LayerRx Mapping ID
268
Slot System
Featured Buckets
Featured Buckets Admin

A COVID-19 Clinical Management Committee to Standardize Care in a 2-Hospital System

Article Type
Changed
Wed, 02/09/2022 - 12:46
Display Headline
A COVID-19 Clinical Management Committee to Standardize Care in a 2-Hospital System

From the Department of Medicine (Drs. Meisenberg, Muganlinskaya, Sharma, Amjadi, Arnold, Barnes, Clance, Khalil, Miller, Mooradian, O’Connell, Patel, Press, Samaras, Shanmugam, Tavadze, and Thompson), Department of Pharmacy (Drs. Jiang, Jarawan, Sheth, and Trinh), Department of Nursing (Dr. Ohnmacht), and Department of Women and Children’s Services (Dr. Raji), Luminis Health, Annapolis, MD, and Lanham, MD.

Objective: The COVID-19 pandemic has been a challenge for hospital medical staffs worldwide due to high volumes of patients acutely ill with novel syndromes and prevailing uncertainty regarding optimum supportive and therapeutic interventions. Additionally, the response to this crisis was driven by a plethora of nontraditional information sources, such as email chains, websites, non–peer-reviewed preprints, and press releases. Care patterns became idiosyncratic and often incorporated unproven interventions driven by these nontraditional information sources. This report evaluates the efforts of a health system to create and empower a multidisciplinary committee to develop, implement, and monitor evidence-based, standardized protocols for patients with COVID-19.

Methods: This report describes the composition of the committee, its scope, and its important interactions with the health system pharmacy and therapeutics committee, research teams, and other work groups planning other aspects of COVID-19 management. It illustrates how the committee was used to demonstrate for trainees the process and value of critically examining evidence, even in a chaotic environment.

Results: Data show successful interventions in reducing excessive ordering of certain laboratory tests, reduction of nonrecommended therapies, and rapid uptake of evidence-based or guidelines-supported interventions.

Conclusions: A multidisciplinary committee dedicated solely to planning, implementing, and monitoring standard approaches that eventually became evidence-based decision-making led to an improved focus on treatment options and outcomes for COVID-19 patients. Data presented illustrate the attainable success that is both adaptable and suitable for similar emergencies in the future.

Keywords: COVID-19; clinical management; pharmacy and therapeutics; treatment; therapy.

The COVID-19 pandemic has spread to nearly all countries, carrying with it high morbidity, mortality, and severe impacts on both well-developed and less-well-developed health systems. Media reports of chaos within overwhelmed hospitals have been prominent.1,2 As of January 5, 2022, SARS-CoV-2 has infected more than 295 million people globally and directly caused the death of more than 5.4 million,3 though this number is likely an undercount even in countries with well-developed mortality tracking.4

Throughout the COVID-19 pandemic, hospital-based medical teams have been confronted with a flood of severely ill patients with novel syndromes. Initially, there were no standards for therapy or supportive care except for treatments borrowed from similar syndromes. In the setting of high volumes, high acuity, and public dismay, it is unsurprising that the usual deliberative methods for weighing evidence and initiating interventions were often pushed aside in favor of the solace of active intervention.5 In this milieu of limited evidence, there was a lamentable, if understandable, tendency to seek guidance from “nontraditional” sources,6 including email chains from colleagues, hospital websites, non–peer-reviewed manuscripts, advanced publication by medical journals,7 and nonscientific media presentations. In many localities, practitioners responded in idiosyncratic ways. For example, findings of high cytokine levels in COVID-19,8 along with reports of in-vitro antiviral activity with drugs like hydroxychloroquine against both SARS9 and SARS-CoV-2,10 drove laboratory test ordering and therapeutic interventions, respectively, carving shortcuts into the traditional clinical trial–dependent standards. Clinical trial results eventually emerged.11COVID-19 created a clinical dilemma for hospital medical staffs in terms of how to organize, standardize, and rapidly adapt to a flood of new information. In this report, we describe how 1 health system responded to these challenges by forming a COVID-19 Clinical Management Committee (CCMC) and empowering this interdisciplinary team to review evidence, create and adjust order sets, educate practitioners, oversee care, and collaborate across teams addressing other aspects of the COVID-19 response.

 

 

Program Overview

Health System Description

Luminis Health is a health system with 2 acute care hospitals that was formed in 2019 just before the start of the pandemic. Anne Arundel Medical Center (hospital A) is a 385-bed teaching hospital in Annapolis, MD. It has more than 23 000 discharges annually. Patients with COVID-19 were cared for by either an internal medicine teaching service or nonteaching hospitalist services on cohorted nursing units. Doctor’s Community Medical Center, in Lanham, MD (hospital B), is a 206-bed acute care hospital with more than 10 350 annual discharges. COVID-19 patients were cared for by hospitalist groups, initially in noncohorted units with transition to cohorted nursing units after a few months. The medical staffs are generally distinct, with different leadership structures, though the Luminis Health Department of Medicine has oversight responsibilities at both hospitals. More than 47 physicians attended COVID-19 patients at hospital A (with medical residents) and 30 individual physicians at hospital B, respectively, including intensivists. The nursing and pharmacy staffs are distinct, but there is a shared oversight Pharmacy and Therapeutics (P&T) Committee.

The 2 hospitals had distinct electronic medical records (EMR) until January 2021, when hospital B adopted the same EMR as hospital A (Epic).

Mission and Formation of CCMC

In order to coordinate the therapeutic approach across the health system, it was important for the CCMC to be designated by the health system P&T committee as an official subcommittee so that decisions on restrictions of medications and/or new or revised order sets could be rapidly initiated across the system without waiting for the subsequent P&T meetings. The full committee retained oversight of the CCMC. Some P&T members were also on the CCMC.

The committee reviewed new reports in medical journals and prepublication servers and consulted recommendations of professional societies, such as the National Institutes of Health (NIH) COVID-19 guidelines, Infectious Diseases Society of America, Society of Critical Care Medicine, and US Food and Drug Administration (FDA) Emergency Use Authorizations (EUA), among other sources.

Composition of the CCMC

Physician leaders from both hospitals in the following specialties were solicited for participation: critical care, epidemiology, hospital medicine (internal medicine), emergency medicine, infectious diseases, nephrology, women and children’s services, and medical informatics. Specialists in other areas, such as hematology, were invited for topic-specific discussions. Hospital pharmacists with different specialties and nursing leadership were essential contributors. The committee members were expected to use various communication channels to inform frontline clinicians of new care standards and the existence of new order sets, which were embedded in the EMR.

Clinical Research

An important connection for the CCMC was with theCOVID-19 clinical research team. Three members of the research team were also members of the CCMC. All new study proposals for therapeutics were discussed with the CCMC as they were being considered by the research team. In this way, feedback on the feasibility and acceptance of new study opportunities could be discussed with the CCMC. Occasionally, CCMC decisions impacted clinical research accrual strategies. For example, new data from randomized trials about tocilizumab1,2 demonstrated benefits in some subsets of patients and resulted in a recommendation for use by the NIH guideline committee in these populations.1 The CCMC quickly adopted this recommendation, which required a reprioritization of clinical research enrollment for studies testing other immune-modulating agents. This important dialogue was mediated within the CCMC.

Guideline Distribution, Reinforcement, and Platform for Feedback

New guidelines were disseminated to clinicians via daily brief patient huddles held on COVID units, with participation by nursing and pharmacy, and by weekly meetings with hospitalist leaders and frontline hospital physicians. Order sets and guidelines were maintained on the intranet. Adherence was reinforced by unit-based and central pharmacists. Order sets, including admission order sets, could be created only by designated informatics personnel, thus enforcing standardization. Feedback on the utility of the order sets was obtained during the weekly meetings or huddles, as described above. To ensure a sense of transparency, physicians who had interest in commenting on a particular therapy, or who wished to discuss a particular manuscript, news article, or website, were invited to attend CCMC meetings.

Scope of CCMC

In order to be effective and timely, we limited the scope of our work to the report to the inpatient therapeutic environment, allowing other committees to work on other aspects of the pandemic response. In addition to issuing guidance and creating order sets to direct clinical practice, the CCMC also monitored COVID-19 therapeutic shortages15,16 and advised on prioritization of such treatments as convalescent plasma, remdesivir (prioritization and duration of therapy, 5 vs 10 days), baricitinib, and tocilizumab, depending upon the location of the patient (critical care or not). The CCMC was not involved in the management of non–COVID-19 shortages brought about by supply chain deficiencies.

Table 1 shows some aspects of the health system pandemic-response planning and the committee workforce that undertook that work. Though many items were out of scope for the CCMC, members of the CCMC did participate in the planning work of these other committees and therefore stayed connected to this complementary work.

A Teaching Opportunity About Making Thoughtful Choices

Another important feature of the CCMC was the contributions of residents from both pharmacy and internal medicine. The purpose and operations of the committee were recognized as an opportunity to involve learners in a curriculum based on Kern’s 6-step approach.17 Though the problem identification and general needs assessment were easily defined, the targeted needs assessment, extracted from individual and group interviews with learners and the committee members, pointed at the need to learn how to assess and analyze a rapidly growing body of literature on several relevant clinical aspects of SARS-CoV-2 and COVID-19. To achieve goals and objectives, residents were assigned to present current literature on a particular intervention during a committee meeting, specifically commenting on the merit or deficiencies of the study design, the strength of the data, and applicability to the local context with a recommendation. Prior to the presentations, the residents worked with faculty to identify the best studies or systematic analyses with potential to alter current practices. We thus used the CCMC process as a teaching tool about evidence-based medicine and the dilemma of clinical equipoise. This was imperative, since trainees thrust into the COVID-19 response have often keenly observed a movement away from deliberative decision-making.18 Indeed, including residents in the process of deliberative responses to COVID-19 addresses a recent call to adjust medical education during COVID-19 to “adapt curriculum to current issues in real time.”19

Interventions and Therapies Considered

Table 2 shows the topics reviewed by the CCMC. By the time of the first meeting, nonstandardization of care was already a source of concern for clinicians. Dialogue often continued outside of the formal meetings. Many topics were considered more than once as new guidance developed, changes to EUAs occurred, and new data or new publicity arose.

 

 

Methods

The Human Protections Administrator determined that this work constituted “quality improvement, and not research” and was therefore exempt from institutional review board review.

Quantitative Analysis

All admitted patients from March 10, 2020, through April 20, 2021, were considered in the quantitative aspects of this report except as noted. Patients diagnosed with COVID-19 were identified by searching our internal data base using diagnostic codes. Patient admissions with the following diagnostic codes were included (prior to April 1, 2020): J12.89, J20.8, J40, J22, J98.8, J80, each with the additional code of B97.29. After April 1, 2020, the guideline for coding COVID-19 was U07.1.

Descriptive statistics were used to measure utilization rates of certain medications and laboratory tests of interest over time. These data were adjusted for number of unique admissions. In a few cases, not all data elements were available from both hospitals due to differences in the EMR.

Case fatality rate was calculated based upon whether the patient died or was admitted to inpatient hospice as a result of COVID-19. Four patients transferred out of hospital A and 18 transferred out of hospital B were censored from case-fatality-rate determination.

Figure 1 shows the number of admissions for each acute care hospital in the health system and the combined COVID-19 case-fatality rate over time.

Results

A total of 5955 consecutive COVID-19 patients admitted from March 10, 2020, through April 30, 2021, were analyzed. Patients with International Statistical Classification of Diseases, Tenth Revision codes J12.89. J20.8, J40, J22, J98.8, J80, each with the additional code of B97.29 (or the code UO7.1 after April 1, 2020), were included in the analysis. The median age of admitted patients was 65 years (range 19-91 years). Using the NIH classification system for severity,20 the distribution of severity during the first 24 hours after the time of hospital admission was as follows: asymptomatic/presymptomatic, 0.5%; mild illness, 5.3%; moderate illness, 37.1%; severe illness, 55.5%; and critical illness, 1.1%.

The impact of the CCMC can be estimated by looking at care patterns over time. Since the work of the CCMC was aimed at influencing and standardizing physician ordering and therapy choices through order set creation and other forms of oversight, we measured the use of the CCMC-approved order sets at both hospitals and the use of certain laboratory tests and therapies that the CCMC sought either to limit or increase. These counts were adjusted for number of unique COVID-19 admissions. But the limits of the case collection tool meant it also collected cases that were not eligible for some of the interventions. For example, COVID-19 admissions without hypoxemia would not have been eligible for remdesivir or glucocorticoids. When admitted, some patients were already on steroids for other medical indications and did not receive the prescribed dexamethasone dose that we measured in pharmacy databases. Similarly, a few patients were hospitalized for indications unrelated to COVID-19, such as surgery or childbirth, and were found to be SARS-CoV-2-positive on routine screening.


Figure 2 shows the utilization of CCMC-approved standard COVID-19 admission order sets as a proportion of all COVID-19 admissions over time. The trend reveals a modest increase in usage (R2 = 0.34), but these data do not reflect the progressive build of content into order sets over time. One of the goals of the order sets was to standardize and reduce the ordering of certain biomarkers: C-reactive protein, serum ferritin, and D-dimer, which were ordered frequently in many early patients. Orders for these 3 laboratory tests are combined and expressed as an average number of labs per COVID-19 admission in Figure 2. A downward trend, with an R2 value of 0.65, is suggestive of impact from the order sets, though other explanations are possible.

Medication guidance was also a goal of the CCMC, simultaneously discouraging poorly supported interventions and driving uptake of the recommended evidence-based interventions in appropriate patients. Figure 3 shows the utilization pattern for some drugs of interest over the course of the pandemic, specifically the proportion of patients receiving at least 1 dose of medication among all COVID-19 admissions by month. (Data for hospital B was excluded from this analysis because it did not include all admitted patients.)

Hydroxychloroquine, which enjoyed a wave of popularity early on during the pandemic, was a target of successful order stewardship through the CCMC. Use of hydroxychloroquine as a COVID-19 therapeutic option after the first 2 months of the pandemic stopped, and subsequent use at low levels likely represented continuation therapy for outpatients who took hydroxychloroquine for rheumatologic indications.

Dexamethasone, as used in the RECOVERY trial,21 had a swift uptake among physicians after it was incorporated into order sets and its use encouraged. Similarly, uptake was immediate for remdesivir when, in May 2020, preliminary reports showed at least some benefits, confirmed by later analysis,22 and it received an FDA EUA.

Our data also show successful stewardship of the interleukin-6 antagonist toclilizumab, which was discouraged early on by the CCMC due to lack of data or negative results. But in March 2021, with new studies releasing data12,13 and new recommendations14 for its use in some subsets of patients with COVID-19, this drug was encouraged in appropriate subsets. A new order set with qualifying indications was prepared by the CCMC and new educational efforts made to encourage its use in appropriate patients.

Ivermectin was nonformulary at the start of the pandemic. This drug enjoyed much publicity from media sources and was promoted by certain physicians and on websites,23 based on in-vitro activity against coronaviruses. Eventually, the World Health Organization24 and the FDA25 found it necessary to issue advisory statements to the public against its use outside of clinical trials. The CCMC had requests from physicians to incorporate ivermectin but declined to add it to the formulary and recommended not approving nonformulary requests due to lack of data. As a result, ivermectin was not used at either hospital.

 

 

Discussion

COVID-19 represents many challenges to health systems all over the world. For Luminis Health, the high volume of acutely ill patients with novel syndromes was a particular challenge for the hospital-based care teams. A flood of information from preprints, press releases, preliminary reports, and many other nontraditional sources made deliberative management decisions difficult for individual physicians. Much commentary has appeared around the phenomenon but with less practical advice about how to make day-to-day care decisions at a time of scientific uncertainty and intense pressure to intervene.26,27 The CCMC was designed to overcome the information management dilemma. The need to coordinate, standardize, and oversee care was necessary given the large number of physicians who cared for COVID-19 patients on inpatient services.

It should be noted that creating order sets and issuing guidance is necessary, but not sufficient, to achieve our goals of being updated and consistent. This is especially true with large cadres of health care workers attending COVID-19 patients. Guidelines and recommendations were reinforced by unit-based oversight and stewardship from pharmacy and other leaders who constituted the CCMC.

The reduction in COVID-19 mortality over time experienced in this health care system was not unique and cannot necessarily be attributed to standardization of care. Similar improvements in mortality have been reported at many US hospitals in aggregate.28 Many other factors, including changes in patient characteristics, may be responsible for reduction in mortality over time.

Throughout this report we have relied upon an implicit assumption that standardization of medical therapeutics is desirable and leads to better outcomes as compared with allowing unlimited empiricism by individual physicians, either consultants or hospitalists. Our program represents a single health system with 2 acute care hospitals located 25 miles apart and which thus were similarly impacted by the different phases of the pandemic. Generalizability to health systems either smaller or larger, or in different geographical areas, has not been established. Data limitations have already been discussed.

We did not measure user satisfaction with the program either from physicians or nurses. However, the high rate of compliance suggests general agreement with the content and process.

We cannot definitively ascribe reduction in utilization of some nonrecommended treatments and increased utilization of the recommended therapies to the work of the CCMC. Individual physicians may have made these adjustments on their own or under the influence of other sources.

Finally, it should be noted that the mission to rapidly respond to data from well-conducted trials might be thwarted by too rigid a process or a committee’s lack of a sense of urgency. Organizing a committee and then empowering it to act is no guarantee of success; commitment to the mission is.

Conclusion

COVID-19 represented a challenge to medical staffs everywhere, inundating them with high volumes of acutely ill patients presenting with unfamiliar syndromes. Initial responses were characterized by idiosyncratic management approaches based on nontraditional sources of opinion and influences.

This report describes how a complex medical system brought order and standardization through a deliberative, but urgent, multidisciplinary committee with responsibility for planning, implementing, and monitoring standard approaches that eventually became evidence based. The composition of the committee and its scope of influence, limited to inpatient management, were important elements of success, allowing for better focus on the many treatment decisions. The important connection between the management committee and the system P&T committee, the clinical research effort, and teaching programs in both medicine and pharmacy are offered as exemplars of coordination. The data presented show success in achieving standardized, guideline-directed care. The approach is adoptable and suitable for similar emergencies in the future.

Acknowledgments: The authors thank Gary Scabis, Kip Waite, John Moxley, Angela Clubb, and David Woodley for their assistance in gathering data. We express appreciation and admiration for all our colleagues at the bedside.

Corresponding author: Barry R. Meisenberg, MD, Department of Medicine, Luminis Health, 2001 Medical Pkwy, Annapolis, MD 21401; meisenberg@AAHS.org.

Financial disclosures: None.

References

1. Gettleman J, Raj S, Kumar H. India’s health system cracks under the strain as coronavirus cases surge. The New York Times. April 22, 2021. https://www.nytimes.com/2021/04/21/world/asia/india-coronavirus-oxygen.html

2. Rappleye H, Lehren AW, Strickler L, Fitzpatrick S. ‘This system is doomed’: doctors, nurses sound off in NBC News coronavirus survey. NBC News. March 20, 2020. https://www.nbcnews.com/news/us-news/system-doomed-doctors-nurses-sound-nbc-news-coronavirus-survey-n1164841

3. Johns Hopkins Coronavirus Resource Center. Accessed January 5, 2022. https://coronavirus.jhu.edu/map.html

4. Fineberg HV. The toll of COVID-19. JAMA. 2020;324(15):1502-1503. doi:10.1001/jama.2020.20019

5. Meisenberg BR. Medical staffs response to COVID-19 ‘data’: have we misplaced our skeptic’s eye? Am J Med. 2021;134(2):151-152. doi:10.1016/j.amjmed.2020.09.013

6. McMahon JH, Lydeamore MH, Stewardson AJ. Bringing evidence from press release to the clinic in the era of COVID-19. J Antimicrob Chemother. 2021;76(3):547-549. doi:10.1093/jac/dkaa506

7. Rubin EJ, Baden LR, Morrissey S, Campion EW. Medical journals and the 2019-nCoV outbreak. N Engl J Med. 2020;382(9):866. doi:10.1056/NEJMe2001329

8. Liu F, Li L, Xu M, et al. Prognostic value of interleukin-6, C-reactive protein, and procalcitonin in patients with COVID-19. J Clin Virol. 2020;127:104370. doi:10.1016/j.jcv.2020.104370

9. Vincent MJ, Bergeron E, Benjannet S, et al. Chloroquine is a potent inhibitor of SARS coronavirus infection and spread. Virol J. 2005;2:69. doi:10.1186/1743-422X-2-69

10. Wang M, Cao R, Zhang L, et al. Remdesivir and chloroquine effectively inhibit the recently emerged novel coronavirus (2019-nCoV) in vitro. Cell Res. 2020;30:269-271. doi:10.1038/s41422-020-0282-0

11. RECOVERY Collaborative Group. Effect of hydroxychloroquine in hospitalized patients with Covid-19. N Engl J Med. 2020;383:2030-2040. doi:10.1056/NEJMoa2022926

12. RECOVERY Collaborative Group. Tocilizumab in patients admitted to hospital with COVID-19 (RECOVERY): preliminary results of a randomised, controlled, open-label, platform trial [preprint]. February 11, 2021. doi:10.1101/2021.02.11.21249258 https://www.medrxiv.org/content/10.1101/2021.02.11.21249258v1

13. REMAP-CAP Investigators. Interleukin-6 receptor antagonists in critically ill patients with COVID-19. N Engl J Med. 2021;384(16):1491-1502. doi:10.1056/NEJMoa2100433

14. National Institutes of Health. COVID-19 treatment guidelines: interleukin-6 inhibitors. https://www.covid19treatmentguidelines.nih.gov/immunomodulators/interleukin-6-inhibitors/

15. Deana C, Vetrugno L, Tonizzo A, et al. Drug supply during COVID-19 pandemic: remember not to run with your tank empty. Hosp Pharm. 2021;56(5):405-407. doi:10.1177/0018578720931749

16. Choe J, Crane M, Greene J, et al. The Pandemic and the Supply Chain: Addressing Gaps in Pharmaceutical Production and Distribution. Johns Hopkins University, November 2020. https://www.jhsph.edu/research/affiliated-programs/johns-hopkins-drug-access-and-affordability-initiative/publications/Pandemic_Supply_Chain.pdf

17. Kern DE. Overview: a six-step approach to curriculum development. In: Kern DE, Thornton PA, Hughes MT, eds. Curriculum Development for Medical Education: A Six-Step Approach. 3rd ed. Johns Hopkins University Press; 2016.

18. Rice TW, Janz DR. In defense of evidence-based medicine for the treatment of COVID-19 acute respiratory distress syndrome. Ann Am Thorac Soc. 2020;17(7):787-789. doi:10.1513/AnnalsATS.202004-325IP

19. Lucey CR, Johnston SC. The transformational effects of COVID-19 on medical education. JAMA. 2020;324(11):1033-1034. doi:10.1001/jama.2020.14136

20. National Institutes of Health. COVID-19 treatment guidelines: clinical spectrum of SARS-CoV-2 infection. https://www.covid19treatmentguidelines.nih.gov/overview/clinical-spectrum/

21. RECOVERY Collaborative Group. Dexamethasone in hospitalized patients with Covid-19. N Engl J Med. 2021;384:693-704. doi:10.1056/NEJMoa2021436

22. Beigel JH, Tomashek KM, Dodd LE, et al. Remdesivir for the treatment of Covid-19—final report. N Engl J Med. 2020;383:1813-1826. doi:10.1056/NEJMoa2007764

23. Jiminez D. Ivermectin and Covid-19: how a cheap antiparasitic became political. April 19, 2021. https://www.pharmaceutical-technology.com/features/ivermectin-covid-19-antiparasitic-political/

24. World Health Organization. WHO advises that ivermectin only be used to treat COVID-19 within clinical trials. March 31, 2021. https://www.who.int/news-room/feature-stories/detail/who-advises-that-ivermectin-only-be-used-to-treat-covid-19-within-clinical-trials

25. U.S. Food and Drug Administration. Why you should not use ivermectin to treat or prevent COVID-19. March 5, 2021. https://www.fda.gov/consumers/consumer-updates/why-you-should-not-use-ivermectin-treat-or-prevent-covid-19

26. Seymour CW, McCreary EK, Stegenga J. Sensible medicine-balancing intervention and inaction during the COVID-19 pandemic. JAMA. 2020;324(18):1827-1828. doi:10.1001/jama.2020.20271

27. Flanagin A, Fontanarosa PB, Bauchner H. Preprints involving medical research—do the benefits outweigh the challenges? JAMA. 2020;324(18):1840-1843. doi:10.1001/jama.2020.20674

28. Asch DA, Shells NE, Islam N, et al. Variation in US hospital mortality rates for patients admitted with COVID-19 during the first 6 months of the pandemic. JAMA Intern Med. 2021;181(4):471-478. doi:10.1001/jamainternmed.2020.8193

Article PDF
Issue
Journal of Clinical Outcomes Management - 29(1)
Publications
Topics
Page Number
39-48
Sections
Article PDF
Article PDF

From the Department of Medicine (Drs. Meisenberg, Muganlinskaya, Sharma, Amjadi, Arnold, Barnes, Clance, Khalil, Miller, Mooradian, O’Connell, Patel, Press, Samaras, Shanmugam, Tavadze, and Thompson), Department of Pharmacy (Drs. Jiang, Jarawan, Sheth, and Trinh), Department of Nursing (Dr. Ohnmacht), and Department of Women and Children’s Services (Dr. Raji), Luminis Health, Annapolis, MD, and Lanham, MD.

Objective: The COVID-19 pandemic has been a challenge for hospital medical staffs worldwide due to high volumes of patients acutely ill with novel syndromes and prevailing uncertainty regarding optimum supportive and therapeutic interventions. Additionally, the response to this crisis was driven by a plethora of nontraditional information sources, such as email chains, websites, non–peer-reviewed preprints, and press releases. Care patterns became idiosyncratic and often incorporated unproven interventions driven by these nontraditional information sources. This report evaluates the efforts of a health system to create and empower a multidisciplinary committee to develop, implement, and monitor evidence-based, standardized protocols for patients with COVID-19.

Methods: This report describes the composition of the committee, its scope, and its important interactions with the health system pharmacy and therapeutics committee, research teams, and other work groups planning other aspects of COVID-19 management. It illustrates how the committee was used to demonstrate for trainees the process and value of critically examining evidence, even in a chaotic environment.

Results: Data show successful interventions in reducing excessive ordering of certain laboratory tests, reduction of nonrecommended therapies, and rapid uptake of evidence-based or guidelines-supported interventions.

Conclusions: A multidisciplinary committee dedicated solely to planning, implementing, and monitoring standard approaches that eventually became evidence-based decision-making led to an improved focus on treatment options and outcomes for COVID-19 patients. Data presented illustrate the attainable success that is both adaptable and suitable for similar emergencies in the future.

Keywords: COVID-19; clinical management; pharmacy and therapeutics; treatment; therapy.

