthcare team.11 It also demonstrates the value of keeping those individuals most important to the patient informed and has been shown to increase satisfaction with care and perceived effectiveness of meeting informational needs.12
Regarding discharge planning, physicians should engage the patient and family/caregivers in developing a patient-centered plan as early in the hospital stay as possible. The adage “discharge planning starts at admission” has never been more relevant. The team should avoid assumptions about patient/family sophistication for understanding complex healthcare concepts. Rather, physicians should assess patients’ and caregivers’ health literacy at the beginning of a hospital stay by asking simple, validated questions in a nonjudgmental way.13,14 This valuable information then allows the team to tailor medical information and discharge education appropriately for both patients and caregivers.
COMMUNICATION WITHIN THE HEALTHCARE TEAM
Challenges
As a result of the COVID-19 pandemic, various members of the healthcare team may be working remotely, and therefore, team members may feel less connected with each other. This could lead to a loss of camaraderie and fellowship within the team, as well as depersonalization, one of the main facets of burnout.15 Even if colocalized in the same area, those wearing face masks may experience disconnection and depersonalization. In an anecdote at our medical center, one clinician did not know what her team members’ faces looked like until they removed their masks for a moment to have a snack just before the end of the rotation.
In addition, healthcare systems have witnessed an increase in the volume of electronic consultations in which faculty and house staff review the patient’s medical record and render medical decision-making and recommendations without physically examining or interviewing the patient at the bedside. The purpose of this is twofold: to reduce the risk of transmitting SARS-CoV-2 and to conserve PPE. Electronic consultations could threaten to reduce collaborative communication and teaching among primary and consulting teams, which may lead to greater misunderstanding, less-effective patient care, and decreased satisfaction within the healthcare team.
Opportunities
Now more than ever, physicians should purposefully engage in regular communication with the multidisciplinary healthcare team that includes nurses, pharmacists, social workers, and other critical members. Because many of these individuals may now be working remotely or not joining in-person rounds, several strategies are needed to ensure care coordination within the primary healthcare team. For example, all members should “huddle” at least once daily to review each patient’s care and progress in meeting discharge goals. Team members who are working remotely should be dialed into these huddles and included in coordinating the plan for the day. While in-person multidisciplinary rounds may be temporarily halted to allow for physical distancing of staff, physician leaders can still encourage regular check-ins and updates throughout the day with multidisciplinary team members by other means, such as discussions by phone or a secure instant messenger, if available.
Another strategy to improve care coordination is to engage consulting teams in direct patient/family communication at critical junctures. For example, when a patient’s renal failure has gotten severe enough that dialysis is a consideration, the primary team may ask the nephrology consult service to participate in a joint telephone discussion with the family about risks, benefits, and alternatives to renal replacement therapy. Additionally, our palliative care consult service volunteered to be automatically consulted for all COVID-19 patients in the intensive care unit and high-risk COVID-19 patients on the acute care wards because of the disease’s high potential morbidity and mortality. Their roles included proactively confirming the patient’s surrogate decision maker, reviewing the patient’s decision-making capacity, eliciting specific goals of care and life-sustaining treatment preferences, and establishing relationships with the family. They also conducted daily huddles with the respective teams, another approach that fostered high-quality, collaborative care.
CONCLUSION
The COVID-19 pandemic has forced us to change the approaches we usually employ to interact with patients and their loved ones, as well as healthcare team members, but it has not changed the heart of medicine, which is to heal. Here we provide tangible and discrete strategies to achieve this goal through clear and compassionate communication, including shifting nonverbal to verbal communication with patients, speaking at least daily to one family point of contact, ensuring early and tailored discharge planning, emphasizing continued close care coordination among the multidisciplinary team, and thoughtfully engaging consultants in patient/family communication. We hope this guidance will assist us in striving to cultivate connection with our patients, their loved ones, and each other, just as we have always sought to do. With these strategies in mind, coupled with a continued focus on patient- and family-centered care for hospitalized patients, no amount of distance or PPE will diminish the power of human connection.
Acknowledgments
The authors wish to thank their colleagues—the physicians, nurses, respiratory therapists, clerks, custodial staff, security, and administrative professionals, to name a few—of the VA Ann Arbor Healthcare System for their collaboration, dedication, and grace in this time of crisis. The authors are indebted to the patients and their loved ones for putting their trust in their team, for teaching team members, and for providing the privilege of being a part of their lives.
Disclosures
The authors reported having nothing to disclose.
1. Ross JE. Resident response during pandemic: this is our time [online first]. Ann Intern Med. 2020. https://doi.org/10.7326/M20-1240
2. Berwick DM. Choices for the “new normal” [online first]. JAMA. 2020. https://doi.org/10.1001/jama.2020.6949.
3. Centers for Medicare & Medicaid Services. President Trump expands telehealth benefits for Medicare beneficiaries during COVID-19 outbreak. CMS.gov. Mar 17, 2020. https://www.cms.gov/newsroom/press-releases/president-trump-expands-telehealth-benefits-medicare-beneficiaries-during-covid-19-outbreak. Accessed May 09, 2020.
4. Zulman DM, Haverfield MC, Shaw JG, et al. Practices to foster physician presence and connection with patients in the clinical encounter. JAMA. 2020;323(1):70‐81. https://doi.org/10.1001/jama.2019.19003.
5. Haverfield MC, Tierney A, Schwartz R, et al. Can patient-provider interpersonal interventions achieve the quadruple aim of healthcare? a systematic review [online first]. J Gen Intern Med. 2020. https://doi.org/10.1007/s11606-019-05525-2.
6. Roter DL, Frankel RM, Hall JA, Sluyter D. The expression of emotion through nonverbal behavior in medical visits: mechanisms and outcomes. J Gen Intern Med. 2006;21(Suppl 1):S28-S34. https://doi.org/10.1111/j.1525-1497.2006.00306.x.
7. Mast MS. On the importance of nonverbal communication in the physician-patient interaction. Patient Educ Couns. 2007;67(3):315-318. https://doi.org/10.1016/j.pec.2007.03.005.
8. Wong CK, Yip BH, Mercer S, et al. Effect of facemasks on empathy and relational continuity: a randomised controlled trial in primary care. BMC Fam Pract. 2013;14:200. https://doi.org/10.1186/1471-2296-14-200.
9. Talevski J, Wong Shee A, Rasmussen B, Kemp G, Beauchamp A. Teach-back: a systematic review of implementation and impacts. PLoS One. 2020;15(4):e0231350. https://doi.org/10.1371/journal.pone.0231350.
10. Kripalani S, Jackson AT, Schnipper JL, Coleman EA. Promoting effective transitions of care at hospital discharge: a review of key issues for hospitalists. J Hosp Med. 2007;2(5):314-323. https://doi.org/10.1002/jhm.228.
11. Ahrens T, Yancey V, Kollef M. Improving family communications at the end of life: implications for length of stay in the intensive care unit and resource use. Am J Crit Care. 2003;12(4):317-324.
12. Medland JJ, Ferrans CE. Effectiveness of a structured communication program for family members of patients in an ICU. Am J Crit Care. 1998;7(1):24-29.
13. Chew LD, Bradley KA, Boyko EJ. Brief questions to identify patients with inadequate health literacy. Fam Med. 2004;36(8):588-594.
14. Wallace LS, Rogers ES, Roskos SE, Holiday DB, Weiss BD. Brief report: screening items to identify patients with limited health literacy skills. J Gen Intern Med. 2006;21:874-877. https://doi.org/10.1111/j.1525-1497.2006.00532.x.
15. West CP, Dyrbye LN, Shanafelt TD. Physician burnout: contributors, consequences and solutions. J Intern Med. 2018;283(6):516‐529. https://doi.org/10.1111/joim.12752.
1. Ross JE. Resident response during pandemic: this is our time [online first]. Ann Intern Med. 2020. https://doi.org/10.7326/M20-1240
2. Berwick DM. Choices for the “new normal” [online first]. JAMA. 2020. https://doi.org/10.1001/jama.2020.6949.
3. Centers for Medicare & Medicaid Services. President Trump expands telehealth benefits for Medicare beneficiaries during COVID-19 outbreak. CMS.gov. Mar 17, 2020. https://www.cms.gov/newsroom/press-releases/president-trump-expands-telehealth-benefits-medicare-beneficiaries-during-covid-19-outbreak. Accessed May 09, 2020.
4. Zulman DM, Haverfield MC, Shaw JG, et al. Practices to foster physician presence and connection with patients in the clinical encounter. JAMA. 2020;323(1):70‐81. https://doi.org/10.1001/jama.2019.19003.
5. Haverfield MC, Tierney A, Schwartz R, et al. Can patient-provider interpersonal interventions achieve the quadruple aim of healthcare? a systematic review [online first]. J Gen Intern Med. 2020. https://doi.org/10.1007/s11606-019-05525-2.
6. Roter DL, Frankel RM, Hall JA, Sluyter D. The expression of emotion through nonverbal behavior in medical visits: mechanisms and outcomes. J Gen Intern Med. 2006;21(Suppl 1):S28-S34. https://doi.org/10.1111/j.1525-1497.2006.00306.x.
7. Mast MS. On the importance of nonverbal communication in the physician-patient interaction. Patient Educ Couns. 2007;67(3):315-318. https://doi.org/10.1016/j.pec.2007.03.005.
8. Wong CK, Yip BH, Mercer S, et al. Effect of facemasks on empathy and relational continuity: a randomised controlled trial in primary care. BMC Fam Pract. 2013;14:200. https://doi.org/10.1186/1471-2296-14-200.
9. Talevski J, Wong Shee A, Rasmussen B, Kemp G, Beauchamp A. Teach-back: a systematic review of implementation and impacts. PLoS One. 2020;15(4):e0231350. https://doi.org/10.1371/journal.pone.0231350.
10. Kripalani S, Jackson AT, Schnipper JL, Coleman EA. Promoting effective transitions of care at hospital discharge: a review of key issues for hospitalists. J Hosp Med. 2007;2(5):314-323. https://doi.org/10.1002/jhm.228.
11. Ahrens T, Yancey V, Kollef M. Improving family communications at the end of life: implications for length of stay in the intensive care unit and resource use. Am J Crit Care. 2003;12(4):317-324.
12. Medland JJ, Ferrans CE. Effectiveness of a structured communication program for family members of patients in an ICU. Am J Crit Care. 1998;7(1):24-29.
13. Chew LD, Bradley KA, Boyko EJ. Brief questions to identify patients with inadequate health literacy. Fam Med. 2004;36(8):588-594.
14. Wallace LS, Rogers ES, Roskos SE, Holiday DB, Weiss BD. Brief report: screening items to identify patients with limited health literacy skills. J Gen Intern Med. 2006;21:874-877. https://doi.org/10.1111/j.1525-1497.2006.00532.x.
15. West CP, Dyrbye LN, Shanafelt TD. Physician burnout: contributors, consequences and solutions. J Intern Med. 2018;283(6):516‐529. https://doi.org/10.1111/joim.12752.
© 2020 Society of Hospital Medicine
SECTION 4: HEALTHCARE SYSTEMS: SUPPORTING AND ADVANCING CHILD HEALTH
Alvarez F, Alverson B, Balighian E, Beauchamp-Walters J, Biondi E, Blankenberg R, Bridgeman C, Brown J, Buchanan AO, Carlson D, Chang P, Coon E, Daud YN, Denniston S, DeWolfe CC, Deutsch SA, Doshi A, Fisher E, Gage S, Gallagher MP, Gill A, Goel Jones V, Grill J, Gupta A, Herbst BF Jr, Hershey D, Hoang K, Holmes AV, Hopkins A, Jones Y, Khan A, Lee V, Li ST, Lye, PS, Maginot T, Maloney C, Maniscalco J, Mannino Avila E, Markowsky A, Marks M, Matheny Antommaria AH, Maul E, McCulloh R, Melwani A, Miller C, Mittal V, Natt B, O'Toole J, Ottolini M, Percelay J, Phillips S, Pressel D, Quinonez R, Ralston S, Rappaport DI, Rauch D, Rhee K, Riese J, Roberts K, Rogers A, Rosenberg RE, Ruhlen M, Russell CJ, Russo C, Schwenk KM, Sekaran A, Shadman KA, Shah SS, Shen M, Simon T, Singh A, Smith K, Srinivas N Srivastava R, Sterni L, Thompson ED Jr, Thomson J, Tieder J, Tremoulet A, Wang ME, Williams R, Wu S. Name of Chapter. In: Pediatric Hospital Medicine Core Competencies: 2020 Revision. Section 4: Healthcare Systems: Supporting and Advancing Child Health. J Hosp Med. 2020;15(S1):xxx-xxx (insert page numbers). https://doi.org/jhm.3400
Alvarez F, Alverson B, Balighian E, Beauchamp-Walters J, Biondi E, Blankenberg R, Bridgeman C, Brown J, Buchanan AO, Carlson D, Chang P, Coon E, Daud YN, Denniston S, DeWolfe CC, Deutsch SA, Doshi A, Fisher E, Gage S, Gallagher MP, Gill A, Goel Jones V, Grill J, Gupta A, Herbst BF Jr, Hershey D, Hoang K, Holmes AV, Hopkins A, Jones Y, Khan A, Lee V, Li ST, Lye, PS, Maginot T, Maloney C, Maniscalco J, Mannino Avila E, Markowsky A, Marks M, Matheny Antommaria AH, Maul E, McCulloh R, Melwani A, Miller C, Mittal V, Natt B, O'Toole J, Ottolini M, Percelay J, Phillips S, Pressel D, Quinonez R, Ralston S, Rappaport DI, Rauch D, Rhee K, Riese J, Roberts K, Rogers A, Rosenberg RE, Ruhlen M, Russell CJ, Russo C, Schwenk KM, Sekaran A, Shadman KA, Shah SS, Shen M, Simon T, Singh A, Smith K, Srinivas N Srivastava R, Sterni L, Thompson ED Jr, Thomson J, Tieder J, Tremoulet A, Wang ME, Williams R, Wu S. Name of Chapter. In: Pediatric Hospital Medicine Core Competencies: 2020 Revision. Section 4: Healthcare Systems: Supporting and Advancing Child Health. J Hosp Med. 2020;15(S1):xxx-xxx (insert page numbers). https://doi.org/jhm.3400
Alvarez F, Alverson B, Balighian E, Beauchamp-Walters J, Biondi E, Blankenberg R, Bridgeman C, Brown J, Buchanan AO, Carlson D, Chang P, Coon E, Daud YN, Denniston S, DeWolfe CC, Deutsch SA, Doshi A, Fisher E, Gage S, Gallagher MP, Gill A, Goel Jones V, Grill J, Gupta A, Herbst BF Jr, Hershey D, Hoang K, Holmes AV, Hopkins A, Jones Y, Khan A, Lee V, Li ST, Lye, PS, Maginot T, Maloney C, Maniscalco J, Mannino Avila E, Markowsky A, Marks M, Matheny Antommaria AH, Maul E, McCulloh R, Melwani A, Miller C, Mittal V, Natt B, O'Toole J, Ottolini M, Percelay J, Phillips S, Pressel D, Quinonez R, Ralston S, Rappaport DI, Rauch D, Rhee K, Riese J, Roberts K, Rogers A, Rosenberg RE, Ruhlen M, Russell CJ, Russo C, Schwenk KM, Sekaran A, Shadman KA, Shah SS, Shen M, Simon T, Singh A, Smith K, Srinivas N Srivastava R, Sterni L, Thompson ED Jr, Thomson J, Tieder J, Tremoulet A, Wang ME, Williams R, Wu S. Name of Chapter. In: Pediatric Hospital Medicine Core Competencies: 2020 Revision. Section 4: Healthcare Systems: Supporting and Advancing Child Health. J Hosp Med. 2020;15(S1):xxx-xxx (insert page numbers). https://doi.org/jhm.3400
SECTION 3: SPECIALIZED SERVICES
How to cite articles within Section 2
Alvarez F, Alverson B, Balighian E, Beauchamp-Walters J, Biondi E, Blankenberg R, Bridgeman C, Brown J, Buchanan AO, Carlson D, Chang P, Coon E, Daud YN, Denniston S, DeWolfe CC, Deutsch SA, Doshi A, Fisher E, Gage S, Gallagher MP, Gill A, Goel Jones V, Grill J, Gupta A, Herbst BF Jr, Hershey D, Hoang K, Holmes AV, Hopkins A, Jones Y, Khan A, Lee V, Li ST, Lye, PS, Maginot T, Maloney C, Maniscalco J, Mannino Avila E, Markowsky A, Marks M, Matheny Antommaria AH, Maul E, McCulloh R, Melwani A, Miller C, Mittal V, Natt B, O'Toole J, Ottolini M, Percelay J, Phillips S, Pressel D, Quinonez R, Ralston S, Rappaport DI, Rauch D, Rhee K, Riese J, Roberts K, Rogers A, Rosenberg RE, Ruhlen M, Russell CJ, Russo C, Schwenk KM, Sekaran A, Shadman KA, Shah SS, Shen M, Simon T, Singh A, Smith K, Srinivas N Srivastava R, Sterni L, Thompson ED Jr, Thomson J, Tieder J, Tremoulet A, Wang ME, Williams R, Wu S. Name of Chapter. In: Pediatric Hospital Medicine Core Competencies: 2020 Revision. Section 3: Specialized Services. J Hosp Med. 2020;15(S1):xx-xxx (insert page numbers). https://doi.org/10.12788/jhm.3399
How to cite articles within Section 2
Alvarez F, Alverson B, Balighian E, Beauchamp-Walters J, Biondi E, Blankenberg R, Bridgeman C, Brown J, Buchanan AO, Carlson D, Chang P, Coon E, Daud YN, Denniston S, DeWolfe CC, Deutsch SA, Doshi A, Fisher E, Gage S, Gallagher MP, Gill A, Goel Jones V, Grill J, Gupta A, Herbst BF Jr, Hershey D, Hoang K, Holmes AV, Hopkins A, Jones Y, Khan A, Lee V, Li ST, Lye, PS, Maginot T, Maloney C, Maniscalco J, Mannino Avila E, Markowsky A, Marks M, Matheny Antommaria AH, Maul E, McCulloh R, Melwani A, Miller C, Mittal V, Natt B, O'Toole J, Ottolini M, Percelay J, Phillips S, Pressel D, Quinonez R, Ralston S, Rappaport DI, Rauch D, Rhee K, Riese J, Roberts K, Rogers A, Rosenberg RE, Ruhlen M, Russell CJ, Russo C, Schwenk KM, Sekaran A, Shadman KA, Shah SS, Shen M, Simon T, Singh A, Smith K, Srinivas N Srivastava R, Sterni L, Thompson ED Jr, Thomson J, Tieder J, Tremoulet A, Wang ME, Williams R, Wu S. Name of Chapter. In: Pediatric Hospital Medicine Core Competencies: 2020 Revision. Section 3: Specialized Services. J Hosp Med. 2020;15(S1):xx-xxx (insert page numbers). https://doi.org/10.12788/jhm.3399
How to cite articles within Section 2
Alvarez F, Alverson B, Balighian E, Beauchamp-Walters J, Biondi E, Blankenberg R, Bridgeman C, Brown J, Buchanan AO, Carlson D, Chang P, Coon E, Daud YN, Denniston S, DeWolfe CC, Deutsch SA, Doshi A, Fisher E, Gage S, Gallagher MP, Gill A, Goel Jones V, Grill J, Gupta A, Herbst BF Jr, Hershey D, Hoang K, Holmes AV, Hopkins A, Jones Y, Khan A, Lee V, Li ST, Lye, PS, Maginot T, Maloney C, Maniscalco J, Mannino Avila E, Markowsky A, Marks M, Matheny Antommaria AH, Maul E, McCulloh R, Melwani A, Miller C, Mittal V, Natt B, O'Toole J, Ottolini M, Percelay J, Phillips S, Pressel D, Quinonez R, Ralston S, Rappaport DI, Rauch D, Rhee K, Riese J, Roberts K, Rogers A, Rosenberg RE, Ruhlen M, Russell CJ, Russo C, Schwenk KM, Sekaran A, Shadman KA, Shah SS, Shen M, Simon T, Singh A, Smith K, Srinivas N Srivastava R, Sterni L, Thompson ED Jr, Thomson J, Tieder J, Tremoulet A, Wang ME, Williams R, Wu S. Name of Chapter. In: Pediatric Hospital Medicine Core Competencies: 2020 Revision. Section 3: Specialized Services. J Hosp Med. 2020;15(S1):xx-xxx (insert page numbers). https://doi.org/10.12788/jhm.3399
SECTION 2: CORE SKILLS
How to cite articles within Section 2
Alvarez F, Alverson B, Balighian E, Beauchamp-Walters J, Biondi E, Blankenberg R, Bridgeman C, Brown J, Buchanan AO, Carlson D, Chang P, Coon E, Daud YN, Denniston S, DeWolfe CC, Deutsch SA, Doshi A, Fisher E, Gage S, Gallagher MP, Gill A, Goel Jones V, Grill J, Gupta A, Herbst BF Jr, Hershey D, Hoang K, Holmes AV, Hopkins A, Jones Y, Khan A, Lee V, Li ST, Lye, PS, Maginot T, Maloney C, Maniscalco J, Mannino Avila E, Markowsky A, Marks M, Matheny Antommaria AH, Maul E, McCulloh R, Melwani A, Miller C, Mittal V, Natt B, O'Toole J, Ottolini M, Percelay J, Phillips S, Pressel D, Quinonez R, Ralston S, Rappaport DI, Rauch D, Rhee K, Riese J, Roberts K, Rogers A, Rosenberg RE, Ruhlen M, Russell CJ, Russo C, Schwenk KM, Sekaran A, Shadman KA, Shah SS, Shen M, Simon T, Singh A, Smith K, Srinivas N Srivastava R, Sterni L, Thompson ED Jr, Thomson J, Tieder J, Tremoulet A, Wang ME, Williams R, Wu S. Name of Chapter. In: Pediatric Hospital Medicine Core Competencies: 2020 Revision. Section 2: Core Skills. J Hosp Med. 2020;15(S1):XX-XX (insert page numbers). https://doi.org/10.12788/jhm.3398
How to cite articles within Section 2
Alvarez F, Alverson B, Balighian E, Beauchamp-Walters J, Biondi E, Blankenberg R, Bridgeman C, Brown J, Buchanan AO, Carlson D, Chang P, Coon E, Daud YN, Denniston S, DeWolfe CC, Deutsch SA, Doshi A, Fisher E, Gage S, Gallagher MP, Gill A, Goel Jones V, Grill J, Gupta A, Herbst BF Jr, Hershey D, Hoang K, Holmes AV, Hopkins A, Jones Y, Khan A, Lee V, Li ST, Lye, PS, Maginot T, Maloney C, Maniscalco J, Mannino Avila E, Markowsky A, Marks M, Matheny Antommaria AH, Maul E, McCulloh R, Melwani A, Miller C, Mittal V, Natt B, O'Toole J, Ottolini M, Percelay J, Phillips S, Pressel D, Quinonez R, Ralston S, Rappaport DI, Rauch D, Rhee K, Riese J, Roberts K, Rogers A, Rosenberg RE, Ruhlen M, Russell CJ, Russo C, Schwenk KM, Sekaran A, Shadman KA, Shah SS, Shen M, Simon T, Singh A, Smith K, Srinivas N Srivastava R, Sterni L, Thompson ED Jr, Thomson J, Tieder J, Tremoulet A, Wang ME, Williams R, Wu S. Name of Chapter. In: Pediatric Hospital Medicine Core Competencies: 2020 Revision. Section 2: Core Skills. J Hosp Med. 2020;15(S1):XX-XX (insert page numbers). https://doi.org/10.12788/jhm.3398
How to cite articles within Section 2
Alvarez F, Alverson B, Balighian E, Beauchamp-Walters J, Biondi E, Blankenberg R, Bridgeman C, Brown J, Buchanan AO, Carlson D, Chang P, Coon E, Daud YN, Denniston S, DeWolfe CC, Deutsch SA, Doshi A, Fisher E, Gage S, Gallagher MP, Gill A, Goel Jones V, Grill J, Gupta A, Herbst BF Jr, Hershey D, Hoang K, Holmes AV, Hopkins A, Jones Y, Khan A, Lee V, Li ST, Lye, PS, Maginot T, Maloney C, Maniscalco J, Mannino Avila E, Markowsky A, Marks M, Matheny Antommaria AH, Maul E, McCulloh R, Melwani A, Miller C, Mittal V, Natt B, O'Toole J, Ottolini M, Percelay J, Phillips S, Pressel D, Quinonez R, Ralston S, Rappaport DI, Rauch D, Rhee K, Riese J, Roberts K, Rogers A, Rosenberg RE, Ruhlen M, Russell CJ, Russo C, Schwenk KM, Sekaran A, Shadman KA, Shah SS, Shen M, Simon T, Singh A, Smith K, Srinivas N Srivastava R, Sterni L, Thompson ED Jr, Thomson J, Tieder J, Tremoulet A, Wang ME, Williams R, Wu S. Name of Chapter. In: Pediatric Hospital Medicine Core Competencies: 2020 Revision. Section 2: Core Skills. J Hosp Med. 2020;15(S1):XX-XX (insert page numbers). https://doi.org/10.12788/jhm.3398
SECTION 1: COMMON CLINICAL DIAGNOSES AND CONDITIONS
How to cite articles within Section 1
Alvarez F, Alverson B, Balighian E, Beauchamp-Walters J, Biondi E, Blankenberg R, Bridgeman C, Brown J, Buchanan AO, Carlson D, Chang P, Coon E, Daud YN, Denniston S, DeWolfe CC, Deutsch SA, Doshi A, Fisher E, Gage S, Gallagher MP, Gill A, Goel Jones V, Grill J, Gupta A, Herbst BF Jr, Hershey D, Hoang K, Holmes AV, Hopkins A, Jones Y, Khan A, Lee V, Li ST, Lye, PS, Maginot T, Maloney C, Maniscalco J, Mannino Avila E, Markowsky A, Marks M, Matheny Antommaria AH, Maul E, McCulloh R, Melwani A, Miller C, Mittal V, Natt B, O'Toole J, Ottolini M, Percelay J, Phillips S, Pressel D, Quinonez R, Ralston S, Rappaport DI, Rauch D, Rhee K, Riese J, Roberts K, Rogers A, Rosenberg RE, Ruhlen M, Russell CJ, Russo C, Schwenk KM, Sekaran A, Shadman KA, Shah SS, Shen M, Simon T, Singh A, Smith K, Srinivas N Srivastava R, Sterni L, Thompson ED Jr, Thomson J, Tieder J, Tremoulet A, Wang ME, Williams R, Wu S. Name of Chapter. In: Pediatric Hospital Medicine Core Competencies: 2020 Revision. Section 1: Common Clinical Diagnoses and Conditions. J Hosp Med. 2020;15(S1):xx-xx (insert page numbers). https://doi.org/10.12788/jhm.3397
How to cite articles within Section 1
Alvarez F, Alverson B, Balighian E, Beauchamp-Walters J, Biondi E, Blankenberg R, Bridgeman C, Brown J, Buchanan AO, Carlson D, Chang P, Coon E, Daud YN, Denniston S, DeWolfe CC, Deutsch SA, Doshi A, Fisher E, Gage S, Gallagher MP, Gill A, Goel Jones V, Grill J, Gupta A, Herbst BF Jr, Hershey D, Hoang K, Holmes AV, Hopkins A, Jones Y, Khan A, Lee V, Li ST, Lye, PS, Maginot T, Maloney C, Maniscalco J, Mannino Avila E, Markowsky A, Marks M, Matheny Antommaria AH, Maul E, McCulloh R, Melwani A, Miller C, Mittal V, Natt B, O'Toole J, Ottolini M, Percelay J, Phillips S, Pressel D, Quinonez R, Ralston S, Rappaport DI, Rauch D, Rhee K, Riese J, Roberts K, Rogers A, Rosenberg RE, Ruhlen M, Russell CJ, Russo C, Schwenk KM, Sekaran A, Shadman KA, Shah SS, Shen M, Simon T, Singh A, Smith K, Srinivas N Srivastava R, Sterni L, Thompson ED Jr, Thomson J, Tieder J, Tremoulet A, Wang ME, Williams R, Wu S. Name of Chapter. In: Pediatric Hospital Medicine Core Competencies: 2020 Revision. Section 1: Common Clinical Diagnoses and Conditions. J Hosp Med. 2020;15(S1):xx-xx (insert page numbers). https://doi.org/10.12788/jhm.3397
How to cite articles within Section 1
Alvarez F, Alverson B, Balighian E, Beauchamp-Walters J, Biondi E, Blankenberg R, Bridgeman C, Brown J, Buchanan AO, Carlson D, Chang P, Coon E, Daud YN, Denniston S, DeWolfe CC, Deutsch SA, Doshi A, Fisher E, Gage S, Gallagher MP, Gill A, Goel Jones V, Grill J, Gupta A, Herbst BF Jr, Hershey D, Hoang K, Holmes AV, Hopkins A, Jones Y, Khan A, Lee V, Li ST, Lye, PS, Maginot T, Maloney C, Maniscalco J, Mannino Avila E, Markowsky A, Marks M, Matheny Antommaria AH, Maul E, McCulloh R, Melwani A, Miller C, Mittal V, Natt B, O'Toole J, Ottolini M, Percelay J, Phillips S, Pressel D, Quinonez R, Ralston S, Rappaport DI, Rauch D, Rhee K, Riese J, Roberts K, Rogers A, Rosenberg RE, Ruhlen M, Russell CJ, Russo C, Schwenk KM, Sekaran A, Shadman KA, Shah SS, Shen M, Simon T, Singh A, Smith K, Srinivas N Srivastava R, Sterni L, Thompson ED Jr, Thomson J, Tieder J, Tremoulet A, Wang ME, Williams R, Wu S. Name of Chapter. In: Pediatric Hospital Medicine Core Competencies: 2020 Revision. Section 1: Common Clinical Diagnoses and Conditions. J Hosp Med. 2020;15(S1):xx-xx (insert page numbers). https://doi.org/10.12788/jhm.3397
Gap Analysis for the Conversion to Area Under the Curve Vancomycin Monitoring in a Small Rural Hospital
The use of weight-based dosing with trough-based monitoring of vancomycin has been in clinical practice for more than a decade. The American Society of Health-System Pharmacists (ASHP), the Infectious Diseases Society of America (IDSA), and the Society of Infectious Diseases Pharmacists (SIDP) published the first guidelines for vancomycin monitoring in 2009.1 Although it has been well established that area under the curve (AUC) over the minimal inhibitory concentration (MIC) ratio > 400 mg.h/L is the best predictor of clinical efficacy, obtaining this value in clinical practice was not pragmatic. Therefore, the 2009 guidelines recommended a goal vancomycin trough of 15 to 20 mcg/ml as a surrogate marker for AUC/MIC > 400 mg.hr/L. This has since become a common practice despite little data that support this recommendation.
The efficacy and safety of trough-based monitoring has been evaluated extensively over the past several years and more recent data suggest that there is wide patient variability in AUC with this method and higher trough levels are associated with more nephrotoxicity.2,3 ASHP, IDSA, SIDP, and the Pediatric Infectious Diseases Society (PIDS) updated the consensus guidelines in 2020.4 Trough-based monitoring is no longer recommended. Instead AUC24 monitoring should be implemented with a goal range of 400 to 600 mg.h/L for efficacy and safety. Given concerns for vancomycin penetration into the central nervous system (CNS), many facility protocols utilize higher targets (> 600 mg.h/L) for CNS infections.
Some hospitals have been utilizing AUC-based monitoring for years. There are strategies from tertiary care centers that drive this practice change in the medical literature.5,6 However, it is important to reproduce these implementation practices in small, rural facilities that may face unique challenges with limited resources and may be slower to implement consensus guidelines.7,8 As this is a major practice change, it is imperative to evaluate the extent of transition and identify areas of needed improvement.
Accurate therapeutic drug monitoring ensures both the safety and efficacy of vancomycin therapy. Unfortunately, research shows that inappropriate laboratory tests are common in medical facilities.9 Drug levels taken inappropriately can lead to delays in therapeutic decision-making, inappropriate dosage adjustments and create a need for repeated drug levels, which increases the overall cost of admission.
Given the multiple affected services needed to make successful practice transitions, it is paramount that facilities evaluate progress during the transition phase. The Agency for Healthcare Research and Quality and the Institute for Healthcare Improvement provide guidance in the Plan-Do-Study-Act Cycle for quality assessment and improvement of new initiatives.10,11 A gap analysis can be used as a simple tool for evaluating the transition of research into practice and to identify areas of needed improvement.
The Veterans Health Care System of the Ozarks (VHSO) in Fayetteville, Arkansas made the transition from trough-based monitoring to 2-level AUC-based monitoring on April 1, 2019. The purpose of this study was to evaluate the effectiveness of transition methods used to implement AUC-monitoring for vancomycin treated patients in a small, primary facility. A further goal of the study was to identify areas of needed improvement and education and whether the problems derived from deficiencies in knowledge and ordering (medical and pharmacy services) or execution (nursing and laboratory services).