The COVID-19 pandemic has spread to nearly all countries, carrying with it high morbidity, mortality, and severe impacts on both well-developed and less-well-developed health systems. Media reports of chaos within overwhelmed hospitals have been prominent.1,2 As of January 5, 2022, SARS-CoV-2 has infected more than 295 million people globally and directly caused the death of more than 5.4 million,3 though this number is likely an undercount even in countries with well-developed mortality tracking.4

Throughout the COVID-19 pandemic, hospital-based medical teams have been confronted with a flood of severely ill patients with novel syndromes. Initially, there were no standards for therapy or supportive care except for treatments borrowed from similar syndromes. In the setting of high volumes, high acuity, and public dismay, it is unsurprising that the usual deliberative methods for weighing evidence and initiating interventions were often pushed aside in favor of the solace of active intervention.5 In this milieu of limited evidence, there was a lamentable, if understandable, tendency to seek guidance from “nontraditional” sources,6 including email chains from colleagues, hospital websites, non–peer-reviewed manuscripts, advanced publication by medical journals,7 and nonscientific media presentations. In many localities, practitioners responded in idiosyncratic ways. For example, findings of high cytokine levels in COVID-19,8 along with reports of in-vitro antiviral activity with drugs like hydroxychloroquine against both SARS9 and SARS-CoV-2,10 drove laboratory test ordering and therapeutic interventions, respectively, carving shortcuts into the traditional clinical trial–dependent standards. Clinical trial results eventually emerged.11COVID-19 created a clinical dilemma for hospital medical staffs in terms of how to organize, standardize, and rapidly adapt to a flood of new information. In this report, we describe how 1 health system responded to these challenges by forming a COVID-19 Clinical Management Committee (CCMC) and empowering this interdisciplinary team to review evidence, create and adjust order sets, educate practitioners, oversee care, and collaborate across teams addressing other aspects of the COVID-19 response.

 

 

Program Overview

Health System Description

Luminis Health is a health system with 2 acute care hospitals that was formed in 2019 just before the start of the pandemic. Anne Arundel Medical Center (hospital A) is a 385-bed teaching hospital in Annapolis, MD. It has more than 23 000 discharges annually. Patients with COVID-19 were cared for by either an internal medicine teaching service or nonteaching hospitalist services on cohorted nursing units. Doctor’s Community Medical Center, in Lanham, MD (hospital B), is a 206-bed acute care hospital with more than 10 350 annual discharges. COVID-19 patients were cared for by hospitalist groups, initially in noncohorted units with transition to cohorted nursing units after a few months. The medical staffs are generally distinct, with different leadership structures, though the Luminis Health Department of Medicine has oversight responsibilities at both hospitals. More than 47 physicians attended COVID-19 patients at hospital A (with medical residents) and 30 individual physicians at hospital B, respectively, including intensivists. The nursing and pharmacy staffs are distinct, but there is a shared oversight Pharmacy and Therapeutics (P&T) Committee.

The 2 hospitals had distinct electronic medical records (EMR) until January 2021, when hospital B adopted the same EMR as hospital A (Epic).

Mission and Formation of CCMC

In order to coordinate the therapeutic approach across the health system, it was important for the CCMC to be designated by the health system P&T committee as an official subcommittee so that decisions on restrictions of medications and/or new or revised order sets could be rapidly initiated across the system without waiting for the subsequent P&T meetings. The full committee retained oversight of the CCMC. Some P&T members were also on the CCMC.

The committee reviewed new reports in medical journals and prepublication servers and consulted recommendations of professional societies, such as the National Institutes of Health (NIH) COVID-19 guidelines, Infectious Diseases Society of America, Society of Critical Care Medicine, and US Food and Drug Administration (FDA) Emergency Use Authorizations (EUA), among other sources.

Composition of the CCMC

Physician leaders from both hospitals in the following specialties were solicited for participation: critical care, epidemiology, hospital medicine (internal medicine), emergency medicine, infectious diseases, nephrology, women and children’s services, and medical informatics. Specialists in other areas, such as hematology, were invited for topic-specific discussions. Hospital pharmacists with different specialties and nursing leadership were essential contributors. The committee members were expected to use various communication channels to inform frontline clinicians of new care standards and the existence of new order sets, which were embedded in the EMR.

Clinical Research

An important connection for the CCMC was with theCOVID-19 clinical research team. Three members of the research team were also members of the CCMC. All new study proposals for therapeutics were discussed with the CCMC as they were being considered by the research team. In this way, feedback on the feasibility and acceptance of new study opportunities could be discussed with the CCMC. Occasionally, CCMC decisions impacted clinical research accrual strategies. For example, new data from randomized trials about tocilizumab1,2 demonstrated benefits in some subsets of patients and resulted in a recommendation for use by the NIH guideline committee in these populations.1 The CCMC quickly adopted this recommendation, which required a reprioritization of clinical research enrollment for studies testing other immune-modulating agents. This important dialogue was mediated within the CCMC.

Guideline Distribution, Reinforcement, and Platform for Feedback

New guidelines were disseminated to clinicians via daily brief patient huddles held on COVID units, with participation by nursing and pharmacy, and by weekly meetings with hospitalist leaders and frontline hospital physicians. Order sets and guidelines were maintained on the intranet. Adherence was reinforced by unit-based and central pharmacists. Order sets, including admission order sets, could be created only by designated informatics personnel, thus enforcing standardization. Feedback on the utility of the order sets was obtained during the weekly meetings or huddles, as described above. To ensure a sense of transparency, physicians who had interest in commenting on a particular therapy, or who wished to discuss a particular manuscript, news article, or website, were invited to attend CCMC meetings.

Scope of CCMC

In order to be effective and timely, we limited the scope of our work to the report to the inpatient therapeutic environment, allowing other committees to work on other aspects of the pandemic response. In addition to issuing guidance and creating order sets to direct clinical practice, the CCMC also monitored COVID-19 therapeutic shortages15,16 and advised on prioritization of such treatments as convalescent plasma, remdesivir (prioritization and duration of therapy, 5 vs 10 days), baricitinib, and tocilizumab, depending upon the location of the patient (critical care or not). The CCMC was not involved in the management of non–COVID-19 shortages brought about by supply chain deficiencies.

Table 1 shows some aspects of the health system pandemic-response planning and the committee workforce that undertook that work. Though many items were out of scope for the CCMC, members of the CCMC did participate in the planning work of these other committees and therefore stayed connected to this complementary work.

A Teaching Opportunity About Making Thoughtful Choices

Another important feature of the CCMC was the contributions of residents from both pharmacy and internal medicine. The purpose and operations of the committee were recognized as an opportunity to involve learners in a curriculum based on Kern’s 6-step approach.17 Though the problem identification and general needs assessment were easily defined, the targeted needs assessment, extracted from individual and group interviews with learners and the committee members, pointed at the need to learn how to assess and analyze a rapidly growing body of literature on several relevant clinical aspects of SARS-CoV-2 and COVID-19. To achieve goals and objectives, residents were assigned to present current literature on a particular intervention during a committee meeting, specifically commenting on the merit or deficiencies of the study design, the strength of the data, and applicability to the local context with a recommendation. Prior to the presentations, the residents worked with faculty to identify the best studies or systematic analyses with potential to alter current practices. We thus used the CCMC process as a teaching tool about evidence-based medicine and the dilemma of clinical equipoise. This was imperative, since trainees thrust into the COVID-19 response have often keenly observed a movement away from deliberative decision-making.18 Indeed, including residents in the process of deliberative responses to COVID-19 addresses a recent call to adjust medical education during COVID-19 to “adapt curriculum to current issues in real time.”19

Interventions and Therapies Considered

Table 2 shows the topics reviewed by the CCMC. By the time of the first meeting, nonstandardization of care was already a source of concern for clinicians. Dialogue often continued outside of the formal meetings. Many topics were considered more than once as new guidance developed, changes to EUAs occurred, and new data or new publicity arose.

 

 

Methods

The Human Protections Administrator determined that this work constituted “quality improvement, and not research” and was therefore exempt from institutional review board review.

Quantitative Analysis

All admitted patients from March 10, 2020, through April 20, 2021, were considered in the quantitative aspects of this report except as noted. Patients diagnosed with COVID-19 were identified by searching our internal data base using diagnostic codes. Patient admissions with the following diagnostic codes were included (prior to April 1, 2020): J12.89, J20.8, J40, J22, J98.8, J80, each with the additional code of B97.29. After April 1, 2020, the guideline for coding COVID-19 was U07.1.

Descriptive statistics were used to measure utilization rates of certain medications and laboratory tests of interest over time. These data were adjusted for number of unique admissions. In a few cases, not all data elements were available from both hospitals due to differences in the EMR.

Case fatality rate was calculated based upon whether the patient died or was admitted to inpatient hospice as a result of COVID-19. Four patients transferred out of hospital A and 18 transferred out of hospital B were censored from case-fatality-rate determination.

Figure 1 shows the number of admissions for each acute care hospital in the health system and the combined COVID-19 case-fatality rate over time.

Results

A total of 5955 consecutive COVID-19 patients admitted from March 10, 2020, through April 30, 2021, were analyzed. Patients with International Statistical Classification of Diseases, Tenth Revision codes J12.89. J20.8, J40, J22, J98.8, J80, each with the additional code of B97.29 (or the code UO7.1 after April 1, 2020), were included in the analysis. The median age of admitted patients was 65 years (range 19-91 years). Using the NIH classification system for severity,20 the distribution of severity during the first 24 hours after the time of hospital admission was as follows: asymptomatic/presymptomatic, 0.5%; mild illness, 5.3%; moderate illness, 37.1%; severe illness, 55.5%; and critical illness, 1.1%.

The impact of the CCMC can be estimated by looking at care patterns over time. Since the work of the CCMC was aimed at influencing and standardizing physician ordering and therapy choices through order set creation and other forms of oversight, we measured the use of the CCMC-approved order sets at both hospitals and the use of certain laboratory tests and therapies that the CCMC sought either to limit or increase. These counts were adjusted for number of unique COVID-19 admissions. But the limits of the case collection tool meant it also collected cases that were not eligible for some of the interventions. For example, COVID-19 admissions without hypoxemia would not have been eligible for remdesivir or glucocorticoids. When admitted, some patients were already on steroids for other medical indications and did not receive the prescribed dexamethasone dose that we measured in pharmacy databases. Similarly, a few patients were hospitalized for indications unrelated to COVID-19, such as surgery or childbirth, and were found to be SARS-CoV-2-positive on routine screening.


Figure 2 shows the utilization of CCMC-approved standard COVID-19 admission order sets as a proportion of all COVID-19 admissions over time. The trend reveals a modest increase in usage (R2 = 0.34), but these data do not reflect the progressive build of content into order sets over time. One of the goals of the order sets was to standardize and reduce the ordering of certain biomarkers: C-reactive protein, serum ferritin, and D-dimer, which were ordered frequently in many early patients. Orders for these 3 laboratory tests are combined and expressed as an average number of labs per COVID-19 admission in Figure 2. A downward trend, with an R2 value of 0.65, is suggestive of impact from the order sets, though other explanations are possible.

Medication guidance was also a goal of the CCMC, simultaneously discouraging poorly supported interventions and driving uptake of the recommended evidence-based interventions in appropriate patients. Figure 3 shows the utilization pattern for some drugs of interest over the course of the pandemic, specifically the proportion of patients receiving at least 1 dose of medication among all COVID-19 admissions by month. (Data for hospital B was excluded from this analysis because it did not include all admitted patients.)

Hydroxychloroquine, which enjoyed a wave of popularity early on during the pandemic, was a target of successful order stewardship through the CCMC. Use of hydroxychloroquine as a COVID-19 therapeutic option after the first 2 months of the pandemic stopped, and subsequent use at low levels likely represented continuation therapy for outpatients who took hydroxychloroquine for rheumatologic indications.

Dexamethasone, as used in the RECOVERY trial,21 had a swift uptake among physicians after it was incorporated into order sets and its use encouraged. Similarly, uptake was immediate for remdesivir when, in May 2020, preliminary reports showed at least some benefits, confirmed by later analysis,22 and it received an FDA EUA.

Our data also show successful stewardship of the interleukin-6 antagonist toclilizumab, which was discouraged early on by the CCMC due to lack of data or negative results. But in March 2021, with new studies releasing data12,13 and new recommendations14 for its use in some subsets of patients with COVID-19, this drug was encouraged in appropriate subsets. A new order set with qualifying indications was prepared by the CCMC and new educational efforts made to encourage its use in appropriate patients.

Ivermectin was nonformulary at the start of the pandemic. This drug enjoyed much publicity from media sources and was promoted by certain physicians and on websites,23 based on in-vitro activity against coronaviruses. Eventually, the World Health Organization24 and the FDA25 found it necessary to issue advisory statements to the public against its use outside of clinical trials. The CCMC had requests from physicians to incorporate ivermectin but declined to add it to the formulary and recommended not approving nonformulary requests due to lack of data. As a result, ivermectin was not used at either hospital.

 

 

Discussion

COVID-19 represents many challenges to health systems all over the world. For Luminis Health, the high volume of acutely ill patients with novel syndromes was a particular challenge for the hospital-based care teams. A flood of information from preprints, press releases, preliminary reports, and many other nontraditional sources made deliberative management decisions difficult for individual physicians. Much commentary has appeared around the phenomenon but with less practical advice about how to make day-to-day care decisions at a time of scientific uncertainty and intense pressure to intervene.26,27 The CCMC was designed to overcome the information management dilemma. The need to coordinate, standardize, and oversee care was necessary given the large number of physicians who cared for COVID-19 patients on inpatient services.

It should be noted that creating order sets and issuing guidance is necessary, but not sufficient, to achieve our goals of being updated and consistent. This is especially true with large cadres of health care workers attending COVID-19 patients. Guidelines and recommendations were reinforced by unit-based oversight and stewardship from pharmacy and other leaders who constituted the CCMC.

The reduction in COVID-19 mortality over time experienced in this health care system was not unique and cannot necessarily be attributed to standardization of care. Similar improvements in mortality have been reported at many US hospitals in aggregate.28 Many other factors, including changes in patient characteristics, may be responsible for reduction in mortality over time.

Throughout this report we have relied upon an implicit assumption that standardization of medical therapeutics is desirable and leads to better outcomes as compared with allowing unlimited empiricism by individual physicians, either consultants or hospitalists. Our program represents a single health system with 2 acute care hospitals located 25 miles apart and which thus were similarly impacted by the different phases of the pandemic. Generalizability to health systems either smaller or larger, or in different geographical areas, has not been established. Data limitations have already been discussed.

We did not measure user satisfaction with the program either from physicians or nurses. However, the high rate of compliance suggests general agreement with the content and process.

We cannot definitively ascribe reduction in utilization of some nonrecommended treatments and increased utilization of the recommended therapies to the work of the CCMC. Individual physicians may have made these adjustments on their own or under the influence of other sources.

Finally, it should be noted that the mission to rapidly respond to data from well-conducted trials might be thwarted by too rigid a process or a committee’s lack of a sense of urgency. Organizing a committee and then empowering it to act is no guarantee of success; commitment to the mission is.

Conclusion

COVID-19 represented a challenge to medical staffs everywhere, inundating them with high volumes of acutely ill patients presenting with unfamiliar syndromes. Initial responses were characterized by idiosyncratic management approaches based on nontraditional sources of opinion and influences.

This report describes how a complex medical system brought order and standardization through a deliberative, but urgent, multidisciplinary committee with responsibility for planning, implementing, and monitoring standard approaches that eventually became evidence based. The composition of the committee and its scope of influence, limited to inpatient management, were important elements of success, allowing for better focus on the many treatment decisions. The important connection between the management committee and the system P&T committee, the clinical research effort, and teaching programs in both medicine and pharmacy are offered as exemplars of coordination. The data presented show success in achieving standardized, guideline-directed care. The approach is adoptable and suitable for similar emergencies in the future.

Acknowledgments: The authors thank Gary Scabis, Kip Waite, John Moxley, Angela Clubb, and David Woodley for their assistance in gathering data. We express appreciation and admiration for all our colleagues at the bedside.

Corresponding author: Barry R. Meisenberg, MD, Department of Medicine, Luminis Health, 2001 Medical Pkwy, Annapolis, MD 21401; meisenberg@AAHS.org.

Financial disclosures: None.

From the Department of Medicine (Drs. Meisenberg, Muganlinskaya, Sharma, Amjadi, Arnold, Barnes, Clance, Khalil, Miller, Mooradian, O’Connell, Patel, Press, Samaras, Shanmugam, Tavadze, and Thompson), Department of Pharmacy (Drs. Jiang, Jarawan, Sheth, and Trinh), Department of Nursing (Dr. Ohnmacht), and Department of Women and Children’s Services (Dr. Raji), Luminis Health, Annapolis, MD, and Lanham, MD.

Objective: The COVID-19 pandemic has been a challenge for hospital medical staffs worldwide due to high volumes of patients acutely ill with novel syndromes and prevailing uncertainty regarding optimum supportive and therapeutic interventions. Additionally, the response to this crisis was driven by a plethora of nontraditional information sources, such as email chains, websites, non–peer-reviewed preprints, and press releases. Care patterns became idiosyncratic and often incorporated unproven interventions driven by these nontraditional information sources. This report evaluates the efforts of a health system to create and empower a multidisciplinary committee to develop, implement, and monitor evidence-based, standardized protocols for patients with COVID-19.

Methods: This report describes the composition of the committee, its scope, and its important interactions with the health system pharmacy and therapeutics committee, research teams, and other work groups planning other aspects of COVID-19 management. It illustrates how the committee was used to demonstrate for trainees the process and value of critically examining evidence, even in a chaotic environment.

Results: Data show successful interventions in reducing excessive ordering of certain laboratory tests, reduction of nonrecommended therapies, and rapid uptake of evidence-based or guidelines-supported interventions.

Conclusions: A multidisciplinary committee dedicated solely to planning, implementing, and monitoring standard approaches that eventually became evidence-based decision-making led to an improved focus on treatment options and outcomes for COVID-19 patients. Data presented illustrate the attainable success that is both adaptable and suitable for similar emergencies in the future.

Keywords: COVID-19; clinical management; pharmacy and therapeutics; treatment; therapy.

The COVID-19 pandemic has spread to nearly all countries, carrying with it high morbidity, mortality, and severe impacts on both well-developed and less-well-developed health systems. Media reports of chaos within overwhelmed hospitals have been prominent.1,2 As of January 5, 2022, SARS-CoV-2 has infected more than 295 million people globally and directly caused the death of more than 5.4 million,3 though this number is likely an undercount even in countries with well-developed mortality tracking.4

Throughout the COVID-19 pandemic, hospital-based medical teams have been confronted with a flood of severely ill patients with novel syndromes. Initially, there were no standards for therapy or supportive care except for treatments borrowed from similar syndromes. In the setting of high volumes, high acuity, and public dismay, it is unsurprising that the usual deliberative methods for weighing evidence and initiating interventions were often pushed aside in favor of the solace of active intervention.5 In this milieu of limited evidence, there was a lamentable, if understandable, tendency to seek guidance from “nontraditional” sources,6 including email chains from colleagues, hospital websites, non–peer-reviewed manuscripts, advanced publication by medical journals,7 and nonscientific media presentations. In many localities, practitioners responded in idiosyncratic ways. For example, findings of high cytokine levels in COVID-19,8 along with reports of in-vitro antiviral activity with drugs like hydroxychloroquine against both SARS9 and SARS-CoV-2,10 drove laboratory test ordering and therapeutic interventions, respectively, carving shortcuts into the traditional clinical trial–dependent standards. Clinical trial results eventually emerged.11COVID-19 created a clinical dilemma for hospital medical staffs in terms of how to organize, standardize, and rapidly adapt to a flood of new information. In this report, we describe how 1 health system responded to these challenges by forming a COVID-19 Clinical Management Committee (CCMC) and empowering this interdisciplinary team to review evidence, create and adjust order sets, educate practitioners, oversee care, and collaborate across teams addressing other aspects of the COVID-19 response.

 

 

Program Overview

Health System Description

Luminis Health is a health system with 2 acute care hospitals that was formed in 2019 just before the start of the pandemic. Anne Arundel Medical Center (hospital A) is a 385-bed teaching hospital in Annapolis, MD. It has more than 23 000 discharges annually. Patients with COVID-19 were cared for by either an internal medicine teaching service or nonteaching hospitalist services on cohorted nursing units. Doctor’s Community Medical Center, in Lanham, MD (hospital B), is a 206-bed acute care hospital with more than 10 350 annual discharges. COVID-19 patients were cared for by hospitalist groups, initially in noncohorted units with transition to cohorted nursing units after a few months. The medical staffs are generally distinct, with different leadership structures, though the Luminis Health Department of Medicine has oversight responsibilities at both hospitals. More than 47 physicians attended COVID-19 patients at hospital A (with medical residents) and 30 individual physicians at hospital B, respectively, including intensivists. The nursing and pharmacy staffs are distinct, but there is a shared oversight Pharmacy and Therapeutics (P&T) Committee.

The 2 hospitals had distinct electronic medical records (EMR) until January 2021, when hospital B adopted the same EMR as hospital A (Epic).

Mission and Formation of CCMC

In order to coordinate the therapeutic approach across the health system, it was important for the CCMC to be designated by the health system P&T committee as an official subcommittee so that decisions on restrictions of medications and/or new or revised order sets could be rapidly initiated across the system without waiting for the subsequent P&T meetings. The full committee retained oversight of the CCMC. Some P&T members were also on the CCMC.

The committee reviewed new reports in medical journals and prepublication servers and consulted recommendations of professional societies, such as the National Institutes of Health (NIH) COVID-19 guidelines, Infectious Diseases Society of America, Society of Critical Care Medicine, and US Food and Drug Administration (FDA) Emergency Use Authorizations (EUA), among other sources.

Composition of the CCMC

Physician leaders from both hospitals in the following specialties were solicited for participation: critical care, epidemiology, hospital medicine (internal medicine), emergency medicine, infectious diseases, nephrology, women and children’s services, and medical informatics. Specialists in other areas, such as hematology, were invited for topic-specific discussions. Hospital pharmacists with different specialties and nursing leadership were essential contributors. The committee members were expected to use various communication channels to inform frontline clinicians of new care standards and the existence of new order sets, which were embedded in the EMR.

Clinical Research

An important connection for the CCMC was with theCOVID-19 clinical research team. Three members of the research team were also members of the CCMC. All new study proposals for therapeutics were discussed with the CCMC as they were being considered by the research team. In this way, feedback on the feasibility and acceptance of new study opportunities could be discussed with the CCMC. Occasionally, CCMC decisions impacted clinical research accrual strategies. For example, new data from randomized trials about tocilizumab1,2 demonstrated benefits in some subsets of patients and resulted in a recommendation for use by the NIH guideline committee in these populations.1 The CCMC quickly adopted this recommendation, which required a reprioritization of clinical research enrollment for studies testing other immune-modulating agents. This important dialogue was mediated within the CCMC.

Guideline Distribution, Reinforcement, and Platform for Feedback

New guidelines were disseminated to clinicians via daily brief patient huddles held on COVID units, with participation by nursing and pharmacy, and by weekly meetings with hospitalist leaders and frontline hospital physicians. Order sets and guidelines were maintained on the intranet. Adherence was reinforced by unit-based and central pharmacists. Order sets, including admission order sets, could be created only by designated informatics personnel, thus enforcing standardization. Feedback on the utility of the order sets was obtained during the weekly meetings or huddles, as described above. To ensure a sense of transparency, physicians who had interest in commenting on a particular therapy, or who wished to discuss a particular manuscript, news article, or website, were invited to attend CCMC meetings.

Scope of CCMC

In order to be effective and timely, we limited the scope of our work to the report to the inpatient therapeutic environment, allowing other committees to work on other aspects of the pandemic response. In addition to issuing guidance and creating order sets to direct clinical practice, the CCMC also monitored COVID-19 therapeutic shortages15,16 and advised on prioritization of such treatments as convalescent plasma, remdesivir (prioritization and duration of therapy, 5 vs 10 days), baricitinib, and tocilizumab, depending upon the location of the patient (critical care or not). The CCMC was not involved in the management of non–COVID-19 shortages brought about by supply chain deficiencies.

Table 1 shows some aspects of the health system pandemic-response planning and the committee workforce that undertook that work. Though many items were out of scope for the CCMC, members of the CCMC did participate in the planning work of these other committees and therefore stayed connected to this complementary work.

A Teaching Opportunity About Making Thoughtful Choices

Another important feature of the CCMC was the contributions of residents from both pharmacy and internal medicine. The purpose and operations of the committee were recognized as an opportunity to involve learners in a curriculum based on Kern’s 6-step approach.17 Though the problem identification and general needs assessment were easily defined, the targeted needs assessment, extracted from individual and group interviews with learners and the committee members, pointed at the need to learn how to assess and analyze a rapidly growing body of literature on several relevant clinical aspects of SARS-CoV-2 and COVID-19. To achieve goals and objectives, residents were assigned to present current literature on a particular intervention during a committee meeting, specifically commenting on the merit or deficiencies of the study design, the strength of the data, and applicability to the local context with a recommendation. Prior to the presentations, the residents worked with faculty to identify the best studies or systematic analyses with potential to alter current practices. We thus used the CCMC process as a teaching tool about evidence-based medicine and the dilemma of clinical equipoise. This was imperative, since trainees thrust into the COVID-19 response have often keenly observed a movement away from deliberative decision-making.18 Indeed, including residents in the process of deliberative responses to COVID-19 addresses a recent call to adjust medical education during COVID-19 to “adapt curriculum to current issues in real time.”19

Interventions and Therapies Considered

Table 2 shows the topics reviewed by the CCMC. By the time of the first meeting, nonstandardization of care was already a source of concern for clinicians. Dialogue often continued outside of the formal meetings. Many topics were considered more than once as new guidance developed, changes to EUAs occurred, and new data or new publicity arose.

 

 

Methods

The Human Protections Administrator determined that this work constituted “quality improvement, and not research” and was therefore exempt from institutional review board review.

Quantitative Analysis

All admitted patients from March 10, 2020, through April 20, 2021, were considered in the quantitative aspects of this report except as noted. Patients diagnosed with COVID-19 were identified by searching our internal data base using diagnostic codes. Patient admissions with the following diagnostic codes were included (prior to April 1, 2020): J12.89, J20.8, J40, J22, J98.8, J80, each with the additional code of B97.29. After April 1, 2020, the guideline for coding COVID-19 was U07.1.

Descriptive statistics were used to measure utilization rates of certain medications and laboratory tests of interest over time. These data were adjusted for number of unique admissions. In a few cases, not all data elements were available from both hospitals due to differences in the EMR.

Case fatality rate was calculated based upon whether the patient died or was admitted to inpatient hospice as a result of COVID-19. Four patients transferred out of hospital A and 18 transferred out of hospital B were censored from case-fatality-rate determination.

Figure 1 shows the number of admissions for each acute care hospital in the health system and the combined COVID-19 case-fatality rate over time.

Results

A total of 5955 consecutive COVID-19 patients admitted from March 10, 2020, through April 30, 2021, were analyzed. Patients with International Statistical Classification of Diseases, Tenth Revision codes J12.89. J20.8, J40, J22, J98.8, J80, each with the additional code of B97.29 (or the code UO7.1 after April 1, 2020), were included in the analysis. The median age of admitted patients was 65 years (range 19-91 years). Using the NIH classification system for severity,20 the distribution of severity during the first 24 hours after the time of hospital admission was as follows: asymptomatic/presymptomatic, 0.5%; mild illness, 5.3%; moderate illness, 37.1%; severe illness, 55.5%; and critical illness, 1.1%.

The impact of the CCMC can be estimated by looking at care patterns over time. Since the work of the CCMC was aimed at influencing and standardizing physician ordering and therapy choices through order set creation and other forms of oversight, we measured the use of the CCMC-approved order sets at both hospitals and the use of certain laboratory tests and therapies that the CCMC sought either to limit or increase. These counts were adjusted for number of unique COVID-19 admissions. But the limits of the case collection tool meant it also collected cases that were not eligible for some of the interventions. For example, COVID-19 admissions without hypoxemia would not have been eligible for remdesivir or glucocorticoids. When admitted, some patients were already on steroids for other medical indications and did not receive the prescribed dexamethasone dose that we measured in pharmacy databases. Similarly, a few patients were hospitalized for indications unrelated to COVID-19, such as surgery or childbirth, and were found to be SARS-CoV-2-positive on routine screening.


Figure 2 shows the utilization of CCMC-approved standard COVID-19 admission order sets as a proportion of all COVID-19 admissions over time. The trend reveals a modest increase in usage (R2 = 0.34), but these data do not reflect the progressive build of content into order sets over time. One of the goals of the order sets was to standardize and reduce the ordering of certain biomarkers: C-reactive protein, serum ferritin, and D-dimer, which were ordered frequently in many early patients. Orders for these 3 laboratory tests are combined and expressed as an average number of labs per COVID-19 admission in Figure 2. A downward trend, with an R2 value of 0.65, is suggestive of impact from the order sets, though other explanations are possible.

Medication guidance was also a goal of the CCMC, simultaneously discouraging poorly supported interventions and driving uptake of the recommended evidence-based interventions in appropriate patients. Figure 3 shows the utilization pattern for some drugs of interest over the course of the pandemic, specifically the proportion of patients receiving at least 1 dose of medication among all COVID-19 admissions by month. (Data for hospital B was excluded from this analysis because it did not include all admitted patients.)

Hydroxychloroquine, which enjoyed a wave of popularity early on during the pandemic, was a target of successful order stewardship through the CCMC. Use of hydroxychloroquine as a COVID-19 therapeutic option after the first 2 months of the pandemic stopped, and subsequent use at low levels likely represented continuation therapy for outpatients who took hydroxychloroquine for rheumatologic indications.

Dexamethasone, as used in the RECOVERY trial,21 had a swift uptake among physicians after it was incorporated into order sets and its use encouraged. Similarly, uptake was immediate for remdesivir when, in May 2020, preliminary reports showed at least some benefits, confirmed by later analysis,22 and it received an FDA EUA.

Our data also show successful stewardship of the interleukin-6 antagonist toclilizumab, which was discouraged early on by the CCMC due to lack of data or negative results. But in March 2021, with new studies releasing data12,13 and new recommendations14 for its use in some subsets of patients with COVID-19, this drug was encouraged in appropriate subsets. A new order set with qualifying indications was prepared by the CCMC and new educational efforts made to encourage its use in appropriate patients.

Ivermectin was nonformulary at the start of the pandemic. This drug enjoyed much publicity from media sources and was promoted by certain physicians and on websites,23 based on in-vitro activity against coronaviruses. Eventually, the World Health Organization24 and the FDA25 found it necessary to issue advisory statements to the public against its use outside of clinical trials. The CCMC had requests from physicians to incorporate ivermectin but declined to add it to the formulary and recommended not approving nonformulary requests due to lack of data. As a result, ivermectin was not used at either hospital.

 

 

Discussion

COVID-19 represents many challenges to health systems all over the world. For Luminis Health, the high volume of acutely ill patients with novel syndromes was a particular challenge for the hospital-based care teams. A flood of information from preprints, press releases, preliminary reports, and many other nontraditional sources made deliberative management decisions difficult for individual physicians. Much commentary has appeared around the phenomenon but with less practical advice about how to make day-to-day care decisions at a time of scientific uncertainty and intense pressure to intervene.26,27 The CCMC was designed to overcome the information management dilemma. The need to coordinate, standardize, and oversee care was necessary given the large number of physicians who cared for COVID-19 patients on inpatient services.

It should be noted that creating order sets and issuing guidance is necessary, but not sufficient, to achieve our goals of being updated and consistent. This is especially true with large cadres of health care workers attending COVID-19 patients. Guidelines and recommendations were reinforced by unit-based oversight and stewardship from pharmacy and other leaders who constituted the CCMC.

The reduction in COVID-19 mortality over time experienced in this health care system was not unique and cannot necessarily be attributed to standardization of care. Similar improvements in mortality have been reported at many US hospitals in aggregate.28 Many other factors, including changes in patient characteristics, may be responsible for reduction in mortality over time.

Throughout this report we have relied upon an implicit assumption that standardization of medical therapeutics is desirable and leads to better outcomes as compared with allowing unlimited empiricism by individual physicians, either consultants or hospitalists. Our program represents a single health system with 2 acute care hospitals located 25 miles apart and which thus were similarly impacted by the different phases of the pandemic. Generalizability to health systems either smaller or larger, or in different geographical areas, has not been established. Data limitations have already been discussed.

We did not measure user satisfaction with the program either from physicians or nurses. However, the high rate of compliance suggests general agreement with the content and process.

We cannot definitively ascribe reduction in utilization of some nonrecommended treatments and increased utilization of the recommended therapies to the work of the CCMC. Individual physicians may have made these adjustments on their own or under the influence of other sources.

Finally, it should be noted that the mission to rapidly respond to data from well-conducted trials might be thwarted by too rigid a process or a committee’s lack of a sense of urgency. Organizing a committee and then empowering it to act is no guarantee of success; commitment to the mission is.

Conclusion

COVID-19 represented a challenge to medical staffs everywhere, inundating them with high volumes of acutely ill patients presenting with unfamiliar syndromes. Initial responses were characterized by idiosyncratic management approaches based on nontraditional sources of opinion and influences.

This report describes how a complex medical system brought order and standardization through a deliberative, but urgent, multidisciplinary committee with responsibility for planning, implementing, and monitoring standard approaches that eventually became evidence based. The composition of the committee and its scope of influence, limited to inpatient management, were important elements of success, allowing for better focus on the many treatment decisions. The important connection between the management committee and the system P&T committee, the clinical research effort, and teaching programs in both medicine and pharmacy are offered as exemplars of coordination. The data presented show success in achieving standardized, guideline-directed care. The approach is adoptable and suitable for similar emergencies in the future.

Acknowledgments: The authors thank Gary Scabis, Kip Waite, John Moxley, Angela Clubb, and David Woodley for their assistance in gathering data. We express appreciation and admiration for all our colleagues at the bedside.

Corresponding author: Barry R. Meisenberg, MD, Department of Medicine, Luminis Health, 2001 Medical Pkwy, Annapolis, MD 21401; meisenberg@AAHS.org.

Financial disclosures: None.

References

1. Gettleman J, Raj S, Kumar H. India’s health system cracks under the strain as coronavirus cases surge. The New York Times. April 22, 2021. https://www.nytimes.com/2021/04/21/world/asia/india-coronavirus-oxygen.html

2. Rappleye H, Lehren AW, Strickler L, Fitzpatrick S. ‘This system is doomed’: doctors, nurses sound off in NBC News coronavirus survey. NBC News. March 20, 2020. https://www.nbcnews.com/news/us-news/system-doomed-doctors-nurses-sound-nbc-news-coronavirus-survey-n1164841

3. Johns Hopkins Coronavirus Resource Center. Accessed January 5, 2022. https://coronavirus.jhu.edu/map.html

4. Fineberg HV. The toll of COVID-19. JAMA. 2020;324(15):1502-1503. doi:10.1001/jama.2020.20019

5. Meisenberg BR. Medical staffs response to COVID-19 ‘data’: have we misplaced our skeptic’s eye? Am J Med. 2021;134(2):151-152. doi:10.1016/j.amjmed.2020.09.013

6. McMahon JH, Lydeamore MH, Stewardson AJ. Bringing evidence from press release to the clinic in the era of COVID-19. J Antimicrob Chemother. 2021;76(3):547-549. doi:10.1093/jac/dkaa506

7. Rubin EJ, Baden LR, Morrissey S, Campion EW. Medical journals and the 2019-nCoV outbreak. N Engl J Med. 2020;382(9):866. doi:10.1056/NEJMe2001329

8. Liu F, Li L, Xu M, et al. Prognostic value of interleukin-6, C-reactive protein, and procalcitonin in patients with COVID-19. J Clin Virol. 2020;127:104370. doi:10.1016/j.jcv.2020.104370

9. Vincent MJ, Bergeron E, Benjannet S, et al. Chloroquine is a potent inhibitor of SARS coronavirus infection and spread. Virol J. 2005;2:69. doi:10.1186/1743-422X-2-69

10. Wang M, Cao R, Zhang L, et al. Remdesivir and chloroquine effectively inhibit the recently emerged novel coronavirus (2019-nCoV) in vitro. Cell Res. 2020;30:269-271. doi:10.1038/s41422-020-0282-0

11. RECOVERY Collaborative Group. Effect of hydroxychloroquine in hospitalized patients with Covid-19. N Engl J Med. 2020;383:2030-2040. doi:10.1056/NEJMoa2022926

12. RECOVERY Collaborative Group. Tocilizumab in patients admitted to hospital with COVID-19 (RECOVERY): preliminary results of a randomised, controlled, open-label, platform trial [preprint]. February 11, 2021. doi:10.1101/2021.02.11.21249258 https://www.medrxiv.org/content/10.1101/2021.02.11.21249258v1

13. REMAP-CAP Investigators. Interleukin-6 receptor antagonists in critically ill patients with COVID-19. N Engl J Med. 2021;384(16):1491-1502. doi:10.1056/NEJMoa2100433

14. National Institutes of Health. COVID-19 treatment guidelines: interleukin-6 inhibitors. https://www.covid19treatmentguidelines.nih.gov/immunomodulators/interleukin-6-inhibitors/

15. Deana C, Vetrugno L, Tonizzo A, et al. Drug supply during COVID-19 pandemic: remember not to run with your tank empty. Hosp Pharm. 2021;56(5):405-407. doi:10.1177/0018578720931749

16. Choe J, Crane M, Greene J, et al. The Pandemic and the Supply Chain: Addressing Gaps in Pharmaceutical Production and Distribution. Johns Hopkins University, November 2020. https://www.jhsph.edu/research/affiliated-programs/johns-hopkins-drug-access-and-affordability-initiative/publications/Pandemic_Supply_Chain.pdf

17. Kern DE. Overview: a six-step approach to curriculum development. In: Kern DE, Thornton PA, Hughes MT, eds. Curriculum Development for Medical Education: A Six-Step Approach. 3rd ed. Johns Hopkins University Press; 2016.

18. Rice TW, Janz DR. In defense of evidence-based medicine for the treatment of COVID-19 acute respiratory distress syndrome. Ann Am Thorac Soc. 2020;17(7):787-789. doi:10.1513/AnnalsATS.202004-325IP

19. Lucey CR, Johnston SC. The transformational effects of COVID-19 on medical education. JAMA. 2020;324(11):1033-1034. doi:10.1001/jama.2020.14136

20. National Institutes of Health. COVID-19 treatment guidelines: clinical spectrum of SARS-CoV-2 infection. https://www.covid19treatmentguidelines.nih.gov/overview/clinical-spectrum/

21. RECOVERY Collaborative Group. Dexamethasone in hospitalized patients with Covid-19. N Engl J Med. 2021;384:693-704. doi:10.1056/NEJMoa2021436

22. Beigel JH, Tomashek KM, Dodd LE, et al. Remdesivir for the treatment of Covid-19—final report. N Engl J Med. 2020;383:1813-1826. doi:10.1056/NEJMoa2007764

23. Jiminez D. Ivermectin and Covid-19: how a cheap antiparasitic became political. April 19, 2021. https://www.pharmaceutical-technology.com/features/ivermectin-covid-19-antiparasitic-political/

24. World Health Organization. WHO advises that ivermectin only be used to treat COVID-19 within clinical trials. March 31, 2021. https://www.who.int/news-room/feature-stories/detail/who-advises-that-ivermectin-only-be-used-to-treat-covid-19-within-clinical-trials

25. U.S. Food and Drug Administration. Why you should not use ivermectin to treat or prevent COVID-19. March 5, 2021. https://www.fda.gov/consumers/consumer-updates/why-you-should-not-use-ivermectin-treat-or-prevent-covid-19

26. Seymour CW, McCreary EK, Stegenga J. Sensible medicine-balancing intervention and inaction during the COVID-19 pandemic. JAMA. 2020;324(18):1827-1828. doi:10.1001/jama.2020.20271

27. Flanagin A, Fontanarosa PB, Bauchner H. Preprints involving medical research—do the benefits outweigh the challenges? JAMA. 2020;324(18):1840-1843. doi:10.1001/jama.2020.20674

28. Asch DA, Shells NE, Islam N, et al. Variation in US hospital mortality rates for patients admitted with COVID-19 during the first 6 months of the pandemic. JAMA Intern Med. 2021;181(4):471-478. doi:10.1001/jamainternmed.2020.8193

References

1. Gettleman J, Raj S, Kumar H. India’s health system cracks under the strain as coronavirus cases surge. The New York Times. April 22, 2021. https://www.nytimes.com/2021/04/21/world/asia/india-coronavirus-oxygen.html

2. Rappleye H, Lehren AW, Strickler L, Fitzpatrick S. ‘This system is doomed’: doctors, nurses sound off in NBC News coronavirus survey. NBC News. March 20, 2020. https://www.nbcnews.com/news/us-news/system-doomed-doctors-nurses-sound-nbc-news-coronavirus-survey-n1164841

3. Johns Hopkins Coronavirus Resource Center. Accessed January 5, 2022. https://coronavirus.jhu.edu/map.html

4. Fineberg HV. The toll of COVID-19. JAMA. 2020;324(15):1502-1503. doi:10.1001/jama.2020.20019

5. Meisenberg BR. Medical staffs response to COVID-19 ‘data’: have we misplaced our skeptic’s eye? Am J Med. 2021;134(2):151-152. doi:10.1016/j.amjmed.2020.09.013

6. McMahon JH, Lydeamore MH, Stewardson AJ. Bringing evidence from press release to the clinic in the era of COVID-19. J Antimicrob Chemother. 2021;76(3):547-549. doi:10.1093/jac/dkaa506

7. Rubin EJ, Baden LR, Morrissey S, Campion EW. Medical journals and the 2019-nCoV outbreak. N Engl J Med. 2020;382(9):866. doi:10.1056/NEJMe2001329

8. Liu F, Li L, Xu M, et al. Prognostic value of interleukin-6, C-reactive protein, and procalcitonin in patients with COVID-19. J Clin Virol. 2020;127:104370. doi:10.1016/j.jcv.2020.104370

9. Vincent MJ, Bergeron E, Benjannet S, et al. Chloroquine is a potent inhibitor of SARS coronavirus infection and spread. Virol J. 2005;2:69. doi:10.1186/1743-422X-2-69

10. Wang M, Cao R, Zhang L, et al. Remdesivir and chloroquine effectively inhibit the recently emerged novel coronavirus (2019-nCoV) in vitro. Cell Res. 2020;30:269-271. doi:10.1038/s41422-020-0282-0

11. RECOVERY Collaborative Group. Effect of hydroxychloroquine in hospitalized patients with Covid-19. N Engl J Med. 2020;383:2030-2040. doi:10.1056/NEJMoa2022926

12. RECOVERY Collaborative Group. Tocilizumab in patients admitted to hospital with COVID-19 (RECOVERY): preliminary results of a randomised, controlled, open-label, platform trial [preprint]. February 11, 2021. doi:10.1101/2021.02.11.21249258 https://www.medrxiv.org/content/10.1101/2021.02.11.21249258v1

13. REMAP-CAP Investigators. Interleukin-6 receptor antagonists in critically ill patients with COVID-19. N Engl J Med. 2021;384(16):1491-1502. doi:10.1056/NEJMoa2100433

14. National Institutes of Health. COVID-19 treatment guidelines: interleukin-6 inhibitors. https://www.covid19treatmentguidelines.nih.gov/immunomodulators/interleukin-6-inhibitors/

15. Deana C, Vetrugno L, Tonizzo A, et al. Drug supply during COVID-19 pandemic: remember not to run with your tank empty. Hosp Pharm. 2021;56(5):405-407. doi:10.1177/0018578720931749

16. Choe J, Crane M, Greene J, et al. The Pandemic and the Supply Chain: Addressing Gaps in Pharmaceutical Production and Distribution. Johns Hopkins University, November 2020. https://www.jhsph.edu/research/affiliated-programs/johns-hopkins-drug-access-and-affordability-initiative/publications/Pandemic_Supply_Chain.pdf

17. Kern DE. Overview: a six-step approach to curriculum development. In: Kern DE, Thornton PA, Hughes MT, eds. Curriculum Development for Medical Education: A Six-Step Approach. 3rd ed. Johns Hopkins University Press; 2016.

18. Rice TW, Janz DR. In defense of evidence-based medicine for the treatment of COVID-19 acute respiratory distress syndrome. Ann Am Thorac Soc. 2020;17(7):787-789. doi:10.1513/AnnalsATS.202004-325IP

19. Lucey CR, Johnston SC. The transformational effects of COVID-19 on medical education. JAMA. 2020;324(11):1033-1034. doi:10.1001/jama.2020.14136

20. National Institutes of Health. COVID-19 treatment guidelines: clinical spectrum of SARS-CoV-2 infection. https://www.covid19treatmentguidelines.nih.gov/overview/clinical-spectrum/

21. RECOVERY Collaborative Group. Dexamethasone in hospitalized patients with Covid-19. N Engl J Med. 2021;384:693-704. doi:10.1056/NEJMoa2021436

22. Beigel JH, Tomashek KM, Dodd LE, et al. Remdesivir for the treatment of Covid-19—final report. N Engl J Med. 2020;383:1813-1826. doi:10.1056/NEJMoa2007764

23. Jiminez D. Ivermectin and Covid-19: how a cheap antiparasitic became political. April 19, 2021. https://www.pharmaceutical-technology.com/features/ivermectin-covid-19-antiparasitic-political/

24. World Health Organization. WHO advises that ivermectin only be used to treat COVID-19 within clinical trials. March 31, 2021. https://www.who.int/news-room/feature-stories/detail/who-advises-that-ivermectin-only-be-used-to-treat-covid-19-within-clinical-trials

25. U.S. Food and Drug Administration. Why you should not use ivermectin to treat or prevent COVID-19. March 5, 2021. https://www.fda.gov/consumers/consumer-updates/why-you-should-not-use-ivermectin-treat-or-prevent-covid-19

26. Seymour CW, McCreary EK, Stegenga J. Sensible medicine-balancing intervention and inaction during the COVID-19 pandemic. JAMA. 2020;324(18):1827-1828. doi:10.1001/jama.2020.20271

27. Flanagin A, Fontanarosa PB, Bauchner H. Preprints involving medical research—do the benefits outweigh the challenges? JAMA. 2020;324(18):1840-1843. doi:10.1001/jama.2020.20674

28. Asch DA, Shells NE, Islam N, et al. Variation in US hospital mortality rates for patients admitted with COVID-19 during the first 6 months of the pandemic. JAMA Intern Med. 2021;181(4):471-478. doi:10.1001/jamainternmed.2020.8193

Issue
Journal of Clinical Outcomes Management - 29(1)
Issue
Journal of Clinical Outcomes Management - 29(1)
Page Number
39-48
Page Number
39-48
Publications
Publications
Topics
Article Type
Display Headline
A COVID-19 Clinical Management Committee to Standardize Care in a 2-Hospital System
Display Headline
A COVID-19 Clinical Management Committee to Standardize Care in a 2-Hospital System
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

Assessment of Same-Day Naloxone Availability in New Mexico Pharmacies

Article Type
Changed
Thu, 12/02/2021 - 10:55
Display Headline
Assessment of Same-Day Naloxone Availability in New Mexico Pharmacies

From the Department of Medicine, University of California San Diego (Dr. Haponyuk), Department of Emergency Medicine, University of Tennessee (Dr. Dejong), the Department of Family Medicine, University of New Mexico (Dr. Gutfrucht), and the Department of Internal Medicine, University of New Mexico (Dr. Barrett)

Objective: Naloxone availability can reduce the risk of death from opioid overdoses, although prescriber, legislative, and payment barriers to accessing this life-saving medication exist. A previously underreported barrier involves same-day availability, the lack of which may force patients to travel to multiple pharmacies and having delays in access or risking not filling their prescription. This study sought to determine same-day availability of naloxone in pharmacies in the state of New Mexico.

Methods: Same-day availability of naloxone was assessed via an audit survey.

Results: Of the 183 pharamacies screened, only 84.7% had same-day availability, including only 72% in Albuquerque, the state’s most populous city/municipality.

Conclusion: These results highlight the extent of a previously underexplored challenge to patient care and barrier to patient safety, and future directions for more patient-centered care.

Keywords: naloxone; barriers to care; opioid overdose prevention.

The US is enduring an ongoing epidemic of deaths due to opioid use, which have increased in frequency since the onset of the COVID-19 pandemic.1 One strategy to reduce the risk of mortality from opioid use is to ensure the widespread availability of naloxone. Individual states have implemented harm reduction strategies to increase access to naloxone, including improving availability via a statewide standing order that it may be dispensed without a prescription.2,3 Such naloxone access laws are being widely adopted and are believed to reduce overdose deaths.4

There are many barriers to patients receiving naloxone despite their clinicians providing a prescription for it, including stigmatization, financial cost, and local availability.5-9 However, the stigma associated with naloxone extends to both patients and pharmacists. Pharmacists in West Virginia, for example, showed widespread concerns about having naloxone available for patients to purchase over the counter, for fear that increasing naloxone access may increase overdoses.6 A study in Tennessee also found pharmacists hesitant to recommend naloxone.7 Another study of rural pharmacies in Georgia found that just over half carried naloxone despite a state law that naloxone be available without a prescription.8 Challenges are not limited to rural areas, however; a study in Philadelphia found that more than one-third of pharmacies required a prescription to dispense naloxone, contrary to state law.9 Thus, in a rapidly changing regulatory environment, there are many evolving barriers to patients receiving naloxone.

 

 

New Mexico has an opioid overdose rate higher than the national average, coming in 15th out of 50 states when last ranked in 2018, with overdose rates that vary across demographic variables.10 Consequently, New Mexico state law added language requiring clinicians prescribing opioids for 5 days or longer to co-prescribe naloxone along with written information on how to administer the opioid antagonist.11 New Mexico is also a geographically large state with a relatively low overall population characterized by striking health disparities, particularly as related to access to care.

The purpose of this study is to describe the same-day availability of naloxone throughout the state of New Mexico after a change in state law requiring co-prescription was enacted, to help identify challenges to patients receiving it. Comprehensive examination of barriers to patients accessing this life-saving medication can advise strategies to both improve patient-centered care and potentially reduce deaths.

Methods

To better understand barriers to patients obtaining naloxone, in July and August of 2019 we performed an audit (“secret shopper”) study of all pharmacies in the state, posing as patients wishing to obtain naloxone. A publicly available list of every pharmacy in New Mexico was used to identify 89 pharmacies in Albuquerque (the most populous city in New Mexico) and 106 pharmacies throughout the rest of the state.12

Every pharmacy was called via a publicly available phone number during business hours (confirmed via an internet search), at least 2 hours prior to closing. One of 3 researchers telephoned pharmacies posing as a patient and inquired whether naloxone would be available for pick up the same day. If the pharmacy confirmed it was available that day, the call concluded. If naloxone was unavailable for same day pick up, researchers asked when it would be next available. Each pharmacy was called once, and neither insurance information nor cost was offered or requested. All questions were asked in English by native English speakers.

All responses were recorded in a secure spreadsheet. Once all responses were received and reviewed, they were characterized in discrete response categories: same day, within 1 to 2 days, within 3 to 4 days, within a week, or unsure/unknown. Naloxone availability was also tracked by city/municipality, and this was compared to the state’s population distribution.

 

 

No personally identifiable information was obtained. This study was Institutional Review Board exempt.

tables and figures for article

Results

Responses were recorded from 183 pharmacies. Seventeen locations were eliminated from our analysis because their phone system was inoperable or the pharmacy was permanently closed. Of the pharmacies reached, 84.7% (155/183) reported they have naloxone available for pick up on the same day (Figure 1). Of the 15.3% (28) pharmacies that did not have same-day availability, 60.7% (17 pharmacies) reported availability in 1 to 2 days, 3.6% had availability in 3 to 4 days, 3.6% had availability in 1 week, and 32.1% were unsure of next availability (Figure 2). More than one-third of the state’s patients reside in municipalities where naloxone is immediately available in at least 72% of pharmacies (Table).13

tables and figures for article

Discussion

Increased access to naloxone at the state and community level is associated with reduced risk for death from overdose, and, consequently, widespread availability is recommended.14-17 Statewide real-time pharmacy availability of naloxone—as patients would experience availability—has not been previously reported. These findings suggest unpredictable same-day availability that may affect experience and care outcomes. That other studies have found similar challenges in naloxone availability in other municipalities and regions suggests this barrier to access is widespread,6-9 and likely affects patients throughout the country.

tables and figures for article

Many patients have misgivings about naloxone, and it places an undue burden on them to travel to multiple pharmacies or take repeated trips to fill prescriptions. Additionally, patients without reliable transportation may be unable to return at a later date. Although we found most pharmacies in New Mexico without immediate availability of naloxone reported they could have it within several days, such a delay may reduce the likelihood that patients will fill their prescription at all. It is also concerning that many pharmacies are unsure of when naloxone will be available, particularly when some of these may be the only pharmacy easily accessible to patients or the one where they regularly fill their prescriptions.

Barriers to naloxone availability requires further study due to possible negative consequences for patient safety and risks for exacerbating health disparities among vulnerable populations. Further research may focus on examining the effects on patients when naloxone dispensing is delayed or impossible, why there is variability in naloxone availability between different pharmacies and municipalities, the reasons for uncertainty when naloxone will be available, and effective solutions. Expanded naloxone distribution in community locations and in clinics offers one potential patient-centered solution that should be explored, but it is likely that more widespread and systemic solutions will require policy and regulatory changes at the state and national levels.

 

 

Limitations of this study include that the findings may be relevant for solely 1 state, such as in the case of state-specific barriers to keeping naloxone in stock that we are unaware of. However, it is unclear why that would be the case, and it is more likely that similar barriers are pervasive. Additionally, repeat phone calls, which we did not follow up with, may have yielded more pharmacies with naloxone availability. However, due to the stigma associated with obtaining naloxone, it may be that patients will not make multiple calls either—highlighting how important real-time availability is.