Methods
VHSO is a 52-bed US Department of Veterans Affairs primary care hospital. The pharmacy and laboratory are staffed 24 hours each day. There is 1 clinical pharmacy specialist (CPS) available for therapeutic drug monitoring consults Monday through Friday between the hours of 7:30 AM and 4:00 PM. No partial full-time equivalent employees were added for this conversion. Pharmacy-driven vancomycin dosing and monitoring is conducted on a collaborative basis, with pharmacy managing the majority of vancomycin treated patients. Night and weekend pharmacy staff provide cross-coverage on vancomycin consultations. Laboratory orders and medication dosage adjustments fall within the CPS scope of practice. Nurses do not perform laboratory draws for therapeutic drug monitoring; this is done solely by phlebotomists. There is no infectious diseases specialist at the facility to champion antibiotic dosing initiatives.
The implementation strategy largely reflected those outlined from tertiary care centers.5,6 First, key personnel from the laboratory department met to discuss this practice change and to add vancomycin peaks to the ordering menu. A critical value was set at 40 mcg/ml. Vancomycin troughs and random levels already were orderable items. A comment field was added to all laboratory orders for further clarification. Verbiage was added to laboratory reports in the computerized medical record to assist clinicians in determining the appropriateness of the level. This was followed by an educational email to both the nursing and laboratory departments explaining the practice change and included a link to the Pharmacy Joe “Vancomycin Dosing by AUC:MIC Instead of Trough-level” podcast (www.pharmacyjoe.com episode 356).
The pharmacy department received an interactive 30-minute presentation, followed immediately by a group activity to discuss practice problems. This presentation was condensed, recorded, and emailed to all VHSO pharmacists. A shared folder contained pertinent material on AUC monitoring.
Finally, an interactive presentation was set up for hospitalists and a video teleconferencing was conducted for rotating medical residents. Both the podcast and recorded presentation were emailed to the entire medical staff with a brief introduction of the practice change. Additionally, the transition process was added as a standing item on the monthly antimicrobial stewardship meeting agenda.
The standardized pharmacokinetic model at the study facility consisted of a vancomycin volume of distribution of 0.7 mg/kg and elimination rate constant (Ke) by Matzke and colleagues for total daily dose calculations.12 Obese patients (BMI ≥ 30) undergo alternative clearance equations described by Crass and colleagues.13 Cockcroft-Gault methods using ideal body weight (or actual body weight if < ideal body weight) are used for determining creatinine clearance. In patients aged ≥ 65 years with a serum creatinine < 1.0 mg/dL, facility guidance was to round serum creatinine up to 1.0 mg/dL. Loading doses were determined on a case-by-case basis with a cap of 2,000 mg, maintenance doses were rounded to the nearest 250 mg.
Vancomycin levels typically are drawn at steady state and analyzed using the logarithmic trapezoidal rule.14 The pharmacy and medical staff were educated to provide details on timing and coordination in nursing and laboratory orders (Table 1). Two-level AUC monitoring typically is not performed in patients with acute renal failure, expected duration of therapy < 72 hours, urinary tract infections, skin and soft tissue infections, or in renal replacement therapy.5
This gap analysis consisted of a retrospective chart review of vancomycin levels ordered after the implementation of AUC-based monitoring to determine the effectiveness of the transition. Three months of data were collected between April 2019 and June 2019. Vancomycin levels were deemed either appropriate or inappropriate based on timing and type (peak, trough, or random) of the laboratory test in relation to the previously administered vancomycin dose. Appropriate peaks were drawn within 2 hours after the end of infusion and troughs at least 1 half-life after the dose or just prior to the next dose and within the same dosing interval as the peak. Tests drawn outside of the specified time range, trough-only laboratory tests, or those drawn after vancomycin had been discontinued were considered inappropriate. Peaks and troughs drawn from separate dosing intervals also were considered inappropriate. Random levels were considered appropriate only if they fit the clinical context in acute renal failure or renal replacement therapy. An effective transition was defined as ≥ 80% of all vancomycin treated patients monitored with AUC methods rather than trough-based methods.
Inclusion criteria included all vancomycin levels ordered during the study period with no exclusions. The primary endpoint was the proportion of vancomycin levels drawn appropriately. Secondary endpoints were the proportion of AUC24 calculations within therapeutic range and a stratification of reasons for inappropriate levels. Descriptive statistics were collected to describe the scope of the project. Levels drawn from various shifts were compared (ie, day, night, or weekend). Calculated AUC24 levels between 400 and 600 mg.h/L were considered therapeutic unless treating CNS infection (600-700 mg.h/L). Given the operational outcomes (rather than clinical outcomes) and no comparator group, patient specific data were not collected.
Descriptive statistics without further analysis were used to describe proportions. The goal level for compliance was set at 100%. These methods were reviewed by the VHSO Institutional Review Board and granted nonresearch status, waiving the requirement for informed consent.
Results
The transition was effective with 97% of all cases utilizing AUC-based methods for monitoring. A total of 65 vancomycin levels were drawn in the study period; 32 peaks, 32 troughs, and 1 random level (drawn appropriately during acute renal failure 24 hours after starting therapy). All shifts were affected proportionately; days (n = 26, 40%), nights (n = 18, 27.7%), and weekends (n = 21, 32.3%). Based on time of dosage administration and laboratory test, there were 9 levels (13.8%) deemed inappropriate, 56 levels (86.1%) were appropriate. Reasons for inappropriate levels gleaned from chart review are presented in Table 2. Four levels had to be repeated for accurate calculations.
From the peak/trough couplets drawn appropriately, calculated AUC24 fell with the desired range in 61% (n = 17) of cases. Of the 11 that fell outside of range, 8 were subtherapeutic (< 400 mg.h/L) and 3 were supratherapeutic (> 600 mg.h/L). All levels were drawn at steady state. Indications for vancomycin monitoring were osteomyelitis (n = 13, 43%), sepsis (n = 10, 33%), pneumonia (n = 6, 20%), and 1 case of meningitis (3%).
Discussion
To the author’s knowledge, this is the first report of a vancomycin AUC24 monitoring conversion in a rural facility. This study adds to the existing medical literature in that it demonstrates that: (1) implementation methods described in large, tertiary centers can be effectively utilized in primary care, rural facilities; (2) the gap analysis used can be duplicated with minimal personnel and resources to ensure effective implementation (Table 3); and (3) the reported improvement needs can serve as a model for preventative measures at other facilities. The incidence of appropriate vancomycin levels was notably better than those reported in other single center studies.15-17 However, given variations in study design and facility operating procedures, it would be difficult to compare incidence among medical facilities. As such, there are no consensus benchmarks for comparison. The majority of inappropriate levels occurred early in the study period and on weekends. Appropriateness of drug levels may have improved with continued feedback and familiarity.
The calculated AUC24 fell within predicted range in 61% of cases. For comparison, a recent study from a large academic medical center reported that 73.5% of 2-level AUC24 cases had initial values within the therapeutic range.18 Of note, the target range used was much wider (400 - 800 mg.h/L) than the present study. Another study reported dose adjustments for subtherapeutic AUC levels in 25% of cases and dose reductions for supratherapeutic levels in 33.3% of cases.19
Of the AUC24 calculations that fell outside of therapeutic range, the majority (n = 8, 73%) were subtherapeutic (< 400 mg.h/L), half of these were for patients who were obese. It was unclear in the medical record which equation was used for initial dosing (Matzke vs Crass), or whether more conservative AUCs were used for calculating the total daily dose. The VHSO policy limiting loading doses also may have played a role; indeed the updated guidelines recommend a maximum loading dose of 3,000 mg depending on the severity of infection.4 Two of the 3 supratherapeutic levels were thought to be due to accumulation with long-term therapy.
Given such a large change from long-standing practices, there was surprisingly little resistance from the various clinical services. A recent survey of academic medical centers reported that the majority (88%) of all respondents who did not currently utilize AUC24 monitoring did not plan on making this immediate transition, largely citing unfamiliarity and training requirements.20 It is conceivable that the transition to AUC monitoring in smaller facilities may have fewer barriers than those seen in tertiary care centers. There are fewer health care providers and pharmacists to educate with the primary responsibilities falling on relatively few clinicians. There is little question as to who will be conducting follow up or whom to contact for questions. A smaller patient load and lesser patient acuity may translate to fewer vancomycin cases that require monitoring.
The interactive meetings were an important element for facility implementation. Research shows that emails alone are not effective for health care provider education, and interactive methods are recommended over passive methods.21,22 Assessing and avoiding barriers up front such as unclear laboratory orders, or communication failures is paramount to successful implementation strategies.23 Additionally, the detailed written ordering communication may have contributed to a smoother transition. The educational recording proved to be helpful in educating new staff and residents. An identified logistical error was that laboratory orders entered while patients were enrolled in sham clinics for electronic workload capture (eg, Pharmacy Inpatient Clinic) created confusion on the physical location of the patient for the phlebotomists, potentially causing delays in specimen collection.
A major development that stemmed from this intervention was that the Medical Service asked that policy changes be made so that the Pharmacy Service take over all vancomycin dosing at the facility. Previously, this had been done on a collaborative basis. Similar facilities with a collaborative practice model may need to anticipate such a request as this may present a new set of challenges. Accordingly, the pharmacy department is in the process of establishing standing operating procedures, pharmacist competencies, and a facility memorandum. Future research should evaluate the safety and efficacy of vancomycin therapy after the switch to AUC-based monitoring.
Limitations
There are several limitations to consider with this study. Operating procedures and implementation processes may vary between facilities, which could limit the generalizability of these results. Given the small facility size, the overall number of laboratory tests drawn was much smaller than those seen in larger facilities. The time needed for AUC calculations is notably longer than older methods of monitoring; however, this was not objectively assessed. It is important to note that clinical outcomes were beyond the scope of this gap analysis and this is an area of future research at the study facility. Vancomycin laboratory tests that were missed due to procedures and subsequently rescheduled were occasionally observed but not accounted for in this analysis. Additionally, vancomycin courses without monitoring (appropriate or otherwise) when indicated were not assessed. However, anecdotally speaking, this would be a very unlikely occurrence.
Conclusion
Conversion to AUC-based vancomycin monitoring is feasible in primary, rural medical centers. Implementation strategies from tertiary facilities can be successfully utilized in smaller hospitals. Quality assessment strategies such as a gap analysis can be utilized with minimal resources for facility uptake of new clinical practices.
1. Rybak M, Lomaestro B, Rotschafer JC, et al. Therapeutic monitoring of vancomycin in adult patients: a consensus review of the American Society of Health-System Pharmacists, the Infectious Diseases Society of America, and the Society of Infectious Diseases Pharmacists [published correction appears in Am J Health Syst Pharm. 2009;66(10):887]. Am J Health Syst Pharm. 2009;66(1):82‐98. doi:10.2146/ajhp080434
2. van Hal SJ, Paterson DL, Lodise TP. Systematic review and meta-analysis of vancomycin-induced nephrotoxicity associated with dosing schedules that maintain troughs between 15 and 20 milligrams per liter. Antimicrob Agents Chemother. 2013;57(2):734‐744. doi:10.1128/AAC.01568-12
3. Pai MP, Neely M, Rodvold KA, Lodise TP. Innovative approaches to optimizing the delivery of vancomycin in individual patients. Adv Drug Deliv Rev. 2014;77:50‐57. doi:10.1016/j.addr.2014.05.016
4. Rybak MJ, Le J, Lodise TP, et al. Therapeutic monitoring of vancomycin for serious methicillin-resistant Staphylococcus aureus infections: a revised consensus guideline and review by the American Society of Health-System Pharmacists, the Infectious Diseases Society of America, the Pediatric Infectious Diseases Society, and the Society of Infectious Diseases Pharmacists [published online ahead of print, 2020 Mar 19]. Am J Health Syst Pharm. 2020;zxaa036. doi:10.1093/ajhp/zxaa036
5. Heil EL, Claeys KC, Mynatt RP, et al. Making the change to area under the curve-based vancomycin dosing. Am J Health Syst Pharm. 2018;75(24):1986‐1995. doi:10.2146/ajhp180034
6. Gregory ER, Burgess DR, Cotner SE, et al. Vancomycin area under the curve dosing and monitoring at an academic medical center: transition strategies and lessons learned [published online ahead of print, 2019 Mar 10]. J Pharm Pract. 2019;897190019834369. doi:10.1177/0897190019834369
7. Septimus EJ, Owens RC Jr. Need and potential of antimicrobial stewardship in community hospitals. Clin Infect Dis. 2011;53 Suppl 1:S8‐S14. doi:10.1093/cid/cir363
8. Goldman LE, Dudley RA. United States rural hospital quality in the Hospital Compare database-accounting for hospital characteristics. Health Policy. 2008;87(1):112‐127. doi:10.1016/j.healthpol.2008.02.002
9. Zhi M, Ding EL, Theisen-Toupal J, Whelan J, Arnaout R. The landscape of inappropriate laboratory testing: a 15-year meta-analysis. PLoS One. 2013;8(11):e78962. doi:10.1371/journal.pone.0078962
10. Institute for Healthcare Improvement. Plan-do-study-act (PDSA) worksheet. http://www.ihi.org/resources/Pages/Tools/PlanDoStudyActWorksheet.aspx. Accessed May 13, 2020.
11. Agency for Healthcare Research and Quality. Plan-do-study-act (PDSA) cycle. https://innovations.ahrq.gov/qualitytools/plan-do-study-act-pdsa-cycle. Updated April 10, 2013. Accessed May 13, 2020.
12. Matzke GR, McGory RW, Halstenson CE, Keane WF. Pharmacokinetics of vancomycin in patients with various degrees of renal function. Antimicrob Agents Chemother. 1984;25(4):433‐437. doi:10.1128/aac.25.4.433
13. Crass RL, Dunn R, Hong J, Krop LC, Pai MP. Dosing vancomycin in the super obese: less is more. J Antimicrob Chemother. 2018;73(11):3081‐3086. doi:10.1093/jac/dky310
14. Pai MP, Russo A, Novelli A, Venditti M, Falcone M. Simplified equations using two concentrations to calculate area under the curve for antimicrobials with concentration-dependent pharmacodynamics: daptomycin as a motivating example. Antimicrob Agents Chemother. 2014;58(6):3162‐3167. doi:10.1128/AAC.02355-14
15. Suryadevara M, Steidl KE, Probst LA, Shaw J. Inappropriate vancomycin therapeutic drug monitoring in hospitalized pediatric patients increases pediatric trauma and hospital costs. J Pediatr Pharmacol Ther. 2012;17(2):159‐165. doi:10.5863/1551-6776-17.2.159
16. Morrison AP, Melanson SE, Carty MG, Bates DW, Szumita PM, Tanasijevic MJ. What proportion of vancomycin trough levels are drawn too early?: frequency and impact on clinical actions. Am J Clin Pathol. 2012;137(3):472‐478. doi:10.1309/AJCPDSYS0DVLKFOH
17. Melanson SE, Mijailovic AS, Wright AP, Szumita PM, Bates DW, Tanasijevic MJ. An intervention to improve the timing of vancomycin levels. Am J Clin Pathol. 2013;140(6):801‐806. doi:10.1309/AJCPKQ6EAH7OYQLB
18. Meng L, Wong T, Huang S, et al. Conversion from vancomycin trough concentration-guided dosing to area under the curve-guided dosing using two sample measurements in adults: implementation at an academic medical center. Pharmacotherapy. 2019;39(4):433‐442. doi:10.1002/phar.2234
19. Stoessel AM, Hale CM, Seabury RW, Miller CD, Steele JM. The impact of AUC-based monitoring on pharmacist-directed vancomycin dose adjustments in complicated methicillin-resistant staphylococcus aureus Infection. J Pharm Pract. 2019;32(4):442‐446. doi:10.1177/0897190018764564
20. Kufel WD, Seabury RW, Mogle BT, Beccari MV, Probst LA, Steele JM. Readiness to implement vancomycin monitoring based on area under the concentration-time curve: a cross-sectional survey of a national health consortium. Am J Health Syst Pharm. 2019;76(12):889‐894. doi:10.1093/ajhp/zxz070
21. Bluestone J, Johnson P, Fullerton J, Carr C, Alderman J, BonTempo J. Effective in-service training design and delivery: evidence from an integrative literature review. Hum Resour Health. 2013;11:51. doi:10.1186/1478-4491-11-51
22. Ebben RHA, Siqeca F, Madsen UR, Vloet LCM, van Achterberg T. Effectiveness of implementation strategies for the improvement of guideline and protocol adherence in emergency care: a systematic review. BMJ Open. 2018;8(11):e017572. doi:10.1136/bmjopen-2017-017572
23. Fischer F, Lange K, Klose K, Greiner W, Kraemer A. Barriers and Strategies in Guideline Implementation-A Scoping Review. Healthcare (Basel). 2016;4(3):36. doi:10.3390/healthcare4030036
The use of weight-based dosing with trough-based monitoring of vancomycin has been in clinical practice for more than a decade. The American Society of Health-System Pharmacists (ASHP), the Infectious Diseases Society of America (IDSA), and the Society of Infectious Diseases Pharmacists (SIDP) published the first guidelines for vancomycin monitoring in 2009.1 Although it has been well established that area under the curve (AUC) over the minimal inhibitory concentration (MIC) ratio > 400 mg.h/L is the best predictor of clinical efficacy, obtaining this value in clinical practice was not pragmatic. Therefore, the 2009 guidelines recommended a goal vancomycin trough of 15 to 20 mcg/ml as a surrogate marker for AUC/MIC > 400 mg.hr/L. This has since become a common practice despite little data that support this recommendation.
The efficacy and safety of trough-based monitoring has been evaluated extensively over the past several years and more recent data suggest that there is wide patient variability in AUC with this method and higher trough levels are associated with more nephrotoxicity.2,3 ASHP, IDSA, SIDP, and the Pediatric Infectious Diseases Society (PIDS) updated the consensus guidelines in 2020.4 Trough-based monitoring is no longer recommended. Instead AUC24 monitoring should be implemented with a goal range of 400 to 600 mg.h/L for efficacy and safety. Given concerns for vancomycin penetration into the central nervous system (CNS), many facility protocols utilize higher targets (> 600 mg.h/L) for CNS infections.
Some hospitals have been utilizing AUC-based monitoring for years. There are strategies from tertiary care centers that drive this practice change in the medical literature.5,6 However, it is important to reproduce these implementation practices in small, rural facilities that may face unique challenges with limited resources and may be slower to implement consensus guidelines.7,8 As this is a major practice change, it is imperative to evaluate the extent of transition and identify areas of needed improvement.
Accurate therapeutic drug monitoring ensures both the safety and efficacy of vancomycin therapy. Unfortunately, research shows that inappropriate laboratory tests are common in medical facilities.9 Drug levels taken inappropriately can lead to delays in therapeutic decision-making, inappropriate dosage adjustments and create a need for repeated drug levels, which increases the overall cost of admission.
Given the multiple affected services needed to make successful practice transitions, it is paramount that facilities evaluate progress during the transition phase. The Agency for Healthcare Research and Quality and the Institute for Healthcare Improvement provide guidance in the Plan-Do-Study-Act Cycle for quality assessment and improvement of new initiatives.10,11 A gap analysis can be used as a simple tool for evaluating the transition of research into practice and to identify areas of needed improvement.
The Veterans Health Care System of the Ozarks (VHSO) in Fayetteville, Arkansas made the transition from trough-based monitoring to 2-level AUC-based monitoring on April 1, 2019. The purpose of this study was to evaluate the effectiveness of transition methods used to implement AUC-monitoring for vancomycin treated patients in a small, primary facility. A further goal of the study was to identify areas of needed improvement and education and whether the problems derived from deficiencies in knowledge and ordering (medical and pharmacy services) or execution (nursing and laboratory services).
Methods
VHSO is a 52-bed US Department of Veterans Affairs primary care hospital. The pharmacy and laboratory are staffed 24 hours each day. There is 1 clinical pharmacy specialist (CPS) available for therapeutic drug monitoring consults Monday through Friday between the hours of 7:30 AM and 4:00 PM. No partial full-time equivalent employees were added for this conversion. Pharmacy-driven vancomycin dosing and monitoring is conducted on a collaborative basis, with pharmacy managing the majority of vancomycin treated patients. Night and weekend pharmacy staff provide cross-coverage on vancomycin consultations. Laboratory orders and medication dosage adjustments fall within the CPS scope of practice. Nurses do not perform laboratory draws for therapeutic drug monitoring; this is done solely by phlebotomists. There is no infectious diseases specialist at the facility to champion antibiotic dosing initiatives.
The implementation strategy largely reflected those outlined from tertiary care centers.5,6 First, key personnel from the laboratory department met to discuss this practice change and to add vancomycin peaks to the ordering menu. A critical value was set at 40 mcg/ml. Vancomycin troughs and random levels already were orderable items. A comment field was added to all laboratory orders for further clarification. Verbiage was added to laboratory reports in the computerized medical record to assist clinicians in determining the appropriateness of the level. This was followed by an educational email to both the nursing and laboratory departments explaining the practice change and included a link to the Pharmacy Joe “Vancomycin Dosing by AUC:MIC Instead of Trough-level” podcast (www.pharmacyjoe.com episode 356).
The pharmacy department received an interactive 30-minute presentation, followed immediately by a group activity to discuss practice problems. This presentation was condensed, recorded, and emailed to all VHSO pharmacists. A shared folder contained pertinent material on AUC monitoring.
Finally, an interactive presentation was set up for hospitalists and a video teleconferencing was conducted for rotating medical residents. Both the podcast and recorded presentation were emailed to the entire medical staff with a brief introduction of the practice change. Additionally, the transition process was added as a standing item on the monthly antimicrobial stewardship meeting agenda.
The standardized pharmacokinetic model at the study facility consisted of a vancomycin volume of distribution of 0.7 mg/kg and elimination rate constant (Ke) by Matzke and colleagues for total daily dose calculations.12 Obese patients (BMI ≥ 30) undergo alternative clearance equations described by Crass and colleagues.13 Cockcroft-Gault methods using ideal body weight (or actual body weight if < ideal body weight) are used for determining creatinine clearance. In patients aged ≥ 65 years with a serum creatinine < 1.0 mg/dL, facility guidance was to round serum creatinine up to 1.0 mg/dL. Loading doses were determined on a case-by-case basis with a cap of 2,000 mg, maintenance doses were rounded to the nearest 250 mg.
Vancomycin levels typically are drawn at steady state and analyzed using the logarithmic trapezoidal rule.14 The pharmacy and medical staff were educated to provide details on timing and coordination in nursing and laboratory orders (Table 1). Two-level AUC monitoring typically is not performed in patients with acute renal failure, expected duration of therapy < 72 hours, urinary tract infections, skin and soft tissue infections, or in renal replacement therapy.5
This gap analysis consisted of a retrospective chart review of vancomycin levels ordered after the implementation of AUC-based monitoring to determine the effectiveness of the transition. Three months of data were collected between April 2019 and June 2019. Vancomycin levels were deemed either appropriate or inappropriate based on timing and type (peak, trough, or random) of the laboratory test in relation to the previously administered vancomycin dose. Appropriate peaks were drawn within 2 hours after the end of infusion and troughs at least 1 half-life after the dose or just prior to the next dose and within the same dosing interval as the peak. Tests drawn outside of the specified time range, trough-only laboratory tests, or those drawn after vancomycin had been discontinued were considered inappropriate. Peaks and troughs drawn from separate dosing intervals also were considered inappropriate. Random levels were considered appropriate only if they fit the clinical context in acute renal failure or renal replacement therapy. An effective transition was defined as ≥ 80% of all vancomycin treated patients monitored with AUC methods rather than trough-based methods.
Inclusion criteria included all vancomycin levels ordered during the study period with no exclusions. The primary endpoint was the proportion of vancomycin levels drawn appropriately. Secondary endpoints were the proportion of AUC24 calculations within therapeutic range and a stratification of reasons for inappropriate levels. Descriptive statistics were collected to describe the scope of the project. Levels drawn from various shifts were compared (ie, day, night, or weekend). Calculated AUC24 levels between 400 and 600 mg.h/L were considered therapeutic unless treating CNS infection (600-700 mg.h/L). Given the operational outcomes (rather than clinical outcomes) and no comparator group, patient specific data were not collected.
Descriptive statistics without further analysis were used to describe proportions. The goal level for compliance was set at 100%. These methods were reviewed by the VHSO Institutional Review Board and granted nonresearch status, waiving the requirement for informed consent.
Results
The transition was effective with 97% of all cases utilizing AUC-based methods for monitoring. A total of 65 vancomycin levels were drawn in the study period; 32 peaks, 32 troughs, and 1 random level (drawn appropriately during acute renal failure 24 hours after starting therapy). All shifts were affected proportionately; days (n = 26, 40%), nights (n = 18, 27.7%), and weekends (n = 21, 32.3%). Based on time of dosage administration and laboratory test, there were 9 levels (13.8%) deemed inappropriate, 56 levels (86.1%) were appropriate. Reasons for inappropriate levels gleaned from chart review are presented in Table 2. Four levels had to be repeated for accurate calculations.
From the peak/trough couplets drawn appropriately, calculated AUC24 fell with the desired range in 61% (n = 17) of cases. Of the 11 that fell outside of range, 8 were subtherapeutic (< 400 mg.h/L) and 3 were supratherapeutic (> 600 mg.h/L). All levels were drawn at steady state. Indications for vancomycin monitoring were osteomyelitis (n = 13, 43%), sepsis (n = 10, 33%), pneumonia (n = 6, 20%), and 1 case of meningitis (3%).
Discussion
To the author’s knowledge, this is the first report of a vancomycin AUC24 monitoring conversion in a rural facility. This study adds to the existing medical literature in that it demonstrates that: (1) implementation methods described in large, tertiary centers can be effectively utilized in primary care, rural facilities; (2) the gap analysis used can be duplicated with minimal personnel and resources to ensure effective implementation (Table 3); and (3) the reported improvement needs can serve as a model for preventative measures at other facilities. The incidence of appropriate vancomycin levels was notably better than those reported in other single center studies.15-17 However, given variations in study design and facility operating procedures, it would be difficult to compare incidence among medical facilities. As such, there are no consensus benchmarks for comparison. The majority of inappropriate levels occurred early in the study period and on weekends. Appropriateness of drug levels may have improved with continued feedback and familiarity.
The calculated AUC24 fell within predicted range in 61% of cases. For comparison, a recent study from a large academic medical center reported that 73.5% of 2-level AUC24 cases had initial values within the therapeutic range.18 Of note, the target range used was much wider (400 - 800 mg.h/L) than the present study. Another study reported dose adjustments for subtherapeutic AUC levels in 25% of cases and dose reductions for supratherapeutic levels in 33.3% of cases.19
Of the AUC24 calculations that fell outside of therapeutic range, the majority (n = 8, 73%) were subtherapeutic (< 400 mg.h/L), half of these were for patients who were obese. It was unclear in the medical record which equation was used for initial dosing (Matzke vs Crass), or whether more conservative AUCs were used for calculating the total daily dose. The VHSO policy limiting loading doses also may have played a role; indeed the updated guidelines recommend a maximum loading dose of 3,000 mg depending on the severity of infection.4 Two of the 3 supratherapeutic levels were thought to be due to accumulation with long-term therapy.
Given such a large change from long-standing practices, there was surprisingly little resistance from the various clinical services. A recent survey of academic medical centers reported that the majority (88%) of all respondents who did not currently utilize AUC24 monitoring did not plan on making this immediate transition, largely citing unfamiliarity and training requirements.20 It is conceivable that the transition to AUC monitoring in smaller facilities may have fewer barriers than those seen in tertiary care centers. There are fewer health care providers and pharmacists to educate with the primary responsibilities falling on relatively few clinicians. There is little question as to who will be conducting follow up or whom to contact for questions. A smaller patient load and lesser patient acuity may translate to fewer vancomycin cases that require monitoring.
The interactive meetings were an important element for facility implementation. Research shows that emails alone are not effective for health care provider education, and interactive methods are recommended over passive methods.21,22 Assessing and avoiding barriers up front such as unclear laboratory orders, or communication failures is paramount to successful implementation strategies.23 Additionally, the detailed written ordering communication may have contributed to a smoother transition. The educational recording proved to be helpful in educating new staff and residents. An identified logistical error was that laboratory orders entered while patients were enrolled in sham clinics for electronic workload capture (eg, Pharmacy Inpatient Clinic) created confusion on the physical location of the patient for the phlebotomists, potentially causing delays in specimen collection.
A major development that stemmed from this intervention was that the Medical Service asked that policy changes be made so that the Pharmacy Service take over all vancomycin dosing at the facility. Previously, this had been done on a collaborative basis. Similar facilities with a collaborative practice model may need to anticipate such a request as this may present a new set of challenges. Accordingly, the pharmacy department is in the process of establishing standing operating procedures, pharmacist competencies, and a facility memorandum. Future research should evaluate the safety and efficacy of vancomycin therapy after the switch to AUC-based monitoring.
Limitations
There are several limitations to consider with this study. Operating procedures and implementation processes may vary between facilities, which could limit the generalizability of these results. Given the small facility size, the overall number of laboratory tests drawn was much smaller than those seen in larger facilities. The time needed for AUC calculations is notably longer than older methods of monitoring; however, this was not objectively assessed. It is important to note that clinical outcomes were beyond the scope of this gap analysis and this is an area of future research at the study facility. Vancomycin laboratory tests that were missed due to procedures and subsequently rescheduled were occasionally observed but not accounted for in this analysis. Additionally, vancomycin courses without monitoring (appropriate or otherwise) when indicated were not assessed. However, anecdotally speaking, this would be a very unlikely occurrence.
Conclusion
Conversion to AUC-based vancomycin monitoring is feasible in primary, rural medical centers. Implementation strategies from tertiary facilities can be successfully utilized in smaller hospitals. Quality assessment strategies such as a gap analysis can be utilized with minimal resources for facility uptake of new clinical practices.
The use of weight-based dosing with trough-based monitoring of vancomycin has been in clinical practice for more than a decade. The American Society of Health-System Pharmacists (ASHP), the Infectious Diseases Society of America (IDSA), and the Society of Infectious Diseases Pharmacists (SIDP) published the first guidelines for vancomycin monitoring in 2009.1 Although it has been well established that area under the curve (AUC) over the minimal inhibitory concentration (MIC) ratio > 400 mg.h/L is the best predictor of clinical efficacy, obtaining this value in clinical practice was not pragmatic. Therefore, the 2009 guidelines recommended a goal vancomycin trough of 15 to 20 mcg/ml as a surrogate marker for AUC/MIC > 400 mg.hr/L. This has since become a common practice despite little data that support this recommendation.
The efficacy and safety of trough-based monitoring has been evaluated extensively over the past several years and more recent data suggest that there is wide patient variability in AUC with this method and higher trough levels are associated with more nephrotoxicity.2,3 ASHP, IDSA, SIDP, and the Pediatric Infectious Diseases Society (PIDS) updated the consensus guidelines in 2020.4 Trough-based monitoring is no longer recommended. Instead AUC24 monitoring should be implemented with a goal range of 400 to 600 mg.h/L for efficacy and safety. Given concerns for vancomycin penetration into the central nervous system (CNS), many facility protocols utilize higher targets (> 600 mg.h/L) for CNS infections.
Some hospitals have been utilizing AUC-based monitoring for years. There are strategies from tertiary care centers that drive this practice change in the medical literature.5,6 However, it is important to reproduce these implementation practices in small, rural facilities that may face unique challenges with limited resources and may be slower to implement consensus guidelines.7,8 As this is a major practice change, it is imperative to evaluate the extent of transition and identify areas of needed improvement.
Accurate therapeutic drug monitoring ensures both the safety and efficacy of vancomycin therapy. Unfortunately, research shows that inappropriate laboratory tests are common in medical facilities.9 Drug levels taken inappropriately can lead to delays in therapeutic decision-making, inappropriate dosage adjustments and create a need for repeated drug levels, which increases the overall cost of admission.
Given the multiple affected services needed to make successful practice transitions, it is paramount that facilities evaluate progress during the transition phase. The Agency for Healthcare Research and Quality and the Institute for Healthcare Improvement provide guidance in the Plan-Do-Study-Act Cycle for quality assessment and improvement of new initiatives.10,11 A gap analysis can be used as a simple tool for evaluating the transition of research into practice and to identify areas of needed improvement.
The Veterans Health Care System of the Ozarks (VHSO) in Fayetteville, Arkansas made the transition from trough-based monitoring to 2-level AUC-based monitoring on April 1, 2019. The purpose of this study was to evaluate the effectiveness of transition methods used to implement AUC-monitoring for vancomycin treated patients in a small, primary facility. A further goal of the study was to identify areas of needed improvement and education and whether the problems derived from deficiencies in knowledge and ordering (medical and pharmacy services) or execution (nursing and laboratory services).