Conclusion

Urgent solutions are needed to address the epidemic of deaths from opioid overdoses. Naloxone availability is an important tool for reducing these deaths, resulting in numerous state laws attempting to increase access. Despite this, there are persistent barriers to patients receiving naloxone, including a lack of same-day availability at pharmacies. Our results suggest that this underexplored barrier is widespread. Improving both availability and accessibility of naloxone may include legislative policy solutions as well as patient-oriented solutions, such as distribution in clinics and hospitals when opioid prescriptions are first written. Further research should be conducted to determine patient-centered, effective solutions that can improve outcomes.

Corresponding author: Eileen Barrett, MD, MPH, Department of Internal Medicine, University of New Mexico; ebarrett@salud.unm.edu.

Financial disclosures: None.

References

1. Mason M, Welch SB, Arunkumar P, et al. Notes from the field: opioid overdose deaths before, during, and after an 11-week COVID-19 stay-at-home order—Cook County, Illinois, January 1, 2018–October 6, 2020. MMWR Morb Mortal Wkly Rep. 2021;70(10):362-363. doi:10.15585/mmwr.mm7010a3

2. Kaiser Family Foundation. Opioid overdose death rates and all drug overdose death rates per 100,000 population (age-adjusted). Accessed October 6, 2021. https://www.kff.org/other/state-indicator/opioid-overdose-death

3. Sohn M, Talbert JC, Huang Z, et al. Association of naloxone coprescription laws with naloxone prescription dispensing in the United States. JAMA Netw Open. 2019;2(6):e196215. doi:10.1001/jamanetworkopen.2019.6215

4. Smart R, Pardo B, Davis CS. Systematic review of the emerging literature on the effectiveness of naloxone access laws in the United States. Addiction. 2021;116(1):6-17. doi:10.1111/add.15163

5. Mueller SR, Koester S, Glanz JM, et al. Attitudes toward naloxone prescribing in clinical settings: a qualitative study of patients prescribed high dose opioids for chronic non-cancer pain. J Gen Intern Med. 2017;32(3):277-283. doi:10.1007/s11606-016-3895-8

6. Thornton JD, Lyvers E, Scott VGG, Dwibedi N. Pharmacists’ readiness to provide naloxone in community pharmacies in West Virginia. J Am Pharm Assoc (2003). 2017;57(2S):S12-S18.e4. doi:10.1016/j.japh.2016.12.070

7. Spivey C, Wilder A, Chisholm-Burns MA, et al. Evaluation of naloxone access, pricing, and barriers to dispensing in Tennessee retail community pharmacies. J Am Pharm Assoc (2003). 2020;60(5):694-701.e1. doi:10.1016/j.japh.2020.01.030

8. Nguyen JL, Gilbert LR, Beasley L, et al. Availability of naloxone at rural Georgia pharmacies, 2019. JAMA Netw Open. 2020;3(2):e1921227. doi:10.1001/jamanetworkopen.2019.21227

9. Guadamuz JS, Alexander GC, Chaudhri T, et al. Availability and cost of naloxone nasal spray at pharmacies in Philadelphia, Pennsylvania. JAMA Netw Open. 2019;2(6):e195388. doi:10.1001/jamanetworkopen.2019.5388

10. Edge K. Changes in drug overdose mortality in New Mexico. New Mexico Epidemiology. July 2020 (3). https://www.nmhealth.org/data/view/report/2402/

11. Senate Bill 221. 54th Legislature, State of New Mexico, First Session, 2019 (introduced by William P. Soules). Accessed October 6, 2021. https://nmlegis.gov/Sessions/19%20Regular/bills/senate/SB0221.pdf

12. GoodRx. Find pharmacies in New Mexico. Accessed October 6, 2021. https://www.goodrx.com/pharmacy-near-me/all/nm

13. U.S. Census Bureau. QuickFacts: New Mexico. Accessed October 6, 2021. https://www.census.gov/quickfacts/NM

14. Linas BP, Savinkina A, Madushani RWMA, et al. Projected estimates of opioid mortality after community-level interventions. JAMA Netw Open. 2021;4(2):e2037259. doi:10.1001/jamanetworkopen.2020.37259

15. You HS, Ha J, Kang CY, et al. Regional variation in states’ naloxone accessibility laws in association with opioid overdose death rates—observational study (STROBE compliant). Medicine (Baltimore). 2020;99(22):e20033. doi:10.1097/MD.0000000000020033

16. Pew Charitable Trusts. Expanded access to naloxone can curb opioid overdose deaths. October 20, 2020. Accessed October 6, 2021. https://www.pewtrusts.org/en/research-and-analysis/issue-briefs/2020/10/expanded-access-to-naloxone-can-curb-opioid-overdose-deaths

17. Centers for Disease Control and Prevention. Still not enough naloxone where it’s most needed. August 6, 2019. Accessed October 6, 2021. https://www.cdc.gov/media/releases/2019/p0806-naloxone.html

Article PDF
Issue
Journal of Clinical Outcomes Management - 28(6)
Publications
Topics
Page Number
268-271
Sections
Article PDF
Article PDF

From the Department of Medicine, University of California San Diego (Dr. Haponyuk), Department of Emergency Medicine, University of Tennessee (Dr. Dejong), the Department of Family Medicine, University of New Mexico (Dr. Gutfrucht), and the Department of Internal Medicine, University of New Mexico (Dr. Barrett)

Objective: Naloxone availability can reduce the risk of death from opioid overdoses, although prescriber, legislative, and payment barriers to accessing this life-saving medication exist. A previously underreported barrier involves same-day availability, the lack of which may force patients to travel to multiple pharmacies and having delays in access or risking not filling their prescription. This study sought to determine same-day availability of naloxone in pharmacies in the state of New Mexico.

Methods: Same-day availability of naloxone was assessed via an audit survey.

Results: Of the 183 pharamacies screened, only 84.7% had same-day availability, including only 72% in Albuquerque, the state’s most populous city/municipality.

Conclusion: These results highlight the extent of a previously underexplored challenge to patient care and barrier to patient safety, and future directions for more patient-centered care.

Keywords: naloxone; barriers to care; opioid overdose prevention.

The US is enduring an ongoing epidemic of deaths due to opioid use, which have increased in frequency since the onset of the COVID-19 pandemic.1 One strategy to reduce the risk of mortality from opioid use is to ensure the widespread availability of naloxone. Individual states have implemented harm reduction strategies to increase access to naloxone, including improving availability via a statewide standing order that it may be dispensed without a prescription.2,3 Such naloxone access laws are being widely adopted and are believed to reduce overdose deaths.4

There are many barriers to patients receiving naloxone despite their clinicians providing a prescription for it, including stigmatization, financial cost, and local availability.5-9 However, the stigma associated with naloxone extends to both patients and pharmacists. Pharmacists in West Virginia, for example, showed widespread concerns about having naloxone available for patients to purchase over the counter, for fear that increasing naloxone access may increase overdoses.6 A study in Tennessee also found pharmacists hesitant to recommend naloxone.7 Another study of rural pharmacies in Georgia found that just over half carried naloxone despite a state law that naloxone be available without a prescription.8 Challenges are not limited to rural areas, however; a study in Philadelphia found that more than one-third of pharmacies required a prescription to dispense naloxone, contrary to state law.9 Thus, in a rapidly changing regulatory environment, there are many evolving barriers to patients receiving naloxone.

 

 

New Mexico has an opioid overdose rate higher than the national average, coming in 15th out of 50 states when last ranked in 2018, with overdose rates that vary across demographic variables.10 Consequently, New Mexico state law added language requiring clinicians prescribing opioids for 5 days or longer to co-prescribe naloxone along with written information on how to administer the opioid antagonist.11 New Mexico is also a geographically large state with a relatively low overall population characterized by striking health disparities, particularly as related to access to care.

The purpose of this study is to describe the same-day availability of naloxone throughout the state of New Mexico after a change in state law requiring co-prescription was enacted, to help identify challenges to patients receiving it. Comprehensive examination of barriers to patients accessing this life-saving medication can advise strategies to both improve patient-centered care and potentially reduce deaths.

Methods

To better understand barriers to patients obtaining naloxone, in July and August of 2019 we performed an audit (“secret shopper”) study of all pharmacies in the state, posing as patients wishing to obtain naloxone. A publicly available list of every pharmacy in New Mexico was used to identify 89 pharmacies in Albuquerque (the most populous city in New Mexico) and 106 pharmacies throughout the rest of the state.12

Every pharmacy was called via a publicly available phone number during business hours (confirmed via an internet search), at least 2 hours prior to closing. One of 3 researchers telephoned pharmacies posing as a patient and inquired whether naloxone would be available for pick up the same day. If the pharmacy confirmed it was available that day, the call concluded. If naloxone was unavailable for same day pick up, researchers asked when it would be next available. Each pharmacy was called once, and neither insurance information nor cost was offered or requested. All questions were asked in English by native English speakers.

All responses were recorded in a secure spreadsheet. Once all responses were received and reviewed, they were characterized in discrete response categories: same day, within 1 to 2 days, within 3 to 4 days, within a week, or unsure/unknown. Naloxone availability was also tracked by city/municipality, and this was compared to the state’s population distribution.

 

 

No personally identifiable information was obtained. This study was Institutional Review Board exempt.

tables and figures for article

Results

Responses were recorded from 183 pharmacies. Seventeen locations were eliminated from our analysis because their phone system was inoperable or the pharmacy was permanently closed. Of the pharmacies reached, 84.7% (155/183) reported they have naloxone available for pick up on the same day (Figure 1). Of the 15.3% (28) pharmacies that did not have same-day availability, 60.7% (17 pharmacies) reported availability in 1 to 2 days, 3.6% had availability in 3 to 4 days, 3.6% had availability in 1 week, and 32.1% were unsure of next availability (Figure 2). More than one-third of the state’s patients reside in municipalities where naloxone is immediately available in at least 72% of pharmacies (Table).13

tables and figures for article

Discussion

Increased access to naloxone at the state and community level is associated with reduced risk for death from overdose, and, consequently, widespread availability is recommended.14-17 Statewide real-time pharmacy availability of naloxone—as patients would experience availability—has not been previously reported. These findings suggest unpredictable same-day availability that may affect experience and care outcomes. That other studies have found similar challenges in naloxone availability in other municipalities and regions suggests this barrier to access is widespread,6-9 and likely affects patients throughout the country.

tables and figures for article

Many patients have misgivings about naloxone, and it places an undue burden on them to travel to multiple pharmacies or take repeated trips to fill prescriptions. Additionally, patients without reliable transportation may be unable to return at a later date. Although we found most pharmacies in New Mexico without immediate availability of naloxone reported they could have it within several days, such a delay may reduce the likelihood that patients will fill their prescription at all. It is also concerning that many pharmacies are unsure of when naloxone will be available, particularly when some of these may be the only pharmacy easily accessible to patients or the one where they regularly fill their prescriptions.

Barriers to naloxone availability requires further study due to possible negative consequences for patient safety and risks for exacerbating health disparities among vulnerable populations. Further research may focus on examining the effects on patients when naloxone dispensing is delayed or impossible, why there is variability in naloxone availability between different pharmacies and municipalities, the reasons for uncertainty when naloxone will be available, and effective solutions. Expanded naloxone distribution in community locations and in clinics offers one potential patient-centered solution that should be explored, but it is likely that more widespread and systemic solutions will require policy and regulatory changes at the state and national levels.

 

 

Limitations of this study include that the findings may be relevant for solely 1 state, such as in the case of state-specific barriers to keeping naloxone in stock that we are unaware of. However, it is unclear why that would be the case, and it is more likely that similar barriers are pervasive. Additionally, repeat phone calls, which we did not follow up with, may have yielded more pharmacies with naloxone availability. However, due to the stigma associated with obtaining naloxone, it may be that patients will not make multiple calls either—highlighting how important real-time availability is.

Conclusion

Urgent solutions are needed to address the epidemic of deaths from opioid overdoses. Naloxone availability is an important tool for reducing these deaths, resulting in numerous state laws attempting to increase access. Despite this, there are persistent barriers to patients receiving naloxone, including a lack of same-day availability at pharmacies. Our results suggest that this underexplored barrier is widespread. Improving both availability and accessibility of naloxone may include legislative policy solutions as well as patient-oriented solutions, such as distribution in clinics and hospitals when opioid prescriptions are first written. Further research should be conducted to determine patient-centered, effective solutions that can improve outcomes.

Corresponding author: Eileen Barrett, MD, MPH, Department of Internal Medicine, University of New Mexico; ebarrett@salud.unm.edu.

Financial disclosures: None.

From the Department of Medicine, University of California San Diego (Dr. Haponyuk), Department of Emergency Medicine, University of Tennessee (Dr. Dejong), the Department of Family Medicine, University of New Mexico (Dr. Gutfrucht), and the Department of Internal Medicine, University of New Mexico (Dr. Barrett)

Objective: Naloxone availability can reduce the risk of death from opioid overdoses, although prescriber, legislative, and payment barriers to accessing this life-saving medication exist. A previously underreported barrier involves same-day availability, the lack of which may force patients to travel to multiple pharmacies and having delays in access or risking not filling their prescription. This study sought to determine same-day availability of naloxone in pharmacies in the state of New Mexico.

Methods: Same-day availability of naloxone was assessed via an audit survey.

Results: Of the 183 pharamacies screened, only 84.7% had same-day availability, including only 72% in Albuquerque, the state’s most populous city/municipality.

Conclusion: These results highlight the extent of a previously underexplored challenge to patient care and barrier to patient safety, and future directions for more patient-centered care.

Keywords: naloxone; barriers to care; opioid overdose prevention.

The US is enduring an ongoing epidemic of deaths due to opioid use, which have increased in frequency since the onset of the COVID-19 pandemic.1 One strategy to reduce the risk of mortality from opioid use is to ensure the widespread availability of naloxone. Individual states have implemented harm reduction strategies to increase access to naloxone, including improving availability via a statewide standing order that it may be dispensed without a prescription.2,3 Such naloxone access laws are being widely adopted and are believed to reduce overdose deaths.4

There are many barriers to patients receiving naloxone despite their clinicians providing a prescription for it, including stigmatization, financial cost, and local availability.5-9 However, the stigma associated with naloxone extends to both patients and pharmacists. Pharmacists in West Virginia, for example, showed widespread concerns about having naloxone available for patients to purchase over the counter, for fear that increasing naloxone access may increase overdoses.6 A study in Tennessee also found pharmacists hesitant to recommend naloxone.7 Another study of rural pharmacies in Georgia found that just over half carried naloxone despite a state law that naloxone be available without a prescription.8 Challenges are not limited to rural areas, however; a study in Philadelphia found that more than one-third of pharmacies required a prescription to dispense naloxone, contrary to state law.9 Thus, in a rapidly changing regulatory environment, there are many evolving barriers to patients receiving naloxone.

 

 

New Mexico has an opioid overdose rate higher than the national average, coming in 15th out of 50 states when last ranked in 2018, with overdose rates that vary across demographic variables.10 Consequently, New Mexico state law added language requiring clinicians prescribing opioids for 5 days or longer to co-prescribe naloxone along with written information on how to administer the opioid antagonist.11 New Mexico is also a geographically large state with a relatively low overall population characterized by striking health disparities, particularly as related to access to care.

The purpose of this study is to describe the same-day availability of naloxone throughout the state of New Mexico after a change in state law requiring co-prescription was enacted, to help identify challenges to patients receiving it. Comprehensive examination of barriers to patients accessing this life-saving medication can advise strategies to both improve patient-centered care and potentially reduce deaths.

Methods

To better understand barriers to patients obtaining naloxone, in July and August of 2019 we performed an audit (“secret shopper”) study of all pharmacies in the state, posing as patients wishing to obtain naloxone. A publicly available list of every pharmacy in New Mexico was used to identify 89 pharmacies in Albuquerque (the most populous city in New Mexico) and 106 pharmacies throughout the rest of the state.12

Every pharmacy was called via a publicly available phone number during business hours (confirmed via an internet search), at least 2 hours prior to closing. One of 3 researchers telephoned pharmacies posing as a patient and inquired whether naloxone would be available for pick up the same day. If the pharmacy confirmed it was available that day, the call concluded. If naloxone was unavailable for same day pick up, researchers asked when it would be next available. Each pharmacy was called once, and neither insurance information nor cost was offered or requested. All questions were asked in English by native English speakers.

All responses were recorded in a secure spreadsheet. Once all responses were received and reviewed, they were characterized in discrete response categories: same day, within 1 to 2 days, within 3 to 4 days, within a week, or unsure/unknown. Naloxone availability was also tracked by city/municipality, and this was compared to the state’s population distribution.

 

 

No personally identifiable information was obtained. This study was Institutional Review Board exempt.

tables and figures for article

Results

Responses were recorded from 183 pharmacies. Seventeen locations were eliminated from our analysis because their phone system was inoperable or the pharmacy was permanently closed. Of the pharmacies reached, 84.7% (155/183) reported they have naloxone available for pick up on the same day (Figure 1). Of the 15.3% (28) pharmacies that did not have same-day availability, 60.7% (17 pharmacies) reported availability in 1 to 2 days, 3.6% had availability in 3 to 4 days, 3.6% had availability in 1 week, and 32.1% were unsure of next availability (Figure 2). More than one-third of the state’s patients reside in municipalities where naloxone is immediately available in at least 72% of pharmacies (Table).13

tables and figures for article

Discussion

Increased access to naloxone at the state and community level is associated with reduced risk for death from overdose, and, consequently, widespread availability is recommended.14-17 Statewide real-time pharmacy availability of naloxone—as patients would experience availability—has not been previously reported. These findings suggest unpredictable same-day availability that may affect experience and care outcomes. That other studies have found similar challenges in naloxone availability in other municipalities and regions suggests this barrier to access is widespread,6-9 and likely affects patients throughout the country.

tables and figures for article

Many patients have misgivings about naloxone, and it places an undue burden on them to travel to multiple pharmacies or take repeated trips to fill prescriptions. Additionally, patients without reliable transportation may be unable to return at a later date. Although we found most pharmacies in New Mexico without immediate availability of naloxone reported they could have it within several days, such a delay may reduce the likelihood that patients will fill their prescription at all. It is also concerning that many pharmacies are unsure of when naloxone will be available, particularly when some of these may be the only pharmacy easily accessible to patients or the one where they regularly fill their prescriptions.

Barriers to naloxone availability requires further study due to possible negative consequences for patient safety and risks for exacerbating health disparities among vulnerable populations. Further research may focus on examining the effects on patients when naloxone dispensing is delayed or impossible, why there is variability in naloxone availability between different pharmacies and municipalities, the reasons for uncertainty when naloxone will be available, and effective solutions. Expanded naloxone distribution in community locations and in clinics offers one potential patient-centered solution that should be explored, but it is likely that more widespread and systemic solutions will require policy and regulatory changes at the state and national levels.

 

 

Limitations of this study include that the findings may be relevant for solely 1 state, such as in the case of state-specific barriers to keeping naloxone in stock that we are unaware of. However, it is unclear why that would be the case, and it is more likely that similar barriers are pervasive. Additionally, repeat phone calls, which we did not follow up with, may have yielded more pharmacies with naloxone availability. However, due to the stigma associated with obtaining naloxone, it may be that patients will not make multiple calls either—highlighting how important real-time availability is.

Conclusion

Urgent solutions are needed to address the epidemic of deaths from opioid overdoses. Naloxone availability is an important tool for reducing these deaths, resulting in numerous state laws attempting to increase access. Despite this, there are persistent barriers to patients receiving naloxone, including a lack of same-day availability at pharmacies. Our results suggest that this underexplored barrier is widespread. Improving both availability and accessibility of naloxone may include legislative policy solutions as well as patient-oriented solutions, such as distribution in clinics and hospitals when opioid prescriptions are first written. Further research should be conducted to determine patient-centered, effective solutions that can improve outcomes.

Corresponding author: Eileen Barrett, MD, MPH, Department of Internal Medicine, University of New Mexico; ebarrett@salud.unm.edu.

Financial disclosures: None.

References

1. Mason M, Welch SB, Arunkumar P, et al. Notes from the field: opioid overdose deaths before, during, and after an 11-week COVID-19 stay-at-home order—Cook County, Illinois, January 1, 2018–October 6, 2020. MMWR Morb Mortal Wkly Rep. 2021;70(10):362-363. doi:10.15585/mmwr.mm7010a3

2. Kaiser Family Foundation. Opioid overdose death rates and all drug overdose death rates per 100,000 population (age-adjusted). Accessed October 6, 2021. https://www.kff.org/other/state-indicator/opioid-overdose-death

3. Sohn M, Talbert JC, Huang Z, et al. Association of naloxone coprescription laws with naloxone prescription dispensing in the United States. JAMA Netw Open. 2019;2(6):e196215. doi:10.1001/jamanetworkopen.2019.6215

4. Smart R, Pardo B, Davis CS. Systematic review of the emerging literature on the effectiveness of naloxone access laws in the United States. Addiction. 2021;116(1):6-17. doi:10.1111/add.15163

5. Mueller SR, Koester S, Glanz JM, et al. Attitudes toward naloxone prescribing in clinical settings: a qualitative study of patients prescribed high dose opioids for chronic non-cancer pain. J Gen Intern Med. 2017;32(3):277-283. doi:10.1007/s11606-016-3895-8

6. Thornton JD, Lyvers E, Scott VGG, Dwibedi N. Pharmacists’ readiness to provide naloxone in community pharmacies in West Virginia. J Am Pharm Assoc (2003). 2017;57(2S):S12-S18.e4. doi:10.1016/j.japh.2016.12.070

7. Spivey C, Wilder A, Chisholm-Burns MA, et al. Evaluation of naloxone access, pricing, and barriers to dispensing in Tennessee retail community pharmacies. J Am Pharm Assoc (2003). 2020;60(5):694-701.e1. doi:10.1016/j.japh.2020.01.030

8. Nguyen JL, Gilbert LR, Beasley L, et al. Availability of naloxone at rural Georgia pharmacies, 2019. JAMA Netw Open. 2020;3(2):e1921227. doi:10.1001/jamanetworkopen.2019.21227

9. Guadamuz JS, Alexander GC, Chaudhri T, et al. Availability and cost of naloxone nasal spray at pharmacies in Philadelphia, Pennsylvania. JAMA Netw Open. 2019;2(6):e195388. doi:10.1001/jamanetworkopen.2019.5388

10. Edge K. Changes in drug overdose mortality in New Mexico. New Mexico Epidemiology. July 2020 (3). https://www.nmhealth.org/data/view/report/2402/

11. Senate Bill 221. 54th Legislature, State of New Mexico, First Session, 2019 (introduced by William P. Soules). Accessed October 6, 2021. https://nmlegis.gov/Sessions/19%20Regular/bills/senate/SB0221.pdf

12. GoodRx. Find pharmacies in New Mexico. Accessed October 6, 2021. https://www.goodrx.com/pharmacy-near-me/all/nm

13. U.S. Census Bureau. QuickFacts: New Mexico. Accessed October 6, 2021. https://www.census.gov/quickfacts/NM

14. Linas BP, Savinkina A, Madushani RWMA, et al. Projected estimates of opioid mortality after community-level interventions. JAMA Netw Open. 2021;4(2):e2037259. doi:10.1001/jamanetworkopen.2020.37259

15. You HS, Ha J, Kang CY, et al. Regional variation in states’ naloxone accessibility laws in association with opioid overdose death rates—observational study (STROBE compliant). Medicine (Baltimore). 2020;99(22):e20033. doi:10.1097/MD.0000000000020033

16. Pew Charitable Trusts. Expanded access to naloxone can curb opioid overdose deaths. October 20, 2020. Accessed October 6, 2021. https://www.pewtrusts.org/en/research-and-analysis/issue-briefs/2020/10/expanded-access-to-naloxone-can-curb-opioid-overdose-deaths

17. Centers for Disease Control and Prevention. Still not enough naloxone where it’s most needed. August 6, 2019. Accessed October 6, 2021. https://www.cdc.gov/media/releases/2019/p0806-naloxone.html

References

1. Mason M, Welch SB, Arunkumar P, et al. Notes from the field: opioid overdose deaths before, during, and after an 11-week COVID-19 stay-at-home order—Cook County, Illinois, January 1, 2018–October 6, 2020. MMWR Morb Mortal Wkly Rep. 2021;70(10):362-363. doi:10.15585/mmwr.mm7010a3

2. Kaiser Family Foundation. Opioid overdose death rates and all drug overdose death rates per 100,000 population (age-adjusted). Accessed October 6, 2021. https://www.kff.org/other/state-indicator/opioid-overdose-death

3. Sohn M, Talbert JC, Huang Z, et al. Association of naloxone coprescription laws with naloxone prescription dispensing in the United States. JAMA Netw Open. 2019;2(6):e196215. doi:10.1001/jamanetworkopen.2019.6215

4. Smart R, Pardo B, Davis CS. Systematic review of the emerging literature on the effectiveness of naloxone access laws in the United States. Addiction. 2021;116(1):6-17. doi:10.1111/add.15163

5. Mueller SR, Koester S, Glanz JM, et al. Attitudes toward naloxone prescribing in clinical settings: a qualitative study of patients prescribed high dose opioids for chronic non-cancer pain. J Gen Intern Med. 2017;32(3):277-283. doi:10.1007/s11606-016-3895-8

6. Thornton JD, Lyvers E, Scott VGG, Dwibedi N. Pharmacists’ readiness to provide naloxone in community pharmacies in West Virginia. J Am Pharm Assoc (2003). 2017;57(2S):S12-S18.e4. doi:10.1016/j.japh.2016.12.070

7. Spivey C, Wilder A, Chisholm-Burns MA, et al. Evaluation of naloxone access, pricing, and barriers to dispensing in Tennessee retail community pharmacies. J Am Pharm Assoc (2003). 2020;60(5):694-701.e1. doi:10.1016/j.japh.2020.01.030

8. Nguyen JL, Gilbert LR, Beasley L, et al. Availability of naloxone at rural Georgia pharmacies, 2019. JAMA Netw Open. 2020;3(2):e1921227. doi:10.1001/jamanetworkopen.2019.21227

9. Guadamuz JS, Alexander GC, Chaudhri T, et al. Availability and cost of naloxone nasal spray at pharmacies in Philadelphia, Pennsylvania. JAMA Netw Open. 2019;2(6):e195388. doi:10.1001/jamanetworkopen.2019.5388

10. Edge K. Changes in drug overdose mortality in New Mexico. New Mexico Epidemiology. July 2020 (3). https://www.nmhealth.org/data/view/report/2402/

11. Senate Bill 221. 54th Legislature, State of New Mexico, First Session, 2019 (introduced by William P. Soules). Accessed October 6, 2021. https://nmlegis.gov/Sessions/19%20Regular/bills/senate/SB0221.pdf

12. GoodRx. Find pharmacies in New Mexico. Accessed October 6, 2021. https://www.goodrx.com/pharmacy-near-me/all/nm

13. U.S. Census Bureau. QuickFacts: New Mexico. Accessed October 6, 2021. https://www.census.gov/quickfacts/NM

14. Linas BP, Savinkina A, Madushani RWMA, et al. Projected estimates of opioid mortality after community-level interventions. JAMA Netw Open. 2021;4(2):e2037259. doi:10.1001/jamanetworkopen.2020.37259

15. You HS, Ha J, Kang CY, et al. Regional variation in states’ naloxone accessibility laws in association with opioid overdose death rates—observational study (STROBE compliant). Medicine (Baltimore). 2020;99(22):e20033. doi:10.1097/MD.0000000000020033

16. Pew Charitable Trusts. Expanded access to naloxone can curb opioid overdose deaths. October 20, 2020. Accessed October 6, 2021. https://www.pewtrusts.org/en/research-and-analysis/issue-briefs/2020/10/expanded-access-to-naloxone-can-curb-opioid-overdose-deaths

17. Centers for Disease Control and Prevention. Still not enough naloxone where it’s most needed. August 6, 2019. Accessed October 6, 2021. https://www.cdc.gov/media/releases/2019/p0806-naloxone.html

Issue
Journal of Clinical Outcomes Management - 28(6)
Issue
Journal of Clinical Outcomes Management - 28(6)
Page Number
268-271
Page Number
268-271
Publications
Publications
Topics
Article Type
Display Headline
Assessment of Same-Day Naloxone Availability in New Mexico Pharmacies
Display Headline
Assessment of Same-Day Naloxone Availability in New Mexico Pharmacies
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

Adjuvant Olaparib Improves Outcomes in High-Risk, HER2-Negative Early Breast Cancer Patients With Germline BRCA1 and BRCA2 Mutations

Article Type
Changed
Tue, 11/23/2021 - 15:03
Display Headline
Adjuvant Olaparib Improves Outcomes in High-Risk, HER2-Negative Early Breast Cancer Patients With Germline BRCA1 and BRCA2 Mutations

Study Overview

Objective. To assess the efficacy and safety of olaparib as an adjuvant treatment in patients with BRCA1 or BRCA2 germline mutations who are at a high-risk for relapse.