Methods
VHSO is a 52-bed US Department of Veterans Affairs primary care hospital. The pharmacy and laboratory are staffed 24 hours each day. There is 1 clinical pharmacy specialist (CPS) available for therapeutic drug monitoring consults Monday through Friday between the hours of 7:30 AM and 4:00 PM. No partial full-time equivalent employees were added for this conversion. Pharmacy-driven vancomycin dosing and monitoring is conducted on a collaborative basis, with pharmacy managing the majority of vancomycin treated patients. Night and weekend pharmacy staff provide cross-coverage on vancomycin consultations. Laboratory orders and medication dosage adjustments fall within the CPS scope of practice. Nurses do not perform laboratory draws for therapeutic drug monitoring; this is done solely by phlebotomists. There is no infectious diseases specialist at the facility to champion antibiotic dosing initiatives.
The implementation strategy largely reflected those outlined from tertiary care centers.5,6 First, key personnel from the laboratory department met to discuss this practice change and to add vancomycin peaks to the ordering menu. A critical value was set at 40 mcg/ml. Vancomycin troughs and random levels already were orderable items. A comment field was added to all laboratory orders for further clarification. Verbiage was added to laboratory reports in the computerized medical record to assist clinicians in determining the appropriateness of the level. This was followed by an educational email to both the nursing and laboratory departments explaining the practice change and included a link to the Pharmacy Joe “Vancomycin Dosing by AUC:MIC Instead of Trough-level” podcast (www.pharmacyjoe.com episode 356).
The pharmacy department received an interactive 30-minute presentation, followed immediately by a group activity to discuss practice problems. This presentation was condensed, recorded, and emailed to all VHSO pharmacists. A shared folder contained pertinent material on AUC monitoring.
Finally, an interactive presentation was set up for hospitalists and a video teleconferencing was conducted for rotating medical residents. Both the podcast and recorded presentation were emailed to the entire medical staff with a brief introduction of the practice change. Additionally, the transition process was added as a standing item on the monthly antimicrobial stewardship meeting agenda.
The standardized pharmacokinetic model at the study facility consisted of a vancomycin volume of distribution of 0.7 mg/kg and elimination rate constant (Ke) by Matzke and colleagues for total daily dose calculations.12 Obese patients (BMI ≥ 30) undergo alternative clearance equations described by Crass and colleagues.13 Cockcroft-Gault methods using ideal body weight (or actual body weight if < ideal body weight) are used for determining creatinine clearance. In patients aged ≥ 65 years with a serum creatinine < 1.0 mg/dL, facility guidance was to round serum creatinine up to 1.0 mg/dL. Loading doses were determined on a case-by-case basis with a cap of 2,000 mg, maintenance doses were rounded to the nearest 250 mg.
Vancomycin levels typically are drawn at steady state and analyzed using the logarithmic trapezoidal rule.14 The pharmacy and medical staff were educated to provide details on timing and coordination in nursing and laboratory orders (Table 1). Two-level AUC monitoring typically is not performed in patients with acute renal failure, expected duration of therapy < 72 hours, urinary tract infections, skin and soft tissue infections, or in renal replacement therapy.5
This gap analysis consisted of a retrospective chart review of vancomycin levels ordered after the implementation of AUC-based monitoring to determine the effectiveness of the transition. Three months of data were collected between April 2019 and June 2019. Vancomycin levels were deemed either appropriate or inappropriate based on timing and type (peak, trough, or random) of the laboratory test in relation to the previously administered vancomycin dose. Appropriate peaks were drawn within 2 hours after the end of infusion and troughs at least 1 half-life after the dose or just prior to the next dose and within the same dosing interval as the peak. Tests drawn outside of the specified time range, trough-only laboratory tests, or those drawn after vancomycin had been discontinued were considered inappropriate. Peaks and troughs drawn from separate dosing intervals also were considered inappropriate. Random levels were considered appropriate only if they fit the clinical context in acute renal failure or renal replacement therapy. An effective transition was defined as ≥ 80% of all vancomycin treated patients monitored with AUC methods rather than trough-based methods.
Inclusion criteria included all vancomycin levels ordered during the study period with no exclusions. The primary endpoint was the proportion of vancomycin levels drawn appropriately. Secondary endpoints were the proportion of AUC24 calculations within therapeutic range and a stratification of reasons for inappropriate levels. Descriptive statistics were collected to describe the scope of the project. Levels drawn from various shifts were compared (ie, day, night, or weekend). Calculated AUC24 levels between 400 and 600 mg.h/L were considered therapeutic unless treating CNS infection (600-700 mg.h/L). Given the operational outcomes (rather than clinical outcomes) and no comparator group, patient specific data were not collected.
Descriptive statistics without further analysis were used to describe proportions. The goal level for compliance was set at 100%. These methods were reviewed by the VHSO Institutional Review Board and granted nonresearch status, waiving the requirement for informed consent.
Results
The transition was effective with 97% of all cases utilizing AUC-based methods for monitoring. A total of 65 vancomycin levels were drawn in the study period; 32 peaks, 32 troughs, and 1 random level (drawn appropriately during acute renal failure 24 hours after starting therapy). All shifts were affected proportionately; days (n = 26, 40%), nights (n = 18, 27.7%), and weekends (n = 21, 32.3%). Based on time of dosage administration and laboratory test, there were 9 levels (13.8%) deemed inappropriate, 56 levels (86.1%) were appropriate. Reasons for inappropriate levels gleaned from chart review are presented in Table 2. Four levels had to be repeated for accurate calculations.
From the peak/trough couplets drawn appropriately, calculated AUC24 fell with the desired range in 61% (n = 17) of cases. Of the 11 that fell outside of range, 8 were subtherapeutic (< 400 mg.h/L) and 3 were supratherapeutic (> 600 mg.h/L). All levels were drawn at steady state. Indications for vancomycin monitoring were osteomyelitis (n = 13, 43%), sepsis (n = 10, 33%), pneumonia (n = 6, 20%), and 1 case of meningitis (3%).
Discussion
To the author’s knowledge, this is the first report of a vancomycin AUC24 monitoring conversion in a rural facility. This study adds to the existing medical literature in that it demonstrates that: (1) implementation methods described in large, tertiary centers can be effectively utilized in primary care, rural facilities; (2) the gap analysis used can be duplicated with minimal personnel and resources to ensure effective implementation (Table 3); and (3) the reported improvement needs can serve as a model for preventative measures at other facilities. The incidence of appropriate vancomycin levels was notably better than those reported in other single center studies.15-17 However, given variations in study design and facility operating procedures, it would be difficult to compare incidence among medical facilities. As such, there are no consensus benchmarks for comparison. The majority of inappropriate levels occurred early in the study period and on weekends. Appropriateness of drug levels may have improved with continued feedback and familiarity.
The calculated AUC24 fell within predicted range in 61% of cases. For comparison, a recent study from a large academic medical center reported that 73.5% of 2-level AUC24 cases had initial values within the therapeutic range.18 Of note, the target range used was much wider (400 - 800 mg.h/L) than the present study. Another study reported dose adjustments for subtherapeutic AUC levels in 25% of cases and dose reductions for supratherapeutic levels in 33.3% of cases.19
Of the AUC24 calculations that fell outside of therapeutic range, the majority (n = 8, 73%) were subtherapeutic (< 400 mg.h/L), half of these were for patients who were obese. It was unclear in the medical record which equation was used for initial dosing (Matzke vs Crass), or whether more conservative AUCs were used for calculating the total daily dose. The VHSO policy limiting loading doses also may have played a role; indeed the updated guidelines recommend a maximum loading dose of 3,000 mg depending on the severity of infection.4 Two of the 3 supratherapeutic levels were thought to be due to accumulation with long-term therapy.
Given such a large change from long-standing practices, there was surprisingly little resistance from the various clinical services. A recent survey of academic medical centers reported that the majority (88%) of all respondents who did not currently utilize AUC24 monitoring did not plan on making this immediate transition, largely citing unfamiliarity and training requirements.20 It is conceivable that the transition to AUC monitoring in smaller facilities may have fewer barriers than those seen in tertiary care centers. There are fewer health care providers and pharmacists to educate with the primary responsibilities falling on relatively few clinicians. There is little question as to who will be conducting follow up or whom to contact for questions. A smaller patient load and lesser patient acuity may translate to fewer vancomycin cases that require monitoring.
The interactive meetings were an important element for facility implementation. Research shows that emails alone are not effective for health care provider education, and interactive methods are recommended over passive methods.21,22 Assessing and avoiding barriers up front such as unclear laboratory orders, or communication failures is paramount to successful implementation strategies.23 Additionally, the detailed written ordering communication may have contributed to a smoother transition. The educational recording proved to be helpful in educating new staff and residents. An identified logistical error was that laboratory orders entered while patients were enrolled in sham clinics for electronic workload capture (eg, Pharmacy Inpatient Clinic) created confusion on the physical location of the patient for the phlebotomists, potentially causing delays in specimen collection.
A major development that stemmed from this intervention was that the Medical Service asked that policy changes be made so that the Pharmacy Service take over all vancomycin dosing at the facility. Previously, this had been done on a collaborative basis. Similar facilities with a collaborative practice model may need to anticipate such a request as this may present a new set of challenges. Accordingly, the pharmacy department is in the process of establishing standing operating procedures, pharmacist competencies, and a facility memorandum. Future research should evaluate the safety and efficacy of vancomycin therapy after the switch to AUC-based monitoring.
Limitations
There are several limitations to consider with this study. Operating procedures and implementation processes may vary between facilities, which could limit the generalizability of these results. Given the small facility size, the overall number of laboratory tests drawn was much smaller than those seen in larger facilities. The time needed for AUC calculations is notably longer than older methods of monitoring; however, this was not objectively assessed. It is important to note that clinical outcomes were beyond the scope of this gap analysis and this is an area of future research at the study facility. Vancomycin laboratory tests that were missed due to procedures and subsequently rescheduled were occasionally observed but not accounted for in this analysis. Additionally, vancomycin courses without monitoring (appropriate or otherwise) when indicated were not assessed. However, anecdotally speaking, this would be a very unlikely occurrence.
Conclusion
Conversion to AUC-based vancomycin monitoring is feasible in primary, rural medical centers. Implementation strategies from tertiary facilities can be successfully utilized in smaller hospitals. Quality assessment strategies such as a gap analysis can be utilized with minimal resources for facility uptake of new clinical practices.
1. Rybak M, Lomaestro B, Rotschafer JC, et al. Therapeutic monitoring of vancomycin in adult patients: a consensus review of the American Society of Health-System Pharmacists, the Infectious Diseases Society of America, and the Society of Infectious Diseases Pharmacists [published correction appears in Am J Health Syst Pharm. 2009;66(10):887]. Am J Health Syst Pharm. 2009;66(1):82‐98. doi:10.2146/ajhp080434
2. van Hal SJ, Paterson DL, Lodise TP. Systematic review and meta-analysis of vancomycin-induced nephrotoxicity associated with dosing schedules that maintain troughs between 15 and 20 milligrams per liter. Antimicrob Agents Chemother. 2013;57(2):734‐744. doi:10.1128/AAC.01568-12
3. Pai MP, Neely M, Rodvold KA, Lodise TP. Innovative approaches to optimizing the delivery of vancomycin in individual patients. Adv Drug Deliv Rev. 2014;77:50‐57. doi:10.1016/j.addr.2014.05.016
4. Rybak MJ, Le J, Lodise TP, et al. Therapeutic monitoring of vancomycin for serious methicillin-resistant Staphylococcus aureus infections: a revised consensus guideline and review by the American Society of Health-System Pharmacists, the Infectious Diseases Society of America, the Pediatric Infectious Diseases Society, and the Society of Infectious Diseases Pharmacists [published online ahead of print, 2020 Mar 19]. Am J Health Syst Pharm. 2020;zxaa036. doi:10.1093/ajhp/zxaa036
5. Heil EL, Claeys KC, Mynatt RP, et al. Making the change to area under the curve-based vancomycin dosing. Am J Health Syst Pharm. 2018;75(24):1986‐1995. doi:10.2146/ajhp180034
6. Gregory ER, Burgess DR, Cotner SE, et al. Vancomycin area under the curve dosing and monitoring at an academic medical center: transition strategies and lessons learned [published online ahead of print, 2019 Mar 10]. J Pharm Pract. 2019;897190019834369. doi:10.1177/0897190019834369
7. Septimus EJ, Owens RC Jr. Need and potential of antimicrobial stewardship in community hospitals. Clin Infect Dis. 2011;53 Suppl 1:S8‐S14. doi:10.1093/cid/cir363
8. Goldman LE, Dudley RA. United States rural hospital quality in the Hospital Compare database-accounting for hospital characteristics. Health Policy. 2008;87(1):112‐127. doi:10.1016/j.healthpol.2008.02.002
9. Zhi M, Ding EL, Theisen-Toupal J, Whelan J, Arnaout R. The landscape of inappropriate laboratory testing: a 15-year meta-analysis. PLoS One. 2013;8(11):e78962. doi:10.1371/journal.pone.0078962
10. Institute for Healthcare Improvement. Plan-do-study-act (PDSA) worksheet. http://www.ihi.org/resources/Pages/Tools/PlanDoStudyActWorksheet.aspx. Accessed May 13, 2020.
11. Agency for Healthcare Research and Quality. Plan-do-study-act (PDSA) cycle. https://innovations.ahrq.gov/qualitytools/plan-do-study-act-pdsa-cycle. Updated April 10, 2013. Accessed May 13, 2020.
12. Matzke GR, McGory RW, Halstenson CE, Keane WF. Pharmacokinetics of vancomycin in patients with various degrees of renal function. Antimicrob Agents Chemother. 1984;25(4):433‐437. doi:10.1128/aac.25.4.433
13. Crass RL, Dunn R, Hong J, Krop LC, Pai MP. Dosing vancomycin in the super obese: less is more. J Antimicrob Chemother. 2018;73(11):3081‐3086. doi:10.1093/jac/dky310
14. Pai MP, Russo A, Novelli A, Venditti M, Falcone M. Simplified equations using two concentrations to calculate area under the curve for antimicrobials with concentration-dependent pharmacodynamics: daptomycin as a motivating example. Antimicrob Agents Chemother. 2014;58(6):3162‐3167. doi:10.1128/AAC.02355-14
15. Suryadevara M, Steidl KE, Probst LA, Shaw J. Inappropriate vancomycin therapeutic drug monitoring in hospitalized pediatric patients increases pediatric trauma and hospital costs. J Pediatr Pharmacol Ther. 2012;17(2):159‐165. doi:10.5863/1551-6776-17.2.159
16. Morrison AP, Melanson SE, Carty MG, Bates DW, Szumita PM, Tanasijevic MJ. What proportion of vancomycin trough levels are drawn too early?: frequency and impact on clinical actions. Am J Clin Pathol. 2012;137(3):472‐478. doi:10.1309/AJCPDSYS0DVLKFOH
17. Melanson SE, Mijailovic AS, Wright AP, Szumita PM, Bates DW, Tanasijevic MJ. An intervention to improve the timing of vancomycin levels. Am J Clin Pathol. 2013;140(6):801‐806. doi:10.1309/AJCPKQ6EAH7OYQLB
18. Meng L, Wong T, Huang S, et al. Conversion from vancomycin trough concentration-guided dosing to area under the curve-guided dosing using two sample measurements in adults: implementation at an academic medical center. Pharmacotherapy. 2019;39(4):433‐442. doi:10.1002/phar.2234
19. Stoessel AM, Hale CM, Seabury RW, Miller CD, Steele JM. The impact of AUC-based monitoring on pharmacist-directed vancomycin dose adjustments in complicated methicillin-resistant staphylococcus aureus Infection. J Pharm Pract. 2019;32(4):442‐446. doi:10.1177/0897190018764564
20. Kufel WD, Seabury RW, Mogle BT, Beccari MV, Probst LA, Steele JM. Readiness to implement vancomycin monitoring based on area under the concentration-time curve: a cross-sectional survey of a national health consortium. Am J Health Syst Pharm. 2019;76(12):889‐894. doi:10.1093/ajhp/zxz070
21. Bluestone J, Johnson P, Fullerton J, Carr C, Alderman J, BonTempo J. Effective in-service training design and delivery: evidence from an integrative literature review. Hum Resour Health. 2013;11:51. doi:10.1186/1478-4491-11-51
22. Ebben RHA, Siqeca F, Madsen UR, Vloet LCM, van Achterberg T. Effectiveness of implementation strategies for the improvement of guideline and protocol adherence in emergency care: a systematic review. BMJ Open. 2018;8(11):e017572. doi:10.1136/bmjopen-2017-017572
23. Fischer F, Lange K, Klose K, Greiner W, Kraemer A. Barriers and Strategies in Guideline Implementation-A Scoping Review. Healthcare (Basel). 2016;4(3):36. doi:10.3390/healthcare4030036
1. Rybak M, Lomaestro B, Rotschafer JC, et al. Therapeutic monitoring of vancomycin in adult patients: a consensus review of the American Society of Health-System Pharmacists, the Infectious Diseases Society of America, and the Society of Infectious Diseases Pharmacists [published correction appears in Am J Health Syst Pharm. 2009;66(10):887]. Am J Health Syst Pharm. 2009;66(1):82‐98. doi:10.2146/ajhp080434
2. van Hal SJ, Paterson DL, Lodise TP. Systematic review and meta-analysis of vancomycin-induced nephrotoxicity associated with dosing schedules that maintain troughs between 15 and 20 milligrams per liter. Antimicrob Agents Chemother. 2013;57(2):734‐744. doi:10.1128/AAC.01568-12
3. Pai MP, Neely M, Rodvold KA, Lodise TP. Innovative approaches to optimizing the delivery of vancomycin in individual patients. Adv Drug Deliv Rev. 2014;77:50‐57. doi:10.1016/j.addr.2014.05.016
4. Rybak MJ, Le J, Lodise TP, et al. Therapeutic monitoring of vancomycin for serious methicillin-resistant Staphylococcus aureus infections: a revised consensus guideline and review by the American Society of Health-System Pharmacists, the Infectious Diseases Society of America, the Pediatric Infectious Diseases Society, and the Society of Infectious Diseases Pharmacists [published online ahead of print, 2020 Mar 19]. Am J Health Syst Pharm. 2020;zxaa036. doi:10.1093/ajhp/zxaa036
5. Heil EL, Claeys KC, Mynatt RP, et al. Making the change to area under the curve-based vancomycin dosing. Am J Health Syst Pharm. 2018;75(24):1986‐1995. doi:10.2146/ajhp180034
6. Gregory ER, Burgess DR, Cotner SE, et al. Vancomycin area under the curve dosing and monitoring at an academic medical center: transition strategies and lessons learned [published online ahead of print, 2019 Mar 10]. J Pharm Pract. 2019;897190019834369. doi:10.1177/0897190019834369
7. Septimus EJ, Owens RC Jr. Need and potential of antimicrobial stewardship in community hospitals. Clin Infect Dis. 2011;53 Suppl 1:S8‐S14. doi:10.1093/cid/cir363
8. Goldman LE, Dudley RA. United States rural hospital quality in the Hospital Compare database-accounting for hospital characteristics. Health Policy. 2008;87(1):112‐127. doi:10.1016/j.healthpol.2008.02.002
9. Zhi M, Ding EL, Theisen-Toupal J, Whelan J, Arnaout R. The landscape of inappropriate laboratory testing: a 15-year meta-analysis. PLoS One. 2013;8(11):e78962. doi:10.1371/journal.pone.0078962
10. Institute for Healthcare Improvement. Plan-do-study-act (PDSA) worksheet. http://www.ihi.org/resources/Pages/Tools/PlanDoStudyActWorksheet.aspx. Accessed May 13, 2020.
11. Agency for Healthcare Research and Quality. Plan-do-study-act (PDSA) cycle. https://innovations.ahrq.gov/qualitytools/plan-do-study-act-pdsa-cycle. Updated April 10, 2013. Accessed May 13, 2020.
12. Matzke GR, McGory RW, Halstenson CE, Keane WF. Pharmacokinetics of vancomycin in patients with various degrees of renal function. Antimicrob Agents Chemother. 1984;25(4):433‐437. doi:10.1128/aac.25.4.433
13. Crass RL, Dunn R, Hong J, Krop LC, Pai MP. Dosing vancomycin in the super obese: less is more. J Antimicrob Chemother. 2018;73(11):3081‐3086. doi:10.1093/jac/dky310
14. Pai MP, Russo A, Novelli A, Venditti M, Falcone M. Simplified equations using two concentrations to calculate area under the curve for antimicrobials with concentration-dependent pharmacodynamics: daptomycin as a motivating example. Antimicrob Agents Chemother. 2014;58(6):3162‐3167. doi:10.1128/AAC.02355-14
15. Suryadevara M, Steidl KE, Probst LA, Shaw J. Inappropriate vancomycin therapeutic drug monitoring in hospitalized pediatric patients increases pediatric trauma and hospital costs. J Pediatr Pharmacol Ther. 2012;17(2):159‐165. doi:10.5863/1551-6776-17.2.159
16. Morrison AP, Melanson SE, Carty MG, Bates DW, Szumita PM, Tanasijevic MJ. What proportion of vancomycin trough levels are drawn too early?: frequency and impact on clinical actions. Am J Clin Pathol. 2012;137(3):472‐478. doi:10.1309/AJCPDSYS0DVLKFOH
17. Melanson SE, Mijailovic AS, Wright AP, Szumita PM, Bates DW, Tanasijevic MJ. An intervention to improve the timing of vancomycin levels. Am J Clin Pathol. 2013;140(6):801‐806. doi:10.1309/AJCPKQ6EAH7OYQLB
18. Meng L, Wong T, Huang S, et al. Conversion from vancomycin trough concentration-guided dosing to area under the curve-guided dosing using two sample measurements in adults: implementation at an academic medical center. Pharmacotherapy. 2019;39(4):433‐442. doi:10.1002/phar.2234
19. Stoessel AM, Hale CM, Seabury RW, Miller CD, Steele JM. The impact of AUC-based monitoring on pharmacist-directed vancomycin dose adjustments in complicated methicillin-resistant staphylococcus aureus Infection. J Pharm Pract. 2019;32(4):442‐446. doi:10.1177/0897190018764564
20. Kufel WD, Seabury RW, Mogle BT, Beccari MV, Probst LA, Steele JM. Readiness to implement vancomycin monitoring based on area under the concentration-time curve: a cross-sectional survey of a national health consortium. Am J Health Syst Pharm. 2019;76(12):889‐894. doi:10.1093/ajhp/zxz070
21. Bluestone J, Johnson P, Fullerton J, Carr C, Alderman J, BonTempo J. Effective in-service training design and delivery: evidence from an integrative literature review. Hum Resour Health. 2013;11:51. doi:10.1186/1478-4491-11-51
22. Ebben RHA, Siqeca F, Madsen UR, Vloet LCM, van Achterberg T. Effectiveness of implementation strategies for the improvement of guideline and protocol adherence in emergency care: a systematic review. BMJ Open. 2018;8(11):e017572. doi:10.1136/bmjopen-2017-017572
23. Fischer F, Lange K, Klose K, Greiner W, Kraemer A. Barriers and Strategies in Guideline Implementation-A Scoping Review. Healthcare (Basel). 2016;4(3):36. doi:10.3390/healthcare4030036
Open Clinical Trials for Patients With COVID-19
Finding effective treatment or a vaccine for COVID-19, the disease caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has placed significant strains on the global health care system. The National Library of Medicine database lists > 1,800 trials that are aimed at addressing COVID-19-related health care. Already, trials developed by the US Department of Veterans Affairs (VA), US Department of Defense (DoD), and the National Institute of Allergy and Infectious Diseases have provided important data on effective treatment options. The clinical trials listed below are all open as of May 31, 2020 and have trial sites at VA and DoD facilities. For additional information and full inclusion/exclusion criteria, please consult clinicaltrials.gov.
Adaptive COVID-19 Treatment Trial (ACTT)
This study is an adaptive, randomized, double-blind, placebo-controlled trial to evaluate the safety and efficacy of novel therapeutic agents in hospitalized adults diagnosed with COVID-19. The study will compare different investigational therapeutic agents to a control arm. ID: NCT04280705
Sponsor: National Institute of Allergy and Infectious Diseases
Contact: Central Contact ([email protected])
Locations: VA Palo Alto Health Care System, California; Naval Medical Center San Diego, California; Southeast Louisiana Veterans Health Care System, New Orleans; Walter Reed National Military Medical Center, Bethesda, Maryland; National Institutes of Health - Clinical Center, National Institute of Allergy and Infectious Diseases Laboratory Of Immunoregulation, Bethesda, Maryland; Brooke Army Medical Center, Fort Sam Houston, Texas; Madigan Army Medical Center, Tacoma, Washington
Study to Evaluate the Safety and Antiviral Activity of Remdesivir (GS-5734) in Participants With Severe Coronavirus Disease (COVID-19)
The primary objective of this study is to evaluate the efficacy of 2 remdesivir (RDV) regimens with respect to clinical status assessed by a 7-point ordinal scale on Day 11 (NCT04292730) or Day 14 (NCT04292899).
ID: NCT04292730/NCT04292899
Sponsor: Gilead Sciences
Contact: Gilead Clinical Study Information Center (833-445-3230)
Location: James J. Peters VA Medical Center, Bronx, New York
Expanded Access Remdesivir (RDV; GS-5734)
The treatment of communicable Novel Coronavirus of 2019 with Remdesivir (RDV; GS-5734) also known as severe acute respiratory syndrome coronavirus 2.
ID: NCT04302766
Sponsor: US Army Medical Research and Development Command
Contact: Sandi Parriott ([email protected])
A Study to Evaluate the Safety and Efficacy of Tocilizumab in Patients With Severe COVID-19 Pneumonia (COVACTA)
This study will evaluate the efficacy, safety, pharmacodynamics, and pharmacokinetics of tocilizumab (TCZ) compared with a matching placebo in combination with standard of care (SOC) in hospitalized patients with severe COVID-19 pneumonia.
ID: NCT04320615
Sponsor: Hoffmann-La Roche
Location: James J Peters VA Medical Center, Bronx, New York
Administration of Intravenous Vitamin C in Novel Coronavirus Infection (COVID-19) and Decreased Oxygenation (AVoCaDO)
Previous research has shown that high dose intravenous vitamin C (HDIVC) may benefit patients with sepsis, acute lung injury (ALI), and the acute respiratory distress syndrome (ARDS). However, it is not known if early administration of HDIVC could prevent progression to ARDS. We hypothesize that HDIVC is safe and tolerable in COVID-19 subjects given early or late in the disease course and may reduce the risk of respiratory failure requiring mechanical ventilation and development of ARDS along with reductions in supplemental oxygen demand and inflammatory markers.
ID: NCT04357782
Sponsor: Hunter Holmes Mcguire VA Medical CenterContact: Brian Davis ([email protected])
Location: Hunter Holmes Mcguire VA Medical Center, Richmond, Virginia
Treatment Of CORONAVIRUS DISEASE 2019 (COVID-19) With Anti-Sars-CoV-2 Convalescent Plasma (ASCoV2CP)
This is an expanded access open-label, single-arm, multi-site protocol to provide convalescent plasma as a treatment for patients diagnosed with severe, or life-threatening COVID-19.
ID: NCT04360486
Sponsor: US Army Medical Research and Development Command
Contact: Andrew Cap ([email protected])
VA Remote and Equitable Access to COVID-19 Healthcare Delivery (VA-REACH TRIAL) (VA-REACH)
We propose a 3-arm randomized control trial to determine the efficacy of hydroxychloroquine or azithromycin in treating mild to moderate COVID-19 among veterans in the outpatient setting.
ID: NCT04363203
Sponsor: Salomeh Keyhani
Location: San Francisco VA Health Care System, California
A Study to Evaluate the Safety and Efficacy of MSTT1041A (Astegolimab) or UTTR1147A in Patients With Severe COVID-19 Pneumonia (COVASTIL)
This is a Phase II, randomized, double-blind, placebo-controlled, multicenter study to assess the efficacy and safety of MSTT1041A (astegolimab) or UTTR1147A in combination with standard of care (SOC) compared with matching placebo in combination with SOC in patients hospitalized with severe coronavirus disease 2019 (COVID-19) pneumonia.
ID: NCT04386616
Sponsor: Genentech
Contact: Study ID Number: GA42469 ([email protected])
Location: Southeast Louisiana Veterans Health Care System, New Orleans
Hormonal Intervention for the Treatment in Veterans With COVID-19 Requiring Hospitalization (HITCH)
The purpose of this study is to determine if temporary androgen suppression improves the clinical outcomes of veterans who are hospitalized to an acute care ward due to COVID-19.ID: NCT04397718
Sponsor: VA Office of Research and Development
Contact: Matthew B Rettig ([email protected]), Nicholas Nickols ([email protected])
Locations: VA Greater Los Angeles Healthcare System, California; VA NY Harbor Healthcare System, New York; VA Puget Sound Health Care System, Seattle, Washington
Adaptive COVID-19 Treatment Trial 2 (ACTT-II)
ACTT-II will evaluate the combination of baricitinib and remdesivir compared to remdesivir alone. Subjects will be assessed daily while hospitalized. If the subjects are discharged from the hospital, they will have a study visit at Days 15, 22, and 29.
ID: NCT04401579
Sponsor: National Institute of Allergy and Infectious Diseases (NIAID)
Contact: Central Contact ([email protected])
Locations: VA Palo Alto Health Care System, California; Naval Medical Center San Diego, California; Rocky Mountain Regional Veteran Affairs Medical Center, Aurora, Colorado; Southeast Louisiana Veterans Health Care System, New Orleans; Walter Reed National Military Medical Center, Bethesda, Maryland; National Institutes of Health - Clinical Center, National Institute of Allergy and Infectious Diseases Laboratory Of Immunoregulation, Bethesda, Maryland; Brooke Army Medical Center, Fort Sam Houston, Texas; Madigan Army Medical Center, Tacoma, Washington
Finding effective treatment or a vaccine for COVID-19, the disease caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has placed significant strains on the global health care system. The National Library of Medicine database lists > 1,800 trials that are aimed at addressing COVID-19-related health care. Already, trials developed by the US Department of Veterans Affairs (VA), US Department of Defense (DoD), and the National Institute of Allergy and Infectious Diseases have provided important data on effective treatment options. The clinical trials listed below are all open as of May 31, 2020 and have trial sites at VA and DoD facilities. For additional information and full inclusion/exclusion criteria, please consult clinicaltrials.gov.
Adaptive COVID-19 Treatment Trial (ACTT)
This study is an adaptive, randomized, double-blind, placebo-controlled trial to evaluate the safety and efficacy of novel therapeutic agents in hospitalized adults diagnosed with COVID-19. The study will compare different investigational therapeutic agents to a control arm. ID: NCT04280705
Sponsor: National Institute of Allergy and Infectious Diseases
Contact: Central Contact ([email protected])
Locations: VA Palo Alto Health Care System, California; Naval Medical Center San Diego, California; Southeast Louisiana Veterans Health Care System, New Orleans; Walter Reed National Military Medical Center, Bethesda, Maryland; National Institutes of Health - Clinical Center, National Institute of Allergy and Infectious Diseases Laboratory Of Immunoregulation, Bethesda, Maryland; Brooke Army Medical Center, Fort Sam Houston, Texas; Madigan Army Medical Center, Tacoma, Washington
Study to Evaluate the Safety and Antiviral Activity of Remdesivir (GS-5734) in Participants With Severe Coronavirus Disease (COVID-19)
The primary objective of this study is to evaluate the efficacy of 2 remdesivir (RDV) regimens with respect to clinical status assessed by a 7-point ordinal scale on Day 11 (NCT04292730) or Day 14 (NCT04292899).
ID: NCT04292730/NCT04292899
Sponsor: Gilead Sciences
Contact: Gilead Clinical Study Information Center (833-445-3230)
Location: James J. Peters VA Medical Center, Bronx, New York
Expanded Access Remdesivir (RDV; GS-5734)
The treatment of communicable Novel Coronavirus of 2019 with Remdesivir (RDV; GS-5734) also known as severe acute respiratory syndrome coronavirus 2.
ID: NCT04302766
Sponsor: US Army Medical Research and Development Command
Contact: Sandi Parriott ([email protected])
A Study to Evaluate the Safety and Efficacy of Tocilizumab in Patients With Severe COVID-19 Pneumonia (COVACTA)
This study will evaluate the efficacy, safety, pharmacodynamics, and pharmacokinetics of tocilizumab (TCZ) compared with a matching placebo in combination with standard of care (SOC) in hospitalized patients with severe COVID-19 pneumonia.
ID: NCT04320615
Sponsor: Hoffmann-La Roche
Location: James J Peters VA Medical Center, Bronx, New York
Administration of Intravenous Vitamin C in Novel Coronavirus Infection (COVID-19) and Decreased Oxygenation (AVoCaDO)
Previous research has shown that high dose intravenous vitamin C (HDIVC) may benefit patients with sepsis, acute lung injury (ALI), and the acute respiratory distress syndrome (ARDS). However, it is not known if early administration of HDIVC could prevent progression to ARDS. We hypothesize that HDIVC is safe and tolerable in COVID-19 subjects given early or late in the disease course and may reduce the risk of respiratory failure requiring mechanical ventilation and development of ARDS along with reductions in supplemental oxygen demand and inflammatory markers.
ID: NCT04357782
Sponsor: Hunter Holmes Mcguire VA Medical CenterContact: Brian Davis ([email protected])
Location: Hunter Holmes Mcguire VA Medical Center, Richmond, Virginia
Treatment Of CORONAVIRUS DISEASE 2019 (COVID-19) With Anti-Sars-CoV-2 Convalescent Plasma (ASCoV2CP)
This is an expanded access open-label, single-arm, multi-site protocol to provide convalescent plasma as a treatment for patients diagnosed with severe, or life-threatening COVID-19.
ID: NCT04360486
Sponsor: US Army Medical Research and Development Command
Contact: Andrew Cap ([email protected])
VA Remote and Equitable Access to COVID-19 Healthcare Delivery (VA-REACH TRIAL) (VA-REACH)
We propose a 3-arm randomized control trial to determine the efficacy of hydroxychloroquine or azithromycin in treating mild to moderate COVID-19 among veterans in the outpatient setting.
ID: NCT04363203
Sponsor: Salomeh Keyhani
Location: San Francisco VA Health Care System, California
A Study to Evaluate the Safety and Efficacy of MSTT1041A (Astegolimab) or UTTR1147A in Patients With Severe COVID-19 Pneumonia (COVASTIL)
This is a Phase II, randomized, double-blind, placebo-controlled, multicenter study to assess the efficacy and safety of MSTT1041A (astegolimab) or UTTR1147A in combination with standard of care (SOC) compared with matching placebo in combination with SOC in patients hospitalized with severe coronavirus disease 2019 (COVID-19) pneumonia.
ID: NCT04386616
Sponsor: Genentech
Contact: Study ID Number: GA42469 ([email protected])
Location: Southeast Louisiana Veterans Health Care System, New Orleans
Hormonal Intervention for the Treatment in Veterans With COVID-19 Requiring Hospitalization (HITCH)
The purpose of this study is to determine if temporary androgen suppression improves the clinical outcomes of veterans who are hospitalized to an acute care ward due to COVID-19.ID: NCT04397718
Sponsor: VA Office of Research and Development
Contact: Matthew B Rettig ([email protected]), Nicholas Nickols ([email protected])
Locations: VA Greater Los Angeles Healthcare System, California; VA NY Harbor Healthcare System, New York; VA Puget Sound Health Care System, Seattle, Washington
Adaptive COVID-19 Treatment Trial 2 (ACTT-II)
ACTT-II will evaluate the combination of baricitinib and remdesivir compared to remdesivir alone. Subjects will be assessed daily while hospitalized. If the subjects are discharged from the hospital, they will have a study visit at Days 15, 22, and 29.
ID: NCT04401579
Sponsor: National Institute of Allergy and Infectious Diseases (NIAID)
Contact: Central Contact ([email protected])
Locations: VA Palo Alto Health Care System, California; Naval Medical Center San Diego, California; Rocky Mountain Regional Veteran Affairs Medical Center, Aurora, Colorado; Southeast Louisiana Veterans Health Care System, New Orleans; Walter Reed National Military Medical Center, Bethesda, Maryland; National Institutes of Health - Clinical Center, National Institute of Allergy and Infectious Diseases Laboratory Of Immunoregulation, Bethesda, Maryland; Brooke Army Medical Center, Fort Sam Houston, Texas; Madigan Army Medical Center, Tacoma, Washington
Finding effective treatment or a vaccine for COVID-19, the disease caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has placed significant strains on the global health care system. The National Library of Medicine database lists > 1,800 trials that are aimed at addressing COVID-19-related health care. Already, trials developed by the US Department of Veterans Affairs (VA), US Department of Defense (DoD), and the National Institute of Allergy and Infectious Diseases have provided important data on effective treatment options. The clinical trials listed below are all open as of May 31, 2020 and have trial sites at VA and DoD facilities. For additional information and full inclusion/exclusion criteria, please consult clinicaltrials.gov.
Adaptive COVID-19 Treatment Trial (ACTT)
This study is an adaptive, randomized, double-blind, placebo-controlled trial to evaluate the safety and efficacy of novel therapeutic agents in hospitalized adults diagnosed with COVID-19. The study will compare different investigational therapeutic agents to a control arm. ID: NCT04280705
Sponsor: National Institute of Allergy and Infectious Diseases
Contact: Central Contact ([email protected])
Locations: VA Palo Alto Health Care System, California; Naval Medical Center San Diego, California; Southeast Louisiana Veterans Health Care System, New Orleans; Walter Reed National Military Medical Center, Bethesda, Maryland; National Institutes of Health - Clinical Center, National Institute of Allergy and Infectious Diseases Laboratory Of Immunoregulation, Bethesda, Maryland; Brooke Army Medical Center, Fort Sam Houston, Texas; Madigan Army Medical Center, Tacoma, Washington
Study to Evaluate the Safety and Antiviral Activity of Remdesivir (GS-5734) in Participants With Severe Coronavirus Disease (COVID-19)
The primary objective of this study is to evaluate the efficacy of 2 remdesivir (RDV) regimens with respect to clinical status assessed by a 7-point ordinal scale on Day 11 (NCT04292730) or Day 14 (NCT04292899).
ID: NCT04292730/NCT04292899
Sponsor: Gilead Sciences
Contact: Gilead Clinical Study Information Center (833-445-3230)
Location: James J. Peters VA Medical Center, Bronx, New York
Expanded Access Remdesivir (RDV; GS-5734)
The treatment of communicable Novel Coronavirus of 2019 with Remdesivir (RDV; GS-5734) also known as severe acute respiratory syndrome coronavirus 2.
ID: NCT04302766
Sponsor: US Army Medical Research and Development Command
Contact: Sandi Parriott ([email protected])
A Study to Evaluate the Safety and Efficacy of Tocilizumab in Patients With Severe COVID-19 Pneumonia (COVACTA)
This study will evaluate the efficacy, safety, pharmacodynamics, and pharmacokinetics of tocilizumab (TCZ) compared with a matching placebo in combination with standard of care (SOC) in hospitalized patients with severe COVID-19 pneumonia.
ID: NCT04320615
Sponsor: Hoffmann-La Roche
Location: James J Peters VA Medical Center, Bronx, New York
Administration of Intravenous Vitamin C in Novel Coronavirus Infection (COVID-19) and Decreased Oxygenation (AVoCaDO)
Previous research has shown that high dose intravenous vitamin C (HDIVC) may benefit patients with sepsis, acute lung injury (ALI), and the acute respiratory distress syndrome (ARDS). However, it is not known if early administration of HDIVC could prevent progression to ARDS. We hypothesize that HDIVC is safe and tolerable in COVID-19 subjects given early or late in the disease course and may reduce the risk of respiratory failure requiring mechanical ventilation and development of ARDS along with reductions in supplemental oxygen demand and inflammatory markers.
ID: NCT04357782
Sponsor: Hunter Holmes Mcguire VA Medical CenterContact: Brian Davis ([email protected])
Location: Hunter Holmes Mcguire VA Medical Center, Richmond, Virginia
Treatment Of CORONAVIRUS DISEASE 2019 (COVID-19) With Anti-Sars-CoV-2 Convalescent Plasma (ASCoV2CP)
This is an expanded access open-label, single-arm, multi-site protocol to provide convalescent plasma as a treatment for patients diagnosed with severe, or life-threatening COVID-19.
ID: NCT04360486
Sponsor: US Army Medical Research and Development Command
Contact: Andrew Cap ([email protected])
VA Remote and Equitable Access to COVID-19 Healthcare Delivery (VA-REACH TRIAL) (VA-REACH)
We propose a 3-arm randomized control trial to determine the efficacy of hydroxychloroquine or azithromycin in treating mild to moderate COVID-19 among veterans in the outpatient setting.
ID: NCT04363203
Sponsor: Salomeh Keyhani
Location: San Francisco VA Health Care System, California
A Study to Evaluate the Safety and Efficacy of MSTT1041A (Astegolimab) or UTTR1147A in Patients With Severe COVID-19 Pneumonia (COVASTIL)
This is a Phase II, randomized, double-blind, placebo-controlled, multicenter study to assess the efficacy and safety of MSTT1041A (astegolimab) or UTTR1147A in combination with standard of care (SOC) compared with matching placebo in combination with SOC in patients hospitalized with severe coronavirus disease 2019 (COVID-19) pneumonia.
ID: NCT04386616
Sponsor: Genentech
Contact: Study ID Number: GA42469 ([email protected])
Location: Southeast Louisiana Veterans Health Care System, New Orleans
Hormonal Intervention for the Treatment in Veterans With COVID-19 Requiring Hospitalization (HITCH)
The purpose of this study is to determine if temporary androgen suppression improves the clinical outcomes of veterans who are hospitalized to an acute care ward due to COVID-19.ID: NCT04397718
Sponsor: VA Office of Research and Development
Contact: Matthew B Rettig ([email protected]), Nicholas Nickols ([email protected])
Locations: VA Greater Los Angeles Healthcare System, California; VA NY Harbor Healthcare System, New York; VA Puget Sound Health Care System, Seattle, Washington
Adaptive COVID-19 Treatment Trial 2 (ACTT-II)
ACTT-II will evaluate the combination of baricitinib and remdesivir compared to remdesivir alone. Subjects will be assessed daily while hospitalized. If the subjects are discharged from the hospital, they will have a study visit at Days 15, 22, and 29.
ID: NCT04401579
Sponsor: National Institute of Allergy and Infectious Diseases (NIAID)
Contact: Central Contact ([email protected])
Locations: VA Palo Alto Health Care System, California; Naval Medical Center San Diego, California; Rocky Mountain Regional Veteran Affairs Medical Center, Aurora, Colorado; Southeast Louisiana Veterans Health Care System, New Orleans; Walter Reed National Military Medical Center, Bethesda, Maryland; National Institutes of Health - Clinical Center, National Institute of Allergy and Infectious Diseases Laboratory Of Immunoregulation, Bethesda, Maryland; Brooke Army Medical Center, Fort Sam Houston, Texas; Madigan Army Medical Center, Tacoma, Washington
Analysis of Pharmacist Interventions Used to Resolve Safety Target of Polypharmacy (STOP) Drug Interactions
Statins are one of the most common medications dispensed in the US and are associated with clinically significant drug interactions.1,2 The most common adverse drug reaction (ADR) of statin drug interactions is muscle-related toxicities.2 Despite technology advances to alert clinicians to drug interactions, updated statin manufacturer labeling, and guideline recommendations, inappropriate prescribing and dispensing of statin drug interactions continues to occur in health care systems.2-10
The medical literature has demonstrated many opportunities for pharmacists to prevent and mitigate drug interactions. At the points of prescribing and dispensing, pharmacists can reduce the number of potential drug interactions for the patient.11-13 Pharmacists also have identified and resolved drug interactions through quality assurance review after dispensing to a patient.7,8
Regardless of the time point of an intervention, the most common method pharmacists used to resolve drug interactions was through recommendations to a prescriber. The recommendations were generated through academic detailing, clinical decision support algorithms, drug conversions, or the pharmacist’s expertise. Regardless of the method the pharmacist used, the prescriber had the final authority to accept or decline the recommendation.7,8,11-13 Although these interventions were effective, pharmacists could further streamline the process by autonomously resolving drug interactions. However, these types of interventions are not well described in the medical literature.
Background
The US Department of Veterans Affairs (VA) Veterans Integrated Service Network (VISN), established the Safety Target of Polypharmacy (STOP) report in 2015. At each facility in the network, the report identified patients who were dispensed medications known to have drug interactions. The interactions were chosen by the VISN, and the severity of the interactions was based on coding parameters within the VA computerized order entry system, which uses a severity score based on First Databank data. At the Harry S. Truman Memorial Veterans’ Hospital (Truman VA) in Columbia, Missouri, > 500 drug interactions were initially active on the STOP report. The most common drug interactions were statins with gemfibrozil and statins with niacin.14-18 The Truman VA Pharmacy Service was charged with resolving the interactions for the facility.
The Truman VA employs 3 Patient Aligned Care Team (PACT) Clinical Pharmacy Specialists (CPS) practicing within primary care clinics. PACT is the patientcentered medical home model used by the VA. PACT CPS are ambulatory care pharmacists who assist providers in managing diseases using a scope of practice. Having a scope of practice would have allowed the PACT CPS to manage drug interactions with independent prescribing authority. However, due to the high volume of STOP report interactions and limited PACT CPS resources, the Pharmacy Service needed to develop an efficient, patient-centered method to resolve them. The intervention also needed to allow pharmacists, both with and without a scope of practice, to address the interactions.
Methods
The Truman VA Pharmacy Service developed protocols, approved by the Pharmacy and Therapeutics (P&T) Committee, to manage the specific gemfibrozil-statin and niacinstatin interactions chosen for the VISN 15 STOP report (Figures 1 and 2). The protocols were designed to identify patients who did not have a clear indication for gemfibrozil or niacin, were likely to maintain triglycerides (TGs) < 500 mg/dL without these medications, and would not likely require close monitoring after discontinuation.19 The protocols allowed pharmacists to autonomously discontinue gemfibrozil or niacin if patients did not have a history of pancreatitis, TGs ≥ 400 mg/dL or a nonlipid indication for niacin (eg, pellagra) after establishing care at Truman VA. Additionally, both interacting medications had to be dispensed by the VA. When pharmacists discontinued a medication, it was documented in a note in the patient electronic health record. The prescriber was notified through the note and the patient received a notification letter. Follow-up laboratory monitoring was not required as part of the protocol.
If patients met any of the exclusion criteria for discontinuation, the primary care provider (PCP) was notified to place a consult to the PACT Pharmacy Clinic for individualized interventions and close monitoring. Patients prescribed niacin for nonlipid indications were allowed to continue with their current drug regimen. At each encounter, the PACT CPS assessed for ADRs, made individualized medication changes, and arranged follow-up appointments. Once the interaction was resolved and treatment goals met, the PCP resumed monitoring of the patient’s lipid therapy.
Following all pharmacist interventions, a retrospective quality improvement analysis was conducted. The primary outcome was to evaluate the impact of discontinuing gemfibrozil and niacin by protocol on patients’ laboratory results. The coprimary endpoints were to describe the change in TG levels and the percentage of patients with TGs ≥ 500 mg/dL at least 5 weeks following the pharmacist-directed discontinuation by protocol. Secondary outcomes included the time required to resolve the interactions and a description of the PACT CPS pharmacologic interventions. Additionally, a quality assurance peer review was used to ensure the pharmacists appropriately utilized the protocols.
Data were collected from August 2016 to September 2017 for patients prescribed gemfibrozil and from May 2017 to January 2018 for patients prescribed niacin. The time spent resolving interactions was quantified based on encounter data. Descriptive statistics were used to analyze demographic information and the endpoints associated with each outcome. The project was reviewed by the University of Missouri Institutional Review Board, Truman VA privacy and information security officers, and was determined to meet guidelines for quality improvement.
Results
The original STOP report included 397 drug interactions involving statins with gemfibrozil or niacin (Table 1). The majority of patients were white and male aged 60 to 79 years. Gemfibrozil was the most common drug involved in all interactions (79.8%). The most common statins were atorvastatin (40%) and simvastatin (36.5%).
Gemfibrozil-Statin Interactions
Pharmacists discontinued gemfibrozil by protocol for 94 patients (29.6%), and 107 patients (33.8%) were referred to the PACT Pharmacy Clinic (Figure 3). For the remaining 116 patients (36.6%), the drug interaction was addressed outside of the protocol for the following reasons: the drug interaction was resolved prior to pharmacist review; an interacting prescription was expired and not to be continued; the patient self-discontinued ≥ 1 interacting medications; the patient was deceased; the patient moved; the patient was receiving ≥ 1 interacting medications outside of the VA; or the prescriber resolved the interaction following notification by the pharmacist.
Ultimately, the interaction was resolved for all patients with a gemfibrozil-statin interaction on the STOP report. Following gemfibrozil discontinuation by protocol, 76 patients (80.9%) had TG laboratory results available and were included in the analysis. Sixty-two patients’ (82%) TG levels decreased or increased by < 100 mg/dL (Figure 4), and the TG levels of 1 patient (1.3%) increased above the threshold of 500 mg/dL. The mean (SD) time to the first laboratory result after the pharmacists mailed the notification letter was 6.5 (3.6) months (range, 1-17). The pharmacists spent a mean of 16 minutes per patient resolving each interaction.
Of the 107 patients referred to the PACT Pharmacy Clinic, 80 (74.8%) had TG laboratory results available and were included in the analysis. These patients were followed by the PACT CPS until the drug interaction was resolved and confirmed to have TG levels at goal (< 500 mg/dL). Gemfibrozil doses ranged from 300 mg daily to 600 mg twice daily, with 70% (n = 56) of patients taking 600 mg twice daily. The PACT CPS made 148 interventions (Table 2). Twenty-three (29%) patients required only gemfibrozil discontinuation. The remaining 57 patients (71%) required at least 2 medication interventions. The PACT CPS generated 213 encounters for resolving drug interactions with a median of 2 encounters per patient.
Quality assurance review identified 5 patients (5.3%) who underwent gemfibrozil discontinuation by protocol, despite having criteria that would have recommended against discontinuation. In accordance with the protocol criteria, these patients were later referred to the PACT Pharmacy Clinic. None of these patients experienced a TG increase at or above the threshold of 500 mg/dL after gemfibrozil was initially discontinued but were excluded from the earlier analysis.
Niacin-Statin Interactions
Pharmacists discontinued niacin by protocol for 48 patients (60.0%), and 22 patients (27.5%) were referred to the PACT Pharmacy Clinic (Figure 5). For the remaining 5 patients (6.3%), the interaction was either addressed outside the protocol prior to pharmacist review, or an interacting prescription was expired and not to be continued. Additionally, niacin was continued per prescriber preference in 5 patients (6.3%).
Thirty-six patients (75%) had TG laboratory results available following niacin discontinuation by protocol and were included in the analysis. Most patients’ (n = 33, 91.7%) TG levels decreased or increased by < 100 mg/dL. No patient had a TG level that increased higher than the threshold of 500 mg/dL. The mean (SD) time to the first laboratory result after the pharmacists mailed the notification letter, was 5.3 (2.5) months (range, 1.2-9.8). The pharmacists spent a mean of 15 minutes per patient resolving each interaction. The quality assurance review found no discrepancies in the pharmacists’ application of the protocol.
Of the 22 patients referred to the PACT Pharmacy Clinic, 16 (72.7%) patients had TG laboratory results available and were included in the analysis. As with the gemfibrozil interactions, these patients were followed by the PACT Pharmacy Clinic until the drug interaction was resolved and confirmed to have TGs at goal (< 500 mg/dL). Niacin doses ranged from 500 mg daily to 2,000 mg daily, with the majority of patients taking 1,000 mg daily. The PACT CPS made 23 interventions. The PACT CPS generated 46 encounters for resolving drug interactions with a median of 2 encounters per patient.
Discussion
Following gemfibrozil or niacin discontinuation by protocol, most patients with available laboratory results experienced either a decrease or modest TG elevation. The proportion of patients experiencing a decrease in TGs was unexpected but potentially multifactorial. Individual causes for the decrease in TGs were beyond the scope of this analysis. The retrospective design limited the ability to identify variables that could have impacted TG levels when gemfibrozil or niacin were started and discontinued. Although the treatment of TG levels is not indicated until it is ≥ 500 mg/dL, due to an increased risk of pancreatitis, both protocols excluded patients with a history of TGs ≥ 400 mg/dL.19 The lower threshold was set to compensate for anticipated increase in TG levels, following gemfibrozil or niacin discontinuation, and to minimize the number of patients with TG levels ≥ 500 mg/dL. The actual impact on patients’ TG levels supports the use of this lower threshold in the protocol.
When TG levels increased by 200 to 249 mg/dL after gemfibrozil or niacin discontinuation, patients were evaluated for possible underlying causes, which occurred for 4 gemfibrozil and 1 niacin patient. One patient started a β-blocker after gemfibrozil was initiated, and 3 patients were taking gemfibrozil prior to establishing care at the VA. The TG levels of the patient taking niacin correlated with an increased hemoglobin A1c. The TG level for only 1 patient taking gemfibrozil increased above the 500 mg/dL threshold. The patient had several comorbidities known to increase TG levels, but the comorbidities were previously well controlled. No additional medication changes were made at that time, and the TG levels on the next fasting lipid panel decreased to goal. The patient did not experience any negative clinical sequelae from the elevated TG levels.
Thirty-five patients (36%) who were referred to the PACT Pharmacy Clinic required only either gemfibrozil or niacin discontinuation. These patients were evaluated to identify whether adjustments to the protocols would have allowed for pharmacist discontinuation without referral to the PACT Pharmacy Clinic. Twenty-four of these patients (69%) had repeated TG levels ≥ 400 mg/dL prior to referral to the PACT Pharmacy Clinic. Additionally, there was no correlation between the gemfibrozil or niacin doses and the change in TG levels following discontinuation. These data indicate the protocols appropriately identified patients who did not have an indication for gemfibrozil or niacin.
In addition to drug interactions identified on the STOP report, the PACT CPS resolved 12 additional interactions involving simvastatin and gemfibrozil. Additionally, unnecessary lipid medications were deprescribed. The PACT CPS identified 13 patients who experienced myalgias, an ADR attributed to the gemfibrozil- statin interaction. Of those, 9 patients’ ADRs resolved after discontinuing gemfibrozil alone. For the remaining 4 patients, additional interventions to convert the patient to another statin were required to resolve the ADR.
Using pharmacists to address the drug interactions shifted workload from the prescribers and other primary care team members. The mean time spent to resolve both gemfibrozil and niacin interactions by protocol was 15.5 minutes. One hundred fortytwo patients (35.8%) had drug interactions resolved by protocol, saving the PACT CPS’ expertise for patients requiring individualized interventions. Drug interactions were resolved within 4 PACT CPS encounters for 93.8% of the patients taking gemfibrozil and within 3 PACT CPS encounters for 93.8% of the patients taking niacin.
The protocols allowed 12 additional pharmacists who did not have an ambulatory care scope of practice to assist the PACT CPS in mitigating the STOP drug interactions. These pharmacists otherwise would have been limited to making consultative recommendations. Simultaneously, the design allowed for the PACT pharmacists’ expertise to be allocated for patients most likely to require interventions beyond the protocols. This type of intraprofessional referral process is not well described in the medical literature. To the authors’ knowledge, the only studies described referrals from hospital pharmacists to community pharmacists during transitions of care on hospital discharge.20,21
Limitations
The results of this study are derived from a retrospective chart review at a single VA facility. The autonomous nature of PACT CPS interventions may be difficult to replicate in other settings that do not permit pharmacists the same prescriptive authority. This analysis was designed to demonstrate the impact of the pharmacist in resolving major drug interactions. Patients referred to the PACT Pharmacy Clinic who also had their lipid medications adjusted by a nonpharmacist provider were excluded. However, this may have minimized the impact of the PACT CPS on the patient care provided. As postintervention laboratory results were not available for all patients, some patients’ TG levels could have increased above the 500 mg/dL threshold but were not identified. The time investment was extensive and likely underestimates the true cost of implementing the interventions.
Because notification letters were used to instruct patients to stop gemfibrozil or niacin, several considerations need to be addressed when interpreting the follow-up laboratory results. First, we cannot confirm whether the patients received the letter or the exact date the letter was received. Additionally, we cannot confirm whether the patients followed the instructions to stop the interacting medications or the date the medications were stopped. It is possible some patients were still taking the interacting medication when the first laboratory was drawn. Should a patient have continued the interacting medication, most would have run out and been unable to obtain a refill within 90 days of receiving the letter, as this is the maximum amount dispensed at one time. The mean time to the first laboratory result for both gemfibrozil and niacin was 6.5 and 5.3 months, respectively. Approximately 85% of patients completed the first laboratory test at least 3 months after the letter was mailed.
The protocols were designed to assess whether gemfibrozil or niacin was indicated and did not assess whether the statin was indicated. Therefore, discontinuing the statin also could have resolved the interaction appropriately. However, due to characteristics of the patient population and recommendations in current lipid guidelines, it was more likely the statin would be indicated.22,23 The protocols also assumed that patients eligible for gemfibrozil or niacin discontinuation would not need additional changes to their lipid medications. The medication changes made by the PACT CPS may have gone beyond those minimally necessary to resolve the drug interaction and maintain TG goals. Patients who had gemfibrozil or niacin discontinued by protocol also may have benefited from additional optimization of their lipid medications.
Conclusions
This quality improvement analysis supports further evaluation of the complementary use of protocols and PACT CPS prescriptive authority to resolve statin drug interactions. The gemfibrozil and niacin protocols appropriately identified patients who were less likely to experience an adverse change in TG laboratory results. Patients more likely to require additional medication interventions were appropriately referred to the PACT Pharmacy Clinics for individualized care. These data support expanded roles for pharmacists, across various settings, to mitigate select drug interactions at the Truman VA.
Acknowledgments
This quality improvement project is the result of work supported with resources and use of the Harry S. Truman Memorial Veterans’ Hospital in Columbia, Missouri.
1. The top 200 drugs of 2020 Provided by the ClinCalc DrugStats Database. http://clincalc.com/DrugStats /Top200Drugs.aspx. Updated February 11, 2017. Accessed May 12, 2020.
2. Wiggins BS, Saseen JJ, Page RL 2nd, et al; American Heart Association Clinical Pharmacology Committee of the Council on Clinical Cardiology; Council on Hypertension; Council on Quality of Care and Outcomes Research; and Council on Functional Genomics and Translational Biology. Recommendations for management of clinically significant drug-drug interactions with statins and select agents used in patients with cardiovascular disease: a scientific statement from the American Heart Association. Circulation. 2016;134(21):e468‐e495. doi:10.1161/CIR.0000000000000456
3. Smithburger PL, Buckley MS, Bejian S, Burenheide K, Kane-Gill SL. A critical evaluation of clinical decision support for the detection of drug-drug interactions. Expert Opin Drug Saf. 2011;10(6):871‐882. doi:10.1517/14740338.2011.583916
4. US Food and Drug Administration. FDA drug safety communication: new restrictions, contraindications, and dose limitations for Zocor (simvastatin) to reduce the risk of muscle injury. https://www.fda.gov/Drugs/DrugSafety /ucm256581.htm. Updated December 15, 2017. Accessed May 12, 2020.
5. US Food and Drug Administration. FDA drug safety communication: important safety label changes to cholesterol-lowering statin drugs. https://www.fda.gov /Drugs/DrugSafety/ucm293101.htm. Updated January 19, 2016. Accessed May 12, 2020.
6. US Food and Drug Administration Federal Register. AbbVie Inc. et al; withdrawal of approval of indications related to the coadministration with statins in applications for niacin extended-release tablets and fenofibric acid delayed-release capsules. https://www.federalregister .gov/documents/2016/04/18/2016-08887/abbvie-inc -et-al-withdrawal-of-approval-of-indications-related -to-the-coadministration-with-statins. Published April 18, 2016. Accessed May 12, 2020.
7. Lamprecht DG Jr, Todd BA, Denham AM, Ruppe LK, Stadler SL. Clinical pharmacist patient-safety initiative to reduce against-label prescribing of statins with cyclosporine. Ann Pharmacother. 2017;51(2):140‐145. doi:10.1177/1060028016675352
8. Roblek T, Deticek A, Leskovar B, et al. Clinical-pharmacist intervention reduces clinically relevant drugdrug interactions in patients with heart failure: A randomized, double-blind, controlled trial. Int J Cardiol. 2016;203:647‐652. doi:10.1016/j.ijcard.2015.10.206
9. Tuchscherer RM, Nair K, Ghushchyan V, Saseen JJ. Simvastatin prescribing patterns before and after FDA dosing restrictions: a retrospective analysis of a large healthcare claims database. Am J Cardiovasc Drugs. 2015;15(1):27‐34. doi:10.1007/s40256-014-0096-x
10. Alford JC, Saseen JJ, Allen RR, Nair KV. Persistent use of against-label statin-fibrate combinations from 2003-2009 despite United States Food and Drug Administration dose restrictions. Pharmacotherapy. 2012;32(7):623‐630. doi:10.1002/j.1875-9114.2011.01090.x
11. Leape LL, Cullen DJ, Clapp MD, et al. Pharmacist participation on physician rounds and adverse drug events in the intensive care unit [published correction appears in JAMA 2000 Mar 8;283(10):1293]. JAMA. 1999;282(3):267‐270. doi:10.1001/jama.282.3.267
12. Kucukarslan SN, Peters M, Mlynarek M, Nafziger DA. Pharmacists on rounding teams reduce preventable adverse drug events in hospital general medicine units. Arch Intern Med. 2003;163(17):2014‐2018. doi:10.1001/archinte.163.17.2014
13. Humphries TL, Carroll N, Chester EA, Magid D, Rocho B. Evaluation of an electronic critical drug interaction program coupled with active pharmacist intervention. Ann Pharmacother. 2007;41(12):1979‐1985. doi:10.1345/aph.1K349
14. Zocor [package insert]. Whitehouse Station, NJ: Merck & Co, Inc; 2018.
15. Lipitor [package insert]. New York, NY: Pfizer; 2017.
16. Crestor [package insert]. Wilmington, DE: AstraZeneca; 2018.
17. Mevacor [package insert]. Whitehouse Station, NJ: Merck & Co, Inc; 2012.
18. Wolters Kluwer Health, Lexi-Drugs, Lexicomp. Pravastatin. www.online.lexi.com. [Source not verified.]
19. Miller M, Stone NJ, Ballantyne C, et al; American Heart Association Clinical Lipidology, Thrombosis, and Prevention Committee of the Council on Nutrition, Physical Activity, and Metabolism; Council on Arteriosclerosis, Thrombosis and Vascular Biology; Council on Cardiovascular Nursing; Council on the Kidney in Cardiovascular Disease. Triglycerides and cardiovascular disease: a scientific statement from the American Heart Association. Circulation. 2011;123(20):2292-2333. doi: 10.1161/CIR.0b013e3182160726
20. Ferguson J, Seston L, Ashcroft DM. Refer-to-pharmacy: a qualitative study exploring the implementation of an electronic transfer of care initiative to improve medicines optimisation following hospital discharge. BMC Health Serv Res. 2018;18(1):424. doi:10.1186/s12913-018-3262-z
21. Ensing HT, Koster ES, Dubero DJ, van Dooren AA, Bouvy ML. Collaboration between hospital and community pharmacists to address drug-related problems: the HomeCoMe-program. Res Social Adm Pharm. 2019;15(3):267‐278. doi:10.1016/j.sapharm.2018.05.001
22. US Department of Defense, US Department of Veterans Affairs. VA/DoD clinical practice guideline for the management of dyslipidemia for cardiovascular risk reduction guideline summary. https://www.healthquality.va.gov /guidelines/CD/lipids/LipidSumOptSinglePg31Aug15.pdf. Published 2014. Accessed May 14, 2020.
23. Stone NJ, Robinson JG, Lichtenstein AH, et al. 2013 ACC/AHA guideline on the treatment of blood cholesterol to reduce atherosclerotic cardiovascular risk in adults: a report of the American College of Cardiology/ American Heart Association Task Force on Practice Guidelines [published correction appears in Circulation. 2014 Jun 24;129(25) (suppl 2):S46-48] [published correction appears in Circulation. 2015 Dec 22;132(25):e396]. Circulation. 2014;129(25)(suppl 2): S1‐S45. doi:10.1161/01.cir.0000437738.63853.7a
Statins are one of the most common medications dispensed in the US and are associated with clinically significant drug interactions.1,2 The most common adverse drug reaction (ADR) of statin drug interactions is muscle-related toxicities.2 Despite technology advances to alert clinicians to drug interactions, updated statin manufacturer labeling, and guideline recommendations, inappropriate prescribing and dispensing of statin drug interactions continues to occur in health care systems.2-10
The medical literature has demonstrated many opportunities for pharmacists to prevent and mitigate drug interactions. At the points of prescribing and dispensing, pharmacists can reduce the number of potential drug interactions for the patient.11-13 Pharmacists also have identified and resolved drug interactions through quality assurance review after dispensing to a patient.7,8
Regardless of the time point of an intervention, the most common method pharmacists used to resolve drug interactions was through recommendations to a prescriber. The recommendations were generated through academic detailing, clinical decision support algorithms, drug conversions, or the pharmacist’s expertise. Regardless of the method the pharmacist used, the prescriber had the final authority to accept or decline the recommendation.7,8,11-13 Although these interventions were effective, pharmacists could further streamline the process by autonomously resolving drug interactions. However, these types of interventions are not well described in the medical literature.