Design. A randomized, double-blind, placebo-controlled, multicenter phase III study. The published results are from the prespecified interim analysis.

Intervention. Patients were randomized in 1:1 ratio to either receive 300 mg of olaparib orally twice daily or to receive a matching placebo. Randomization was stratified by hormone receptor status (estrogen receptor and/or progesterone receptor positive/HER2-negative vs triple negative), prior neoadjuvant vs adjuvant chemotherapy, and prior platinum use for breast cancer. Treatment was continued for 52 weeks.

Setting and participants. A total of 1836 patients were randomized in a 1:1 fashion to receive olaparib or a placebo. Eligible patients had a germline BRCA1 or BRCA1 pathogenic or likely pathogenic variant. Patients had high-risk, HER2-negative primary breast cancers and all had received definitive local therapy and neoadjuvant or adjuvant chemotherapy. Patients were enrolled between 2 to 12 weeks after completion of all local therapy. Platinum chemotherapy was allowed. Patients received adjuvant endocrine therapy for hormone receptor positive disease as well as adjuvant bisphosphonates per institutional guidelines. Patients with triple negative disease who received adjuvant chemotherapy were required to be lymph node positive or have at least 2 cm invasive disease. Patients who received neoadjuvant chemotherapy were required to have residual invasive disease to be eligible. For hormone receptor positive patients receiving adjuvant chemotherapy to be eligible they had to have at least 4 pathologically confirmed lymph nodes involved. Hormone receptor positive patients who had neoadjuvant chemotherapy were required to have had residual invasive disease.

Main outcome measures. The primary endpoint for the study was invasive disease-free survival which was defined as time from randomization to date of recurrence or death from any cause. The secondary endpoints included overall survival (OS), distant disease-free survival, safety, and tolerability of olaparib.

Main results. At the time of data cutoff, 284 events had occurred with a median follow-up of 2.5 years in the intention to treat population. A total of 81% of patients had triple negative breast cancer. Most patients (94% in the olaparib group and 92% in the placebo group) received both taxane and anthracycline based chemotherapy regimens. Platinum based chemotherapy was used in 26% of patients in each group. The groups were otherwise well balanced. Germline mutations in BRCA1 were present in 72% of patients and BRCA2 in 27% of patients. These were balanced between groups.

At the time of this analysis, adjuvant olaparib reduced the risk of invasive disease-free survival by 42% compared with placebo (P < .001). At 3 years, invasive disease-free survival was 85.9% in the olaparib group and 77.1% in the placebo group (difference, 8.8 percentage points; 95% CI, 4.5-13.0; hazard ratio [HR], 0.58; 99.5% CI, 0.41-0.82; P < .001). The 3-year distant disease-free survival was 87.5% in the olaparib group and 80.4% in the placebo group (HR 0.57; 99.5% CI, 0.39-0.83; P < .001). Results also showed that olaparib was associated with fewer deaths than placebo (59 and 86, respectively) (HR, 0.68; 99% CI, 0.44-1.05; P = .02); however, there was no significant difference between treatment arms at the time of this interim analysis. Subgroup analysis showed a consistent benefit across all groups with no difference noted regarding BRCA mutation, hormone receptor status or use of neoadjuvant vs adjuvant chemotherapy.

 

 

The side effects were consistent with the safety profile of olaparib. Adverse events of grade 3 or higher more common with olaparib included anemia (8.7%), leukopenia (3%), and fatigue (1.8%). Early discontinuation of trial regimen due to adverse events of disease recurrence occurred in 25.9% in the olaparib group and 20.7% in the placebo group. Blood transfusions were required in 5.8% of patients in the olaparib group. Myelodysplasia or acute myleoid leukemia was observed in 2 patients in the olaparib group and 3 patients in the placebo group. Adverse events leading to death occurred in 1 patient in the olaparib group and 2 patients in the placebo group.

Conclusion. Among patients with high-risk, HER2-negative early breast cancer and germline BRCA1 or BRCA2 pathogenic or likely pathogenic variants, adjuvant olaparib after completion of local treatment and neoadjuvant or adjuvant chemotherapy was associated with significantly longer invasive disease-free and distant disease-free survival compared with placebo.

Commentary

The results from the current OlympiA trial provide the first evidence that adjuvant therapy with poly adenosine diphosphate-ribose polymerase (PARP) inhibitors can improve outcomes in high-risk, HER2-negative breast cancer in patients with pathogenic BRCA1 and BRCA2 mutations. The OS, while favoring olaparib, is not yet mature at the time of this analysis. Nevertheless, these results represent an important step forward in improving outcomes in this patient population. The efficacy and safety of PARP inhibitors in BRCA-mutated breast cancer has previously been shown in patients with advanced disease leading to FDA approval of both olaparib and talazoparib in this setting.1,2 With the current results, PARP inhibitors will certainly play an important role in the adjuvant setting in patients with deleterious BRCA1 or BRCA2 mutations at high risk for relapse. Importantly, the side effect profile appears acceptable with no unexpected events and a very low rate of secondary myeloid malignancies.

Subgroup analysis appears to indicate a benefit across all groups including hormone receptor–positive disease and triple negative breast cancer. Interestingly, approximately 25% of patients in both cohorts received platinum-based chemotherapy. The efficacy of adjuvant olaparib did not appear to be impacted by prior use of platinum-containing chemotherapy regimens. It is important to consider that postneoadjuvant capecitabine, per the results of the CREATE-X trial, in triple-negative patients was not permitted in the current study. Although, this has been widely adopted in clinical practice.3 The CREATE-X trial did not specify the benefit of adjuvant capecitabine in the BRCA-mutated cohort, thus, it is not clear how this subgroup fares with this approach. Thus, one cannot extrapolate the relative efficacy of olaparib compared with capecitabine, as pointed out by the authors, and whether we consider the use of capecitabine and/or olaparib in triple-negative patients with residual invasive disease after neoadjuvant chemotherapy is not clear at this time.

Nevertheless, the magnitude of benefit seen in this trial certainly provide clinically relevant and potentially practice changing results. It will be imperative to follow these results as the survival data matures and ensure no further long-term toxicity, particularly secondary myeloid malignancies, develop. These results should be discussed with each patient and informed decisions regarding the use of adjuvant olaparib should be considered for this patient population. Lastly, these results highlight the importance of germline testing for patients with breast cancer in accordance with national guideline recommendations. Moreover, these results certainly call into question whether it is time to consider expansion of our current germline testing guidelines to detect all potential patients who may benefit from this therapy.

Application for Clinical Practice

Adjuvant olaparib in high-risk patients with germline BRCA1 or BRCA2 mutations improves invasive and distant disease-free survival and should be considered in patients who meet the enrollment criteria of the current study. Furthermore, this highlights the importance of appropriate germline genetic testing in patients with breast cancer.

Financial disclosures: None.

References

1. Robson M, Im SA, Senkus E, et al. Olaparib for metastatic breast cancer in patients with a germline BRCA mutation. N Engl J Med. 2017;377(6):523-533. doi:10.1056/NEJMoa1706450

2. Litton JK, Rugo HS, Ettl J, et al. Talazoparib in Patients with Advanced Breast Cancer and a Germline BRCA Mutation. N Engl J Med. 2018;379(8):753-763. doi:10.1056/NEJMoa1802905

3. Masuda N, Lee SJ, Ohtani S, et al. Adjuvant Capecitabine for Breast Cancer after Preoperative Chemotherapy. N Engl J Med. 2017;376(22):2147-2159. doi:10.1056/NEJMoa1612645

Article PDF
Issue
Journal of Clinical Outcomes Management - 28(6)
Publications
Topics
Page Number
249-251
Sections
Article PDF
Article PDF

Study Overview

Objective. To assess the efficacy and safety of olaparib as an adjuvant treatment in patients with BRCA1 or BRCA2 germline mutations who are at a high-risk for relapse.

Design. A randomized, double-blind, placebo-controlled, multicenter phase III study. The published results are from the prespecified interim analysis.

Intervention. Patients were randomized in 1:1 ratio to either receive 300 mg of olaparib orally twice daily or to receive a matching placebo. Randomization was stratified by hormone receptor status (estrogen receptor and/or progesterone receptor positive/HER2-negative vs triple negative), prior neoadjuvant vs adjuvant chemotherapy, and prior platinum use for breast cancer. Treatment was continued for 52 weeks.

Setting and participants. A total of 1836 patients were randomized in a 1:1 fashion to receive olaparib or a placebo. Eligible patients had a germline BRCA1 or BRCA1 pathogenic or likely pathogenic variant. Patients had high-risk, HER2-negative primary breast cancers and all had received definitive local therapy and neoadjuvant or adjuvant chemotherapy. Patients were enrolled between 2 to 12 weeks after completion of all local therapy. Platinum chemotherapy was allowed. Patients received adjuvant endocrine therapy for hormone receptor positive disease as well as adjuvant bisphosphonates per institutional guidelines. Patients with triple negative disease who received adjuvant chemotherapy were required to be lymph node positive or have at least 2 cm invasive disease. Patients who received neoadjuvant chemotherapy were required to have residual invasive disease to be eligible. For hormone receptor positive patients receiving adjuvant chemotherapy to be eligible they had to have at least 4 pathologically confirmed lymph nodes involved. Hormone receptor positive patients who had neoadjuvant chemotherapy were required to have had residual invasive disease.

Main outcome measures. The primary endpoint for the study was invasive disease-free survival which was defined as time from randomization to date of recurrence or death from any cause. The secondary endpoints included overall survival (OS), distant disease-free survival, safety, and tolerability of olaparib.

Main results. At the time of data cutoff, 284 events had occurred with a median follow-up of 2.5 years in the intention to treat population. A total of 81% of patients had triple negative breast cancer. Most patients (94% in the olaparib group and 92% in the placebo group) received both taxane and anthracycline based chemotherapy regimens. Platinum based chemotherapy was used in 26% of patients in each group. The groups were otherwise well balanced. Germline mutations in BRCA1 were present in 72% of patients and BRCA2 in 27% of patients. These were balanced between groups.

At the time of this analysis, adjuvant olaparib reduced the risk of invasive disease-free survival by 42% compared with placebo (P < .001). At 3 years, invasive disease-free survival was 85.9% in the olaparib group and 77.1% in the placebo group (difference, 8.8 percentage points; 95% CI, 4.5-13.0; hazard ratio [HR], 0.58; 99.5% CI, 0.41-0.82; P < .001). The 3-year distant disease-free survival was 87.5% in the olaparib group and 80.4% in the placebo group (HR 0.57; 99.5% CI, 0.39-0.83; P < .001). Results also showed that olaparib was associated with fewer deaths than placebo (59 and 86, respectively) (HR, 0.68; 99% CI, 0.44-1.05; P = .02); however, there was no significant difference between treatment arms at the time of this interim analysis. Subgroup analysis showed a consistent benefit across all groups with no difference noted regarding BRCA mutation, hormone receptor status or use of neoadjuvant vs adjuvant chemotherapy.

 

 

The side effects were consistent with the safety profile of olaparib. Adverse events of grade 3 or higher more common with olaparib included anemia (8.7%), leukopenia (3%), and fatigue (1.8%). Early discontinuation of trial regimen due to adverse events of disease recurrence occurred in 25.9% in the olaparib group and 20.7% in the placebo group. Blood transfusions were required in 5.8% of patients in the olaparib group. Myelodysplasia or acute myleoid leukemia was observed in 2 patients in the olaparib group and 3 patients in the placebo group. Adverse events leading to death occurred in 1 patient in the olaparib group and 2 patients in the placebo group.

Conclusion. Among patients with high-risk, HER2-negative early breast cancer and germline BRCA1 or BRCA2 pathogenic or likely pathogenic variants, adjuvant olaparib after completion of local treatment and neoadjuvant or adjuvant chemotherapy was associated with significantly longer invasive disease-free and distant disease-free survival compared with placebo.

Commentary

The results from the current OlympiA trial provide the first evidence that adjuvant therapy with poly adenosine diphosphate-ribose polymerase (PARP) inhibitors can improve outcomes in high-risk, HER2-negative breast cancer in patients with pathogenic BRCA1 and BRCA2 mutations. The OS, while favoring olaparib, is not yet mature at the time of this analysis. Nevertheless, these results represent an important step forward in improving outcomes in this patient population. The efficacy and safety of PARP inhibitors in BRCA-mutated breast cancer has previously been shown in patients with advanced disease leading to FDA approval of both olaparib and talazoparib in this setting.1,2 With the current results, PARP inhibitors will certainly play an important role in the adjuvant setting in patients with deleterious BRCA1 or BRCA2 mutations at high risk for relapse. Importantly, the side effect profile appears acceptable with no unexpected events and a very low rate of secondary myeloid malignancies.

Subgroup analysis appears to indicate a benefit across all groups including hormone receptor–positive disease and triple negative breast cancer. Interestingly, approximately 25% of patients in both cohorts received platinum-based chemotherapy. The efficacy of adjuvant olaparib did not appear to be impacted by prior use of platinum-containing chemotherapy regimens. It is important to consider that postneoadjuvant capecitabine, per the results of the CREATE-X trial, in triple-negative patients was not permitted in the current study. Although, this has been widely adopted in clinical practice.3 The CREATE-X trial did not specify the benefit of adjuvant capecitabine in the BRCA-mutated cohort, thus, it is not clear how this subgroup fares with this approach. Thus, one cannot extrapolate the relative efficacy of olaparib compared with capecitabine, as pointed out by the authors, and whether we consider the use of capecitabine and/or olaparib in triple-negative patients with residual invasive disease after neoadjuvant chemotherapy is not clear at this time.

Nevertheless, the magnitude of benefit seen in this trial certainly provide clinically relevant and potentially practice changing results. It will be imperative to follow these results as the survival data matures and ensure no further long-term toxicity, particularly secondary myeloid malignancies, develop. These results should be discussed with each patient and informed decisions regarding the use of adjuvant olaparib should be considered for this patient population. Lastly, these results highlight the importance of germline testing for patients with breast cancer in accordance with national guideline recommendations. Moreover, these results certainly call into question whether it is time to consider expansion of our current germline testing guidelines to detect all potential patients who may benefit from this therapy.

Application for Clinical Practice

Adjuvant olaparib in high-risk patients with germline BRCA1 or BRCA2 mutations improves invasive and distant disease-free survival and should be considered in patients who meet the enrollment criteria of the current study. Furthermore, this highlights the importance of appropriate germline genetic testing in patients with breast cancer.

Financial disclosures: None.

Study Overview

Objective. To assess the efficacy and safety of olaparib as an adjuvant treatment in patients with BRCA1 or BRCA2 germline mutations who are at a high-risk for relapse.

Design. A randomized, double-blind, placebo-controlled, multicenter phase III study. The published results are from the prespecified interim analysis.

Intervention. Patients were randomized in 1:1 ratio to either receive 300 mg of olaparib orally twice daily or to receive a matching placebo. Randomization was stratified by hormone receptor status (estrogen receptor and/or progesterone receptor positive/HER2-negative vs triple negative), prior neoadjuvant vs adjuvant chemotherapy, and prior platinum use for breast cancer. Treatment was continued for 52 weeks.

Setting and participants. A total of 1836 patients were randomized in a 1:1 fashion to receive olaparib or a placebo. Eligible patients had a germline BRCA1 or BRCA1 pathogenic or likely pathogenic variant. Patients had high-risk, HER2-negative primary breast cancers and all had received definitive local therapy and neoadjuvant or adjuvant chemotherapy. Patients were enrolled between 2 to 12 weeks after completion of all local therapy. Platinum chemotherapy was allowed. Patients received adjuvant endocrine therapy for hormone receptor positive disease as well as adjuvant bisphosphonates per institutional guidelines. Patients with triple negative disease who received adjuvant chemotherapy were required to be lymph node positive or have at least 2 cm invasive disease. Patients who received neoadjuvant chemotherapy were required to have residual invasive disease to be eligible. For hormone receptor positive patients receiving adjuvant chemotherapy to be eligible they had to have at least 4 pathologically confirmed lymph nodes involved. Hormone receptor positive patients who had neoadjuvant chemotherapy were required to have had residual invasive disease.

Main outcome measures. The primary endpoint for the study was invasive disease-free survival which was defined as time from randomization to date of recurrence or death from any cause. The secondary endpoints included overall survival (OS), distant disease-free survival, safety, and tolerability of olaparib.

Main results. At the time of data cutoff, 284 events had occurred with a median follow-up of 2.5 years in the intention to treat population. A total of 81% of patients had triple negative breast cancer. Most patients (94% in the olaparib group and 92% in the placebo group) received both taxane and anthracycline based chemotherapy regimens. Platinum based chemotherapy was used in 26% of patients in each group. The groups were otherwise well balanced. Germline mutations in BRCA1 were present in 72% of patients and BRCA2 in 27% of patients. These were balanced between groups.

At the time of this analysis, adjuvant olaparib reduced the risk of invasive disease-free survival by 42% compared with placebo (P < .001). At 3 years, invasive disease-free survival was 85.9% in the olaparib group and 77.1% in the placebo group (difference, 8.8 percentage points; 95% CI, 4.5-13.0; hazard ratio [HR], 0.58; 99.5% CI, 0.41-0.82; P < .001). The 3-year distant disease-free survival was 87.5% in the olaparib group and 80.4% in the placebo group (HR 0.57; 99.5% CI, 0.39-0.83; P < .001). Results also showed that olaparib was associated with fewer deaths than placebo (59 and 86, respectively) (HR, 0.68; 99% CI, 0.44-1.05; P = .02); however, there was no significant difference between treatment arms at the time of this interim analysis. Subgroup analysis showed a consistent benefit across all groups with no difference noted regarding BRCA mutation, hormone receptor status or use of neoadjuvant vs adjuvant chemotherapy.

 

 

The side effects were consistent with the safety profile of olaparib. Adverse events of grade 3 or higher more common with olaparib included anemia (8.7%), leukopenia (3%), and fatigue (1.8%). Early discontinuation of trial regimen due to adverse events of disease recurrence occurred in 25.9% in the olaparib group and 20.7% in the placebo group. Blood transfusions were required in 5.8% of patients in the olaparib group. Myelodysplasia or acute myleoid leukemia was observed in 2 patients in the olaparib group and 3 patients in the placebo group. Adverse events leading to death occurred in 1 patient in the olaparib group and 2 patients in the placebo group.

Conclusion. Among patients with high-risk, HER2-negative early breast cancer and germline BRCA1 or BRCA2 pathogenic or likely pathogenic variants, adjuvant olaparib after completion of local treatment and neoadjuvant or adjuvant chemotherapy was associated with significantly longer invasive disease-free and distant disease-free survival compared with placebo.

Commentary

The results from the current OlympiA trial provide the first evidence that adjuvant therapy with poly adenosine diphosphate-ribose polymerase (PARP) inhibitors can improve outcomes in high-risk, HER2-negative breast cancer in patients with pathogenic BRCA1 and BRCA2 mutations. The OS, while favoring olaparib, is not yet mature at the time of this analysis. Nevertheless, these results represent an important step forward in improving outcomes in this patient population. The efficacy and safety of PARP inhibitors in BRCA-mutated breast cancer has previously been shown in patients with advanced disease leading to FDA approval of both olaparib and talazoparib in this setting.1,2 With the current results, PARP inhibitors will certainly play an important role in the adjuvant setting in patients with deleterious BRCA1 or BRCA2 mutations at high risk for relapse. Importantly, the side effect profile appears acceptable with no unexpected events and a very low rate of secondary myeloid malignancies.

Subgroup analysis appears to indicate a benefit across all groups including hormone receptor–positive disease and triple negative breast cancer. Interestingly, approximately 25% of patients in both cohorts received platinum-based chemotherapy. The efficacy of adjuvant olaparib did not appear to be impacted by prior use of platinum-containing chemotherapy regimens. It is important to consider that postneoadjuvant capecitabine, per the results of the CREATE-X trial, in triple-negative patients was not permitted in the current study. Although, this has been widely adopted in clinical practice.3 The CREATE-X trial did not specify the benefit of adjuvant capecitabine in the BRCA-mutated cohort, thus, it is not clear how this subgroup fares with this approach. Thus, one cannot extrapolate the relative efficacy of olaparib compared with capecitabine, as pointed out by the authors, and whether we consider the use of capecitabine and/or olaparib in triple-negative patients with residual invasive disease after neoadjuvant chemotherapy is not clear at this time.

Nevertheless, the magnitude of benefit seen in this trial certainly provide clinically relevant and potentially practice changing results. It will be imperative to follow these results as the survival data matures and ensure no further long-term toxicity, particularly secondary myeloid malignancies, develop. These results should be discussed with each patient and informed decisions regarding the use of adjuvant olaparib should be considered for this patient population. Lastly, these results highlight the importance of germline testing for patients with breast cancer in accordance with national guideline recommendations. Moreover, these results certainly call into question whether it is time to consider expansion of our current germline testing guidelines to detect all potential patients who may benefit from this therapy.

Application for Clinical Practice

Adjuvant olaparib in high-risk patients with germline BRCA1 or BRCA2 mutations improves invasive and distant disease-free survival and should be considered in patients who meet the enrollment criteria of the current study. Furthermore, this highlights the importance of appropriate germline genetic testing in patients with breast cancer.

Financial disclosures: None.

References

1. Robson M, Im SA, Senkus E, et al. Olaparib for metastatic breast cancer in patients with a germline BRCA mutation. N Engl J Med. 2017;377(6):523-533. doi:10.1056/NEJMoa1706450

2. Litton JK, Rugo HS, Ettl J, et al. Talazoparib in Patients with Advanced Breast Cancer and a Germline BRCA Mutation. N Engl J Med. 2018;379(8):753-763. doi:10.1056/NEJMoa1802905

3. Masuda N, Lee SJ, Ohtani S, et al. Adjuvant Capecitabine for Breast Cancer after Preoperative Chemotherapy. N Engl J Med. 2017;376(22):2147-2159. doi:10.1056/NEJMoa1612645

References

1. Robson M, Im SA, Senkus E, et al. Olaparib for metastatic breast cancer in patients with a germline BRCA mutation. N Engl J Med. 2017;377(6):523-533. doi:10.1056/NEJMoa1706450

2. Litton JK, Rugo HS, Ettl J, et al. Talazoparib in Patients with Advanced Breast Cancer and a Germline BRCA Mutation. N Engl J Med. 2018;379(8):753-763. doi:10.1056/NEJMoa1802905

3. Masuda N, Lee SJ, Ohtani S, et al. Adjuvant Capecitabine for Breast Cancer after Preoperative Chemotherapy. N Engl J Med. 2017;376(22):2147-2159. doi:10.1056/NEJMoa1612645

Issue
Journal of Clinical Outcomes Management - 28(6)
Issue
Journal of Clinical Outcomes Management - 28(6)
Page Number
249-251
Page Number
249-251
Publications
Publications
Topics
Article Type
Display Headline
Adjuvant Olaparib Improves Outcomes in High-Risk, HER2-Negative Early Breast Cancer Patients With Germline BRCA1 and BRCA2 Mutations
Display Headline
Adjuvant Olaparib Improves Outcomes in High-Risk, HER2-Negative Early Breast Cancer Patients With Germline BRCA1 and BRCA2 Mutations
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

Nivolumab Plus Cabozantinib Improves Outcomes Compared With Sunitinib for Advanced Renal Cell Carcinoma

Article Type
Changed
Fri, 07/30/2021 - 01:15
Display Headline
Nivolumab Plus Cabozantinib Improves Outcomes Compared With Sunitinib for Advanced Renal Cell Carcinoma

Study Overview

Objective. To evaluate the efficacy and safety of the combination of nivolumab plus cabozantinib as compared with sunitinib monotherapy in the treatment of previously untreated advanced renal cell carcinoma (RCC).

Design. Multicenter, international, open-label, randomized, phase 3 trial.

Intervention. Patients were randomized in a 1:1 fashion to 1 of 2 treatment arms:

  • Arm A: Nivolumab intravenously 240 mg every 2 weeks plus cabozantinib orally 40 mg once daily.
  • Arm B: Sunitinib orally 50 mg daily for 4 weeks, followed by 2 weeks off therapy (6-week cycle).

Randomization was stratified by the International Metastatic RCC Database Consortium prognostic risk score (low-, intermediate-, and high-risk). Treatment was continued until disease progression or development of unacceptable toxic side effects with a maximum of 2-year duration of Nivolumab therapy.

Settings and participants. Adults with previously untreated advanced RCC with a clear cell component were eligible for enrollment. Subjects were excluded if they had active central nervous system metastases or active autoimmune disease.

Main outcome measures. The primary outcome of this study was progression-free survival (PFS) as assessed by an independent review committee. Secondary endpoints included overall survival, objective response rate, safety, and PFS as assessed by investigators. All subgroup analyses were prespecified. Efficacy was assessed in the intention-to-treat population, including all patients who underwent randomization.

Main results. A total of 651 patients underwent randomization: 323 to the nivolumab plus cabozantinib group, and 328 to the sunitinib group. Baseline demographics were balanced. The median follow-up period for overall survival (OS) was 18.1 months. The primary reason for treatment discontinuation in any group was disease progression. PFS as indicated by an independent review committee was significantly longer in the nivolumab plus cabozantinib group compared to the sunitinib group (median 16.6 months vs 8.2 months; hazard ratio [HR] 0.51, P < .001). The median OS was not reached for any group. Overall survival was longer in the nivolumab plus cabozantinib group compared to the sunitinib group (HR 0.60, 95% CI: 0.40-0.89; P = .001). The objective response rate was 55.7% with the nivolumab plus cabozantinib group versus 27.1% with sunitinib (P < .001). The complete response rate was 8% in the nivolumab plus cabozantinib group compared to 4.6% in the sunitinib group. The median time to response was 2.8 months with nivolumab plus cabozantinib and 4.2 months in the sunitinib group, while the median duration of response was 20.2 months and 11.5 months, respectively.