Background
The US Department of Veterans Affairs (VA) Veterans Integrated Service Network (VISN), established the Safety Target of Polypharmacy (STOP) report in 2015. At each facility in the network, the report identified patients who were dispensed medications known to have drug interactions. The interactions were chosen by the VISN, and the severity of the interactions was based on coding parameters within the VA computerized order entry system, which uses a severity score based on First Databank data. At the Harry S. Truman Memorial Veterans’ Hospital (Truman VA) in Columbia, Missouri, > 500 drug interactions were initially active on the STOP report. The most common drug interactions were statins with gemfibrozil and statins with niacin.14-18 The Truman VA Pharmacy Service was charged with resolving the interactions for the facility.
The Truman VA employs 3 Patient Aligned Care Team (PACT) Clinical Pharmacy Specialists (CPS) practicing within primary care clinics. PACT is the patientcentered medical home model used by the VA. PACT CPS are ambulatory care pharmacists who assist providers in managing diseases using a scope of practice. Having a scope of practice would have allowed the PACT CPS to manage drug interactions with independent prescribing authority. However, due to the high volume of STOP report interactions and limited PACT CPS resources, the Pharmacy Service needed to develop an efficient, patient-centered method to resolve them. The intervention also needed to allow pharmacists, both with and without a scope of practice, to address the interactions.
Methods
The Truman VA Pharmacy Service developed protocols, approved by the Pharmacy and Therapeutics (P&T) Committee, to manage the specific gemfibrozil-statin and niacinstatin interactions chosen for the VISN 15 STOP report (Figures 1 and 2). The protocols were designed to identify patients who did not have a clear indication for gemfibrozil or niacin, were likely to maintain triglycerides (TGs) < 500 mg/dL without these medications, and would not likely require close monitoring after discontinuation.19 The protocols allowed pharmacists to autonomously discontinue gemfibrozil or niacin if patients did not have a history of pancreatitis, TGs ≥ 400 mg/dL or a nonlipid indication for niacin (eg, pellagra) after establishing care at Truman VA. Additionally, both interacting medications had to be dispensed by the VA. When pharmacists discontinued a medication, it was documented in a note in the patient electronic health record. The prescriber was notified through the note and the patient received a notification letter. Follow-up laboratory monitoring was not required as part of the protocol.
If patients met any of the exclusion criteria for discontinuation, the primary care provider (PCP) was notified to place a consult to the PACT Pharmacy Clinic for individualized interventions and close monitoring. Patients prescribed niacin for nonlipid indications were allowed to continue with their current drug regimen. At each encounter, the PACT CPS assessed for ADRs, made individualized medication changes, and arranged follow-up appointments. Once the interaction was resolved and treatment goals met, the PCP resumed monitoring of the patient’s lipid therapy.
Following all pharmacist interventions, a retrospective quality improvement analysis was conducted. The primary outcome was to evaluate the impact of discontinuing gemfibrozil and niacin by protocol on patients’ laboratory results. The coprimary endpoints were to describe the change in TG levels and the percentage of patients with TGs ≥ 500 mg/dL at least 5 weeks following the pharmacist-directed discontinuation by protocol. Secondary outcomes included the time required to resolve the interactions and a description of the PACT CPS pharmacologic interventions. Additionally, a quality assurance peer review was used to ensure the pharmacists appropriately utilized the protocols.
Data were collected from August 2016 to September 2017 for patients prescribed gemfibrozil and from May 2017 to January 2018 for patients prescribed niacin. The time spent resolving interactions was quantified based on encounter data. Descriptive statistics were used to analyze demographic information and the endpoints associated with each outcome. The project was reviewed by the University of Missouri Institutional Review Board, Truman VA privacy and information security officers, and was determined to meet guidelines for quality improvement.
Results
The original STOP report included 397 drug interactions involving statins with gemfibrozil or niacin (Table 1). The majority of patients were white and male aged 60 to 79 years. Gemfibrozil was the most common drug involved in all interactions (79.8%). The most common statins were atorvastatin (40%) and simvastatin (36.5%).
Gemfibrozil-Statin Interactions
Pharmacists discontinued gemfibrozil by protocol for 94 patients (29.6%), and 107 patients (33.8%) were referred to the PACT Pharmacy Clinic (Figure 3). For the remaining 116 patients (36.6%), the drug interaction was addressed outside of the protocol for the following reasons: the drug interaction was resolved prior to pharmacist review; an interacting prescription was expired and not to be continued; the patient self-discontinued ≥ 1 interacting medications; the patient was deceased; the patient moved; the patient was receiving ≥ 1 interacting medications outside of the VA; or the prescriber resolved the interaction following notification by the pharmacist.
Ultimately, the interaction was resolved for all patients with a gemfibrozil-statin interaction on the STOP report. Following gemfibrozil discontinuation by protocol, 76 patients (80.9%) had TG laboratory results available and were included in the analysis. Sixty-two patients’ (82%) TG levels decreased or increased by < 100 mg/dL (Figure 4), and the TG levels of 1 patient (1.3%) increased above the threshold of 500 mg/dL. The mean (SD) time to the first laboratory result after the pharmacists mailed the notification letter was 6.5 (3.6) months (range, 1-17). The pharmacists spent a mean of 16 minutes per patient resolving each interaction.
Of the 107 patients referred to the PACT Pharmacy Clinic, 80 (74.8%) had TG laboratory results available and were included in the analysis. These patients were followed by the PACT CPS until the drug interaction was resolved and confirmed to have TG levels at goal (< 500 mg/dL). Gemfibrozil doses ranged from 300 mg daily to 600 mg twice daily, with 70% (n = 56) of patients taking 600 mg twice daily. The PACT CPS made 148 interventions (Table 2). Twenty-three (29%) patients required only gemfibrozil discontinuation. The remaining 57 patients (71%) required at least 2 medication interventions. The PACT CPS generated 213 encounters for resolving drug interactions with a median of 2 encounters per patient.
Quality assurance review identified 5 patients (5.3%) who underwent gemfibrozil discontinuation by protocol, despite having criteria that would have recommended against discontinuation. In accordance with the protocol criteria, these patients were later referred to the PACT Pharmacy Clinic. None of these patients experienced a TG increase at or above the threshold of 500 mg/dL after gemfibrozil was initially discontinued but were excluded from the earlier analysis.
Niacin-Statin Interactions
Pharmacists discontinued niacin by protocol for 48 patients (60.0%), and 22 patients (27.5%) were referred to the PACT Pharmacy Clinic (Figure 5). For the remaining 5 patients (6.3%), the interaction was either addressed outside the protocol prior to pharmacist review, or an interacting prescription was expired and not to be continued. Additionally, niacin was continued per prescriber preference in 5 patients (6.3%).
Thirty-six patients (75%) had TG laboratory results available following niacin discontinuation by protocol and were included in the analysis. Most patients’ (n = 33, 91.7%) TG levels decreased or increased by < 100 mg/dL. No patient had a TG level that increased higher than the threshold of 500 mg/dL. The mean (SD) time to the first laboratory result after the pharmacists mailed the notification letter, was 5.3 (2.5) months (range, 1.2-9.8). The pharmacists spent a mean of 15 minutes per patient resolving each interaction. The quality assurance review found no discrepancies in the pharmacists’ application of the protocol.
Of the 22 patients referred to the PACT Pharmacy Clinic, 16 (72.7%) patients had TG laboratory results available and were included in the analysis. As with the gemfibrozil interactions, these patients were followed by the PACT Pharmacy Clinic until the drug interaction was resolved and confirmed to have TGs at goal (< 500 mg/dL). Niacin doses ranged from 500 mg daily to 2,000 mg daily, with the majority of patients taking 1,000 mg daily. The PACT CPS made 23 interventions. The PACT CPS generated 46 encounters for resolving drug interactions with a median of 2 encounters per patient.
Discussion
Following gemfibrozil or niacin discontinuation by protocol, most patients with available laboratory results experienced either a decrease or modest TG elevation. The proportion of patients experiencing a decrease in TGs was unexpected but potentially multifactorial. Individual causes for the decrease in TGs were beyond the scope of this analysis. The retrospective design limited the ability to identify variables that could have impacted TG levels when gemfibrozil or niacin were started and discontinued. Although the treatment of TG levels is not indicated until it is ≥ 500 mg/dL, due to an increased risk of pancreatitis, both protocols excluded patients with a history of TGs ≥ 400 mg/dL.19 The lower threshold was set to compensate for anticipated increase in TG levels, following gemfibrozil or niacin discontinuation, and to minimize the number of patients with TG levels ≥ 500 mg/dL. The actual impact on patients’ TG levels supports the use of this lower threshold in the protocol.
When TG levels increased by 200 to 249 mg/dL after gemfibrozil or niacin discontinuation, patients were evaluated for possible underlying causes, which occurred for 4 gemfibrozil and 1 niacin patient. One patient started a β-blocker after gemfibrozil was initiated, and 3 patients were taking gemfibrozil prior to establishing care at the VA. The TG levels of the patient taking niacin correlated with an increased hemoglobin A1c. The TG level for only 1 patient taking gemfibrozil increased above the 500 mg/dL threshold. The patient had several comorbidities known to increase TG levels, but the comorbidities were previously well controlled. No additional medication changes were made at that time, and the TG levels on the next fasting lipid panel decreased to goal. The patient did not experience any negative clinical sequelae from the elevated TG levels.
Thirty-five patients (36%) who were referred to the PACT Pharmacy Clinic required only either gemfibrozil or niacin discontinuation. These patients were evaluated to identify whether adjustments to the protocols would have allowed for pharmacist discontinuation without referral to the PACT Pharmacy Clinic. Twenty-four of these patients (69%) had repeated TG levels ≥ 400 mg/dL prior to referral to the PACT Pharmacy Clinic. Additionally, there was no correlation between the gemfibrozil or niacin doses and the change in TG levels following discontinuation. These data indicate the protocols appropriately identified patients who did not have an indication for gemfibrozil or niacin.
In addition to drug interactions identified on the STOP report, the PACT CPS resolved 12 additional interactions involving simvastatin and gemfibrozil. Additionally, unnecessary lipid medications were deprescribed. The PACT CPS identified 13 patients who experienced myalgias, an ADR attributed to the gemfibrozil- statin interaction. Of those, 9 patients’ ADRs resolved after discontinuing gemfibrozil alone. For the remaining 4 patients, additional interventions to convert the patient to another statin were required to resolve the ADR.
Using pharmacists to address the drug interactions shifted workload from the prescribers and other primary care team members. The mean time spent to resolve both gemfibrozil and niacin interactions by protocol was 15.5 minutes. One hundred fortytwo patients (35.8%) had drug interactions resolved by protocol, saving the PACT CPS’ expertise for patients requiring individualized interventions. Drug interactions were resolved within 4 PACT CPS encounters for 93.8% of the patients taking gemfibrozil and within 3 PACT CPS encounters for 93.8% of the patients taking niacin.
The protocols allowed 12 additional pharmacists who did not have an ambulatory care scope of practice to assist the PACT CPS in mitigating the STOP drug interactions. These pharmacists otherwise would have been limited to making consultative recommendations. Simultaneously, the design allowed for the PACT pharmacists’ expertise to be allocated for patients most likely to require interventions beyond the protocols. This type of intraprofessional referral process is not well described in the medical literature. To the authors’ knowledge, the only studies described referrals from hospital pharmacists to community pharmacists during transitions of care on hospital discharge.20,21
Limitations
The results of this study are derived from a retrospective chart review at a single VA facility. The autonomous nature of PACT CPS interventions may be difficult to replicate in other settings that do not permit pharmacists the same prescriptive authority. This analysis was designed to demonstrate the impact of the pharmacist in resolving major drug interactions. Patients referred to the PACT Pharmacy Clinic who also had their lipid medications adjusted by a nonpharmacist provider were excluded. However, this may have minimized the impact of the PACT CPS on the patient care provided. As postintervention laboratory results were not available for all patients, some patients’ TG levels could have increased above the 500 mg/dL threshold but were not identified. The time investment was extensive and likely underestimates the true cost of implementing the interventions.
Because notification letters were used to instruct patients to stop gemfibrozil or niacin, several considerations need to be addressed when interpreting the follow-up laboratory results. First, we cannot confirm whether the patients received the letter or the exact date the letter was received. Additionally, we cannot confirm whether the patients followed the instructions to stop the interacting medications or the date the medications were stopped. It is possible some patients were still taking the interacting medication when the first laboratory was drawn. Should a patient have continued the interacting medication, most would have run out and been unable to obtain a refill within 90 days of receiving the letter, as this is the maximum amount dispensed at one time. The mean time to the first laboratory result for both gemfibrozil and niacin was 6.5 and 5.3 months, respectively. Approximately 85% of patients completed the first laboratory test at least 3 months after the letter was mailed.
The protocols were designed to assess whether gemfibrozil or niacin was indicated and did not assess whether the statin was indicated. Therefore, discontinuing the statin also could have resolved the interaction appropriately. However, due to characteristics of the patient population and recommendations in current lipid guidelines, it was more likely the statin would be indicated.22,23 The protocols also assumed that patients eligible for gemfibrozil or niacin discontinuation would not need additional changes to their lipid medications. The medication changes made by the PACT CPS may have gone beyond those minimally necessary to resolve the drug interaction and maintain TG goals. Patients who had gemfibrozil or niacin discontinued by protocol also may have benefited from additional optimization of their lipid medications.
Conclusions
This quality improvement analysis supports further evaluation of the complementary use of protocols and PACT CPS prescriptive authority to resolve statin drug interactions. The gemfibrozil and niacin protocols appropriately identified patients who were less likely to experience an adverse change in TG laboratory results. Patients more likely to require additional medication interventions were appropriately referred to the PACT Pharmacy Clinics for individualized care. These data support expanded roles for pharmacists, across various settings, to mitigate select drug interactions at the Truman VA.
Acknowledgments
This quality improvement project is the result of work supported with resources and use of the Harry S. Truman Memorial Veterans’ Hospital in Columbia, Missouri.
Statins are one of the most common medications dispensed in the US and are associated with clinically significant drug interactions.1,2 The most common adverse drug reaction (ADR) of statin drug interactions is muscle-related toxicities.2 Despite technology advances to alert clinicians to drug interactions, updated statin manufacturer labeling, and guideline recommendations, inappropriate prescribing and dispensing of statin drug interactions continues to occur in health care systems.2-10
The medical literature has demonstrated many opportunities for pharmacists to prevent and mitigate drug interactions. At the points of prescribing and dispensing, pharmacists can reduce the number of potential drug interactions for the patient.11-13 Pharmacists also have identified and resolved drug interactions through quality assurance review after dispensing to a patient.7,8
Regardless of the time point of an intervention, the most common method pharmacists used to resolve drug interactions was through recommendations to a prescriber. The recommendations were generated through academic detailing, clinical decision support algorithms, drug conversions, or the pharmacist’s expertise. Regardless of the method the pharmacist used, the prescriber had the final authority to accept or decline the recommendation.7,8,11-13 Although these interventions were effective, pharmacists could further streamline the process by autonomously resolving drug interactions. However, these types of interventions are not well described in the medical literature.
Background
The US Department of Veterans Affairs (VA) Veterans Integrated Service Network (VISN), established the Safety Target of Polypharmacy (STOP) report in 2015. At each facility in the network, the report identified patients who were dispensed medications known to have drug interactions. The interactions were chosen by the VISN, and the severity of the interactions was based on coding parameters within the VA computerized order entry system, which uses a severity score based on First Databank data. At the Harry S. Truman Memorial Veterans’ Hospital (Truman VA) in Columbia, Missouri, > 500 drug interactions were initially active on the STOP report. The most common drug interactions were statins with gemfibrozil and statins with niacin.14-18 The Truman VA Pharmacy Service was charged with resolving the interactions for the facility.
The Truman VA employs 3 Patient Aligned Care Team (PACT) Clinical Pharmacy Specialists (CPS) practicing within primary care clinics. PACT is the patientcentered medical home model used by the VA. PACT CPS are ambulatory care pharmacists who assist providers in managing diseases using a scope of practice. Having a scope of practice would have allowed the PACT CPS to manage drug interactions with independent prescribing authority. However, due to the high volume of STOP report interactions and limited PACT CPS resources, the Pharmacy Service needed to develop an efficient, patient-centered method to resolve them. The intervention also needed to allow pharmacists, both with and without a scope of practice, to address the interactions.
Methods
The Truman VA Pharmacy Service developed protocols, approved by the Pharmacy and Therapeutics (P&T) Committee, to manage the specific gemfibrozil-statin and niacinstatin interactions chosen for the VISN 15 STOP report (Figures 1 and 2). The protocols were designed to identify patients who did not have a clear indication for gemfibrozil or niacin, were likely to maintain triglycerides (TGs) < 500 mg/dL without these medications, and would not likely require close monitoring after discontinuation.19 The protocols allowed pharmacists to autonomously discontinue gemfibrozil or niacin if patients did not have a history of pancreatitis, TGs ≥ 400 mg/dL or a nonlipid indication for niacin (eg, pellagra) after establishing care at Truman VA. Additionally, both interacting medications had to be dispensed by the VA. When pharmacists discontinued a medication, it was documented in a note in the patient electronic health record. The prescriber was notified through the note and the patient received a notification letter. Follow-up laboratory monitoring was not required as part of the protocol.
If patients met any of the exclusion criteria for discontinuation, the primary care provider (PCP) was notified to place a consult to the PACT Pharmacy Clinic for individualized interventions and close monitoring. Patients prescribed niacin for nonlipid indications were allowed to continue with their current drug regimen. At each encounter, the PACT CPS assessed for ADRs, made individualized medication changes, and arranged follow-up appointments. Once the interaction was resolved and treatment goals met, the PCP resumed monitoring of the patient’s lipid therapy.
Following all pharmacist interventions, a retrospective quality improvement analysis was conducted. The primary outcome was to evaluate the impact of discontinuing gemfibrozil and niacin by protocol on patients’ laboratory results. The coprimary endpoints were to describe the change in TG levels and the percentage of patients with TGs ≥ 500 mg/dL at least 5 weeks following the pharmacist-directed discontinuation by protocol. Secondary outcomes included the time required to resolve the interactions and a description of the PACT CPS pharmacologic interventions. Additionally, a quality assurance peer review was used to ensure the pharmacists appropriately utilized the protocols.
Data were collected from August 2016 to September 2017 for patients prescribed gemfibrozil and from May 2017 to January 2018 for patients prescribed niacin. The time spent resolving interactions was quantified based on encounter data. Descriptive statistics were used to analyze demographic information and the endpoints associated with each outcome. The project was reviewed by the University of Missouri Institutional Review Board, Truman VA privacy and information security officers, and was determined to meet guidelines for quality improvement.
Results
The original STOP report included 397 drug interactions involving statins with gemfibrozil or niacin (Table 1). The majority of patients were white and male aged 60 to 79 years. Gemfibrozil was the most common drug involved in all interactions (79.8%). The most common statins were atorvastatin (40%) and simvastatin (36.5%).
Gemfibrozil-Statin Interactions
Pharmacists discontinued gemfibrozil by protocol for 94 patients (29.6%), and 107 patients (33.8%) were referred to the PACT Pharmacy Clinic (Figure 3). For the remaining 116 patients (36.6%), the drug interaction was addressed outside of the protocol for the following reasons: the drug interaction was resolved prior to pharmacist review; an interacting prescription was expired and not to be continued; the patient self-discontinued ≥ 1 interacting medications; the patient was deceased; the patient moved; the patient was receiving ≥ 1 interacting medications outside of the VA; or the prescriber resolved the interaction following notification by the pharmacist.
Ultimately, the interaction was resolved for all patients with a gemfibrozil-statin interaction on the STOP report. Following gemfibrozil discontinuation by protocol, 76 patients (80.9%) had TG laboratory results available and were included in the analysis. Sixty-two patients’ (82%) TG levels decreased or increased by < 100 mg/dL (Figure 4), and the TG levels of 1 patient (1.3%) increased above the threshold of 500 mg/dL. The mean (SD) time to the first laboratory result after the pharmacists mailed the notification letter was 6.5 (3.6) months (range, 1-17). The pharmacists spent a mean of 16 minutes per patient resolving each interaction.
Of the 107 patients referred to the PACT Pharmacy Clinic, 80 (74.8%) had TG laboratory results available and were included in the analysis. These patients were followed by the PACT CPS until the drug interaction was resolved and confirmed to have TG levels at goal (< 500 mg/dL). Gemfibrozil doses ranged from 300 mg daily to 600 mg twice daily, with 70% (n = 56) of patients taking 600 mg twice daily. The PACT CPS made 148 interventions (Table 2). Twenty-three (29%) patients required only gemfibrozil discontinuation. The remaining 57 patients (71%) required at least 2 medication interventions. The PACT CPS generated 213 encounters for resolving drug interactions with a median of 2 encounters per patient.
Quality assurance review identified 5 patients (5.3%) who underwent gemfibrozil discontinuation by protocol, despite having criteria that would have recommended against discontinuation. In accordance with the protocol criteria, these patients were later referred to the PACT Pharmacy Clinic. None of these patients experienced a TG increase at or above the threshold of 500 mg/dL after gemfibrozil was initially discontinued but were excluded from the earlier analysis.
Niacin-Statin Interactions
Pharmacists discontinued niacin by protocol for 48 patients (60.0%), and 22 patients (27.5%) were referred to the PACT Pharmacy Clinic (Figure 5). For the remaining 5 patients (6.3%), the interaction was either addressed outside the protocol prior to pharmacist review, or an interacting prescription was expired and not to be continued. Additionally, niacin was continued per prescriber preference in 5 patients (6.3%).
Thirty-six patients (75%) had TG laboratory results available following niacin discontinuation by protocol and were included in the analysis. Most patients’ (n = 33, 91.7%) TG levels decreased or increased by < 100 mg/dL. No patient had a TG level that increased higher than the threshold of 500 mg/dL. The mean (SD) time to the first laboratory result after the pharmacists mailed the notification letter, was 5.3 (2.5) months (range, 1.2-9.8). The pharmacists spent a mean of 15 minutes per patient resolving each interaction. The quality assurance review found no discrepancies in the pharmacists’ application of the protocol.
Of the 22 patients referred to the PACT Pharmacy Clinic, 16 (72.7%) patients had TG laboratory results available and were included in the analysis. As with the gemfibrozil interactions, these patients were followed by the PACT Pharmacy Clinic until the drug interaction was resolved and confirmed to have TGs at goal (< 500 mg/dL). Niacin doses ranged from 500 mg daily to 2,000 mg daily, with the majority of patients taking 1,000 mg daily. The PACT CPS made 23 interventions. The PACT CPS generated 46 encounters for resolving drug interactions with a median of 2 encounters per patient.
Discussion
Following gemfibrozil or niacin discontinuation by protocol, most patients with available laboratory results experienced either a decrease or modest TG elevation. The proportion of patients experiencing a decrease in TGs was unexpected but potentially multifactorial. Individual causes for the decrease in TGs were beyond the scope of this analysis. The retrospective design limited the ability to identify variables that could have impacted TG levels when gemfibrozil or niacin were started and discontinued. Although the treatment of TG levels is not indicated until it is ≥ 500 mg/dL, due to an increased risk of pancreatitis, both protocols excluded patients with a history of TGs ≥ 400 mg/dL.19 The lower threshold was set to compensate for anticipated increase in TG levels, following gemfibrozil or niacin discontinuation, and to minimize the number of patients with TG levels ≥ 500 mg/dL. The actual impact on patients’ TG levels supports the use of this lower threshold in the protocol.
When TG levels increased by 200 to 249 mg/dL after gemfibrozil or niacin discontinuation, patients were evaluated for possible underlying causes, which occurred for 4 gemfibrozil and 1 niacin patient. One patient started a β-blocker after gemfibrozil was initiated, and 3 patients were taking gemfibrozil prior to establishing care at the VA. The TG levels of the patient taking niacin correlated with an increased hemoglobin A1c. The TG level for only 1 patient taking gemfibrozil increased above the 500 mg/dL threshold. The patient had several comorbidities known to increase TG levels, but the comorbidities were previously well controlled. No additional medication changes were made at that time, and the TG levels on the next fasting lipid panel decreased to goal. The patient did not experience any negative clinical sequelae from the elevated TG levels.
Thirty-five patients (36%) who were referred to the PACT Pharmacy Clinic required only either gemfibrozil or niacin discontinuation. These patients were evaluated to identify whether adjustments to the protocols would have allowed for pharmacist discontinuation without referral to the PACT Pharmacy Clinic. Twenty-four of these patients (69%) had repeated TG levels ≥ 400 mg/dL prior to referral to the PACT Pharmacy Clinic. Additionally, there was no correlation between the gemfibrozil or niacin doses and the change in TG levels following discontinuation. These data indicate the protocols appropriately identified patients who did not have an indication for gemfibrozil or niacin.
In addition to drug interactions identified on the STOP report, the PACT CPS resolved 12 additional interactions involving simvastatin and gemfibrozil. Additionally, unnecessary lipid medications were deprescribed. The PACT CPS identified 13 patients who experienced myalgias, an ADR attributed to the gemfibrozil- statin interaction. Of those, 9 patients’ ADRs resolved after discontinuing gemfibrozil alone. For the remaining 4 patients, additional interventions to convert the patient to another statin were required to resolve the ADR.
Using pharmacists to address the drug interactions shifted workload from the prescribers and other primary care team members. The mean time spent to resolve both gemfibrozil and niacin interactions by protocol was 15.5 minutes. One hundred fortytwo patients (35.8%) had drug interactions resolved by protocol, saving the PACT CPS’ expertise for patients requiring individualized interventions. Drug interactions were resolved within 4 PACT CPS encounters for 93.8% of the patients taking gemfibrozil and within 3 PACT CPS encounters for 93.8% of the patients taking niacin.
The protocols allowed 12 additional pharmacists who did not have an ambulatory care scope of practice to assist the PACT CPS in mitigating the STOP drug interactions. These pharmacists otherwise would have been limited to making consultative recommendations. Simultaneously, the design allowed for the PACT pharmacists’ expertise to be allocated for patients most likely to require interventions beyond the protocols. This type of intraprofessional referral process is not well described in the medical literature. To the authors’ knowledge, the only studies described referrals from hospital pharmacists to community pharmacists during transitions of care on hospital discharge.20,21
Limitations
The results of this study are derived from a retrospective chart review at a single VA facility. The autonomous nature of PACT CPS interventions may be difficult to replicate in other settings that do not permit pharmacists the same prescriptive authority. This analysis was designed to demonstrate the impact of the pharmacist in resolving major drug interactions. Patients referred to the PACT Pharmacy Clinic who also had their lipid medications adjusted by a nonpharmacist provider were excluded. However, this may have minimized the impact of the PACT CPS on the patient care provided. As postintervention laboratory results were not available for all patients, some patients’ TG levels could have increased above the 500 mg/dL threshold but were not identified. The time investment was extensive and likely underestimates the true cost of implementing the interventions.
Because notification letters were used to instruct patients to stop gemfibrozil or niacin, several considerations need to be addressed when interpreting the follow-up laboratory results. First, we cannot confirm whether the patients received the letter or the exact date the letter was received. Additionally, we cannot confirm whether the patients followed the instructions to stop the interacting medications or the date the medications were stopped. It is possible some patients were still taking the interacting medication when the first laboratory was drawn. Should a patient have continued the interacting medication, most would have run out and been unable to obtain a refill within 90 days of receiving the letter, as this is the maximum amount dispensed at one time. The mean time to the first laboratory result for both gemfibrozil and niacin was 6.5 and 5.3 months, respectively. Approximately 85% of patients completed the first laboratory test at least 3 months after the letter was mailed.
The protocols were designed to assess whether gemfibrozil or niacin was indicated and did not assess whether the statin was indicated. Therefore, discontinuing the statin also could have resolved the interaction appropriately. However, due to characteristics of the patient population and recommendations in current lipid guidelines, it was more likely the statin would be indicated.22,23 The protocols also assumed that patients eligible for gemfibrozil or niacin discontinuation would not need additional changes to their lipid medications. The medication changes made by the PACT CPS may have gone beyond those minimally necessary to resolve the drug interaction and maintain TG goals. Patients who had gemfibrozil or niacin discontinued by protocol also may have benefited from additional optimization of their lipid medications.
Conclusions
This quality improvement analysis supports further evaluation of the complementary use of protocols and PACT CPS prescriptive authority to resolve statin drug interactions. The gemfibrozil and niacin protocols appropriately identified patients who were less likely to experience an adverse change in TG laboratory results. Patients more likely to require additional medication interventions were appropriately referred to the PACT Pharmacy Clinics for individualized care. These data support expanded roles for pharmacists, across various settings, to mitigate select drug interactions at the Truman VA.
Acknowledgments
This quality improvement project is the result of work supported with resources and use of the Harry S. Truman Memorial Veterans’ Hospital in Columbia, Missouri.
1. The top 200 drugs of 2020 Provided by the ClinCalc DrugStats Database. http://clincalc.com/DrugStats /Top200Drugs.aspx. Updated February 11, 2017. Accessed May 12, 2020.
2. Wiggins BS, Saseen JJ, Page RL 2nd, et al; American Heart Association Clinical Pharmacology Committee of the Council on Clinical Cardiology; Council on Hypertension; Council on Quality of Care and Outcomes Research; and Council on Functional Genomics and Translational Biology. Recommendations for management of clinically significant drug-drug interactions with statins and select agents used in patients with cardiovascular disease: a scientific statement from the American Heart Association. Circulation. 2016;134(21):e468‐e495. doi:10.1161/CIR.0000000000000456
3. Smithburger PL, Buckley MS, Bejian S, Burenheide K, Kane-Gill SL. A critical evaluation of clinical decision support for the detection of drug-drug interactions. Expert Opin Drug Saf. 2011;10(6):871‐882. doi:10.1517/14740338.2011.583916
4. US Food and Drug Administration. FDA drug safety communication: new restrictions, contraindications, and dose limitations for Zocor (simvastatin) to reduce the risk of muscle injury. https://www.fda.gov/Drugs/DrugSafety /ucm256581.htm. Updated December 15, 2017. Accessed May 12, 2020.
5. US Food and Drug Administration. FDA drug safety communication: important safety label changes to cholesterol-lowering statin drugs. https://www.fda.gov /Drugs/DrugSafety/ucm293101.htm. Updated January 19, 2016. Accessed May 12, 2020.
6. US Food and Drug Administration Federal Register. AbbVie Inc. et al; withdrawal of approval of indications related to the coadministration with statins in applications for niacin extended-release tablets and fenofibric acid delayed-release capsules. https://www.federalregister .gov/documents/2016/04/18/2016-08887/abbvie-inc -et-al-withdrawal-of-approval-of-indications-related -to-the-coadministration-with-statins. Published April 18, 2016. Accessed May 12, 2020.