 

 

Nearly all patients (about 99% in each group) had an adverse event (AE). Hypertension was the most common side effect, with grade 3 or higher seen in 12.5% in the nivolumab plus cabzantinib group and 13.1% in the sunitinib group. Other grade 3 or higher side effects occurring in at least 10% of patients in any group were hyponatremia, diarrhea, palmar-plantar erythrodysesthesia, hypothyroidism, and fatigue. AEs of any cause leading to discontinuation of the therapy occurred in 19.7% in the nivolumab plus cabzantinib group vs 16.9% of the sunitinib group. One death was considered to be treatment-related (small intestinal perforation) in the nivolumab plus cabozantinib group vs 2 treatment-related deaths with sunitinib (pneumonia and respiratory distress). In the nivolumab plus cabozantinib group, 57% of the patients had a dose reduction of cabozantinib and 52% had a reduction in sunitinib dosage.

Using the Functional Assessment of Cancer Therapy-Kidney Symptoms Index, patients in the nivolumab plus cabozantinib group reported better health-related quality of life and less disease-related symptoms compared to the sunitinib group.

Commentary

The treatment landscape for frontline therapy for patients with advanced RCC has rapidly expanded over the last several years and has revolutionized cancer care. Ushered in by the results from the CheckMate 214 study highlighting the efficacy of dual checkpoint inhibition with nivolumab and ipilimumab in intermediate and poor risk patients, several subsequent trials have demonstrated improved outcomes with combination therapy with immune checkpoint inhibitors and tyrosine-kinase inhibitors (TKI). To date, data from Keynote-426 (pembrolizumab plus axitinib vs sunitinib), Javelin Renal 101 (avelumab plus axitinib vs sunitinib) and the CLEAR trial (lenvatinib plus pembrolizumab vs levatinib plus everolimus vs sunitinib) have demonstrated superiority of immune checkpoint inhibitor/TKI combinations over sunitinb in the first-line setting.1-5

The current phase 3, CheckMate 9ER trial adds yet another dynamic option for patients with advanced clear cell RCC. While cross-trial comparisons are fraught with important caveats, the median PFS of almost 16.6 months and complete response rate of 8% the nivolumab plus cabozantinib group compares favorably with other combinations. Data from the CLEAR study with the combination of lenvatinib and pembrolizumab showed a complete response rate approaching 16%. Importantly, the current study highlights improved quality of life with the combination of cabozantinib and nivolumab compared to sunitinib alone adding to the efficacy and benefits of this combination treatment.

The selection of first line therapy for patients with advanced RCC should be always guided by individual patient characteristics, and any single immune checkpoint inhibitor/TKI combination is not “superior” to any other. Perhaps more importantly is developing an understanding of the overlapping toxicity profiles of checkpoint inhibitors and TKIs. Again, this trial results are consistent with prior studies in terms of the adverse event profile which were not trivial, and almost all patients (99%) experienced AEs. It is important for oncologists to understand the management of the toxicities with these combinations and dose reductions as appropriate. It is worth noting that 19% of patients with nivolumab plus cabozantinib received glucocorticoids for management of immune-related AEs.

While long-term follow-up data will be needed to further understand the durability of response to this combination, nivolumab-cabozantinib represents an exciting new option for patients with advanced clear cell RCC. As we continue to see improvement in outcomes in clear cell histology, further work must focus on optimization of therapy in non-clear cell RCC as this is a population that is not represented in these data sets. Furthermore, future efforts should begin to explore triplet combinations and biomarker driven patient selection for upfront therapy in ordercontinue to improve outcomes in patients with advanced RCC.

Applications for Clinical Practice

The combination of nivolumab plus cabozantinib adds to the growing list of highly active checkpoint inhibitor/TKI combinations for first-line treatment of advanced RCC. With significant higher response rates, improved outcomes, and improvement in the quality of life, this combination will add another standard treatment option for patients with previously untreated advanced RCC.

References

1. Motzer RJ, Tannir NM, McDermott DF, et al. Nivolumab plus Ipilimumab Versus Sunitinib in Advanced Renal-Cell Carcinoma. N Engl J Med. 2018;378(14)1277-1290. doi:10.1056/NEJMoa1712126

2. Rini BI, Plimack ER, Stus V, et al. Pembrolizumab plus Axitinib versus Sunitinib for Advanced Renal-Cell Carcinoma. N Engl J Med. 2019;380(12):1116-1127. doi:10.1056/NEJMoa1816714

3. Powles T, Plimack ER, Soulières D, et al. Pembrolizumab plus axitinib versus sunitinib monotherapy as first-line treatment of advanced renal cell carcinoma (KEYNOTE-426): extended follow-up from a randomised, open-label, phase 3 trial. Lancet Oncol. 2020;21(12):1563-1573. doi:10.1016/S1470-2045(20)30436-8

4. Choueiri TK, Motzer RJ, Rini BI, et al. Updated efficacy results from the JAVELIN Renal 101 trial: first-line avelumab plus axitinib versus sunitinib in patients with advanced renal cell carcinoma. Ann Oncol. 2020;31:1030-1039. doi:10.1016/j.annonc.2020.04.010

5, Motzer R, Alekseev B, Rha SY, et al. CLEAR Trial Investigators. Lenvatinib plus Pembrolizumab or Everolimus for Advanced Renal Cell Carcinoma. N Engl J Med. 2021;384(14):1289-1300. doi:10.1056/NEJMoa2035716

Article PDF
Issue
Journal of Clinical Outcomes Management - 28(4)
Publications
Topics
Page Number
152-154
Sections
Article PDF
Article PDF

Study Overview

Objective. To evaluate the efficacy and safety of the combination of nivolumab plus cabozantinib as compared with sunitinib monotherapy in the treatment of previously untreated advanced renal cell carcinoma (RCC).

Design. Multicenter, international, open-label, randomized, phase 3 trial.

Intervention. Patients were randomized in a 1:1 fashion to 1 of 2 treatment arms:

  • Arm A: Nivolumab intravenously 240 mg every 2 weeks plus cabozantinib orally 40 mg once daily.
  • Arm B: Sunitinib orally 50 mg daily for 4 weeks, followed by 2 weeks off therapy (6-week cycle).

Randomization was stratified by the International Metastatic RCC Database Consortium prognostic risk score (low-, intermediate-, and high-risk). Treatment was continued until disease progression or development of unacceptable toxic side effects with a maximum of 2-year duration of Nivolumab therapy.

Settings and participants. Adults with previously untreated advanced RCC with a clear cell component were eligible for enrollment. Subjects were excluded if they had active central nervous system metastases or active autoimmune disease.

Main outcome measures. The primary outcome of this study was progression-free survival (PFS) as assessed by an independent review committee. Secondary endpoints included overall survival, objective response rate, safety, and PFS as assessed by investigators. All subgroup analyses were prespecified. Efficacy was assessed in the intention-to-treat population, including all patients who underwent randomization.

Main results. A total of 651 patients underwent randomization: 323 to the nivolumab plus cabozantinib group, and 328 to the sunitinib group. Baseline demographics were balanced. The median follow-up period for overall survival (OS) was 18.1 months. The primary reason for treatment discontinuation in any group was disease progression. PFS as indicated by an independent review committee was significantly longer in the nivolumab plus cabozantinib group compared to the sunitinib group (median 16.6 months vs 8.2 months; hazard ratio [HR] 0.51, P < .001). The median OS was not reached for any group. Overall survival was longer in the nivolumab plus cabozantinib group compared to the sunitinib group (HR 0.60, 95% CI: 0.40-0.89; P = .001). The objective response rate was 55.7% with the nivolumab plus cabozantinib group versus 27.1% with sunitinib (P < .001). The complete response rate was 8% in the nivolumab plus cabozantinib group compared to 4.6% in the sunitinib group. The median time to response was 2.8 months with nivolumab plus cabozantinib and 4.2 months in the sunitinib group, while the median duration of response was 20.2 months and 11.5 months, respectively.

 

 

Nearly all patients (about 99% in each group) had an adverse event (AE). Hypertension was the most common side effect, with grade 3 or higher seen in 12.5% in the nivolumab plus cabzantinib group and 13.1% in the sunitinib group. Other grade 3 or higher side effects occurring in at least 10% of patients in any group were hyponatremia, diarrhea, palmar-plantar erythrodysesthesia, hypothyroidism, and fatigue. AEs of any cause leading to discontinuation of the therapy occurred in 19.7% in the nivolumab plus cabzantinib group vs 16.9% of the sunitinib group. One death was considered to be treatment-related (small intestinal perforation) in the nivolumab plus cabozantinib group vs 2 treatment-related deaths with sunitinib (pneumonia and respiratory distress). In the nivolumab plus cabozantinib group, 57% of the patients had a dose reduction of cabozantinib and 52% had a reduction in sunitinib dosage.

Using the Functional Assessment of Cancer Therapy-Kidney Symptoms Index, patients in the nivolumab plus cabozantinib group reported better health-related quality of life and less disease-related symptoms compared to the sunitinib group.

Commentary

The treatment landscape for frontline therapy for patients with advanced RCC has rapidly expanded over the last several years and has revolutionized cancer care. Ushered in by the results from the CheckMate 214 study highlighting the efficacy of dual checkpoint inhibition with nivolumab and ipilimumab in intermediate and poor risk patients, several subsequent trials have demonstrated improved outcomes with combination therapy with immune checkpoint inhibitors and tyrosine-kinase inhibitors (TKI). To date, data from Keynote-426 (pembrolizumab plus axitinib vs sunitinib), Javelin Renal 101 (avelumab plus axitinib vs sunitinib) and the CLEAR trial (lenvatinib plus pembrolizumab vs levatinib plus everolimus vs sunitinib) have demonstrated superiority of immune checkpoint inhibitor/TKI combinations over sunitinb in the first-line setting.1-5

The current phase 3, CheckMate 9ER trial adds yet another dynamic option for patients with advanced clear cell RCC. While cross-trial comparisons are fraught with important caveats, the median PFS of almost 16.6 months and complete response rate of 8% the nivolumab plus cabozantinib group compares favorably with other combinations. Data from the CLEAR study with the combination of lenvatinib and pembrolizumab showed a complete response rate approaching 16%. Importantly, the current study highlights improved quality of life with the combination of cabozantinib and nivolumab compared to sunitinib alone adding to the efficacy and benefits of this combination treatment.

The selection of first line therapy for patients with advanced RCC should be always guided by individual patient characteristics, and any single immune checkpoint inhibitor/TKI combination is not “superior” to any other. Perhaps more importantly is developing an understanding of the overlapping toxicity profiles of checkpoint inhibitors and TKIs. Again, this trial results are consistent with prior studies in terms of the adverse event profile which were not trivial, and almost all patients (99%) experienced AEs. It is important for oncologists to understand the management of the toxicities with these combinations and dose reductions as appropriate. It is worth noting that 19% of patients with nivolumab plus cabozantinib received glucocorticoids for management of immune-related AEs.

While long-term follow-up data will be needed to further understand the durability of response to this combination, nivolumab-cabozantinib represents an exciting new option for patients with advanced clear cell RCC. As we continue to see improvement in outcomes in clear cell histology, further work must focus on optimization of therapy in non-clear cell RCC as this is a population that is not represented in these data sets. Furthermore, future efforts should begin to explore triplet combinations and biomarker driven patient selection for upfront therapy in ordercontinue to improve outcomes in patients with advanced RCC.

Applications for Clinical Practice

The combination of nivolumab plus cabozantinib adds to the growing list of highly active checkpoint inhibitor/TKI combinations for first-line treatment of advanced RCC. With significant higher response rates, improved outcomes, and improvement in the quality of life, this combination will add another standard treatment option for patients with previously untreated advanced RCC.

Study Overview

Objective. To evaluate the efficacy and safety of the combination of nivolumab plus cabozantinib as compared with sunitinib monotherapy in the treatment of previously untreated advanced renal cell carcinoma (RCC).

Design. Multicenter, international, open-label, randomized, phase 3 trial.

Intervention. Patients were randomized in a 1:1 fashion to 1 of 2 treatment arms:

  • Arm A: Nivolumab intravenously 240 mg every 2 weeks plus cabozantinib orally 40 mg once daily.
  • Arm B: Sunitinib orally 50 mg daily for 4 weeks, followed by 2 weeks off therapy (6-week cycle).

Randomization was stratified by the International Metastatic RCC Database Consortium prognostic risk score (low-, intermediate-, and high-risk). Treatment was continued until disease progression or development of unacceptable toxic side effects with a maximum of 2-year duration of Nivolumab therapy.

Settings and participants. Adults with previously untreated advanced RCC with a clear cell component were eligible for enrollment. Subjects were excluded if they had active central nervous system metastases or active autoimmune disease.

Main outcome measures. The primary outcome of this study was progression-free survival (PFS) as assessed by an independent review committee. Secondary endpoints included overall survival, objective response rate, safety, and PFS as assessed by investigators. All subgroup analyses were prespecified. Efficacy was assessed in the intention-to-treat population, including all patients who underwent randomization.

Main results. A total of 651 patients underwent randomization: 323 to the nivolumab plus cabozantinib group, and 328 to the sunitinib group. Baseline demographics were balanced. The median follow-up period for overall survival (OS) was 18.1 months. The primary reason for treatment discontinuation in any group was disease progression. PFS as indicated by an independent review committee was significantly longer in the nivolumab plus cabozantinib group compared to the sunitinib group (median 16.6 months vs 8.2 months; hazard ratio [HR] 0.51, P < .001). The median OS was not reached for any group. Overall survival was longer in the nivolumab plus cabozantinib group compared to the sunitinib group (HR 0.60, 95% CI: 0.40-0.89; P = .001). The objective response rate was 55.7% with the nivolumab plus cabozantinib group versus 27.1% with sunitinib (P < .001). The complete response rate was 8% in the nivolumab plus cabozantinib group compared to 4.6% in the sunitinib group. The median time to response was 2.8 months with nivolumab plus cabozantinib and 4.2 months in the sunitinib group, while the median duration of response was 20.2 months and 11.5 months, respectively.

 

 

Nearly all patients (about 99% in each group) had an adverse event (AE). Hypertension was the most common side effect, with grade 3 or higher seen in 12.5% in the nivolumab plus cabzantinib group and 13.1% in the sunitinib group. Other grade 3 or higher side effects occurring in at least 10% of patients in any group were hyponatremia, diarrhea, palmar-plantar erythrodysesthesia, hypothyroidism, and fatigue. AEs of any cause leading to discontinuation of the therapy occurred in 19.7% in the nivolumab plus cabzantinib group vs 16.9% of the sunitinib group. One death was considered to be treatment-related (small intestinal perforation) in the nivolumab plus cabozantinib group vs 2 treatment-related deaths with sunitinib (pneumonia and respiratory distress). In the nivolumab plus cabozantinib group, 57% of the patients had a dose reduction of cabozantinib and 52% had a reduction in sunitinib dosage.

Using the Functional Assessment of Cancer Therapy-Kidney Symptoms Index, patients in the nivolumab plus cabozantinib group reported better health-related quality of life and less disease-related symptoms compared to the sunitinib group.

Commentary

The treatment landscape for frontline therapy for patients with advanced RCC has rapidly expanded over the last several years and has revolutionized cancer care. Ushered in by the results from the CheckMate 214 study highlighting the efficacy of dual checkpoint inhibition with nivolumab and ipilimumab in intermediate and poor risk patients, several subsequent trials have demonstrated improved outcomes with combination therapy with immune checkpoint inhibitors and tyrosine-kinase inhibitors (TKI). To date, data from Keynote-426 (pembrolizumab plus axitinib vs sunitinib), Javelin Renal 101 (avelumab plus axitinib vs sunitinib) and the CLEAR trial (lenvatinib plus pembrolizumab vs levatinib plus everolimus vs sunitinib) have demonstrated superiority of immune checkpoint inhibitor/TKI combinations over sunitinb in the first-line setting.1-5

The current phase 3, CheckMate 9ER trial adds yet another dynamic option for patients with advanced clear cell RCC. While cross-trial comparisons are fraught with important caveats, the median PFS of almost 16.6 months and complete response rate of 8% the nivolumab plus cabozantinib group compares favorably with other combinations. Data from the CLEAR study with the combination of lenvatinib and pembrolizumab showed a complete response rate approaching 16%. Importantly, the current study highlights improved quality of life with the combination of cabozantinib and nivolumab compared to sunitinib alone adding to the efficacy and benefits of this combination treatment.

The selection of first line therapy for patients with advanced RCC should be always guided by individual patient characteristics, and any single immune checkpoint inhibitor/TKI combination is not “superior” to any other. Perhaps more importantly is developing an understanding of the overlapping toxicity profiles of checkpoint inhibitors and TKIs. Again, this trial results are consistent with prior studies in terms of the adverse event profile which were not trivial, and almost all patients (99%) experienced AEs. It is important for oncologists to understand the management of the toxicities with these combinations and dose reductions as appropriate. It is worth noting that 19% of patients with nivolumab plus cabozantinib received glucocorticoids for management of immune-related AEs.

While long-term follow-up data will be needed to further understand the durability of response to this combination, nivolumab-cabozantinib represents an exciting new option for patients with advanced clear cell RCC. As we continue to see improvement in outcomes in clear cell histology, further work must focus on optimization of therapy in non-clear cell RCC as this is a population that is not represented in these data sets. Furthermore, future efforts should begin to explore triplet combinations and biomarker driven patient selection for upfront therapy in ordercontinue to improve outcomes in patients with advanced RCC.

Applications for Clinical Practice

The combination of nivolumab plus cabozantinib adds to the growing list of highly active checkpoint inhibitor/TKI combinations for first-line treatment of advanced RCC. With significant higher response rates, improved outcomes, and improvement in the quality of life, this combination will add another standard treatment option for patients with previously untreated advanced RCC.

References

1. Motzer RJ, Tannir NM, McDermott DF, et al. Nivolumab plus Ipilimumab Versus Sunitinib in Advanced Renal-Cell Carcinoma. N Engl J Med. 2018;378(14)1277-1290. doi:10.1056/NEJMoa1712126

2. Rini BI, Plimack ER, Stus V, et al. Pembrolizumab plus Axitinib versus Sunitinib for Advanced Renal-Cell Carcinoma. N Engl J Med. 2019;380(12):1116-1127. doi:10.1056/NEJMoa1816714

3. Powles T, Plimack ER, Soulières D, et al. Pembrolizumab plus axitinib versus sunitinib monotherapy as first-line treatment of advanced renal cell carcinoma (KEYNOTE-426): extended follow-up from a randomised, open-label, phase 3 trial. Lancet Oncol. 2020;21(12):1563-1573. doi:10.1016/S1470-2045(20)30436-8

4. Choueiri TK, Motzer RJ, Rini BI, et al. Updated efficacy results from the JAVELIN Renal 101 trial: first-line avelumab plus axitinib versus sunitinib in patients with advanced renal cell carcinoma. Ann Oncol. 2020;31:1030-1039. doi:10.1016/j.annonc.2020.04.010

5, Motzer R, Alekseev B, Rha SY, et al. CLEAR Trial Investigators. Lenvatinib plus Pembrolizumab or Everolimus for Advanced Renal Cell Carcinoma. N Engl J Med. 2021;384(14):1289-1300. doi:10.1056/NEJMoa2035716

References

1. Motzer RJ, Tannir NM, McDermott DF, et al. Nivolumab plus Ipilimumab Versus Sunitinib in Advanced Renal-Cell Carcinoma. N Engl J Med. 2018;378(14)1277-1290. doi:10.1056/NEJMoa1712126

2. Rini BI, Plimack ER, Stus V, et al. Pembrolizumab plus Axitinib versus Sunitinib for Advanced Renal-Cell Carcinoma. N Engl J Med. 2019;380(12):1116-1127. doi:10.1056/NEJMoa1816714

3. Powles T, Plimack ER, Soulières D, et al. Pembrolizumab plus axitinib versus sunitinib monotherapy as first-line treatment of advanced renal cell carcinoma (KEYNOTE-426): extended follow-up from a randomised, open-label, phase 3 trial. Lancet Oncol. 2020;21(12):1563-1573. doi:10.1016/S1470-2045(20)30436-8

4. Choueiri TK, Motzer RJ, Rini BI, et al. Updated efficacy results from the JAVELIN Renal 101 trial: first-line avelumab plus axitinib versus sunitinib in patients with advanced renal cell carcinoma. Ann Oncol. 2020;31:1030-1039. doi:10.1016/j.annonc.2020.04.010

5, Motzer R, Alekseev B, Rha SY, et al. CLEAR Trial Investigators. Lenvatinib plus Pembrolizumab or Everolimus for Advanced Renal Cell Carcinoma. N Engl J Med. 2021;384(14):1289-1300. doi:10.1056/NEJMoa2035716

Issue
Journal of Clinical Outcomes Management - 28(4)
Issue
Journal of Clinical Outcomes Management - 28(4)
Page Number
152-154
Page Number
152-154
Publications
Publications
Topics
Article Type
Display Headline
Nivolumab Plus Cabozantinib Improves Outcomes Compared With Sunitinib for Advanced Renal Cell Carcinoma
Display Headline
Nivolumab Plus Cabozantinib Improves Outcomes Compared With Sunitinib for Advanced Renal Cell Carcinoma
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

Ticagrelor or Clopidogrel in Elective Percutaneous Coronary Intervention

Article Type
Changed
Fri, 05/28/2021 - 01:15
Display Headline
Ticagrelor or Clopidogrel in Elective Percutaneous Coronary Intervention

Study Overview

Objective: To assess whether ticagrelor was superior to clopidogrel in reducing periprocedural myocardial necrosis in stable coronary patients undergoing elective percutaneous coronary intervention (PCI).

Design: Multicenter, open-label, and prospective randomized control trial. Setting and participants: A total of 1910 patients with indication for PCI and at least 1 high risk characteristic were randomized to either ticagrelor or clopidogrel.

Main outcome measures: The primary outcome was the composite of PCI-related type 4a or 4b myocardial infarction or major myocardial injury. The primary safety outcome was major bleeding, evaluated within 48 hours of PCI.

Main results: At 48 hours, the primary outcome was observed in 334 of 941 patients (35%) in the ticagrelor group and 341 of 942 patients (36%) in the clopidogrel group (odds ratio [OR], 0.97; 95% confidence interval [CI], 0.80-1.17; P = .75). The primary safety outcome did not differ between groups. Minor bleeding events at 30 days were more frequently observed with ticagrelor (11%) than clopidogrel (8%) (1.54; 95% CI 1.12-2.11; P = .007).

Conclusion: Among patients undergoing elective PCI, ticagrelor was not superior to clopidogrel in reducing periprocedural myocardial necrosis. Ticagrelor did not cause increase in major bleeding compared to clopidogrel but did increase the rate of minor bleeding at 30 days.

Commentary

Standard treatment after PCI includes dual antiplatelet therapy combining adenosine diphosphate (ADP) receptor antagonist and aspirin. The newer generation thienopyridine prasugrel and the reversible direct acting oral antagonist of the ADP receptor ticagrelor, provides consistent and greater antiplatelet effect compared to clopidogrel, and are superior in reducing ischemic events when compared to clopidogrel in patients presenting with acute coronary syndrome (ACS).1,2 Therefore, current guidelines recommend ticagrelor and prasugrel in preference to clopidogrel in patients presenting with ACS.3,4 However, whether these findings of improved outcomes with newer agents compared to clopidogrel extends to patients with stable ischemic heart disease presenting for elective PCI is unknown.

In this context, Silvain et al investigated this clinical question and compared ticagrelor and clopidogrel by performing a well-designed multicenter randomized control trial in patients presenting with elective PCI. At 48 hours and at 30 days the composite of PCI-related type 4 myocardial infarction or major myocardial injury defined by the third universal definition5 was similar between the ticagrelor and clopidogrel groups. Although the incidence of major bleeding was not significantly different between the 2 groups, minor bleeding at 30 days was higher in the ticagrelor group (11%) than clopidogrel (8%) (1.54; 95% CI, 1.12-2.11, P = .007).

 

 

The strengths of this current study include the randomized design and the large number of patients enrolled with adequate power to evaluate for superiority of ticagrelor compared to clopidogrel. This was a multicenter trial in Europe with 49 participating centers from France and Czech, and the interventional technique used by the operators reflects contemporary technique with 95% use of radial or ulnar access.

There are a few important points to consider in this study. First, the primary outcome was biomarker assessed myocardial necrosis and myocardial injury, and the study was not powered to assess the hard outcomes such as death and myocardial infarction. Although there have been previous reports describing the relationship between the postprocedural myocardial necrosis with worse outcomes, the definition of myocardial necrosis post-PCI and its relationship with hard outcomes remains controversial. Second, half of the patients enrolled were on chronic clopidogrel therapy which suggests that patients with inadequate platelet inhibition with clopidogrel may be under-represented in this cohort. Third, this was an open-label study and the knowledge of agent used could have affected the study results. Finally, whether the population included represents a true high-risk population is questionable. Some of the prespecified high-risk features necessary to enter the study was relatively light, such as presence of diabetes mellitus or body mass index > 30 kg/m2 compared to other criteria such as bifurcation stenting or left main stenting.

Currently, when treating patients with stable ischemic heart disease with higher risk anatomy, some operators may use ticagrelor over clopidogrel by extrapolating the study results from the ACS population. However, the results from the current study do not support the uniform use of ticagrelor in stable patients and suggests that the use of clopidogrel continues to be the standard of care. This is especially relevant considering the cost difference for the 2 agents studied. Whether there is a subgroup that benefits from ticagrelor use, such as patients with unprotected left main stenting or bifurcation stenting with 2 stent strategies, requires further investigation.

Applications for Clinical Practice

In patients presenting with stable ischemic heart disease undergoing elective PCI, ticagrelor did not lower composite of periprocedural myocardial infarction and myocardial injury at 48 hours. Clopidogrel continues to be a first line treatment after elective PCI.

References

1. Wiviott SD, Braunwald E, McCabe CH, et al. Prasugrel versus clopidogrel in patients with acute coronary syndromes. N Engl J Med. 2007;357(20):2001-15.

2. Wallentin L, Becker RC, Budaj A, et al. Ticagrelor versus Clopidogrel in Patients with Acute Coronary Syndromes. N Engl J Med. 2009;361(11):1045-57.

3. Ibanez B, James S, Agewall S, et al. 2017 ESC Guidelines for the management of acute myocardial infarction in patients presenting with ST-segment elevation: The Task Force for the management of acute myocardial infarction in patients presenting with ST-segment elevation of the European Society of Cardiology (ESC). Eur Heart J. 2018;39(2):119-177.

4. Levine GN, Bates ER, Bittl JA, et al. 2016 ACC/AHA guideline focused update on duration of dual antiplatelet therapy in patients with coronary artery disease: A report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. J Thorac Cardiovasc Surg. 2016;152(5):12432-1275.

5. Thygesen K, Alpert JS, Jaffe AS, et al. Third universal definition of myocardial infarction. J Am Coll Cardiol. 2012;60(16):1581-98.

Article PDF
Issue
Journal of Clinical Outcomes Management - 28(3)
Publications
Topics
Page Number
108-109
Sections
Article PDF
Article PDF

Study Overview

Objective: To assess whether ticagrelor was superior to clopidogrel in reducing periprocedural myocardial necrosis in stable coronary patients undergoing elective percutaneous coronary intervention (PCI).

Design: Multicenter, open-label, and prospective randomized control trial. Setting and participants: A total of 1910 patients with indication for PCI and at least 1 high risk characteristic were randomized to either ticagrelor or clopidogrel.

Main outcome measures: The primary outcome was the composite of PCI-related type 4a or 4b myocardial infarction or major myocardial injury. The primary safety outcome was major bleeding, evaluated within 48 hours of PCI.

Main results: At 48 hours, the primary outcome was observed in 334 of 941 patients (35%) in the ticagrelor group and 341 of 942 patients (36%) in the clopidogrel group (odds ratio [OR], 0.97; 95% confidence interval [CI], 0.80-1.17; P = .75). The primary safety outcome did not differ between groups. Minor bleeding events at 30 days were more frequently observed with ticagrelor (11%) than clopidogrel (8%) (1.54; 95% CI 1.12-2.11; P = .007).

Conclusion: Among patients undergoing elective PCI, ticagrelor was not superior to clopidogrel in reducing periprocedural myocardial necrosis. Ticagrelor did not cause increase in major bleeding compared to clopidogrel but did increase the rate of minor bleeding at 30 days.

Commentary

Standard treatment after PCI includes dual antiplatelet therapy combining adenosine diphosphate (ADP) receptor antagonist and aspirin. The newer generation thienopyridine prasugrel and the reversible direct acting oral antagonist of the ADP receptor ticagrelor, provides consistent and greater antiplatelet effect compared to clopidogrel, and are superior in reducing ischemic events when compared to clopidogrel in patients presenting with acute coronary syndrome (ACS).1,2 Therefore, current guidelines recommend ticagrelor and prasugrel in preference to clopidogrel in patients presenting with ACS.3,4 However, whether these findings of improved outcomes with newer agents compared to clopidogrel extends to patients with stable ischemic heart disease presenting for elective PCI is unknown.

In this context, Silvain et al investigated this clinical question and compared ticagrelor and clopidogrel by performing a well-designed multicenter randomized control trial in patients presenting with elective PCI. At 48 hours and at 30 days the composite of PCI-related type 4 myocardial infarction or major myocardial injury defined by the third universal definition5 was similar between the ticagrelor and clopidogrel groups. Although the incidence of major bleeding was not significantly different between the 2 groups, minor bleeding at 30 days was higher in the ticagrelor group (11%) than clopidogrel (8%) (1.54; 95% CI, 1.12-2.11, P = .007).

 

 

The strengths of this current study include the randomized design and the large number of patients enrolled with adequate power to evaluate for superiority of ticagrelor compared to clopidogrel. This was a multicenter trial in Europe with 49 participating centers from France and Czech, and the interventional technique used by the operators reflects contemporary technique with 95% use of radial or ulnar access.

There are a few important points to consider in this study. First, the primary outcome was biomarker assessed myocardial necrosis and myocardial injury, and the study was not powered to assess the hard outcomes such as death and myocardial infarction. Although there have been previous reports describing the relationship between the postprocedural myocardial necrosis with worse outcomes, the definition of myocardial necrosis post-PCI and its relationship with hard outcomes remains controversial. Second, half of the patients enrolled were on chronic clopidogrel therapy which suggests that patients with inadequate platelet inhibition with clopidogrel may be under-represented in this cohort. Third, this was an open-label study and the knowledge of agent used could have affected the study results. Finally, whether the population included represents a true high-risk population is questionable. Some of the prespecified high-risk features necessary to enter the study was relatively light, such as presence of diabetes mellitus or body mass index > 30 kg/m2 compared to other criteria such as bifurcation stenting or left main stenting.

Currently, when treating patients with stable ischemic heart disease with higher risk anatomy, some operators may use ticagrelor over clopidogrel by extrapolating the study results from the ACS population. However, the results from the current study do not support the uniform use of ticagrelor in stable patients and suggests that the use of clopidogrel continues to be the standard of care. This is especially relevant considering the cost difference for the 2 agents studied. Whether there is a subgroup that benefits from ticagrelor use, such as patients with unprotected left main stenting or bifurcation stenting with 2 stent strategies, requires further investigation.

Applications for Clinical Practice

In patients presenting with stable ischemic heart disease undergoing elective PCI, ticagrelor did not lower composite of periprocedural myocardial infarction and myocardial injury at 48 hours. Clopidogrel continues to be a first line treatment after elective PCI.

Study Overview

Objective: To assess whether ticagrelor was superior to clopidogrel in reducing periprocedural myocardial necrosis in stable coronary patients undergoing elective percutaneous coronary intervention (PCI).

Design: Multicenter, open-label, and prospective randomized control trial. Setting and participants: A total of 1910 patients with indication for PCI and at least 1 high risk characteristic were randomized to either ticagrelor or clopidogrel.

Main outcome measures: The primary outcome was the composite of PCI-related type 4a or 4b myocardial infarction or major myocardial injury. The primary safety outcome was major bleeding, evaluated within 48 hours of PCI.

Main results: At 48 hours, the primary outcome was observed in 334 of 941 patients (35%) in the ticagrelor group and 341 of 942 patients (36%) in the clopidogrel group (odds ratio [OR], 0.97; 95% confidence interval [CI], 0.80-1.17; P = .75). The primary safety outcome did not differ between groups. Minor bleeding events at 30 days were more frequently observed with ticagrelor (11%) than clopidogrel (8%) (1.54; 95% CI 1.12-2.11; P = .007).

Conclusion: Among patients undergoing elective PCI, ticagrelor was not superior to clopidogrel in reducing periprocedural myocardial necrosis. Ticagrelor did not cause increase in major bleeding compared to clopidogrel but did increase the rate of minor bleeding at 30 days.

Commentary

Standard treatment after PCI includes dual antiplatelet therapy combining adenosine diphosphate (ADP) receptor antagonist and aspirin. The newer generation thienopyridine prasugrel and the reversible direct acting oral antagonist of the ADP receptor ticagrelor, provides consistent and greater antiplatelet effect compared to clopidogrel, and are superior in reducing ischemic events when compared to clopidogrel in patients presenting with acute coronary syndrome (ACS).1,2 Therefore, current guidelines recommend ticagrelor and prasugrel in preference to clopidogrel in patients presenting with ACS.3,4 However, whether these findings of improved outcomes with newer agents compared to clopidogrel extends to patients with stable ischemic heart disease presenting for elective PCI is unknown.

In this context, Silvain et al investigated this clinical question and compared ticagrelor and clopidogrel by performing a well-designed multicenter randomized control trial in patients presenting with elective PCI. At 48 hours and at 30 days the composite of PCI-related type 4 myocardial infarction or major myocardial injury defined by the third universal definition5 was similar between the ticagrelor and clopidogrel groups. Although the incidence of major bleeding was not significantly different between the 2 groups, minor bleeding at 30 days was higher in the ticagrelor group (11%) than clopidogrel (8%) (1.54; 95% CI, 1.12-2.11, P = .007).

 

 

The strengths of this current study include the randomized design and the large number of patients enrolled with adequate power to evaluate for superiority of ticagrelor compared to clopidogrel. This was a multicenter trial in Europe with 49 participating centers from France and Czech, and the interventional technique used by the operators reflects contemporary technique with 95% use of radial or ulnar access.

There are a few important points to consider in this study. First, the primary outcome was biomarker assessed myocardial necrosis and myocardial injury, and the study was not powered to assess the hard outcomes such as death and myocardial infarction. Although there have been previous reports describing the relationship between the postprocedural myocardial necrosis with worse outcomes, the definition of myocardial necrosis post-PCI and its relationship with hard outcomes remains controversial. Second, half of the patients enrolled were on chronic clopidogrel therapy which suggests that patients with inadequate platelet inhibition with clopidogrel may be under-represented in this cohort. Third, this was an open-label study and the knowledge of agent used could have affected the study results. Finally, whether the population included represents a true high-risk population is questionable. Some of the prespecified high-risk features necessary to enter the study was relatively light, such as presence of diabetes mellitus or body mass index > 30 kg/m2 compared to other criteria such as bifurcation stenting or left main stenting.

Currently, when treating patients with stable ischemic heart disease with higher risk anatomy, some operators may use ticagrelor over clopidogrel by extrapolating the study results from the ACS population. However, the results from the current study do not support the uniform use of ticagrelor in stable patients and suggests that the use of clopidogrel continues to be the standard of care. This is especially relevant considering the cost difference for the 2 agents studied. Whether there is a subgroup that benefits from ticagrelor use, such as patients with unprotected left main stenting or bifurcation stenting with 2 stent strategies, requires further investigation.

Applications for Clinical Practice

In patients presenting with stable ischemic heart disease undergoing elective PCI, ticagrelor did not lower composite of periprocedural myocardial infarction and myocardial injury at 48 hours. Clopidogrel continues to be a first line treatment after elective PCI.

References

1. Wiviott SD, Braunwald E, McCabe CH, et al. Prasugrel versus clopidogrel in patients with acute coronary syndromes. N Engl J Med. 2007;357(20):2001-15.

2. Wallentin L, Becker RC, Budaj A, et al. Ticagrelor versus Clopidogrel in Patients with Acute Coronary Syndromes. N Engl J Med. 2009;361(11):1045-57.

3. Ibanez B, James S, Agewall S, et al. 2017 ESC Guidelines for the management of acute myocardial infarction in patients presenting with ST-segment elevation: The Task Force for the management of acute myocardial infarction in patients presenting with ST-segment elevation of the European Society of Cardiology (ESC). Eur Heart J. 2018;39(2):119-177.

4. Levine GN, Bates ER, Bittl JA, et al. 2016 ACC/AHA guideline focused update on duration of dual antiplatelet therapy in patients with coronary artery disease: A report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. J Thorac Cardiovasc Surg. 2016;152(5):12432-1275.

5. Thygesen K, Alpert JS, Jaffe AS, et al. Third universal definition of myocardial infarction. J Am Coll Cardiol. 2012;60(16):1581-98.

References

1. Wiviott SD, Braunwald E, McCabe CH, et al. Prasugrel versus clopidogrel in patients with acute coronary syndromes. N Engl J Med. 2007;357(20):2001-15.

2. Wallentin L, Becker RC, Budaj A, et al. Ticagrelor versus Clopidogrel in Patients with Acute Coronary Syndromes. N Engl J Med. 2009;361(11):1045-57.

3. Ibanez B, James S, Agewall S, et al. 2017 ESC Guidelines for the management of acute myocardial infarction in patients presenting with ST-segment elevation: The Task Force for the management of acute myocardial infarction in patients presenting with ST-segment elevation of the European Society of Cardiology (ESC). Eur Heart J. 2018;39(2):119-177.

4. Levine GN, Bates ER, Bittl JA, et al. 2016 ACC/AHA guideline focused update on duration of dual antiplatelet therapy in patients with coronary artery disease: A report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. J Thorac Cardiovasc Surg. 2016;152(5):12432-1275.

5. Thygesen K, Alpert JS, Jaffe AS, et al. Third universal definition of myocardial infarction. J Am Coll Cardiol. 2012;60(16):1581-98.

Issue
Journal of Clinical Outcomes Management - 28(3)
Issue
Journal of Clinical Outcomes Management - 28(3)
Page Number
108-109
Page Number
108-109
Publications
Publications
Topics
Article Type
Display Headline
Ticagrelor or Clopidogrel in Elective Percutaneous Coronary Intervention
Display Headline
Ticagrelor or Clopidogrel in Elective Percutaneous Coronary Intervention
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

Implementation of a Symptom–Triggered Protocol for Severe Alcohol Withdrawal Treatment in a Medical Step-down Unit

Article Type
Changed
Fri, 05/28/2021 - 01:15
Display Headline
Implementation of a Symptom–Triggered Protocol for Severe Alcohol Withdrawal Treatment in a Medical Step-down Unit

From Stamford Hospital, Stamford, CT.

Objective: This single-center, quasi-experimental study of adult patients admitted or transferred to a medical step-down unit with alcohol withdrawal diagnoses sought to determine if symptom–triggered therapy (STT) is more effective than combined fixed-scheduled (FS) and STT in severe alcohol withdrawal.

Methods: In the preintervention group (72 episodes), patients were treated with FS and STT based on physician preference. In the postintervention group (69 episodes), providers were required to utilize only the STT protocol.

Results: Implementation of the intervention was associated with a significant reduction in average (per patient) cumulative benzodiazepine dose, from 250 mg to 96 mg (P < .001) and a decrease in average length of stay from 8.0 days to 5.1 days (P < .001). Secondary safety measures included a reduction in the proportion of patients who experienced delirium tremens from 47.5% to 22.5% (P < .001), and a reduction in intubation rates from 13.8% to 1.3% (P = .003).

Conclusion: The STT protocol proved to be more effective and safer in treating severe alcohol withdrawal patients than usual care employing STT with FS. We believe the successful implementation of a STT protocol in high-acuity patients requires frequent monitoring to assess withdrawal severity combined with appropriate and timely dosing of benzodiazepines.

Keywords: alcohol withdrawal delirium; alcohol withdrawal syndrome; treatment protocol; benzodiazepine; lorazepam.

Management of severe alcohol withdrawal and delirium tremens (DT) is challenging and requires significant resources, including close monitoring and intensive treatment, frequently in an intensive care unit (ICU).1 Early diagnosis and therapeutic intervention are important to limit potential complications associated with DT.2 Benzodiazepines are first-line therapeutic agents, but the definition of optimal use and dosing regimens has been limited, due to a lack of randomized controlled trials. In lower acuity patients admitted to a detoxification unit, systematic symptom–triggered benzodiazepine therapy (STT) has been established to be more effective than fixed-schedule (FS) dosing.3-5 Patients treated using STT require lower total benzodiazepine dosing and achieve shorter treatment durations. However, in higher-acuity patients admitted to general medical services, analyses have not shown an advantage of STT over combined FS and STT.6

 

 

Methods

The purpose of this study was to determine whether implementation of STT is more effective than FS dosing combined with episodic STT in the management of hospitalized high-acuity alcohol withdrawal patients. We conducted a preintervention and postintervention quasi-experimental study in the step-down unit (SDU) of a 305-bed community teaching hospital. The study population consisted of adult inpatients 18 years or older admitted or transferred to the 12-bed SDU with alcohol withdrawal, as defined by primary or secondary International Classification of Diseases, Tenth Revision diagnoses. SDU admission criteria included patients with prior DT or those who had received multiple doses of benzodiazepines in the emergency department. In-hospital transfer to the SDU was at the physician’s discretion, if the patient required escalating doses of benzodiazepines or the use of increasing resources, such as those for behavioral emergencies. The majority of patients admitted or transferred to the SDU were assigned to medical house staff teams under hospitalist supervision, and, on occasion, under community physicians. The nurse-to-patient ratio in the SDU was 1:3.

Study groups

The preintervention group consisted of 80 successive treatment episodes involving patients admitted or transferred to the SDU from December 2, 2015, to July 1, 2017. Patients were treated based upon physician preference, consisting of a scheduled dosing regimen with additional doses as needed. The postintervention group included 80 successive treatment episodes involving patients admitted or transferred to the SDU from October 1, 2017, to March 23, 2019. The STT protocol was used in all patients in the postintervention group.

In the preintervention group, fixed, scheduled doses of lorazepam or chlordiazepoxide and as-needed lorazepam were prescribed and adjusted based upon physician judgment. Monitoring of symptom severity was scored using the revised Clinical Institute Withdrawal Assessment for Alcohol scale (CIWA-Ar). Benzodiazepine dosing occurred if the CIWA-Ar score had increased 2 or more points from the last score.

In the postintervention group, the STT protocol included the creation of a standardized physician order set for benzodiazepine “sliding scale” administration. The STT protocol allowed for escalating doses for higher withdrawal scores. Symptom severity was scored using MINDS (Minnesota Detoxification Scale) criteria.1 Lorazepam as-needed dosing was based upon MINDS scores. A MINDS score less than 10 resulted in no medication, MINDS 10-12 required 2 mg, MINDS 13-16 required 4 mg, MINDS 17-19 required 6 mg, and MINDS 20 required 8 mg and a call to the physician. Transfer to the ICU was recommended if the MINDS score was ≥ 20 for 3 consecutive hours. Monitoring intervals occurred more frequently at 30 minutes unless the MINDS score was less than 10. After 7 days, the MINDS protocol was recommended to be discontinued, as the patient might have had iatrogenic delirium.

The STT protocol was introduced during a didactic session for the hospitalists and a separate session for internal medicine and family residents. Each registered nurse working in the SDU was trained in the use of the STT protocol and MINDS during nursing huddles.

 

 

Patients were excluded from evaluation if they were transferred to the SDU after 7 or more days in the hospital, if they had stayed in the hospital more than 30 days, were chronically on benzodiazepine therapy (to avoid confounding withdrawal symptoms), or if they left the hospital against medical advice (AMA). To avoid bias in the results, the patients with early discontinuation of treatment were included in analyses of secondary outcomes, thus resulting in all 80 episodes analyzed.

Measures and data

The primary outcome measure was benzodiazepine dose intensity, expressed in total lorazepam-equivalents. Secondary measures included average length of stay (including general medical, surgical, and ICU days), seizure incidence, DT incidence, sitter use, behavioral emergency responses, rates of leaving AMA, intubation, transfer to the ICU, and death.

Benzodiazepine dosing and length of stay were obtained from the data warehouse of the hospital’s electronic health record (EHR; Meditech). Benzodiazepine dosing was expressed in total lorazepam-equivalents, with conversion as follows: lorazepam orally and intravenously 1 mg = chlordiazepoxide 25 mg = diazepam 5 mg. All other measures were obtained from chart review of the patients’ EMR entries. The Stamford Hospital Institutional Review Board approved this study.

Analysis

Data analyses for this study were performed using SPSS version 25.0 (IBM). Categorical data were reported as frequency (count) and percent within category. Continuous data were reported as mean (SD). Categorical data were analyzed using χ2 analysis; continuous data were analyzed using t-tests. A P value of .05 was considered significant for each analysis.

Results

During the preintervention period, 72 episodes (58 patients) met inclusion criteria, and 69 episodes (55 patients) met inclusion criteria during the postintervention period. Ten patients were represented in both groups. Eight preintervention episodes were excluded from the primary analysis because the patient left AMA. Eleven postintervention episodes were excluded: 9 due to patients leaving AMA, 1 due to chronic benzodiazepine usage, and 1 due to transfer to the SDU unit after 7 days. Baseline characteristics and medication use profiles of the preintervention and postintervention groups are summarized in Table 1.

Comparison of Demographic Characteristics by Preintervention and Postintervention Group

 

 

Implementation of the intervention was associated with a significant reduction in average (per patient) cumulative benzodiazepine dose, from 250 mg to 96 mg (P < .001), as shown in Table 2. Average length of stay decreased from 8.0 days to 5.1 days (P < .001). Secondary safety measures were notable for a reduction in DT incidence, from 47.5% to 22.5% (P < .001), and lower rates of intubation, from 13.8% to 1.3% (P = .003). Seven-day readmission rates were 0% preintervention and 1.4% postintervention.

Comparison of Treatment Outcomes by Treatment Group

Discussion

We found that hospitalized patients with severe alcohol withdrawal treated with STT required fewer benzodiazepines and had a lower length of stay than patients treated with a conventional combined STT and FS regimen. Implementation of the change from the STT and FS approach to the STT approach in the SDU resulted in concerns that waiting for symptoms to appear could result in more severe withdrawal and prolonged treatment.3 To address this, the intervention included monitoring and dosing every 30 minutes, as compared to monitoring and dosing every 1 hour preintervention. In addition, a sliding-scale approach to match alcohol withdrawal score with dosage was employed in postintervention patients.

Employment of the STT protocol also resulted in decreased complications, including lower rates of DT and transfer to the ICU. This new intervention resulted in significantly decreased time required to control severe symptoms. In the preintervention phase, if a patient’s symptoms escalated despite administration of the as-needed dose of benzodiazepine, there was often a delay in administration of additional doses due to the time needed for nurses to reach a physician and subsequent placement of a new order. In the postintervention phase, the STT protocol allowed nursing staff to give benzodiazepines without delay when needed. We believe this reduced the number of calls by nursing staff to physicians requesting additional medications, and that this improved teamwork when managing these patients.

As part of the intervention, a decision was made to use the MINDS scale rather than the CIWA-Ar scale to assess withdrawal severity. This was because the CIWA-Ar has only been validated in patients with uncomplicated alcohol withdrawal syndrome and has not been researched extensively in patients requiring ICU-level care.1 MINDS assessment has proven to be reliable and reflects severity of withdrawal. Furthermore, MINDS requires less time to administer—3 to 5 minutes vs 5 to 15 minutes for the CIWA-Ar scale. CIWA-Ar, unlike MINDS, requires subjective input from the patient, which is less reliable for higher acuity patients. Our study is unique in that it focused on high-acuity patients and it showed both a significant reduction in quantity of benzodiazepines prescribed and length of stay. Previous studies on lower acuity patients in detoxification units have confirmed that STT is more effective than a FS approach.3-5 In patients of higher acuity, STT has not proven to be superior.

A key lesson learned was the need for proper education of nursing staff. Concurrent nursing audits were necessary to ensure that scoring was performed in an accurate and timely manner. In addition, it was challenging to predict which patients might develop DTs versus those requiring a brief inpatient stay. While there was initial concern that an STT protocol could result in underdosing, we found that patients had fewer DT episodes and fewer ICU transfers.

 

 

This study had several limitations. These include a relatively small sample size and the data being less recent. As there has been no intervening change to the therapeutic paradigm of DT treatment, the findings remain pertinent to the present time. The study employed a simple pre/post design and was conducted in a single setting. We are not aware of any temporal or local trends likely to influence these results. Admissions and transfers to the SDU for severe alcohol withdrawal were based on physician discretion. However, patient characteristics in both groups were similar (Table 1). We note that the postintervention STT protocol allowed for more frequent benzodiazepine dosing, though benzodiazepine use did decrease. Different alcohol withdrawal scores (MINDS vs. CIWA-Ar) were used for postintervention and preintervention, although previous research has shown that MINDS and CIWA-Ar scores correlate well.7 Finally, some patients of higher acuity and complexity were excluded, potentially limiting the generalizability of our results.

Conclusion

Our STT protocol proved to be more effective and safer in treating severe alcohol withdrawal patients than usual care employing STT with FS. We believe the successful implementation of a STT protocol in high-acuity patients also requires frequent monitoring using the MINDS scale, integrated with benzodiazepine sliding-scale dosing to match symptom severity. This bundled approach resulted in a significant reduction of benzodiazepine usage and reduced length of stay. Timely treatment of these patients also reduced the percent of patients developing DTs, and reduced intubation rates and transfers to the ICU. Further studies may be warranted at other sites to confirm the effectiveness of this STT protocol.

Corresponding author: Paul W. Huang, MD, Stamford Hospital, One Hospital Plaza, PO Box 9317, Stamford, CT 06904; phuang@stamhealth.org.

Financial disclosures: None.

References

1. DeCarolis DD, Rice KL, Ho L, et al. Symptom-driven lorazepam protocol for treatment of severe alcohol withdrawal delirium in the intensive care unit. Pharmacotherapy. 2007;27(4):510-518.

2. DeBellis R, Smith BS, Choi S, Malloy M. Management of delirium tremens. J Intensive Care Med. 2005;20(3):164-173.

3. Saitz R, Mayo-Smith MF, Roberts MS, et al. Individualized treatment for alcohol withdrawal. A randomized double-blind controlled trial. JAMA. 1994;272(7):519-523.

4. Sachdeva A, Chandra M, Deshpande SN. A comparative study of fixed tapering dose regimen versus symptom-triggered regimen of lorazepam for alcohol detoxification. Alcohol Alcohol. 2014;49(3):287-291.

5. Daeppen JB, Gache P, Landry U, et al. Symptom-triggered vs fixed-schedule doses of benzodiazepine for alcohol withdrawal: a randomized treatment trial. Arch Intern Med. 2002;162(10):1117-1121.

6. Jaeger TM, Lohr RH, Pankratz VS. Symptom-triggered therapy for alcohol withdrawal syndrome in medical inpatients. Mayo Clin Proc. 2001;76(7):695-701.

7. Littlefield AJ, Heavner MS, Eng CC, et al. Correlation Between mMINDS and CIWA-Ar Scoring Tools in Patients With Alcohol Withdrawal Syndrome. Am J Crit Care. 2018;27(4):280-286.

Article PDF
Issue
Journal of Clinical Outcomes Management - 28(3)
Publications
Topics
Page Number
134-138
Sections
Article PDF
Article PDF

From Stamford Hospital, Stamford, CT.

Objective: This single-center, quasi-experimental study of adult patients admitted or transferred to a medical step-down unit with alcohol withdrawal diagnoses sought to determine if symptom–triggered therapy (STT) is more effective than combined fixed-scheduled (FS) and STT in severe alcohol withdrawal.

Methods: In the preintervention group (72 episodes), patients were treated with FS and STT based on physician preference. In the postintervention group (69 episodes), providers were required to utilize only the STT protocol.

Results: Implementation of the intervention was associated with a significant reduction in average (per patient) cumulative benzodiazepine dose, from 250 mg to 96 mg (P < .001) and a decrease in average length of stay from 8.0 days to 5.1 days (P < .001). Secondary safety measures included a reduction in the proportion of patients who experienced delirium tremens from 47.5% to 22.5% (P < .001), and a reduction in intubation rates from 13.8% to 1.3% (P = .003).

Conclusion: The STT protocol proved to be more effective and safer in treating severe alcohol withdrawal patients than usual care employing STT with FS. We believe the successful implementation of a STT protocol in high-acuity patients requires frequent monitoring to assess withdrawal severity combined with appropriate and timely dosing of benzodiazepines.

Keywords: alcohol withdrawal delirium; alcohol withdrawal syndrome; treatment protocol; benzodiazepine; lorazepam.