7. Lamprecht DG Jr, Todd BA, Denham AM, Ruppe LK, Stadler SL. Clinical pharmacist patient-safety initiative to reduce against-label prescribing of statins with cyclosporine. Ann Pharmacother. 2017;51(2):140‐145. doi:10.1177/1060028016675352
8. Roblek T, Deticek A, Leskovar B, et al. Clinical-pharmacist intervention reduces clinically relevant drugdrug interactions in patients with heart failure: A randomized, double-blind, controlled trial. Int J Cardiol. 2016;203:647‐652. doi:10.1016/j.ijcard.2015.10.206
9. Tuchscherer RM, Nair K, Ghushchyan V, Saseen JJ. Simvastatin prescribing patterns before and after FDA dosing restrictions: a retrospective analysis of a large healthcare claims database. Am J Cardiovasc Drugs. 2015;15(1):27‐34. doi:10.1007/s40256-014-0096-x
10. Alford JC, Saseen JJ, Allen RR, Nair KV. Persistent use of against-label statin-fibrate combinations from 2003-2009 despite United States Food and Drug Administration dose restrictions. Pharmacotherapy. 2012;32(7):623‐630. doi:10.1002/j.1875-9114.2011.01090.x
11. Leape LL, Cullen DJ, Clapp MD, et al. Pharmacist participation on physician rounds and adverse drug events in the intensive care unit [published correction appears in JAMA 2000 Mar 8;283(10):1293]. JAMA. 1999;282(3):267‐270. doi:10.1001/jama.282.3.267
12. Kucukarslan SN, Peters M, Mlynarek M, Nafziger DA. Pharmacists on rounding teams reduce preventable adverse drug events in hospital general medicine units. Arch Intern Med. 2003;163(17):2014‐2018. doi:10.1001/archinte.163.17.2014
13. Humphries TL, Carroll N, Chester EA, Magid D, Rocho B. Evaluation of an electronic critical drug interaction program coupled with active pharmacist intervention. Ann Pharmacother. 2007;41(12):1979‐1985. doi:10.1345/aph.1K349
14. Zocor [package insert]. Whitehouse Station, NJ: Merck & Co, Inc; 2018.
15. Lipitor [package insert]. New York, NY: Pfizer; 2017.
16. Crestor [package insert]. Wilmington, DE: AstraZeneca; 2018.
17. Mevacor [package insert]. Whitehouse Station, NJ: Merck & Co, Inc; 2012.
18. Wolters Kluwer Health, Lexi-Drugs, Lexicomp. Pravastatin. www.online.lexi.com. [Source not verified.]
19. Miller M, Stone NJ, Ballantyne C, et al; American Heart Association Clinical Lipidology, Thrombosis, and Prevention Committee of the Council on Nutrition, Physical Activity, and Metabolism; Council on Arteriosclerosis, Thrombosis and Vascular Biology; Council on Cardiovascular Nursing; Council on the Kidney in Cardiovascular Disease. Triglycerides and cardiovascular disease: a scientific statement from the American Heart Association. Circulation. 2011;123(20):2292-2333. doi: 10.1161/CIR.0b013e3182160726
20. Ferguson J, Seston L, Ashcroft DM. Refer-to-pharmacy: a qualitative study exploring the implementation of an electronic transfer of care initiative to improve medicines optimisation following hospital discharge. BMC Health Serv Res. 2018;18(1):424. doi:10.1186/s12913-018-3262-z
21. Ensing HT, Koster ES, Dubero DJ, van Dooren AA, Bouvy ML. Collaboration between hospital and community pharmacists to address drug-related problems: the HomeCoMe-program. Res Social Adm Pharm. 2019;15(3):267‐278. doi:10.1016/j.sapharm.2018.05.001
22. US Department of Defense, US Department of Veterans Affairs. VA/DoD clinical practice guideline for the management of dyslipidemia for cardiovascular risk reduction guideline summary. https://www.healthquality.va.gov /guidelines/CD/lipids/LipidSumOptSinglePg31Aug15.pdf. Published 2014. Accessed May 14, 2020.
23. Stone NJ, Robinson JG, Lichtenstein AH, et al. 2013 ACC/AHA guideline on the treatment of blood cholesterol to reduce atherosclerotic cardiovascular risk in adults: a report of the American College of Cardiology/ American Heart Association Task Force on Practice Guidelines [published correction appears in Circulation. 2014 Jun 24;129(25) (suppl 2):S46-48] [published correction appears in Circulation. 2015 Dec 22;132(25):e396]. Circulation. 2014;129(25)(suppl 2): S1‐S45. doi:10.1161/01.cir.0000437738.63853.7a
1. The top 200 drugs of 2020 Provided by the ClinCalc DrugStats Database. http://clincalc.com/DrugStats /Top200Drugs.aspx. Updated February 11, 2017. Accessed May 12, 2020.
2. Wiggins BS, Saseen JJ, Page RL 2nd, et al; American Heart Association Clinical Pharmacology Committee of the Council on Clinical Cardiology; Council on Hypertension; Council on Quality of Care and Outcomes Research; and Council on Functional Genomics and Translational Biology. Recommendations for management of clinically significant drug-drug interactions with statins and select agents used in patients with cardiovascular disease: a scientific statement from the American Heart Association. Circulation. 2016;134(21):e468‐e495. doi:10.1161/CIR.0000000000000456
3. Smithburger PL, Buckley MS, Bejian S, Burenheide K, Kane-Gill SL. A critical evaluation of clinical decision support for the detection of drug-drug interactions. Expert Opin Drug Saf. 2011;10(6):871‐882. doi:10.1517/14740338.2011.583916
4. US Food and Drug Administration. FDA drug safety communication: new restrictions, contraindications, and dose limitations for Zocor (simvastatin) to reduce the risk of muscle injury. https://www.fda.gov/Drugs/DrugSafety /ucm256581.htm. Updated December 15, 2017. Accessed May 12, 2020.
5. US Food and Drug Administration. FDA drug safety communication: important safety label changes to cholesterol-lowering statin drugs. https://www.fda.gov /Drugs/DrugSafety/ucm293101.htm. Updated January 19, 2016. Accessed May 12, 2020.
6. US Food and Drug Administration Federal Register. AbbVie Inc. et al; withdrawal of approval of indications related to the coadministration with statins in applications for niacin extended-release tablets and fenofibric acid delayed-release capsules. https://www.federalregister .gov/documents/2016/04/18/2016-08887/abbvie-inc -et-al-withdrawal-of-approval-of-indications-related -to-the-coadministration-with-statins. Published April 18, 2016. Accessed May 12, 2020.
7. Lamprecht DG Jr, Todd BA, Denham AM, Ruppe LK, Stadler SL. Clinical pharmacist patient-safety initiative to reduce against-label prescribing of statins with cyclosporine. Ann Pharmacother. 2017;51(2):140‐145. doi:10.1177/1060028016675352
8. Roblek T, Deticek A, Leskovar B, et al. Clinical-pharmacist intervention reduces clinically relevant drugdrug interactions in patients with heart failure: A randomized, double-blind, controlled trial. Int J Cardiol. 2016;203:647‐652. doi:10.1016/j.ijcard.2015.10.206
9. Tuchscherer RM, Nair K, Ghushchyan V, Saseen JJ. Simvastatin prescribing patterns before and after FDA dosing restrictions: a retrospective analysis of a large healthcare claims database. Am J Cardiovasc Drugs. 2015;15(1):27‐34. doi:10.1007/s40256-014-0096-x
10. Alford JC, Saseen JJ, Allen RR, Nair KV. Persistent use of against-label statin-fibrate combinations from 2003-2009 despite United States Food and Drug Administration dose restrictions. Pharmacotherapy. 2012;32(7):623‐630. doi:10.1002/j.1875-9114.2011.01090.x
11. Leape LL, Cullen DJ, Clapp MD, et al. Pharmacist participation on physician rounds and adverse drug events in the intensive care unit [published correction appears in JAMA 2000 Mar 8;283(10):1293]. JAMA. 1999;282(3):267‐270. doi:10.1001/jama.282.3.267
12. Kucukarslan SN, Peters M, Mlynarek M, Nafziger DA. Pharmacists on rounding teams reduce preventable adverse drug events in hospital general medicine units. Arch Intern Med. 2003;163(17):2014‐2018. doi:10.1001/archinte.163.17.2014
13. Humphries TL, Carroll N, Chester EA, Magid D, Rocho B. Evaluation of an electronic critical drug interaction program coupled with active pharmacist intervention. Ann Pharmacother. 2007;41(12):1979‐1985. doi:10.1345/aph.1K349
14. Zocor [package insert]. Whitehouse Station, NJ: Merck & Co, Inc; 2018.
15. Lipitor [package insert]. New York, NY: Pfizer; 2017.
16. Crestor [package insert]. Wilmington, DE: AstraZeneca; 2018.
17. Mevacor [package insert]. Whitehouse Station, NJ: Merck & Co, Inc; 2012.
18. Wolters Kluwer Health, Lexi-Drugs, Lexicomp. Pravastatin. www.online.lexi.com. [Source not verified.]
19. Miller M, Stone NJ, Ballantyne C, et al; American Heart Association Clinical Lipidology, Thrombosis, and Prevention Committee of the Council on Nutrition, Physical Activity, and Metabolism; Council on Arteriosclerosis, Thrombosis and Vascular Biology; Council on Cardiovascular Nursing; Council on the Kidney in Cardiovascular Disease. Triglycerides and cardiovascular disease: a scientific statement from the American Heart Association. Circulation. 2011;123(20):2292-2333. doi: 10.1161/CIR.0b013e3182160726
20. Ferguson J, Seston L, Ashcroft DM. Refer-to-pharmacy: a qualitative study exploring the implementation of an electronic transfer of care initiative to improve medicines optimisation following hospital discharge. BMC Health Serv Res. 2018;18(1):424. doi:10.1186/s12913-018-3262-z
21. Ensing HT, Koster ES, Dubero DJ, van Dooren AA, Bouvy ML. Collaboration between hospital and community pharmacists to address drug-related problems: the HomeCoMe-program. Res Social Adm Pharm. 2019;15(3):267‐278. doi:10.1016/j.sapharm.2018.05.001
22. US Department of Defense, US Department of Veterans Affairs. VA/DoD clinical practice guideline for the management of dyslipidemia for cardiovascular risk reduction guideline summary. https://www.healthquality.va.gov /guidelines/CD/lipids/LipidSumOptSinglePg31Aug15.pdf. Published 2014. Accessed May 14, 2020.
23. Stone NJ, Robinson JG, Lichtenstein AH, et al. 2013 ACC/AHA guideline on the treatment of blood cholesterol to reduce atherosclerotic cardiovascular risk in adults: a report of the American College of Cardiology/ American Heart Association Task Force on Practice Guidelines [published correction appears in Circulation. 2014 Jun 24;129(25) (suppl 2):S46-48] [published correction appears in Circulation. 2015 Dec 22;132(25):e396]. Circulation. 2014;129(25)(suppl 2): S1‐S45. doi:10.1161/01.cir.0000437738.63853.7a
When Grief and Crises Intersect: Perspectives of a Black Physician in the Time of Two Pandemics
“Hey there—just checking on you and letting you know I’m thinking of you.”
“I know words don’t suffice right now. You are in my thoughts.”
“If there’s any way that I can be of support or if there’s something you need, just let me know.”
The texts and emails have come in waves. Pinging into my already distracted headspace when, like them, I’m supposed to be focused on a Zoom or WebEx department meeting. These somber reminders underscore what I have known for years but struggled to describe with each new “justice for” hashtag accompanying the name of the latest unarmed Black person to die. This is grief.
With every headline in prior years, as Black Americans we have usually found solace in our collective fellowship of suffering. Social media timelines become flooded with our own amen choirs and outrage along with words of comfort and inspiration. We remind ourselves of the prior atrocities survived by our people. And like them, we vow to rally; clinging to one other and praying to make it to shore. Though intermittently joined by a smattering of allies, our suffering has mostly been a private, repetitive mourning.
THE TWO PANDEMICS
The year 2020 ushered in a new decade along with the novel SARS-CoV2 (COVID-19) global pandemic. In addition to the thousands of lives that have been lost in the United States alone, COVID-19 brought with it a disruption of life in ways never seen by most generations. Schools and businesses were closed to mitigate spread. Mandatory shelter-in-place orders coupled with physical distancing recommendations limited human interactions and cancelled everything from hospital visitations to graduations, intergenerational family gatherings, conferences, and weddings.1 As the data expanded, it quickly became apparent that minorities, particularly Black Americans, shouldered a disproportionate burden of COVID-19.2 Known health disparities were amplified.
While caring for our patients as Black physicians in the time of coronavirus, silently we mourned again. The connection and trust once found through racial concordance was now masked figuratively and literally by personal protective equipment (PPE). We ignored the sting of intimations that the staggering numbers of African Americans hospitalized and dying from COVID-19 could be explained by lack of discipline or, worse, genetic differences by race. Years of disenfranchisement and missed economic opportunities forced large numbers of our patients and loved ones out on the front lines to do essential jobs—but without the celebratory cheers or fanfare enjoyed by others. Frantic phone calls from family and acquaintances interrupted our quiet drives home from emotionally grueling shifts in the hospital—each conversation serving as our personal evidence of COVID-19 and her ruthless ravage of the Black community. Add to this trying to serve as cultural bridges between the complexities of medical distrust and patient advocacy along with wrestling with our own vulnerability as potential COVID-19 patients, these have been overwhelming times to say the least.
Then came the acute decompensation of the chronic racism we’d always known in the form of three recent killings of more unarmed African Americans. On March 13, 2020, 26-year-old Breonna Taylor was shot after police forcibly entered her home after midnight on a “no knock” warrant.3 The story was buried in the news of COVID-19—but we knew. Later we’d learn that 26-year-old Ahmaud Arbery was shot and killed by armed neighbors while running through a Brunswick, Georgia, neighborhood. His death on February 23, 2020, initially yielded no criminal charges.4 Then, on May 25, 2020, George Floyd, a 46-year-old father arrested for suspected use of a counterfeit $20 bill, died after a law enforcement official kneeled with his full body weight upon Floyd’s neck for over 8 minutes.5 The deaths of Arbery and Floyd were captured by cell phone cameras which, aided by social media, quickly reached the eyes of the entire world.
At first, it seemed plausible that this would be like it always has been. A Black mother would stand before a podium filled with multiple microphones crying out in anguish. She would be flanked by community leaders and attorneys demanding justice. Hashtags would be formed. Our people would stand up or kneel down in solidarity—holding fast to our historic resilience. Evanescent allies would appear with signs on lawns and held high over heads. A few weeks would pass by and things would go back to normal. Black people would be left with what always remains: heads bowed and praying at dinner tables petitioning a higher power for protection followed by reaffirmations of what, if anything, could be done to keep our own mamas away from that podium. We’ve learned to treat the grief of racism as endemic to us alone, knowing that it has been a pandemic all along.
A TIME OF RECKONING
The intersection of the crisis of the COVID-19 pandemic, complete with its social isolation and inordinate impact on minorities, and the acuity of the grief felt by the most recent events of abject racism have coalesced to form what feels like a pivotal point in the arc of justice. Like the bloated, disfigured face of lynched teenager Emmett Till lying lifeless in an open casket for the entire world to see in 1955,6 footage of these recent deaths typify a level of inhumanity that makes it too hard to turn away or carry on in indifference. The acute-on-chronic grief of racism felt by African Americans has risen into a tsunami, washing open the eyes of privileged persons belonging to all races, ethnicities, faiths, socioeconomic backgrounds, political views, and ages. The bulging neck veins, crackles, and thumping gallop rhythm of our hidden grief has declared itself: The rest of the world now knows that we can’t breathe.
Our moral outrage is pushing us to do something. Marches and demonstrations have occurred in nearly every major city. For those historically disenfranchised and let down by our societal contract, grief has, at times, met rage. Though we all feel an urgency, when we try to imagine ways to dismantle racism in the US it seems insurmountable. But as hospitalists and leaders, we will face black patients, colleagues, and neighbors navigating the pain of this exhausting collective trauma. While we won’t have all the answers immediately, we recognize the peculiar intersection between the COVID-19 crisis and the tipping point of grief felt by Black people with the recent deaths of Ahmaud Arbery, Breonna Taylor, and George Floyd, and it urges us to try.
Where can we start?
This is a time of deep sorrow for Black people. Recognizing it as such is an empathic place to begin. Everyone steers through grief differently, but a few things always hold true:
- Listen more than you talk—even if it’s uncomfortable. This isn’t a time to render opinions or draw suffering comparisons.
- Timely support is always appreciated. Leaders should feel the urgency to speak up early and often. Formal letters from leadership on behalf of organizations may feel like an echo chamber but they are worth the effort. Delays can be misunderstood as indifference and make the pain worse.
- The ministry of presence does not have to be physical. Those awkward text messages and emails create psychological safety in your organization and reduce loneliness. They also afford space to those who are still processing emotions and would prefer not to talk.
- Don’t place an expectation on the grieving to guide you through ways to help them heal. Though well-meaning, it can be overwhelming. This is particularly true in these current times.
- When in doubt, remember that support is a verb. Ultimately, sustained action or inaction will make your position clearer than any text message or email. Be sensitive to the unique intricacy of chronicity and missed opportunity when talking about racism.
Along with the pain we all feel from the impact of COVID-19, this is the time to recognize that your African American colleagues, patients, and friends have been navigating another tenacious and far more destructive pandemic at the same time. It is acute. It is chronic. It is acute-on-chronic. Perhaps 2020 will also be remembered for the opportunity it presented for the centuries old scourge of racism to no longer be our transparent cross to bear alone. Unlike COVID-19, this pandemic of racism is not “unprecedented.” We have been here before. It’s time we all grieve—and act—together.
1. COVID-19: Statewide Shelter in Place Order. https://georgia.gov/covid-19-state-services-georgia/covid-19-statewide-shelter-place-order. Accessed June 2, 2020.
2. Garg S, Kim L, Whitaker M, et al. Hospitalization Rates and Characteristics of Patients Hospitalized with Laboratory-Confirmed Coronavirus Disease 2019—COVID-NET, 14 States, March 1–30, 2020. MMWR Morb Mortal Wkly Rep. 2020;69:458–464. http://dx.doi.org/10.15585/mmwr.mm6915e3.
3. Oppel RA Jr. Here’s what you need to know about Breonna Taylor’s death. May 30, 2020. New York Times. https://www.nytimes.com/article/breonna-taylor-police.html. Accessed June 2, 2020.
4. Fausset R. What we know about the shooting death of Ahmaud Arbery. New York Times. May 22, 2020. https://www.nytimes.com/article/ahmaud-arbery-shooting-georgia.html. Accessed June 2, 2020.
5. Hill E, Tiefenthäler, Triebert C, Jordan D, Willis H, Stein R. 8 minutes and 46 seconds: how George Floyd was killed in police custody. May 31, 2020. New York Times. https://www.nytimes.com/2020/05/31/us/george-floyd-investigation.html. Accessed June 2, 2020.
6. Pilkington E. Will justice finally be done for Emmett Till? Family hope a 65-year wait may soon be over. April 25, 2020. The Guardian. https://www.theguardian.com/us-news/2020/apr/25/emmett-till-long-wait-for-justice. Accessed June 2, 2020.
“Hey there—just checking on you and letting you know I’m thinking of you.”
“I know words don’t suffice right now. You are in my thoughts.”
“If there’s any way that I can be of support or if there’s something you need, just let me know.”
The texts and emails have come in waves. Pinging into my already distracted headspace when, like them, I’m supposed to be focused on a Zoom or WebEx department meeting. These somber reminders underscore what I have known for years but struggled to describe with each new “justice for” hashtag accompanying the name of the latest unarmed Black person to die. This is grief.
With every headline in prior years, as Black Americans we have usually found solace in our collective fellowship of suffering. Social media timelines become flooded with our own amen choirs and outrage along with words of comfort and inspiration. We remind ourselves of the prior atrocities survived by our people. And like them, we vow to rally; clinging to one other and praying to make it to shore. Though intermittently joined by a smattering of allies, our suffering has mostly been a private, repetitive mourning.
THE TWO PANDEMICS
The year 2020 ushered in a new decade along with the novel SARS-CoV2 (COVID-19) global pandemic. In addition to the thousands of lives that have been lost in the United States alone, COVID-19 brought with it a disruption of life in ways never seen by most generations. Schools and businesses were closed to mitigate spread. Mandatory shelter-in-place orders coupled with physical distancing recommendations limited human interactions and cancelled everything from hospital visitations to graduations, intergenerational family gatherings, conferences, and weddings.1 As the data expanded, it quickly became apparent that minorities, particularly Black Americans, shouldered a disproportionate burden of COVID-19.2 Known health disparities were amplified.
While caring for our patients as Black physicians in the time of coronavirus, silently we mourned again. The connection and trust once found through racial concordance was now masked figuratively and literally by personal protective equipment (PPE). We ignored the sting of intimations that the staggering numbers of African Americans hospitalized and dying from COVID-19 could be explained by lack of discipline or, worse, genetic differences by race. Years of disenfranchisement and missed economic opportunities forced large numbers of our patients and loved ones out on the front lines to do essential jobs—but without the celebratory cheers or fanfare enjoyed by others. Frantic phone calls from family and acquaintances interrupted our quiet drives home from emotionally grueling shifts in the hospital—each conversation serving as our personal evidence of COVID-19 and her ruthless ravage of the Black community. Add to this trying to serve as cultural bridges between the complexities of medical distrust and patient advocacy along with wrestling with our own vulnerability as potential COVID-19 patients, these have been overwhelming times to say the least.
Then came the acute decompensation of the chronic racism we’d always known in the form of three recent killings of more unarmed African Americans. On March 13, 2020, 26-year-old Breonna Taylor was shot after police forcibly entered her home after midnight on a “no knock” warrant.3 The story was buried in the news of COVID-19—but we knew. Later we’d learn that 26-year-old Ahmaud Arbery was shot and killed by armed neighbors while running through a Brunswick, Georgia, neighborhood. His death on February 23, 2020, initially yielded no criminal charges.4 Then, on May 25, 2020, George Floyd, a 46-year-old father arrested for suspected use of a counterfeit $20 bill, died after a law enforcement official kneeled with his full body weight upon Floyd’s neck for over 8 minutes.5 The deaths of Arbery and Floyd were captured by cell phone cameras which, aided by social media, quickly reached the eyes of the entire world.
At first, it seemed plausible that this would be like it always has been. A Black mother would stand before a podium filled with multiple microphones crying out in anguish. She would be flanked by community leaders and attorneys demanding justice. Hashtags would be formed. Our people would stand up or kneel down in solidarity—holding fast to our historic resilience. Evanescent allies would appear with signs on lawns and held high over heads. A few weeks would pass by and things would go back to normal. Black people would be left with what always remains: heads bowed and praying at dinner tables petitioning a higher power for protection followed by reaffirmations of what, if anything, could be done to keep our own mamas away from that podium. We’ve learned to treat the grief of racism as endemic to us alone, knowing that it has been a pandemic all along.
A TIME OF RECKONING
The intersection of the crisis of the COVID-19 pandemic, complete with its social isolation and inordinate impact on minorities, and the acuity of the grief felt by the most recent events of abject racism have coalesced to form what feels like a pivotal point in the arc of justice. Like the bloated, disfigured face of lynched teenager Emmett Till lying lifeless in an open casket for the entire world to see in 1955,6 footage of these recent deaths typify a level of inhumanity that makes it too hard to turn away or carry on in indifference. The acute-on-chronic grief of racism felt by African Americans has risen into a tsunami, washing open the eyes of privileged persons belonging to all races, ethnicities, faiths, socioeconomic backgrounds, political views, and ages. The bulging neck veins, crackles, and thumping gallop rhythm of our hidden grief has declared itself: The rest of the world now knows that we can’t breathe.
Our moral outrage is pushing us to do something. Marches and demonstrations have occurred in nearly every major city. For those historically disenfranchised and let down by our societal contract, grief has, at times, met rage. Though we all feel an urgency, when we try to imagine ways to dismantle racism in the US it seems insurmountable. But as hospitalists and leaders, we will face black patients, colleagues, and neighbors navigating the pain of this exhausting collective trauma. While we won’t have all the answers immediately, we recognize the peculiar intersection between the COVID-19 crisis and the tipping point of grief felt by Black people with the recent deaths of Ahmaud Arbery, Breonna Taylor, and George Floyd, and it urges us to try.
Where can we start?
This is a time of deep sorrow for Black people. Recognizing it as such is an empathic place to begin. Everyone steers through grief differently, but a few things always hold true:
- Listen more than you talk—even if it’s uncomfortable. This isn’t a time to render opinions or draw suffering comparisons.
- Timely support is always appreciated. Leaders should feel the urgency to speak up early and often. Formal letters from leadership on behalf of organizations may feel like an echo chamber but they are worth the effort. Delays can be misunderstood as indifference and make the pain worse.
- The ministry of presence does not have to be physical. Those awkward text messages and emails create psychological safety in your organization and reduce loneliness. They also afford space to those who are still processing emotions and would prefer not to talk.
- Don’t place an expectation on the grieving to guide you through ways to help them heal. Though well-meaning, it can be overwhelming. This is particularly true in these current times.
- When in doubt, remember that support is a verb. Ultimately, sustained action or inaction will make your position clearer than any text message or email. Be sensitive to the unique intricacy of chronicity and missed opportunity when talking about racism.
Along with the pain we all feel from the impact of COVID-19, this is the time to recognize that your African American colleagues, patients, and friends have been navigating another tenacious and far more destructive pandemic at the same time. It is acute. It is chronic. It is acute-on-chronic. Perhaps 2020 will also be remembered for the opportunity it presented for the centuries old scourge of racism to no longer be our transparent cross to bear alone. Unlike COVID-19, this pandemic of racism is not “unprecedented.” We have been here before. It’s time we all grieve—and act—together.
“Hey there—just checking on you and letting you know I’m thinking of you.”
“I know words don’t suffice right now. You are in my thoughts.”
“If there’s any way that I can be of support or if there’s something you need, just let me know.”
The texts and emails have come in waves. Pinging into my already distracted headspace when, like them, I’m supposed to be focused on a Zoom or WebEx department meeting. These somber reminders underscore what I have known for years but struggled to describe with each new “justice for” hashtag accompanying the name of the latest unarmed Black person to die. This is grief.
With every headline in prior years, as Black Americans we have usually found solace in our collective fellowship of suffering. Social media timelines become flooded with our own amen choirs and outrage along with words of comfort and inspiration. We remind ourselves of the prior atrocities survived by our people. And like them, we vow to rally; clinging to one other and praying to make it to shore. Though intermittently joined by a smattering of allies, our suffering has mostly been a private, repetitive mourning.
THE TWO PANDEMICS
The year 2020 ushered in a new decade along with the novel SARS-CoV2 (COVID-19) global pandemic. In addition to the thousands of lives that have been lost in the United States alone, COVID-19 brought with it a disruption of life in ways never seen by most generations. Schools and businesses were closed to mitigate spread. Mandatory shelter-in-place orders coupled with physical distancing recommendations limited human interactions and cancelled everything from hospital visitations to graduations, intergenerational family gatherings, conferences, and weddings.1 As the data expanded, it quickly became apparent that minorities, particularly Black Americans, shouldered a disproportionate burden of COVID-19.2 Known health disparities were amplified.
While caring for our patients as Black physicians in the time of coronavirus, silently we mourned again. The connection and trust once found through racial concordance was now masked figuratively and literally by personal protective equipment (PPE). We ignored the sting of intimations that the staggering numbers of African Americans hospitalized and dying from COVID-19 could be explained by lack of discipline or, worse, genetic differences by race. Years of disenfranchisement and missed economic opportunities forced large numbers of our patients and loved ones out on the front lines to do essential jobs—but without the celebratory cheers or fanfare enjoyed by others. Frantic phone calls from family and acquaintances interrupted our quiet drives home from emotionally grueling shifts in the hospital—each conversation serving as our personal evidence of COVID-19 and her ruthless ravage of the Black community. Add to this trying to serve as cultural bridges between the complexities of medical distrust and patient advocacy along with wrestling with our own vulnerability as potential COVID-19 patients, these have been overwhelming times to say the least.
Then came the acute decompensation of the chronic racism we’d always known in the form of three recent killings of more unarmed African Americans. On March 13, 2020, 26-year-old Breonna Taylor was shot after police forcibly entered her home after midnight on a “no knock” warrant.3 The story was buried in the news of COVID-19—but we knew. Later we’d learn that 26-year-old Ahmaud Arbery was shot and killed by armed neighbors while running through a Brunswick, Georgia, neighborhood. His death on February 23, 2020, initially yielded no criminal charges.4 Then, on May 25, 2020, George Floyd, a 46-year-old father arrested for suspected use of a counterfeit $20 bill, died after a law enforcement official kneeled with his full body weight upon Floyd’s neck for over 8 minutes.5 The deaths of Arbery and Floyd were captured by cell phone cameras which, aided by social media, quickly reached the eyes of the entire world.
At first, it seemed plausible that this would be like it always has been. A Black mother would stand before a podium filled with multiple microphones crying out in anguish. She would be flanked by community leaders and attorneys demanding justice. Hashtags would be formed. Our people would stand up or kneel down in solidarity—holding fast to our historic resilience. Evanescent allies would appear with signs on lawns and held high over heads. A few weeks would pass by and things would go back to normal. Black people would be left with what always remains: heads bowed and praying at dinner tables petitioning a higher power for protection followed by reaffirmations of what, if anything, could be done to keep our own mamas away from that podium. We’ve learned to treat the grief of racism as endemic to us alone, knowing that it has been a pandemic all along.
A TIME OF RECKONING
The intersection of the crisis of the COVID-19 pandemic, complete with its social isolation and inordinate impact on minorities, and the acuity of the grief felt by the most recent events of abject racism have coalesced to form what feels like a pivotal point in the arc of justice. Like the bloated, disfigured face of lynched teenager Emmett Till lying lifeless in an open casket for the entire world to see in 1955,6 footage of these recent deaths typify a level of inhumanity that makes it too hard to turn away or carry on in indifference. The acute-on-chronic grief of racism felt by African Americans has risen into a tsunami, washing open the eyes of privileged persons belonging to all races, ethnicities, faiths, socioeconomic backgrounds, political views, and ages. The bulging neck veins, crackles, and thumping gallop rhythm of our hidden grief has declared itself: The rest of the world now knows that we can’t breathe.
Our moral outrage is pushing us to do something. Marches and demonstrations have occurred in nearly every major city. For those historically disenfranchised and let down by our societal contract, grief has, at times, met rage. Though we all feel an urgency, when we try to imagine ways to dismantle racism in the US it seems insurmountable. But as hospitalists and leaders, we will face black patients, colleagues, and neighbors navigating the pain of this exhausting collective trauma. While we won’t have all the answers immediately, we recognize the peculiar intersection between the COVID-19 crisis and the tipping point of grief felt by Black people with the recent deaths of Ahmaud Arbery, Breonna Taylor, and George Floyd, and it urges us to try.
Where can we start?
This is a time of deep sorrow for Black people. Recognizing it as such is an empathic place to begin. Everyone steers through grief differently, but a few things always hold true:
- Listen more than you talk—even if it’s uncomfortable. This isn’t a time to render opinions or draw suffering comparisons.
- Timely support is always appreciated. Leaders should feel the urgency to speak up early and often. Formal letters from leadership on behalf of organizations may feel like an echo chamber but they are worth the effort. Delays can be misunderstood as indifference and make the pain worse.
- The ministry of presence does not have to be physical. Those awkward text messages and emails create psychological safety in your organization and reduce loneliness. They also afford space to those who are still processing emotions and would prefer not to talk.
- Don’t place an expectation on the grieving to guide you through ways to help them heal. Though well-meaning, it can be overwhelming. This is particularly true in these current times.
- When in doubt, remember that support is a verb. Ultimately, sustained action or inaction will make your position clearer than any text message or email. Be sensitive to the unique intricacy of chronicity and missed opportunity when talking about racism.
Along with the pain we all feel from the impact of COVID-19, this is the time to recognize that your African American colleagues, patients, and friends have been navigating another tenacious and far more destructive pandemic at the same time. It is acute. It is chronic. It is acute-on-chronic. Perhaps 2020 will also be remembered for the opportunity it presented for the centuries old scourge of racism to no longer be our transparent cross to bear alone. Unlike COVID-19, this pandemic of racism is not “unprecedented.” We have been here before. It’s time we all grieve—and act—together.
1. COVID-19: Statewide Shelter in Place Order. https://georgia.gov/covid-19-state-services-georgia/covid-19-statewide-shelter-place-order. Accessed June 2, 2020.
2. Garg S, Kim L, Whitaker M, et al. Hospitalization Rates and Characteristics of Patients Hospitalized with Laboratory-Confirmed Coronavirus Disease 2019—COVID-NET, 14 States, March 1–30, 2020. MMWR Morb Mortal Wkly Rep. 2020;69:458–464. http://dx.doi.org/10.15585/mmwr.mm6915e3.
3. Oppel RA Jr. Here’s what you need to know about Breonna Taylor’s death. May 30, 2020. New York Times. https://www.nytimes.com/article/breonna-taylor-police.html. Accessed June 2, 2020.
4. Fausset R. What we know about the shooting death of Ahmaud Arbery. New York Times. May 22, 2020. https://www.nytimes.com/article/ahmaud-arbery-shooting-georgia.html. Accessed June 2, 2020.
5. Hill E, Tiefenthäler, Triebert C, Jordan D, Willis H, Stein R. 8 minutes and 46 seconds: how George Floyd was killed in police custody. May 31, 2020. New York Times. https://www.nytimes.com/2020/05/31/us/george-floyd-investigation.html. Accessed June 2, 2020.
6. Pilkington E. Will justice finally be done for Emmett Till? Family hope a 65-year wait may soon be over. April 25, 2020. The Guardian. https://www.theguardian.com/us-news/2020/apr/25/emmett-till-long-wait-for-justice. Accessed June 2, 2020.
1. COVID-19: Statewide Shelter in Place Order. https://georgia.gov/covid-19-state-services-georgia/covid-19-statewide-shelter-place-order. Accessed June 2, 2020.
2. Garg S, Kim L, Whitaker M, et al. Hospitalization Rates and Characteristics of Patients Hospitalized with Laboratory-Confirmed Coronavirus Disease 2019—COVID-NET, 14 States, March 1–30, 2020. MMWR Morb Mortal Wkly Rep. 2020;69:458–464. http://dx.doi.org/10.15585/mmwr.mm6915e3.
3. Oppel RA Jr. Here’s what you need to know about Breonna Taylor’s death. May 30, 2020. New York Times. https://www.nytimes.com/article/breonna-taylor-police.html. Accessed June 2, 2020.
4. Fausset R. What we know about the shooting death of Ahmaud Arbery. New York Times. May 22, 2020. https://www.nytimes.com/article/ahmaud-arbery-shooting-georgia.html. Accessed June 2, 2020.
5. Hill E, Tiefenthäler, Triebert C, Jordan D, Willis H, Stein R. 8 minutes and 46 seconds: how George Floyd was killed in police custody. May 31, 2020. New York Times. https://www.nytimes.com/2020/05/31/us/george-floyd-investigation.html. Accessed June 2, 2020.
6. Pilkington E. Will justice finally be done for Emmett Till? Family hope a 65-year wait may soon be over. April 25, 2020. The Guardian. https://www.theguardian.com/us-news/2020/apr/25/emmett-till-long-wait-for-justice. Accessed June 2, 2020.
© 2020 Society of Hospital Medicine
Intensive Care Unit Utilization After Adoption of a Ward-Based High-Flow Nasal Cannula Protocol
Children hospitalized for bronchiolitis frequently require admission to the intensive care unit (ICU), with estimates as high as 18%1,2 and 35%3 in two prospective, multicenter studies. The indication for ICU admission is nearly always a need for advanced respiratory support, which historically consisted of continuous or bilevel positive airway pressure (CPAP and BiPAP, respectively) or mechanical ventilation. High-flow nasal cannula (HFNC) is a recent addition to the respiratory support armamentarium, delivering heated and humidified oxygen at rates of up to 60 L/min and allowing for clinicians to titrate both flow rate and fraction of inspired oxygen (FiO2).4
Several studies have demonstrated that HFNC is capable of decreasing a child’s work of breathing,5-8 and it has the potential advantage of being better tolerated than other forms of advanced respiratory support.9,10 These case-series physiologic studies informed early ward-based HFNC protocols for bronchiolitis, which were adopted to decrease ICU utilization. Since then, single center observational studies examining the association between ward-based HFNC protocols and subsequent ICU utilization have come to discordant conclusions.11-14 Studying the effect of employing HFNC outside of the ICU is challenging in the context of a randomized, controlled trial (RCT) because it is difficult to blind healthcare providers to the intervention and because crossover from the control group to HFNC is frequent. Two unblinded RCTs published in 2017 and 2018 found that children randomized to conventional nasal cannula were frequently escalated to HFNC (flow rates of 1-2 L/kg per minute), but neither trial found a difference in ICU admission.15,16 Sample sizes substantially larger than those present in currently published or registered RCTs would be required to evaluate the impact of ward-based HFNC protocols on the outcome that inspired the protocols in the first place, namely ICU utilization.17
Children’s hospitals have adopted ward-based HFNC protocols at different time points over the last decade, which allows for a natural experiment—a promising alternative study design that avoids the challenges of blinding, crossover, and modest sample sizes. In order to have sufficient postadoption data for analyses, the present study is limited to ward-based HFNC protocols adopted prior to 2016, which we have termed “early” ward-based HFNC protocols. Among children with bronchiolitis, our objective was to measure the association between hospital-level adoption of a ward-based HFNC protocol and subsequent ICU utilization, using a multicenter network of children’s hospitals.
METHODS
We conducted a multicenter retrospective cohort study using the Pediatric Health Information System (PHIS) database. The PHIS database is operated by the Children’s Hospital Association (Lenexa, Kansas) and provides deidentified patient-level information for children who receive hospital care at 55 US children’s hospitals. Available data elements include patient demographic data, discharge diagnosis and procedure codes, and detailed billing information, such as laboratory, imaging, pharmacy, and supply charges. At the patient level, the use of HFNC vs standard oxygen therapy circuits cannot be discriminated.
Exposure
The study exposure was a hospital’s first ward-based HFNC protocol, with adoption measured at the hospital level at each PHIS site via direct communication with leaders in hospital medicine. In most cases, first contact was made with the pediatric hospital medicine division chief or fellowship program director, who then, if necessary, connected study investigators to local HFNC champions aware of site-specific historical HFNC protocol details. Contact with a hospital was made only if the hospital had contributed at least 6 consecutive years of data to PHIS. Hospitals were classified as “adopting” hospitals if their HFNC protocol met all of the following criteria: (a) allows initiation of HFNC outside of the ICU (on the floor or in the ED), (b) allows continued care outside of the ICU (on the floor), (c) not limited to a small unit like an intermediate care unit, and (d) adopted during a specific, known respiratory season. Hospitals for which ward-based HFNC protocols were adopted but did not meet these criteria were excluded from further analysis. Our intent was to identify large scale, programmatic protocol launches and exclude hospitals with exceptions that might preclude a sizable portion of our cohort from being eligible for the protocol. Hospitals for which inpatient use of HFNC remains limited to the ICU were defined as “nonadopting” hospitals. Respondents at adopting hospitals were asked to share details about their protocol, including patient eligibility criteria and maximum HFNC rates of flow permitted outside of the ICU.
Patient Characteristics
Patients aged 3 to 24 months who were hospitalized at adopting and nonadopting hospitals were included if an International Classification of Diseases, Ninth Revision (ICD-9) discharge diagnosis code for bronchiolitis (466.XX) was present in any position (not limited to a primary diagnosis). The lower age limit of 3 months was chosen to match the most restrictive age eligibility criteria of provided HFNC protocols (Appendix 1). A crosswalk available from the Centers for Medicare & Medicaid Services18 was used to convert ICD-10 diagnosis and procedure codes from recent years to ICD-9 diagnosis and procedure codes. Patients were excluded if their encounter contained a diagnosis or procedure code signifying a complex chronic condition,19 if their hospitalization involved care in the neonatal ICU, or if their admission date occurred outside of the respiratory season. Respiratory season was defined as November 1 through April 30.
Outcomes
Outcomes were measured during three respiratory seasons leading up to adoption and during three respiratory seasons after adoption. The primary outcome was ICU utilization, including the proportion of patients admitted to the ICU and ICU length of stay, expressed as ICU days per 100 patients. Secondary outcomes included mean total length of stay and the proportion of patients who received mechanical ventilation. Lengths of stay were measured in days, the most granular unit of time provided in PHIS over the entire study period. As such, partial days of care are rounded up to 1 full day. A previously published strict definition for mechanical ventilation that limits false positives was used, requiring that patients have a procedure or supply code for mechanical ventilation and a pharmacy charge for a neuromuscular blocking agent.20
Primary Analysis
The primary analysis was restricted to adopting hospitals. An interrupted time series approach was used to measure two possible types of change associated with HFNC protocol adoption: an immediate intervention effect and a change in the slope of an outcome.21 The immediate intervention effect represents the change in the level of the outcome that occurs in the period immediately following the introduction of the protocol. The change in slope is the extent to which the outcome changes on a per season basis, attributable to the protocol. Interrupted time series estimates were adjusted for patient age, gender, race, ethnicity, and insurance type; linear regression was used for continuous outcomes and logistic regression for dichotomous outcomes. An ordinary least squares time series model was used to adjust for autocorrelation and Newey-West standard errors were employed.22 Analyses were performed using STATA version 14 (Stata-Corp, College Station, Texas).
Supplementary Analyses
Two preplanned supplementary analyses were conducted. Supplementary analysis 1 was identical to the primary analysis, with the exception that the first season after adoption was censored. The rationale for censoring the first adoption season was to account for a potential learning effect and/or delayed start to full protocol implementation. Supplementary analysis 2 used the nonadopting hospitals as a control group and subtracted the effects measured from an interrupted time series analysis among nonadopting hospitals from the effects measured among adopting hospitals. The rationale for this approach was to control for unmeasured secular (eg, availability of ICU beds) and temporal (eg, severity of a given bronchiolitis season) factors that may have coincidentally occurred with HFNC adoption seasons. The only modification to the interrupted time series approach for supplementary analysis 2 was to provide the nonadopting hospitals with an artificial interruption point because nonadopting hospitals, by definition, did not have an adoption season that could be used in an interrupted time series approach. The interruption point for nonadopting hospitals was set at the median adoption season for adopting hospitals.
RESULTS
Exposure
Leaders at 44 hospitals were contacted regarding their hospital’s use of HFNC outside of the ICU (Figure 1). Responses were obtained for 41 hospitals (93% response rate), 18 of which were classified as nonadopting hospitals. Of the 23 hospitals where the presence of ward-based HFNC protocols were reported, 12 met inclusion criteria and were classified as adopting hospitals. HFNC protocols were adopted at these hospitals in a staggered fashion between the 2010-2011 and 2015-2016 respiratory seasons (Figure 2). The median adoption season was the 2013-14 respiratory season.
Nine adopting hospitals were able to provide details about their first HFNC protocols (Appendix 1). No two protocols were identical, but they shared many similarities. Minimum age requirements ranged from birth to a few months of age. Exclusion criteria were particularly variable, with a history of chronic lung disease or apnea being the most common criteria. Maximum allowed rates of flow ranged from 4 to 10 liters per minute. Criteria for transfer to the ICU were consistently based on an elevated FiO2 and duration of HFNC exposure.
Patient Characteristics
A total of 32,809 bronchiolitis encounters occurred at adopting hospitals during qualifying respiratory seasons, of which 6,556 (20%) involved patients with a complex chronic condition and were excluded. Of the 26,253 included bronchiolitis encounters, 12,495 encounters occurred prior to ward-based HFNC protocol adoption and 13,758 encounters occurred after adoption. The median age of patients was 8 months (interquartile range, 5-14 months). Most patients were on government insurance (64%), male (58%), of white (56%) or black (18%) race, and of non-Hispanic ethnicity (72%). Pre- and postadoption patient demographics were similar (Appendix 2).
Primary Analysis
Shifts in the level of ICU use and ICU length of stay were observed at the time of adoption of a ward-based HFNC protocol (Figure 3). Specifically, ward-based HFNC protocol adoption was associated with an immediate 3.1% absolute increase (95% CI, 2.8%-3.4%) in the proportion of patients admitted to the ICU and a 9.1 days per 100 patients increase (95% CI, 5.1-13.2) in ICU length of stay (Table). The slope of ICU admissions per season was increasing after HFNC protocol adoption (1.0% increase per season; 95% CI, 0.8%-1.1%). When examined at the individual-hospital level (Appendix 3), seven hospitals were found to have significant increases in ICU admissions (immediate intervention effect or change in slope) after adoption, and one hospital was found to have a significant decrease in ICU admissions (change in slope only). Neither immediate intervention effects nor changes in the slopes of total length of stay and mechanical ventilation were observed, with mean total length of stay approximately 3 days and just over 1% of patients receiving mechanical ventilation (Figure 3).
Supplementary Analyses
Supplementary analyses were largely consistent with the primary analysis. Associations with increased ICU utilization were again observed, although the immediate change in ICU length of stay for supplementary analysis 1 was not significant and the slope for ICU length of stay in supplementary analysis 2 was down trending (Table). Changes in total length of stay and mechanical ventilation were not observed in either supplementary analysis, with the lone exception being an increase in the proportion of patients receiving mechanical ventilation per season (increase in slope) in supplementary analysis 1.
DISCUSSION
This is the largest multicenter study to date evaluating ICU utilization after adoption of a ward-based HFNC protocol for patients with bronchiolitis. While a principal goal of allowing HFNC use outside of the ICU is to reduce the time that patients with bronchiolitis spend in the ICU, we found that early protocols were, paradoxically, associated with increased ICU utilization. Ward-based HFNC protocols were not associated with changes in hospital length of stay or need for mechanical ventilation. Our findings are particularly relevant given that the majority of children’s hospitals in our sample have adopted ward-based HFNC protocols to care for patients with bronchiolitis.
The increase in ICU utilization measured in our study is a novel finding, seemingly in contradiction to existing literature. Early pilot studies inspired hope that employing HFNC on the general ward might prevent a portion of children from needing ICU care.11,12 Subsequent larger observational studies did not demonstrate decreases in ICU utilization after adoption of ward-based HFNC protocols.13,14 The two RCTs comparing low-flow and high-flow nasal cannula use outside of the ICU did not measure a statistically significant effect on ICU utilization, an exploratory outcome in both trials.15,16 However, the reported point estimates for absolute differences in ICU admission were 2% to 3% higher among patients randomized to HFNC, which is consistent with the 2% to 4% increase in ICU admission measured in the present study.
What might explain this surprising finding? While our observational study cannot speak to mechanism, the protocol details examined in the present study suggest that initial adoption of a ward-based HFNC protocol is often coupled with specific ICU transfer criteria that were unlikely in place prior to protocol initiation. For example, most protocols recommended consideration of ICU transfer for elevated FiO2 or prolonged duration of HFNC. Transfer to the ICU for prolonged HFNC duration is only possible in the setting of a ward-based HFNC protocol and transfer for elevated FiO2 was probably unnecessary prior to protocol adoption given that low-flow nasal cannula generally delivers 100% FiO2. It is also possible that with HFNC comes a perception of increased acuity. For example, medical providers may see patients on HFNC as sicker than patients with the same amount of work of breathing but off HFNC, which makes providers more likely to seek ICU admission for patients on HFNC. The combination of unchanged total length of stay and increased ICU utilization suggests that early ward-based HFNC protocols were an ineffective instrument to improve hospital bed availability during the peak census times that often occur in bronchiolitis season.
The large sample size afforded our study by its multicenter, retrospective design also allowed for a meaningful assessment of the association between a ward-based HFNC protocol and the need for mechanical ventilation. Early indications suggested a lack of substantial association between HFNC use outside of the ICU and rates of mechanical ventilation, but prior studies were limited by small numbers of patients receiving mechanical ventilation (<30 patients in each study).13,14,16 The present study, in which 783 patients received mechanical ventilation, supports the lack of association between early ward-based HFNC protocols and the need for mechanical ventilation. It should be noted that other studies have measured decreases in mechanical ventilation in association with ICU-based HFNC use.23-26 In addition to examining HFNC use in a different clinical context, decreases in mechanical ventilation measured after HFNC implementation in the ICU could be explained by preexisting practice trends to limit invasive ventilation and/or selection bias resulting from an increase in less severely ill patients being admitted to the ICU over time. The interrupted time series approach and the staggered adoption of HFNC protocols make the present study less susceptible to biases from preexisting trends and the inclusion of patients cared for both on the ward and within the ICU reduces selection bias.
Our study has several important limitations. First, all hospitals included in the analysis were US children’s hospitals and these findings may not generalize to other practice environments, including community hospitals and other countries. Second, our cohort and outcomes were defined using administrative billing data, which have been incompletely validated, making some degree of misclassification likely. Third, we measured HFNC exposure at the hospital level, but could not examine the extent to which individual patients were exposed to HFNC because such data are not present in PHIS. Even if we had access to patient-level HFNC exposure data, we would have still compared outcomes among all patients with bronchiolitis (not just those who received HFNC), to avoid selection bias. However, knowing HFNC exposure status at the patient level would have allowed for weighting of the effects measured at each hospital according to the extent of HFNC exposure. Fourth, there are likely other, unmeasured secular and temporal factors that could affect study outcomes. To some degree, the interrupted time series approach, observed staggered adoption of protocols, and nonadopting hospital supplementary analysis mitigate this risk of bias. Fifth, while the pre- and postadoption populations appeared demographically similar, it is possible that the populations might have differed by other unmeasured factors. Finally, early ward-based HFNC protocols have likely undergone iterative changes since adoption. We compared pre- and postadoption outcome slopes and censored the first adoption season in a supplementary analysis to attempt to account for this potential limitation.
In conclusion, our findings suggest that initial implementation of ward-based HFNC protocols were not successful at reducing ICU utilization for children with bronchiolitis. Future research should examine whether more evolved HFNC protocols that use higher flow rates, more generous ICU transfer criteria, and more rapid weaning criteria can reduce ICU utilization.
Acknowledgments
We thank Dr Vineeta Mittal (University of Texas Southwestern Medical Center) for providing feedback regarding the manuscript.
1. Mansbach JM, Piedra PA, Teach SJ, et al. Prospective multicenter study of viral etiology and hospital length of stay in children with severe bronchiolitis. Arch Pediatr Adolesc Med. 2012;166(8):700-706. https://doi.org/10.1001/archpediatrics.2011.1669.
2. Hasegawa K, Pate BM, Mansbach JM, et al. Risk factors for requiring intensive care among children admitted to ward with bronchiolitis. Acad Pediatr. 2015;15(1):77-81. https://doi.org/10.1016/j.acap.2014.06.008.
3. Schroeder AR, Destino LA, Brooks R, Wang CJ, Coon ER. Outcomes of follow-up visits after bronchiolitis hospitalizations. JAMA Pediatr. 2018;172(3):296-297. https://doi.org/10.1001/jamapediatrics.2017.4002.
4. Drake MG. High-flow nasal cannula oxygen in adults: an evidence-based assessment. Ann Am Thorac Soc. 2018;15(2):145-155. https://doi.org/10.1513/AnnalsATS.201707-548FR.
5. Rubin S, Ghuman A, Deakers T, Khemani R, Ross P, Newth CJ. Effort of breathing in children receiving high-flow nasal cannula. Pediatr Crit Care Med. 2014;15(1):1-6. https://doi.org/10.1097/PCC.0000000000000011.
6. Hough JL, Pham TM, Schibler A. Physiologic effect of high-flow nasal cannula in infants with bronchiolitis. Pediatr Crit Care Med. 2014;15(5):e214-e219. https://doi.org/10.1097/PCC.0000000000000112.
7. Pham TM, O’Malley L, Mayfield S, Martin S, Schibler A. The effect of high flow nasal cannula therapy on the work of breathing in infants with bronchiolitis. Pediatr Pulmonol. 2015;50(7):713-720. https://doi.org/10.1002/ppul.23060.
8. Weiler T, Kamerkar A, Hotz J, Ross PA, Newth CJL, Khemani RG. The relationship between high flow nasal cannula flow rate and effort of breathing in children. J Pediatr. 2017;189:66-71.e63. https://doi.org/10.1016/j.jpeds.2017.06.006.
9. Mayfield S, Jauncey-Cooke J, Hough JL, Schibler A, Gibbons K, Bogossian F. High-flow nasal cannula therapy for respiratory support in children. Cochrane Database Syst Rev. 2014(3):CD009850. https://doi.org/10.1002/14651858.CD009850.pub2.
10. Roca O, Riera J, Torres F, Masclans JR. High-flow oxygen therapy in acute respiratory failure. Respir Care. 2010;55(4):408-413.
11. Kallappa C, Hufton M, Millen G, Ninan TK. Use of high flow nasal cannula oxygen (HFNCO) in infants with bronchiolitis on a paediatric ward: a 3-year experience. Arch Dis Child. 2014;99(8):790-791. https://doi.org/10.1136/archdischild-2014-306637.
12. Mayfield S, Bogossian F, O’Malley L, Schibler A. High-flow nasal cannula oxygen therapy for infants with bronchiolitis: pilot study. J Paediatr Child Health. 2014;50(5):373-378. https://doi.org/10.1111/jpc.12509.
13. Riese J, Porter T, Fierce J, Riese A, Richardson T, Alverson BK. Clinical outcomes of bronchiolitis after implementation of a general ward high flow nasal cannula guideline. Hosp Pediatr. 2017;7(4):197-203. https://doi.org/10.1542/hpeds.2016-0195.
14. Mace AO, Gibbons J, Schultz A, Knight G, Martin AC. Humidified high-flow nasal cannula oxygen for bronchiolitis: should we go with the flow? Arch Dis Child. 2018;103(3):303. https://doi.org/10.1136/archdischild-2017-313950.
15. Kepreotes E, Whitehead B, Attia J, et al. High-flow warm humidified oxygen versus standard low-flow nasal cannula oxygen for moderate bronchiolitis (HFWHO RCT): an open, phase 4, randomised controlled trial. Lancet. 2017;389(10072):930-939. https://doi.org/10.1016/S0140-6736(17)30061-2.
16. Franklin D, Babl FE, Schlapbach LJ, et al. A randomized trial of high-flow oxygen therapy in infants with bronchiolitis. N Engl J Med. 2018;378(12):1121-1131. https://doi.org/10.1056/NEJMoa1714855.
17. Coon ER, Mittal V, Brady PW. High flow nasal cannula-just expensive paracetamol? Lancet Child Adolesc Health. 2019;3(9):593-595. https://doi.org/10.1016/S2352-4642(19)30235-4.
18. Roth J. CMS’ ICD-9-CM to and from ICD-10-CM and ICD-10-PCS Crosswalk or General Equivalence Mappings. 2012. http://www.nber.org/data/icd9-icd-10-cm-and-pcs-crosswalk-general-equivalence-mapping.html. Accessed November 19, 2016.
19. Feudtner C, Feinstein JA, Zhong W, Hall M, Dai D. Pediatric complex chronic conditions classification system version 2: updated for ICD-10 and complex medical technology dependence and transplantation. BMC Pediatr. 2014;14:199. https://doi.org/10.1186/1471-2431-14-199.
20. Shein SL, Slain K, Wilson-Costello D, McKee B, Rotta AT. Temporal changes in prescription of neuropharmacologic drugs and utilization of resources related to neurologic morbidity in mechanically ventilated children with bronchiolitis. Pediatr Crit Care Med. 2017;18(12):e606-e614. https://doi.org/10.1097/PCC.0000000000001351.
21. Penfold RB, Zhang F. Use of interrupted time series analysis in evaluating health care quality improvements. Acad Pediatr. 2013;13(6 Suppl):S38-S44. https://doi.org/10.1016/j.acap.2013.08.002.
22. Newey WK, West KD. A simple, positive semi-definite, heteroskedasticity and autocorrelation consistent covariance matrix. Econometrica. 1987;55(3):703-708.
23. McKiernan C, Chua LC, Visintainer PF, Allen H. High flow nasal cannulae therapy in infants with bronchiolitis. J Pediatr. 2010;156(4):634-638. https://doi.org/10.1016/j.jpeds.2009.10.039.
24. Schibler A, Pham TM, Dunster KR, et al. Reduced intubation rates for infants after introduction of high-flow nasal prong oxygen delivery. Intensive Care Med. 2011;37(5):847-852. https://doi.org/10.1007/s00134-011-2177-5.
25. Kawaguchi A, Yasui Y, deCaen A, Garros D. The clinical impact of heated humidified high-flow nasal cannula on pediatric respiratory distress. Pediatr Crit Care Med. 2017;18(2):112-119. https://doi.org/10.1097/PCC.0000000000000985.
26. Schlapbach LJ, Straney L, Gelbart B, et al. Burden of disease and change in practice in critically ill infants with bronchiolitis. Eur Respir J. 2017;49(6):1601648. https://doi.org/10.1183/13993003.01648-2016.
Children hospitalized for bronchiolitis frequently require admission to the intensive care unit (ICU), with estimates as high as 18%1,2 and 35%3 in two prospective, multicenter studies. The indication for ICU admission is nearly always a need for advanced respiratory support, which historically consisted of continuous or bilevel positive airway pressure (CPAP and BiPAP, respectively) or mechanical ventilation. High-flow nasal cannula (HFNC) is a recent addition to the respiratory support armamentarium, delivering heated and humidified oxygen at rates of up to 60 L/min and allowing for clinicians to titrate both flow rate and fraction of inspired oxygen (FiO2).4
Several studies have demonstrated that HFNC is capable of decreasing a child’s work of breathing,5-8 and it has the potential advantage of being better tolerated than other forms of advanced respiratory support.9,10 These case-series physiologic studies informed early ward-based HFNC protocols for bronchiolitis, which were adopted to decrease ICU utilization. Since then, single center observational studies examining the association between ward-based HFNC protocols and subsequent ICU utilization have come to discordant conclusions.11-14 Studying the effect of employing HFNC outside of the ICU is challenging in the context of a randomized, controlled trial (RCT) because it is difficult to blind healthcare providers to the intervention and because crossover from the control group to HFNC is frequent. Two unblinded RCTs published in 2017 and 2018 found that children randomized to conventional nasal cannula were frequently escalated to HFNC (flow rates of 1-2 L/kg per minute), but neither trial found a difference in ICU admission.15,16 Sample sizes substantially larger than those present in currently published or registered RCTs would be required to evaluate the impact of ward-based HFNC protocols on the outcome that inspired the protocols in the first place, namely ICU utilization.17
Children’s hospitals have adopted ward-based HFNC protocols at different time points over the last decade, which allows for a natural experiment—a promising alternative study design that avoids the challenges of blinding, crossover, and modest sample sizes. In order to have sufficient postadoption data for analyses, the present study is limited to ward-based HFNC protocols adopted prior to 2016, which we have termed “early” ward-based HFNC protocols. Among children with bronchiolitis, our objective was to measure the association between hospital-level adoption of a ward-based HFNC protocol and subsequent ICU utilization, using a multicenter network of children’s hospitals.
METHODS
We conducted a multicenter retrospective cohort study using the Pediatric Health Information System (PHIS) database. The PHIS database is operated by the Children’s Hospital Association (Lenexa, Kansas) and provides deidentified patient-level information for children who receive hospital care at 55 US children’s hospitals. Available data elements include patient demographic data, discharge diagnosis and procedure codes, and detailed billing information, such as laboratory, imaging, pharmacy, and supply charges. At the patient level, the use of HFNC vs standard oxygen therapy circuits cannot be discriminated.
Exposure
The study exposure was a hospital’s first ward-based HFNC protocol, with adoption measured at the hospital level at each PHIS site via direct communication with leaders in hospital medicine. In most cases, first contact was made with the pediatric hospital medicine division chief or fellowship program director, who then, if necessary, connected study investigators to local HFNC champions aware of site-specific historical HFNC protocol details. Contact with a hospital was made only if the hospital had contributed at least 6 consecutive years of data to PHIS. Hospitals were classified as “adopting” hospitals if their HFNC protocol met all of the following criteria: (a) allows initiation of HFNC outside of the ICU (on the floor or in the ED), (b) allows continued care outside of the ICU (on the floor), (c) not limited to a small unit like an intermediate care unit, and (d) adopted during a specific, known respiratory season. Hospitals for which ward-based HFNC protocols were adopted but did not meet these criteria were excluded from further analysis. Our intent was to identify large scale, programmatic protocol launches and exclude hospitals with exceptions that might preclude a sizable portion of our cohort from being eligible for the protocol. Hospitals for which inpatient use of HFNC remains limited to the ICU were defined as “nonadopting” hospitals. Respondents at adopting hospitals were asked to share details about their protocol, including patient eligibility criteria and maximum HFNC rates of flow permitted outside of the ICU.
Patient Characteristics
Patients aged 3 to 24 months who were hospitalized at adopting and nonadopting hospitals were included if an International Classification of Diseases, Ninth Revision (ICD-9) discharge diagnosis code for bronchiolitis (466.XX) was present in any position (not limited to a primary diagnosis). The lower age limit of 3 months was chosen to match the most restrictive age eligibility criteria of provided HFNC protocols (Appendix 1). A crosswalk available from the Centers for Medicare & Medicaid Services18 was used to convert ICD-10 diagnosis and procedure codes from recent years to ICD-9 diagnosis and procedure codes. Patients were excluded if their encounter contained a diagnosis or procedure code signifying a complex chronic condition,19 if their hospitalization involved care in the neonatal ICU, or if their admission date occurred outside of the respiratory season. Respiratory season was defined as November 1 through April 30.
Outcomes
Outcomes were measured during three respiratory seasons leading up to adoption and during three respiratory seasons after adoption. The primary outcome was ICU utilization, including the proportion of patients admitted to the ICU and ICU length of stay, expressed as ICU days per 100 patients. Secondary outcomes included mean total length of stay and the proportion of patients who received mechanical ventilation. Lengths of stay were measured in days, the most granular unit of time provided in PHIS over the entire study period. As such, partial days of care are rounded up to 1 full day. A previously published strict definition for mechanical ventilation that limits false positives was used, requiring that patients have a procedure or supply code for mechanical ventilation and a pharmacy charge for a neuromuscular blocking agent.20
Primary Analysis
The primary analysis was restricted to adopting hospitals. An interrupted time series approach was used to measure two possible types of change associated with HFNC protocol adoption: an immediate intervention effect and a change in the slope of an outcome.21 The immediate intervention effect represents the change in the level of the outcome that occurs in the period immediately following the introduction of the protocol. The change in slope is the extent to which the outcome changes on a per season basis, attributable to the protocol. Interrupted time series estimates were adjusted for patient age, gender, race, ethnicity, and insurance type; linear regression was used for continuous outcomes and logistic regression for dichotomous outcomes. An ordinary least squares time series model was used to adjust for autocorrelation and Newey-West standard errors were employed.22 Analyses were performed using STATA version 14 (Stata-Corp, College Station, Texas).
Supplementary Analyses
Two preplanned supplementary analyses were conducted. Supplementary analysis 1 was identical to the primary analysis, with the exception that the first season after adoption was censored. The rationale for censoring the first adoption season was to account for a potential learning effect and/or delayed start to full protocol implementation. Supplementary analysis 2 used the nonadopting hospitals as a control group and subtracted the effects measured from an interrupted time series analysis among nonadopting hospitals from the effects measured among adopting hospitals. The rationale for this approach was to control for unmeasured secular (eg, availability of ICU beds) and temporal (eg, severity of a given bronchiolitis season) factors that may have coincidentally occurred with HFNC adoption seasons. The only modification to the interrupted time series approach for supplementary analysis 2 was to provide the nonadopting hospitals with an artificial interruption point because nonadopting hospitals, by definition, did not have an adoption season that could be used in an interrupted time series approach. The interruption point for nonadopting hospitals was set at the median adoption season for adopting hospitals.
RESULTS
Exposure
Leaders at 44 hospitals were contacted regarding their hospital’s use of HFNC outside of the ICU (Figure 1). Responses were obtained for 41 hospitals (93% response rate), 18 of which were classified as nonadopting hospitals. Of the 23 hospitals where the presence of ward-based HFNC protocols were reported, 12 met inclusion criteria and were classified as adopting hospitals. HFNC protocols were adopted at these hospitals in a staggered fashion between the 2010-2011 and 2015-2016 respiratory seasons (Figure 2). The median adoption season was the 2013-14 respiratory season.
Nine adopting hospitals were able to provide details about their first HFNC protocols (Appendix 1). No two protocols were identical, but they shared many similarities. Minimum age requirements ranged from birth to a few months of age. Exclusion criteria were particularly variable, with a history of chronic lung disease or apnea being the most common criteria. Maximum allowed rates of flow ranged from 4 to 10 liters per minute. Criteria for transfer to the ICU were consistently based on an elevated FiO2 and duration of HFNC exposure.
Patient Characteristics
A total of 32,809 bronchiolitis encounters occurred at adopting hospitals during qualifying respiratory seasons, of which 6,556 (20%) involved patients with a complex chronic condition and were excluded. Of the 26,253 included bronchiolitis encounters, 12,495 encounters occurred prior to ward-based HFNC protocol adoption and 13,758 encounters occurred after adoption. The median age of patients was 8 months (interquartile range, 5-14 months). Most patients were on government insurance (64%), male (58%), of white (56%) or black (18%) race, and of non-Hispanic ethnicity (72%). Pre- and postadoption patient demographics were similar (Appendix 2).
Primary Analysis
Shifts in the level of ICU use and ICU length of stay were observed at the time of adoption of a ward-based HFNC protocol (Figure 3). Specifically, ward-based HFNC protocol adoption was associated with an immediate 3.1% absolute increase (95% CI, 2.8%-3.4%) in the proportion of patients admitted to the ICU and a 9.1 days per 100 patients increase (95% CI, 5.1-13.2) in ICU length of stay (Table). The slope of ICU admissions per season was increasing after HFNC protocol adoption (1.0% increase per season; 95% CI, 0.8%-1.1%). When examined at the individual-hospital level (Appendix 3), seven hospitals were found to have significant increases in ICU admissions (immediate intervention effect or change in slope) after adoption, and one hospital was found to have a significant decrease in ICU admissions (change in slope only). Neither immediate intervention effects nor changes in the slopes of total length of stay and mechanical ventilation were observed, with mean total length of stay approximately 3 days and just over 1% of patients receiving mechanical ventilation (Figure 3).
Supplementary Analyses
Supplementary analyses were largely consistent with the primary analysis. Associations with increased ICU utilization were again observed, although the immediate change in ICU length of stay for supplementary analysis 1 was not significant and the slope for ICU length of stay in supplementary analysis 2 was down trending (Table). Changes in total length of stay and mechanical ventilation were not observed in either supplementary analysis, with the lone exception being an increase in the proportion of patients receiving mechanical ventilation per season (increase in slope) in supplementary analysis 1.
DISCUSSION
This is the largest multicenter study to date evaluating ICU utilization after adoption of a ward-based HFNC protocol for patients with bronchiolitis. While a principal goal of allowing HFNC use outside of the ICU is to reduce the time that patients with bronchiolitis spend in the ICU, we found that early protocols were, paradoxically, associated with increased ICU utilization. Ward-based HFNC protocols were not associated with changes in hospital length of stay or need for mechanical ventilation. Our findings are particularly relevant given that the majority of children’s hospitals in our sample have adopted ward-based HFNC protocols to care for patients with bronchiolitis.
The increase in ICU utilization measured in our study is a novel finding, seemingly in contradiction to existing literature. Early pilot studies inspired hope that employing HFNC on the general ward might prevent a portion of children from needing ICU care.11,12 Subsequent larger observational studies did not demonstrate decreases in ICU utilization after adoption of ward-based HFNC protocols.13,14 The two RCTs comparing low-flow and high-flow nasal cannula use outside of the ICU did not measure a statistically significant effect on ICU utilization, an exploratory outcome in both trials.15,16 However, the reported point estimates for absolute differences in ICU admission were 2% to 3% higher among patients randomized to HFNC, which is consistent with the 2% to 4% increase in ICU admission measured in the present study.
What might explain this surprising finding? While our observational study cannot speak to mechanism, the protocol details examined in the present study suggest that initial adoption of a ward-based HFNC protocol is often coupled with specific ICU transfer criteria that were unlikely in place prior to protocol initiation. For example, most protocols recommended consideration of ICU transfer for elevated FiO2 or prolonged duration of HFNC. Transfer to the ICU for prolonged HFNC duration is only possible in the setting of a ward-based HFNC protocol and transfer for elevated FiO2 was probably unnecessary prior to protocol adoption given that low-flow nasal cannula generally delivers 100% FiO2. It is also possible that with HFNC comes a perception of increased acuity. For example, medical providers may see patients on HFNC as sicker than patients with the same amount of work of breathing but off HFNC, which makes providers more likely to seek ICU admission for patients on HFNC. The combination of unchanged total length of stay and increased ICU utilization suggests that early ward-based HFNC protocols were an ineffective instrument to improve hospital bed availability during the peak census times that often occur in bronchiolitis season.
The large sample size afforded our study by its multicenter, retrospective design also allowed for a meaningful assessment of the association between a ward-based HFNC protocol and the need for mechanical ventilation. Early indications suggested a lack of substantial association between HFNC use outside of the ICU and rates of mechanical ventilation, but prior studies were limited by small numbers of patients receiving mechanical ventilation (<30 patients in each study).13,14,16 The present study, in which 783 patients received mechanical ventilation, supports the lack of association between early ward-based HFNC protocols and the need for mechanical ventilation. It should be noted that other studies have measured decreases in mechanical ventilation in association with ICU-based HFNC use.23-26 In addition to examining HFNC use in a different clinical context, decreases in mechanical ventilation measured after HFNC implementation in the ICU could be explained by preexisting practice trends to limit invasive ventilation and/or selection bias resulting from an increase in less severely ill patients being admitted to the ICU over time. The interrupted time series approach and the staggered adoption of HFNC protocols make the present study less susceptible to biases from preexisting trends and the inclusion of patients cared for both on the ward and within the ICU reduces selection bias.
Our study has several important limitations. First, all hospitals included in the analysis were US children’s hospitals and these findings may not generalize to other practice environments, including community hospitals and other countries. Second, our cohort and outcomes were defined using administrative billing data, which have been incompletely validated, making some degree of misclassification likely. Third, we measured HFNC exposure at the hospital level, but could not examine the extent to which individual patients were exposed to HFNC because such data are not present in PHIS. Even if we had access to patient-level HFNC exposure data, we would have still compared outcomes among all patients with bronchiolitis (not just those who received HFNC), to avoid selection bias. However, knowing HFNC exposure status at the patient level would have allowed for weighting of the effects measured at each hospital according to the extent of HFNC exposure. Fourth, there are likely other, unmeasured secular and temporal factors that could affect study outcomes. To some degree, the interrupted time series approach, observed staggered adoption of protocols, and nonadopting hospital supplementary analysis mitigate this risk of bias. Fifth, while the pre- and postadoption populations appeared demographically similar, it is possible that the populations might have differed by other unmeasured factors. Finally, early ward-based HFNC protocols have likely undergone iterative changes since adoption. We compared pre- and postadoption outcome slopes and censored the first adoption season in a supplementary analysis to attempt to account for this potential limitation.
In conclusion, our findings suggest that initial implementation of ward-based HFNC protocols were not successful at reducing ICU utilization for children with bronchiolitis. Future research should examine whether more evolved HFNC protocols that use higher flow rates, more generous ICU transfer criteria, and more rapid weaning criteria can reduce ICU utilization.
Acknowledgments
We thank Dr Vineeta Mittal (University of Texas Southwestern Medical Center) for providing feedback regarding the manuscript.
Children hospitalized for bronchiolitis frequently require admission to the intensive care unit (ICU), with estimates as high as 18%1,2 and 35%3 in two prospective, multicenter studies. The indication for ICU admission is nearly always a need for advanced respiratory support, which historically consisted of continuous or bilevel positive airway pressure (CPAP and BiPAP, respectively) or mechanical ventilation. High-flow nasal cannula (HFNC) is a recent addition to the respiratory support armamentarium, delivering heated and humidified oxygen at rates of up to 60 L/min and allowing for clinicians to titrate both flow rate and fraction of inspired oxygen (FiO2).4
Several studies have demonstrated that HFNC is capable of decreasing a child’s work of breathing,5-8 and it has the potential advantage of being better tolerated than other forms of advanced respiratory support.9,10 These case-series physiologic studies informed early ward-based HFNC protocols for bronchiolitis, which were adopted to decrease ICU utilization. Since then, single center observational studies examining the association between ward-based HFNC protocols and subsequent ICU utilization have come to discordant conclusions.11-14 Studying the effect of employing HFNC outside of the ICU is challenging in the context of a randomized, controlled trial (RCT) because it is difficult to blind healthcare providers to the intervention and because crossover from the control group to HFNC is frequent. Two unblinded RCTs published in 2017 and 2018 found that children randomized to conventional nasal cannula were frequently escalated to HFNC (flow rates of 1-2 L/kg per minute), but neither trial found a difference in ICU admission.15,16 Sample sizes substantially larger than those present in currently published or registered RCTs would be required to evaluate the impact of ward-based HFNC protocols on the outcome that inspired the protocols in the first place, namely ICU utilization.17
Children’s hospitals have adopted ward-based HFNC protocols at different time points over the last decade, which allows for a natural experiment—a promising alternative study design that avoids the challenges of blinding, crossover, and modest sample sizes. In order to have sufficient postadoption data for analyses, the present study is limited to ward-based HFNC protocols adopted prior to 2016, which we have termed “early” ward-based HFNC protocols. Among children with bronchiolitis, our objective was to measure the association between hospital-level adoption of a ward-based HFNC protocol and subsequent ICU utilization, using a multicenter network of children’s hospitals.
METHODS
We conducted a multicenter retrospective cohort study using the Pediatric Health Information System (PHIS) database. The PHIS database is operated by the Children’s Hospital Association (Lenexa, Kansas) and provides deidentified patient-level information for children who receive hospital care at 55 US children’s hospitals. Available data elements include patient demographic data, discharge diagnosis and procedure codes, and detailed billing information, such as laboratory, imaging, pharmacy, and supply charges. At the patient level, the use of HFNC vs standard oxygen therapy circuits cannot be discriminated.
Exposure
The study exposure was a hospital’s first ward-based HFNC protocol, with adoption measured at the hospital level at each PHIS site via direct communication with leaders in hospital medicine. In most cases, first contact was made with the pediatric hospital medicine division chief or fellowship program director, who then, if necessary, connected study investigators to local HFNC champions aware of site-specific historical HFNC protocol details. Contact with a hospital was made only if the hospital had contributed at least 6 consecutive years of data to PHIS. Hospitals were classified as “adopting” hospitals if their HFNC protocol met all of the following criteria: (a) allows initiation of HFNC outside of the ICU (on the floor or in the ED), (b) allows continued care outside of the ICU (on the floor), (c) not limited to a small unit like an intermediate care unit, and (d) adopted during a specific, known respiratory season. Hospitals for which ward-based HFNC protocols were adopted but did not meet these criteria were excluded from further analysis. Our intent was to identify large scale, programmatic protocol launches and exclude hospitals with exceptions that might preclude a sizable portion of our cohort from being eligible for the protocol. Hospitals for which inpatient use of HFNC remains limited to the ICU were defined as “nonadopting” hospitals. Respondents at adopting hospitals were asked to share details about their protocol, including patient eligibility criteria and maximum HFNC rates of flow permitted outside of the ICU.
Patient Characteristics
Patients aged 3 to 24 months who were hospitalized at adopting and nonadopting hospitals were included if an International Classification of Diseases, Ninth Revision (ICD-9) discharge diagnosis code for bronchiolitis (466.XX) was present in any position (not limited to a primary diagnosis). The lower age limit of 3 months was chosen to match the most restrictive age eligibility criteria of provided HFNC protocols (Appendix 1). A crosswalk available from the Centers for Medicare & Medicaid Services18 was used to convert ICD-10 diagnosis and procedure codes from recent years to ICD-9 diagnosis and procedure codes. Patients were excluded if their encounter contained a diagnosis or procedure code signifying a complex chronic condition,19 if their hospitalization involved care in the neonatal ICU, or if their admission date occurred outside of the respiratory season. Respiratory season was defined as November 1 through April 30.
Outcomes
Outcomes were measured during three respiratory seasons leading up to adoption and during three respiratory seasons after adoption. The primary outcome was ICU utilization, including the proportion of patients admitted to the ICU and ICU length of stay, expressed as ICU days per 100 patients. Secondary outcomes included mean total length of stay and the proportion of patients who received mechanical ventilation. Lengths of stay were measured in days, the most granular unit of time provided in PHIS over the entire study period. As such, partial days of care are rounded up to 1 full day. A previously published strict definition for mechanical ventilation that limits false positives was used, requiring that patients have a procedure or supply code for mechanical ventilation and a pharmacy charge for a neuromuscular blocking agent.20
Primary Analysis
The primary analysis was restricted to adopting hospitals. An interrupted time series approach was used to measure two possible types of change associated with HFNC protocol adoption: an immediate intervention effect and a change in the slope of an outcome.21 The immediate intervention effect represents the change in the level of the outcome that occurs in the period immediately following the introduction of the protocol. The change in slope is the extent to which the outcome changes on a per season basis, attributable to the protocol. Interrupted time series estimates were adjusted for patient age, gender, race, ethnicity, and insurance type; linear regression was used for continuous outcomes and logistic regression for dichotomous outcomes. An ordinary least squares time series model was used to adjust for autocorrelation and Newey-West standard errors were employed.22 Analyses were performed using STATA version 14 (Stata-Corp, College Station, Texas).
Supplementary Analyses
Two preplanned supplementary analyses were conducted. Supplementary analysis 1 was identical to the primary analysis, with the exception that the first season after adoption was censored. The rationale for censoring the first adoption season was to account for a potential learning effect and/or delayed start to full protocol implementation. Supplementary analysis 2 used the nonadopting hospitals as a control group and subtracted the effects measured from an interrupted time series analysis among nonadopting hospitals from the effects measured among adopting hospitals. The rationale for this approach was to control for unmeasured secular (eg, availability of ICU beds) and temporal (eg, severity of a given bronchiolitis season) factors that may have coincidentally occurred with HFNC adoption seasons. The only modification to the interrupted time series approach for supplementary analysis 2 was to provide the nonadopting hospitals with an artificial interruption point because nonadopting hospitals, by definition, did not have an adoption season that could be used in an interrupted time series approach. The interruption point for nonadopting hospitals was set at the median adoption season for adopting hospitals.
RESULTS
Exposure
Leaders at 44 hospitals were contacted regarding their hospital’s use of HFNC outside of the ICU (Figure 1). Responses were obtained for 41 hospitals (93% response rate), 18 of which were classified as nonadopting hospitals. Of the 23 hospitals where the presence of ward-based HFNC protocols were reported, 12 met inclusion criteria and were classified as adopting hospitals. HFNC protocols were adopted at these hospitals in a staggered fashion between the 2010-2011 and 2015-2016 respiratory seasons (Figure 2). The median adoption season was the 2013-14 respiratory season.
Nine adopting hospitals were able to provide details about their first HFNC protocols (Appendix 1). No two protocols were identical, but they shared many similarities. Minimum age requirements ranged from birth to a few months of age. Exclusion criteria were particularly variable, with a history of chronic lung disease or apnea being the most common criteria. Maximum allowed rates of flow ranged from 4 to 10 liters per minute. Criteria for transfer to the ICU were consistently based on an elevated FiO2 and duration of HFNC exposure.
Patient Characteristics
A total of 32,809 bronchiolitis encounters occurred at adopting hospitals during qualifying respiratory seasons, of which 6,556 (20%) involved patients with a complex chronic condition and were excluded. Of the 26,253 included bronchiolitis encounters, 12,495 encounters occurred prior to ward-based HFNC protocol adoption and 13,758 encounters occurred after adoption. The median age of patients was 8 months (interquartile range, 5-14 months). Most patients were on government insurance (64%), male (58%), of white (56%) or black (18%) race, and of non-Hispanic ethnicity (72%). Pre- and postadoption patient demographics were similar (Appendix 2).
Primary Analysis
Shifts in the level of ICU use and ICU length of stay were observed at the time of adoption of a ward-based HFNC protocol (Figure 3). Specifically, ward-based HFNC protocol adoption was associated with an immediate 3.1% absolute increase (95% CI, 2.8%-3.4%) in the proportion of patients admitted to the ICU and a 9.1 days per 100 patients increase (95% CI, 5.1-13.2) in ICU length of stay (Table). The slope of ICU admissions per season was increasing after HFNC protocol adoption (1.0% increase per season; 95% CI, 0.8%-1.1%). When examined at the individual-hospital level (Appendix 3), seven hospitals were found to have significant increases in ICU admissions (immediate intervention effect or change in slope) after adoption, and one hospital was found to have a significant decrease in ICU admissions (change in slope only). Neither immediate intervention effects nor changes in the slopes of total length of stay and mechanical ventilation were observed, with mean total length of stay approximately 3 days and just over 1% of patients receiving mechanical ventilation (Figure 3).
Supplementary Analyses
Supplementary analyses were largely consistent with the primary analysis. Associations with increased ICU utilization were again observed, although the immediate change in ICU length of stay for supplementary analysis 1 was not significant and the slope for ICU length of stay in supplementary analysis 2 was down trending (Table). Changes in total length of stay and mechanical ventilation were not observed in either supplementary analysis, with the lone exception being an increase in the proportion of patients receiving mechanical ventilation per season (increase in slope) in supplementary analysis 1.
DISCUSSION
This is the largest multicenter study to date evaluating ICU utilization after adoption of a ward-based HFNC protocol for patients with bronchiolitis. While a principal goal of allowing HFNC use outside of the ICU is to reduce the time that patients with bronchiolitis spend in the ICU, we found that early protocols were, paradoxically, associated with increased ICU utilization. Ward-based HFNC protocols were not associated with changes in hospital length of stay or need for mechanical ventilation. Our findings are particularly relevant given that the majority of children’s hospitals in our sample have adopted ward-based HFNC protocols to care for patients with bronchiolitis.
The increase in ICU utilization measured in our study is a novel finding, seemingly in contradiction to existing literature. Early pilot studies inspired hope that employing HFNC on the general ward might prevent a portion of children from needing ICU care.11,12 Subsequent larger observational studies did not demonstrate decreases in ICU utilization after adoption of ward-based HFNC protocols.13,14 The two RCTs comparing low-flow and high-flow nasal cannula use outside of the ICU did not measure a statistically significant effect on ICU utilization, an exploratory outcome in both trials.15,16 However, the reported point estimates for absolute differences in ICU admission were 2% to 3% higher among patients randomized to HFNC, which is consistent with the 2% to 4% increase in ICU admission measured in the present study.
What might explain this surprising finding? While our observational study cannot speak to mechanism, the protocol details examined in the present study suggest that initial adoption of a ward-based HFNC protocol is often coupled with specific ICU transfer criteria that were unlikely in place prior to protocol initiation. For example, most protocols recommended consideration of ICU transfer for elevated FiO2 or prolonged duration of HFNC. Transfer to the ICU for prolonged HFNC duration is only possible in the setting of a ward-based HFNC protocol and transfer for elevated FiO2 was probably unnecessary prior to protocol adoption given that low-flow nasal cannula generally delivers 100% FiO2. It is also possible that with HFNC comes a perception of increased acuity. For example, medical providers may see patients on HFNC as sicker than patients with the same amount of work of breathing but off HFNC, which makes providers more likely to seek ICU admission for patients on HFNC. The combination of unchanged total length of stay and increased ICU utilization suggests that early ward-based HFNC protocols were an ineffective instrument to improve hospital bed availability during the peak census times that often occur in bronchiolitis season.
The large sample size afforded our study by its multicenter, retrospective design also allowed for a meaningful assessment of the association between a ward-based HFNC protocol and the need for mechanical ventilation. Early indications suggested a lack of substantial association between HFNC use outside of the ICU and rates of mechanical ventilation, but prior studies were limited by small numbers of patients receiving mechanical ventilation (<30 patients in each study).13,14,16 The present study, in which 783 patients received mechanical ventilation, supports the lack of association between early ward-based HFNC protocols and the need for mechanical ventilation. It should be noted that other studies have measured decreases in mechanical ventilation in association with ICU-based HFNC use.23-26 In addition to examining HFNC use in a different clinical context, decreases in mechanical ventilation measured after HFNC implementation in the ICU could be explained by preexisting practice trends to limit invasive ventilation and/or selection bias resulting from an increase in less severely ill patients being admitted to the ICU over time. The interrupted time series approach and the staggered adoption of HFNC protocols make the present study less susceptible to biases from preexisting trends and the inclusion of patients cared for both on the ward and within the ICU reduces selection bias.
Our study has several important limitations. First, all hospitals included in the analysis were US children’s hospitals and these findings may not generalize to other practice environments, including community hospitals and other countries. Second, our cohort and outcomes were defined using administrative billing data, which have been incompletely validated, making some degree of misclassification likely. Third, we measured HFNC exposure at the hospital level, but could not examine the extent to which individual patients were exposed to HFNC because such data are not present in PHIS. Even if we had access to patient-level HFNC exposure data, we would have still compared outcomes among all patients with bronchiolitis (not just those who received HFNC), to avoid selection bias. However, knowing HFNC exposure status at the patient level would have allowed for weighting of the effects measured at each hospital according to the extent of HFNC exposure. Fourth, there are likely other, unmeasured secular and temporal factors that could affect study outcomes. To some degree, the interrupted time series approach, observed staggered adoption of protocols, and nonadopting hospital supplementary analysis mitigate this risk of bias. Fifth, while the pre- and postadoption populations appeared demographically similar, it is possible that the populations might have differed by other unmeasured factors. Finally, early ward-based HFNC protocols have likely undergone iterative changes since adoption. We compared pre- and postadoption outcome slopes and censored the first adoption season in a supplementary analysis to attempt to account for this potential limitation.
In conclusion, our findings suggest that initial implementation of ward-based HFNC protocols were not successful at reducing ICU utilization for children with bronchiolitis. Future research should examine whether more evolved HFNC protocols that use higher flow rates, more generous ICU transfer criteria, and more rapid weaning criteria can reduce ICU utilization.
Acknowledgments
We thank Dr Vineeta Mittal (University of Texas Southwestern Medical Center) for providing feedback regarding the manuscript.
1. Mansbach JM, Piedra PA, Teach SJ, et al. Prospective multicenter study of viral etiology and hospital length of stay in children with severe bronchiolitis. Arch Pediatr Adolesc Med. 2012;166(8):700-706. https://doi.org/10.1001/archpediatrics.2011.1669.
2. Hasegawa K, Pate BM, Mansbach JM, et al. Risk factors for requiring intensive care among children admitted to ward with bronchiolitis. Acad Pediatr. 2015;15(1):77-81. https://doi.org/10.1016/j.acap.2014.06.008.
3. Schroeder AR, Destino LA, Brooks R, Wang CJ, Coon ER. Outcomes of follow-up visits after bronchiolitis hospitalizations. JAMA Pediatr. 2018;172(3):296-297. https://doi.org/10.1001/jamapediatrics.2017.4002.
4. Drake MG. High-flow nasal cannula oxygen in adults: an evidence-based assessment. Ann Am Thorac Soc. 2018;15(2):145-155. https://doi.org/10.1513/AnnalsATS.201707-548FR.
5. Rubin S, Ghuman A, Deakers T, Khemani R, Ross P, Newth CJ. Effort of breathing in children receiving high-flow nasal cannula. Pediatr Crit Care Med. 2014;15(1):1-6. https://doi.org/10.1097/PCC.0000000000000011.
6. Hough JL, Pham TM, Schibler A. Physiologic effect of high-flow nasal cannula in infants with bronchiolitis. Pediatr Crit Care Med. 2014;15(5):e214-e219. https://doi.org/10.1097/PCC.0000000000000112.
7. Pham TM, O’Malley L, Mayfield S, Martin S, Schibler A. The effect of high flow nasal cannula therapy on the work of breathing in infants with bronchiolitis. Pediatr Pulmonol. 2015;50(7):713-720. https://doi.org/10.1002/ppul.23060.
8. Weiler T, Kamerkar A, Hotz J, Ross PA, Newth CJL, Khemani RG. The relationship between high flow nasal cannula flow rate and effort of breathing in children. J Pediatr. 2017;189:66-71.e63. https://doi.org/10.1016/j.jpeds.2017.06.006.
9. Mayfield S, Jauncey-Cooke J, Hough JL, Schibler A, Gibbons K, Bogossian F. High-flow nasal cannula therapy for respiratory support in children. Cochrane Database Syst Rev. 2014(3):CD009850. https://doi.org/10.1002/14651858.CD009850.pub2.
10. Roca O, Riera J, Torres F, Masclans JR. High-flow oxygen therapy in acute respiratory failure. Respir Care. 2010;55(4):408-413.
11. Kallappa C, Hufton M, Millen G, Ninan TK. Use of high flow nasal cannula oxygen (HFNCO) in infants with bronchiolitis on a paediatric ward: a 3-year experience. Arch Dis Child. 2014;99(8):790-791. https://doi.org/10.1136/archdischild-2014-306637.
12. Mayfield S, Bogossian F, O’Malley L, Schibler A. High-flow nasal cannula oxygen therapy for infants with bronchiolitis: pilot study. J Paediatr Child Health. 2014;50(5):373-378. https://doi.org/10.1111/jpc.12509.
13. Riese J, Porter T, Fierce J, Riese A, Richardson T, Alverson BK. Clinical outcomes of bronchiolitis after implementation of a general ward high flow nasal cannula guideline. Hosp Pediatr. 2017;7(4):197-203. https://doi.org/10.1542/hpeds.2016-0195.
14. Mace AO, Gibbons J, Schultz A, Knight G, Martin AC. Humidified high-flow nasal cannula oxygen for bronchiolitis: should we go with the flow? Arch Dis Child. 2018;103(3):303. https://doi.org/10.1136/archdischild-2017-313950.
15. Kepreotes E, Whitehead B, Attia J, et al. High-flow warm humidified oxygen versus standard low-flow nasal cannula oxygen for moderate bronchiolitis (HFWHO RCT): an open, phase 4, randomised controlled trial. Lancet. 2017;389(10072):930-939. https://doi.org/10.1016/S0140-6736(17)30061-2.
16. Franklin D, Babl FE, Schlapbach LJ, et al. A randomized trial of high-flow oxygen therapy in infants with bronchiolitis. N Engl J Med. 2018;378(12):1121-1131. https://doi.org/10.1056/NEJMoa1714855.
17. Coon ER, Mittal V, Brady PW. High flow nasal cannula-just expensive paracetamol? Lancet Child Adolesc Health. 2019;3(9):593-595. https://doi.org/10.1016/S2352-4642(19)30235-4.
18. Roth J. CMS’ ICD-9-CM to and from ICD-10-CM and ICD-10-PCS Crosswalk or General Equivalence Mappings. 2012. http://www.nber.org/data/icd9-icd-10-cm-and-pcs-crosswalk-general-equivalence-mapping.html. Accessed November 19, 2016.
19. Feudtner C, Feinstein JA, Zhong W, Hall M, Dai D. Pediatric complex chronic conditions classification system version 2: updated for ICD-10 and complex medical technology dependence and transplantation. BMC Pediatr. 2014;14:199. https://doi.org/10.1186/1471-2431-14-199.
20. Shein SL, Slain K, Wilson-Costello D, McKee B, Rotta AT. Temporal changes in prescription of neuropharmacologic drugs and utilization of resources related to neurologic morbidity in mechanically ventilated children with bronchiolitis. Pediatr Crit Care Med. 2017;18(12):e606-e614. https://doi.org/10.1097/PCC.0000000000001351.
21. Penfold RB, Zhang F. Use of interrupted time series analysis in evaluating health care quality improvements. Acad Pediatr. 2013;13(6 Suppl):S38-S44. https://doi.org/10.1016/j.acap.2013.08.002.
22. Newey WK, West KD. A simple, positive semi-definite, heteroskedasticity and autocorrelation consistent covariance matrix. Econometrica. 1987;55(3):703-708.
23. McKiernan C, Chua LC, Visintainer PF, Allen H. High flow nasal cannulae therapy in infants with bronchiolitis. J Pediatr. 2010;156(4):634-638. https://doi.org/10.1016/j.jpeds.2009.10.039.
24. Schibler A, Pham TM, Dunster KR, et al. Reduced intubation rates for infants after introduction of high-flow nasal prong oxygen delivery. Intensive Care Med. 2011;37(5):847-852. https://doi.org/10.1007/s00134-011-2177-5.
25. Kawaguchi A, Yasui Y, deCaen A, Garros D. The clinical impact of heated humidified high-flow nasal cannula on pediatric respiratory distress. Pediatr Crit Care Med. 2017;18(2):112-119. https://doi.org/10.1097/PCC.0000000000000985.
26. Schlapbach LJ, Straney L, Gelbart B, et al. Burden of disease and change in practice in critically ill infants with bronchiolitis. Eur Respir J. 2017;49(6):1601648. https://doi.org/10.1183/13993003.01648-2016.
1. Mansbach JM, Piedra PA, Teach SJ, et al. Prospective multicenter study of viral etiology and hospital length of stay in children with severe bronchiolitis. Arch Pediatr Adolesc Med. 2012;166(8):700-706. https://doi.org/10.1001/archpediatrics.2011.1669.
2. Hasegawa K, Pate BM, Mansbach JM, et al. Risk factors for requiring intensive care among children admitted to ward with bronchiolitis. Acad Pediatr. 2015;15(1):77-81. https://doi.org/10.1016/j.acap.2014.06.008.
3. Schroeder AR, Destino LA, Brooks R, Wang CJ, Coon ER. Outcomes of follow-up visits after bronchiolitis hospitalizations. JAMA Pediatr. 2018;172(3):296-297. https://doi.org/10.1001/jamapediatrics.2017.4002.
4. Drake MG. High-flow nasal cannula oxygen in adults: an evidence-based assessment. Ann Am Thorac Soc. 2018;15(2):145-155. https://doi.org/10.1513/AnnalsATS.201707-548FR.
5. Rubin S, Ghuman A, Deakers T, Khemani R, Ross P, Newth CJ. Effort of breathing in children receiving high-flow nasal cannula. Pediatr Crit Care Med. 2014;15(1):1-6. https://doi.org/10.1097/PCC.0000000000000011.
6. Hough JL, Pham TM, Schibler A. Physiologic effect of high-flow nasal cannula in infants with bronchiolitis. Pediatr Crit Care Med. 2014;15(5):e214-e219. https://doi.org/10.1097/PCC.0000000000000112.
7. Pham TM, O’Malley L, Mayfield S, Martin S, Schibler A. The effect of high flow nasal cannula therapy on the work of breathing in infants with bronchiolitis. Pediatr Pulmonol. 2015;50(7):713-720. https://doi.org/10.1002/ppul.23060.
8. Weiler T, Kamerkar A, Hotz J, Ross PA, Newth CJL, Khemani RG. The relationship between high flow nasal cannula flow rate and effort of breathing in children. J Pediatr. 2017;189:66-71.e63. https://doi.org/10.1016/j.jpeds.2017.06.006.
9. Mayfield S, Jauncey-Cooke J, Hough JL, Schibler A, Gibbons K, Bogossian F. High-flow nasal cannula therapy for respiratory support in children. Cochrane Database Syst Rev. 2014(3):CD009850. https://doi.org/10.1002/14651858.CD009850.pub2.
10. Roca O, Riera J, Torres F, Masclans JR. High-flow oxygen therapy in acute respiratory failure. Respir Care. 2010;55(4):408-413.
11. Kallappa C, Hufton M, Millen G, Ninan TK. Use of high flow nasal cannula oxygen (HFNCO) in infants with bronchiolitis on a paediatric ward: a 3-year experience. Arch Dis Child. 2014;99(8):790-791. https://doi.org/10.1136/archdischild-2014-306637.
12. Mayfield S, Bogossian F, O’Malley L, Schibler A. High-flow nasal cannula oxygen therapy for infants with bronchiolitis: pilot study. J Paediatr Child Health. 2014;50(5):373-378. https://doi.org/10.1111/jpc.12509.
13. Riese J, Porter T, Fierce J, Riese A, Richardson T, Alverson BK. Clinical outcomes of bronchiolitis after implementation of a general ward high flow nasal cannula guideline. Hosp Pediatr. 2017;7(4):197-203. https://doi.org/10.1542/hpeds.2016-0195.
14. Mace AO, Gibbons J, Schultz A, Knight G, Martin AC. Humidified high-flow nasal cannula oxygen for bronchiolitis: should we go with the flow? Arch Dis Child. 2018;103(3):303. https://doi.org/10.1136/archdischild-2017-313950.
15. Kepreotes E, Whitehead B, Attia J, et al. High-flow warm humidified oxygen versus standard low-flow nasal cannula oxygen for moderate bronchiolitis (HFWHO RCT): an open, phase 4, randomised controlled trial. Lancet. 2017;389(10072):930-939. https://doi.org/10.1016/S0140-6736(17)30061-2.
16. Franklin D, Babl FE, Schlapbach LJ, et al. A randomized trial of high-flow oxygen therapy in infants with bronchiolitis. N Engl J Med. 2018;378(12):1121-1131. https://doi.org/10.1056/NEJMoa1714855.
17. Coon ER, Mittal V, Brady PW. High flow nasal cannula-just expensive paracetamol? Lancet Child Adolesc Health. 2019;3(9):593-595. https://doi.org/10.1016/S2352-4642(19)30235-4.
18. Roth J. CMS’ ICD-9-CM to and from ICD-10-CM and ICD-10-PCS Crosswalk or General Equivalence Mappings. 2012. http://www.nber.org/data/icd9-icd-10-cm-and-pcs-crosswalk-general-equivalence-mapping.html. Accessed November 19, 2016.
19. Feudtner C, Feinstein JA, Zhong W, Hall M, Dai D. Pediatric complex chronic conditions classification system version 2: updated for ICD-10 and complex medical technology dependence and transplantation. BMC Pediatr. 2014;14:199. https://doi.org/10.1186/1471-2431-14-199.
20. Shein SL, Slain K, Wilson-Costello D, McKee B, Rotta AT. Temporal changes in prescription of neuropharmacologic drugs and utilization of resources related to neurologic morbidity in mechanically ventilated children with bronchiolitis. Pediatr Crit Care Med. 2017;18(12):e606-e614. https://doi.org/10.1097/PCC.0000000000001351.
21. Penfold RB, Zhang F. Use of interrupted time series analysis in evaluating health care quality improvements. Acad Pediatr. 2013;13(6 Suppl):S38-S44. https://doi.org/10.1016/j.acap.2013.08.002.
22. Newey WK, West KD. A simple, positive semi-definite, heteroskedasticity and autocorrelation consistent covariance matrix. Econometrica. 1987;55(3):703-708.
23. McKiernan C, Chua LC, Visintainer PF, Allen H. High flow nasal cannulae therapy in infants with bronchiolitis. J Pediatr. 2010;156(4):634-638. https://doi.org/10.1016/j.jpeds.2009.10.039.
24. Schibler A, Pham TM, Dunster KR, et al. Reduced intubation rates for infants after introduction of high-flow nasal prong oxygen delivery. Intensive Care Med. 2011;37(5):847-852. https://doi.org/10.1007/s00134-011-2177-5.
25. Kawaguchi A, Yasui Y, deCaen A, Garros D. The clinical impact of heated humidified high-flow nasal cannula on pediatric respiratory distress. Pediatr Crit Care Med. 2017;18(2):112-119. https://doi.org/10.1097/PCC.0000000000000985.
26. Schlapbach LJ, Straney L, Gelbart B, et al. Burden of disease and change in practice in critically ill infants with bronchiolitis. Eur Respir J. 2017;49(6):1601648. https://doi.org/10.1183/13993003.01648-2016.
© 2020 Society of Hospital Medicine