Management of severe alcohol withdrawal and delirium tremens (DT) is challenging and requires significant resources, including close monitoring and intensive treatment, frequently in an intensive care unit (ICU).1 Early diagnosis and therapeutic intervention are important to limit potential complications associated with DT.2 Benzodiazepines are first-line therapeutic agents, but the definition of optimal use and dosing regimens has been limited, due to a lack of randomized controlled trials. In lower acuity patients admitted to a detoxification unit, systematic symptom–triggered benzodiazepine therapy (STT) has been established to be more effective than fixed-schedule (FS) dosing.3-5 Patients treated using STT require lower total benzodiazepine dosing and achieve shorter treatment durations. However, in higher-acuity patients admitted to general medical services, analyses have not shown an advantage of STT over combined FS and STT.6

 

 

Methods

The purpose of this study was to determine whether implementation of STT is more effective than FS dosing combined with episodic STT in the management of hospitalized high-acuity alcohol withdrawal patients. We conducted a preintervention and postintervention quasi-experimental study in the step-down unit (SDU) of a 305-bed community teaching hospital. The study population consisted of adult inpatients 18 years or older admitted or transferred to the 12-bed SDU with alcohol withdrawal, as defined by primary or secondary International Classification of Diseases, Tenth Revision diagnoses. SDU admission criteria included patients with prior DT or those who had received multiple doses of benzodiazepines in the emergency department. In-hospital transfer to the SDU was at the physician’s discretion, if the patient required escalating doses of benzodiazepines or the use of increasing resources, such as those for behavioral emergencies. The majority of patients admitted or transferred to the SDU were assigned to medical house staff teams under hospitalist supervision, and, on occasion, under community physicians. The nurse-to-patient ratio in the SDU was 1:3.

Study groups

The preintervention group consisted of 80 successive treatment episodes involving patients admitted or transferred to the SDU from December 2, 2015, to July 1, 2017. Patients were treated based upon physician preference, consisting of a scheduled dosing regimen with additional doses as needed. The postintervention group included 80 successive treatment episodes involving patients admitted or transferred to the SDU from October 1, 2017, to March 23, 2019. The STT protocol was used in all patients in the postintervention group.

In the preintervention group, fixed, scheduled doses of lorazepam or chlordiazepoxide and as-needed lorazepam were prescribed and adjusted based upon physician judgment. Monitoring of symptom severity was scored using the revised Clinical Institute Withdrawal Assessment for Alcohol scale (CIWA-Ar). Benzodiazepine dosing occurred if the CIWA-Ar score had increased 2 or more points from the last score.

In the postintervention group, the STT protocol included the creation of a standardized physician order set for benzodiazepine “sliding scale” administration. The STT protocol allowed for escalating doses for higher withdrawal scores. Symptom severity was scored using MINDS (Minnesota Detoxification Scale) criteria.1 Lorazepam as-needed dosing was based upon MINDS scores. A MINDS score less than 10 resulted in no medication, MINDS 10-12 required 2 mg, MINDS 13-16 required 4 mg, MINDS 17-19 required 6 mg, and MINDS 20 required 8 mg and a call to the physician. Transfer to the ICU was recommended if the MINDS score was ≥ 20 for 3 consecutive hours. Monitoring intervals occurred more frequently at 30 minutes unless the MINDS score was less than 10. After 7 days, the MINDS protocol was recommended to be discontinued, as the patient might have had iatrogenic delirium.

The STT protocol was introduced during a didactic session for the hospitalists and a separate session for internal medicine and family residents. Each registered nurse working in the SDU was trained in the use of the STT protocol and MINDS during nursing huddles.

 

 

Patients were excluded from evaluation if they were transferred to the SDU after 7 or more days in the hospital, if they had stayed in the hospital more than 30 days, were chronically on benzodiazepine therapy (to avoid confounding withdrawal symptoms), or if they left the hospital against medical advice (AMA). To avoid bias in the results, the patients with early discontinuation of treatment were included in analyses of secondary outcomes, thus resulting in all 80 episodes analyzed.

Measures and data

The primary outcome measure was benzodiazepine dose intensity, expressed in total lorazepam-equivalents. Secondary measures included average length of stay (including general medical, surgical, and ICU days), seizure incidence, DT incidence, sitter use, behavioral emergency responses, rates of leaving AMA, intubation, transfer to the ICU, and death.

Benzodiazepine dosing and length of stay were obtained from the data warehouse of the hospital’s electronic health record (EHR; Meditech). Benzodiazepine dosing was expressed in total lorazepam-equivalents, with conversion as follows: lorazepam orally and intravenously 1 mg = chlordiazepoxide 25 mg = diazepam 5 mg. All other measures were obtained from chart review of the patients’ EMR entries. The Stamford Hospital Institutional Review Board approved this study.

Analysis

Data analyses for this study were performed using SPSS version 25.0 (IBM). Categorical data were reported as frequency (count) and percent within category. Continuous data were reported as mean (SD). Categorical data were analyzed using χ2 analysis; continuous data were analyzed using t-tests. A P value of .05 was considered significant for each analysis.

Results

During the preintervention period, 72 episodes (58 patients) met inclusion criteria, and 69 episodes (55 patients) met inclusion criteria during the postintervention period. Ten patients were represented in both groups. Eight preintervention episodes were excluded from the primary analysis because the patient left AMA. Eleven postintervention episodes were excluded: 9 due to patients leaving AMA, 1 due to chronic benzodiazepine usage, and 1 due to transfer to the SDU unit after 7 days. Baseline characteristics and medication use profiles of the preintervention and postintervention groups are summarized in Table 1.

Comparison of Demographic Characteristics by Preintervention and Postintervention Group

 

 

Implementation of the intervention was associated with a significant reduction in average (per patient) cumulative benzodiazepine dose, from 250 mg to 96 mg (P < .001), as shown in Table 2. Average length of stay decreased from 8.0 days to 5.1 days (P < .001). Secondary safety measures were notable for a reduction in DT incidence, from 47.5% to 22.5% (P < .001), and lower rates of intubation, from 13.8% to 1.3% (P = .003). Seven-day readmission rates were 0% preintervention and 1.4% postintervention.

Comparison of Treatment Outcomes by Treatment Group

Discussion

We found that hospitalized patients with severe alcohol withdrawal treated with STT required fewer benzodiazepines and had a lower length of stay than patients treated with a conventional combined STT and FS regimen. Implementation of the change from the STT and FS approach to the STT approach in the SDU resulted in concerns that waiting for symptoms to appear could result in more severe withdrawal and prolonged treatment.3 To address this, the intervention included monitoring and dosing every 30 minutes, as compared to monitoring and dosing every 1 hour preintervention. In addition, a sliding-scale approach to match alcohol withdrawal score with dosage was employed in postintervention patients.

Employment of the STT protocol also resulted in decreased complications, including lower rates of DT and transfer to the ICU. This new intervention resulted in significantly decreased time required to control severe symptoms. In the preintervention phase, if a patient’s symptoms escalated despite administration of the as-needed dose of benzodiazepine, there was often a delay in administration of additional doses due to the time needed for nurses to reach a physician and subsequent placement of a new order. In the postintervention phase, the STT protocol allowed nursing staff to give benzodiazepines without delay when needed. We believe this reduced the number of calls by nursing staff to physicians requesting additional medications, and that this improved teamwork when managing these patients.

As part of the intervention, a decision was made to use the MINDS scale rather than the CIWA-Ar scale to assess withdrawal severity. This was because the CIWA-Ar has only been validated in patients with uncomplicated alcohol withdrawal syndrome and has not been researched extensively in patients requiring ICU-level care.1 MINDS assessment has proven to be reliable and reflects severity of withdrawal. Furthermore, MINDS requires less time to administer—3 to 5 minutes vs 5 to 15 minutes for the CIWA-Ar scale. CIWA-Ar, unlike MINDS, requires subjective input from the patient, which is less reliable for higher acuity patients. Our study is unique in that it focused on high-acuity patients and it showed both a significant reduction in quantity of benzodiazepines prescribed and length of stay. Previous studies on lower acuity patients in detoxification units have confirmed that STT is more effective than a FS approach.3-5 In patients of higher acuity, STT has not proven to be superior.

A key lesson learned was the need for proper education of nursing staff. Concurrent nursing audits were necessary to ensure that scoring was performed in an accurate and timely manner. In addition, it was challenging to predict which patients might develop DTs versus those requiring a brief inpatient stay. While there was initial concern that an STT protocol could result in underdosing, we found that patients had fewer DT episodes and fewer ICU transfers.

 

 

This study had several limitations. These include a relatively small sample size and the data being less recent. As there has been no intervening change to the therapeutic paradigm of DT treatment, the findings remain pertinent to the present time. The study employed a simple pre/post design and was conducted in a single setting. We are not aware of any temporal or local trends likely to influence these results. Admissions and transfers to the SDU for severe alcohol withdrawal were based on physician discretion. However, patient characteristics in both groups were similar (Table 1). We note that the postintervention STT protocol allowed for more frequent benzodiazepine dosing, though benzodiazepine use did decrease. Different alcohol withdrawal scores (MINDS vs. CIWA-Ar) were used for postintervention and preintervention, although previous research has shown that MINDS and CIWA-Ar scores correlate well.7 Finally, some patients of higher acuity and complexity were excluded, potentially limiting the generalizability of our results.

Conclusion

Our STT protocol proved to be more effective and safer in treating severe alcohol withdrawal patients than usual care employing STT with FS. We believe the successful implementation of a STT protocol in high-acuity patients also requires frequent monitoring using the MINDS scale, integrated with benzodiazepine sliding-scale dosing to match symptom severity. This bundled approach resulted in a significant reduction of benzodiazepine usage and reduced length of stay. Timely treatment of these patients also reduced the percent of patients developing DTs, and reduced intubation rates and transfers to the ICU. Further studies may be warranted at other sites to confirm the effectiveness of this STT protocol.

Corresponding author: Paul W. Huang, MD, Stamford Hospital, One Hospital Plaza, PO Box 9317, Stamford, CT 06904; phuang@stamhealth.org.

Financial disclosures: None.

From Stamford Hospital, Stamford, CT.

Objective: This single-center, quasi-experimental study of adult patients admitted or transferred to a medical step-down unit with alcohol withdrawal diagnoses sought to determine if symptom–triggered therapy (STT) is more effective than combined fixed-scheduled (FS) and STT in severe alcohol withdrawal.

Methods: In the preintervention group (72 episodes), patients were treated with FS and STT based on physician preference. In the postintervention group (69 episodes), providers were required to utilize only the STT protocol.

Results: Implementation of the intervention was associated with a significant reduction in average (per patient) cumulative benzodiazepine dose, from 250 mg to 96 mg (P < .001) and a decrease in average length of stay from 8.0 days to 5.1 days (P < .001). Secondary safety measures included a reduction in the proportion of patients who experienced delirium tremens from 47.5% to 22.5% (P < .001), and a reduction in intubation rates from 13.8% to 1.3% (P = .003).

Conclusion: The STT protocol proved to be more effective and safer in treating severe alcohol withdrawal patients than usual care employing STT with FS. We believe the successful implementation of a STT protocol in high-acuity patients requires frequent monitoring to assess withdrawal severity combined with appropriate and timely dosing of benzodiazepines.

Keywords: alcohol withdrawal delirium; alcohol withdrawal syndrome; treatment protocol; benzodiazepine; lorazepam.

Management of severe alcohol withdrawal and delirium tremens (DT) is challenging and requires significant resources, including close monitoring and intensive treatment, frequently in an intensive care unit (ICU).1 Early diagnosis and therapeutic intervention are important to limit potential complications associated with DT.2 Benzodiazepines are first-line therapeutic agents, but the definition of optimal use and dosing regimens has been limited, due to a lack of randomized controlled trials. In lower acuity patients admitted to a detoxification unit, systematic symptom–triggered benzodiazepine therapy (STT) has been established to be more effective than fixed-schedule (FS) dosing.3-5 Patients treated using STT require lower total benzodiazepine dosing and achieve shorter treatment durations. However, in higher-acuity patients admitted to general medical services, analyses have not shown an advantage of STT over combined FS and STT.6

 

 

Methods

The purpose of this study was to determine whether implementation of STT is more effective than FS dosing combined with episodic STT in the management of hospitalized high-acuity alcohol withdrawal patients. We conducted a preintervention and postintervention quasi-experimental study in the step-down unit (SDU) of a 305-bed community teaching hospital. The study population consisted of adult inpatients 18 years or older admitted or transferred to the 12-bed SDU with alcohol withdrawal, as defined by primary or secondary International Classification of Diseases, Tenth Revision diagnoses. SDU admission criteria included patients with prior DT or those who had received multiple doses of benzodiazepines in the emergency department. In-hospital transfer to the SDU was at the physician’s discretion, if the patient required escalating doses of benzodiazepines or the use of increasing resources, such as those for behavioral emergencies. The majority of patients admitted or transferred to the SDU were assigned to medical house staff teams under hospitalist supervision, and, on occasion, under community physicians. The nurse-to-patient ratio in the SDU was 1:3.

Study groups

The preintervention group consisted of 80 successive treatment episodes involving patients admitted or transferred to the SDU from December 2, 2015, to July 1, 2017. Patients were treated based upon physician preference, consisting of a scheduled dosing regimen with additional doses as needed. The postintervention group included 80 successive treatment episodes involving patients admitted or transferred to the SDU from October 1, 2017, to March 23, 2019. The STT protocol was used in all patients in the postintervention group.

In the preintervention group, fixed, scheduled doses of lorazepam or chlordiazepoxide and as-needed lorazepam were prescribed and adjusted based upon physician judgment. Monitoring of symptom severity was scored using the revised Clinical Institute Withdrawal Assessment for Alcohol scale (CIWA-Ar). Benzodiazepine dosing occurred if the CIWA-Ar score had increased 2 or more points from the last score.

In the postintervention group, the STT protocol included the creation of a standardized physician order set for benzodiazepine “sliding scale” administration. The STT protocol allowed for escalating doses for higher withdrawal scores. Symptom severity was scored using MINDS (Minnesota Detoxification Scale) criteria.1 Lorazepam as-needed dosing was based upon MINDS scores. A MINDS score less than 10 resulted in no medication, MINDS 10-12 required 2 mg, MINDS 13-16 required 4 mg, MINDS 17-19 required 6 mg, and MINDS 20 required 8 mg and a call to the physician. Transfer to the ICU was recommended if the MINDS score was ≥ 20 for 3 consecutive hours. Monitoring intervals occurred more frequently at 30 minutes unless the MINDS score was less than 10. After 7 days, the MINDS protocol was recommended to be discontinued, as the patient might have had iatrogenic delirium.

The STT protocol was introduced during a didactic session for the hospitalists and a separate session for internal medicine and family residents. Each registered nurse working in the SDU was trained in the use of the STT protocol and MINDS during nursing huddles.

 

 

Patients were excluded from evaluation if they were transferred to the SDU after 7 or more days in the hospital, if they had stayed in the hospital more than 30 days, were chronically on benzodiazepine therapy (to avoid confounding withdrawal symptoms), or if they left the hospital against medical advice (AMA). To avoid bias in the results, the patients with early discontinuation of treatment were included in analyses of secondary outcomes, thus resulting in all 80 episodes analyzed.

Measures and data

The primary outcome measure was benzodiazepine dose intensity, expressed in total lorazepam-equivalents. Secondary measures included average length of stay (including general medical, surgical, and ICU days), seizure incidence, DT incidence, sitter use, behavioral emergency responses, rates of leaving AMA, intubation, transfer to the ICU, and death.

Benzodiazepine dosing and length of stay were obtained from the data warehouse of the hospital’s electronic health record (EHR; Meditech). Benzodiazepine dosing was expressed in total lorazepam-equivalents, with conversion as follows: lorazepam orally and intravenously 1 mg = chlordiazepoxide 25 mg = diazepam 5 mg. All other measures were obtained from chart review of the patients’ EMR entries. The Stamford Hospital Institutional Review Board approved this study.

Analysis

Data analyses for this study were performed using SPSS version 25.0 (IBM). Categorical data were reported as frequency (count) and percent within category. Continuous data were reported as mean (SD). Categorical data were analyzed using χ2 analysis; continuous data were analyzed using t-tests. A P value of .05 was considered significant for each analysis.

Results

During the preintervention period, 72 episodes (58 patients) met inclusion criteria, and 69 episodes (55 patients) met inclusion criteria during the postintervention period. Ten patients were represented in both groups. Eight preintervention episodes were excluded from the primary analysis because the patient left AMA. Eleven postintervention episodes were excluded: 9 due to patients leaving AMA, 1 due to chronic benzodiazepine usage, and 1 due to transfer to the SDU unit after 7 days. Baseline characteristics and medication use profiles of the preintervention and postintervention groups are summarized in Table 1.

Comparison of Demographic Characteristics by Preintervention and Postintervention Group

 

 

Implementation of the intervention was associated with a significant reduction in average (per patient) cumulative benzodiazepine dose, from 250 mg to 96 mg (P < .001), as shown in Table 2. Average length of stay decreased from 8.0 days to 5.1 days (P < .001). Secondary safety measures were notable for a reduction in DT incidence, from 47.5% to 22.5% (P < .001), and lower rates of intubation, from 13.8% to 1.3% (P = .003). Seven-day readmission rates were 0% preintervention and 1.4% postintervention.

Comparison of Treatment Outcomes by Treatment Group

Discussion

We found that hospitalized patients with severe alcohol withdrawal treated with STT required fewer benzodiazepines and had a lower length of stay than patients treated with a conventional combined STT and FS regimen. Implementation of the change from the STT and FS approach to the STT approach in the SDU resulted in concerns that waiting for symptoms to appear could result in more severe withdrawal and prolonged treatment.3 To address this, the intervention included monitoring and dosing every 30 minutes, as compared to monitoring and dosing every 1 hour preintervention. In addition, a sliding-scale approach to match alcohol withdrawal score with dosage was employed in postintervention patients.

Employment of the STT protocol also resulted in decreased complications, including lower rates of DT and transfer to the ICU. This new intervention resulted in significantly decreased time required to control severe symptoms. In the preintervention phase, if a patient’s symptoms escalated despite administration of the as-needed dose of benzodiazepine, there was often a delay in administration of additional doses due to the time needed for nurses to reach a physician and subsequent placement of a new order. In the postintervention phase, the STT protocol allowed nursing staff to give benzodiazepines without delay when needed. We believe this reduced the number of calls by nursing staff to physicians requesting additional medications, and that this improved teamwork when managing these patients.

As part of the intervention, a decision was made to use the MINDS scale rather than the CIWA-Ar scale to assess withdrawal severity. This was because the CIWA-Ar has only been validated in patients with uncomplicated alcohol withdrawal syndrome and has not been researched extensively in patients requiring ICU-level care.1 MINDS assessment has proven to be reliable and reflects severity of withdrawal. Furthermore, MINDS requires less time to administer—3 to 5 minutes vs 5 to 15 minutes for the CIWA-Ar scale. CIWA-Ar, unlike MINDS, requires subjective input from the patient, which is less reliable for higher acuity patients. Our study is unique in that it focused on high-acuity patients and it showed both a significant reduction in quantity of benzodiazepines prescribed and length of stay. Previous studies on lower acuity patients in detoxification units have confirmed that STT is more effective than a FS approach.3-5 In patients of higher acuity, STT has not proven to be superior.

A key lesson learned was the need for proper education of nursing staff. Concurrent nursing audits were necessary to ensure that scoring was performed in an accurate and timely manner. In addition, it was challenging to predict which patients might develop DTs versus those requiring a brief inpatient stay. While there was initial concern that an STT protocol could result in underdosing, we found that patients had fewer DT episodes and fewer ICU transfers.

 

 

This study had several limitations. These include a relatively small sample size and the data being less recent. As there has been no intervening change to the therapeutic paradigm of DT treatment, the findings remain pertinent to the present time. The study employed a simple pre/post design and was conducted in a single setting. We are not aware of any temporal or local trends likely to influence these results. Admissions and transfers to the SDU for severe alcohol withdrawal were based on physician discretion. However, patient characteristics in both groups were similar (Table 1). We note that the postintervention STT protocol allowed for more frequent benzodiazepine dosing, though benzodiazepine use did decrease. Different alcohol withdrawal scores (MINDS vs. CIWA-Ar) were used for postintervention and preintervention, although previous research has shown that MINDS and CIWA-Ar scores correlate well.7 Finally, some patients of higher acuity and complexity were excluded, potentially limiting the generalizability of our results.

Conclusion

Our STT protocol proved to be more effective and safer in treating severe alcohol withdrawal patients than usual care employing STT with FS. We believe the successful implementation of a STT protocol in high-acuity patients also requires frequent monitoring using the MINDS scale, integrated with benzodiazepine sliding-scale dosing to match symptom severity. This bundled approach resulted in a significant reduction of benzodiazepine usage and reduced length of stay. Timely treatment of these patients also reduced the percent of patients developing DTs, and reduced intubation rates and transfers to the ICU. Further studies may be warranted at other sites to confirm the effectiveness of this STT protocol.

Corresponding author: Paul W. Huang, MD, Stamford Hospital, One Hospital Plaza, PO Box 9317, Stamford, CT 06904; phuang@stamhealth.org.

Financial disclosures: None.

References

1. DeCarolis DD, Rice KL, Ho L, et al. Symptom-driven lorazepam protocol for treatment of severe alcohol withdrawal delirium in the intensive care unit. Pharmacotherapy. 2007;27(4):510-518.

2. DeBellis R, Smith BS, Choi S, Malloy M. Management of delirium tremens. J Intensive Care Med. 2005;20(3):164-173.

3. Saitz R, Mayo-Smith MF, Roberts MS, et al. Individualized treatment for alcohol withdrawal. A randomized double-blind controlled trial. JAMA. 1994;272(7):519-523.

4. Sachdeva A, Chandra M, Deshpande SN. A comparative study of fixed tapering dose regimen versus symptom-triggered regimen of lorazepam for alcohol detoxification. Alcohol Alcohol. 2014;49(3):287-291.

5. Daeppen JB, Gache P, Landry U, et al. Symptom-triggered vs fixed-schedule doses of benzodiazepine for alcohol withdrawal: a randomized treatment trial. Arch Intern Med. 2002;162(10):1117-1121.

6. Jaeger TM, Lohr RH, Pankratz VS. Symptom-triggered therapy for alcohol withdrawal syndrome in medical inpatients. Mayo Clin Proc. 2001;76(7):695-701.

7. Littlefield AJ, Heavner MS, Eng CC, et al. Correlation Between mMINDS and CIWA-Ar Scoring Tools in Patients With Alcohol Withdrawal Syndrome. Am J Crit Care. 2018;27(4):280-286.

References

1. DeCarolis DD, Rice KL, Ho L, et al. Symptom-driven lorazepam protocol for treatment of severe alcohol withdrawal delirium in the intensive care unit. Pharmacotherapy. 2007;27(4):510-518.

2. DeBellis R, Smith BS, Choi S, Malloy M. Management of delirium tremens. J Intensive Care Med. 2005;20(3):164-173.

3. Saitz R, Mayo-Smith MF, Roberts MS, et al. Individualized treatment for alcohol withdrawal. A randomized double-blind controlled trial. JAMA. 1994;272(7):519-523.

4. Sachdeva A, Chandra M, Deshpande SN. A comparative study of fixed tapering dose regimen versus symptom-triggered regimen of lorazepam for alcohol detoxification. Alcohol Alcohol. 2014;49(3):287-291.

5. Daeppen JB, Gache P, Landry U, et al. Symptom-triggered vs fixed-schedule doses of benzodiazepine for alcohol withdrawal: a randomized treatment trial. Arch Intern Med. 2002;162(10):1117-1121.

6. Jaeger TM, Lohr RH, Pankratz VS. Symptom-triggered therapy for alcohol withdrawal syndrome in medical inpatients. Mayo Clin Proc. 2001;76(7):695-701.

7. Littlefield AJ, Heavner MS, Eng CC, et al. Correlation Between mMINDS and CIWA-Ar Scoring Tools in Patients With Alcohol Withdrawal Syndrome. Am J Crit Care. 2018;27(4):280-286.

Issue
Journal of Clinical Outcomes Management - 28(3)
Issue
Journal of Clinical Outcomes Management - 28(3)
Page Number
134-138
Page Number
134-138
Publications
Publications
Topics
Article Type
Display Headline
Implementation of a Symptom–Triggered Protocol for Severe Alcohol Withdrawal Treatment in a Medical Step-down Unit
Display Headline
Implementation of a Symptom–Triggered Protocol for Severe Alcohol Withdrawal Treatment in a Medical Step-down Unit
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

Lenvatinib Plus Pembrolizumab Improves Outcomes in Previously Untreated Advanced Clear Cell Renal Cell Carcinoma

Article Type
Changed
Fri, 03/26/2021 - 01:15
Display Headline
Lenvatinib Plus Pembrolizumab Improves Outcomes in Previously Untreated Advanced Clear Cell Renal Cell Carcinoma

Study Overview

Objective. To evaluate the efficacy and safety of lenvatinib in combination with everolimus or pembrolizumab compared with sunitinib alone for the treatment of newly diagnosed advanced clear cell renal cell carcinoma (ccRCC).

Design. Global, multicenter, randomized, open-label, phase 3 trial.

Intervention. Patients were randomized in a 1:1:1 ratio to receive treatment with 1 of 3 regimens: lenvatinib 20 mg daily plus pembrolizumab 200 mg on day 1 of each 21-day cycle; lenvatinib 18 mg daily plus everolimus 5 mg once daily for each 21-day cycle; or sunitinib 50 mg daily for 4 weeks followed by 2 weeks off. Patients were stratified according to geographic region and Memorial Sloan Kettering Cancer Center (MSKCC) prognostic risk group.

Setting and participants. A total of 1417 patients were screened, and 1069 patients underwent randomization between October 2016 and July 2019: 355 patients were randomized to the lenvatinib plus pembrolizumab group, 357 were randomized to the lenvatinib plus everolimus group, and 357 were randomized to the sunitinib alone group. The patients must have had a diagnosis of previously untreated advanced renal cell carcinoma with a clear-cell component. All the patients need to have a Karnofsky performance status of at least 70, adequate renal function, and controlled blood pressure with or without antihypertensive medications.

Main outcome measures. The primary endpoint assessed the progression-free survival (PFS) as evaluated by independent review committee using RECIST, version 1.1. Imaging was performed at the time of screening and every 8 weeks thereafter. Secondary endpoints were safety, overall survival (OS), and objective response rate as well as investigator-assessed PFS. Also, they assessed the duration of response. During the treatment period, the safety and adverse events were assessed up to 30 days from the last dose of the trial drug.

Main results. The baseline characteristics were well balanced between the treatment groups. More than 70% of enrolled participants were male. Approximately 60% of participants were MSKCC intermediate risk, 27% were favorable risk, and 9% were poor risk. Patients with a PD-L1 combined positive score of 1% or more represented 30% of the population. The remainder had a PD-L1 combined positive score of <1% (30%) or such data were not available (38%). Liver metastases were present in 17% of patients at baseline in each group, and 70% of patients had a prior nephrectomy. The data cutoff occurred in August 2020 for PFS and the median follow-up for OS was 26.6 months. Around 40% of the participants in the lenvatinib plus pembrolizumab group, 18.8% in the sunitinib group, and 31% in the lenvatinib plus everolimus group were still receiving trial treatment at data cutoff. The leading cause for discontinuing therapy was disease progression. Approximately 50% of patients in the lenvatinib/everolimus group and sunitinib group received subsequent checkpoint inhibitor therapy after progression.

The median PFS in the lenvatinib plus pembrolizumab group was significantly longer than in the sunitinib group, 23.9 months vs 9.2 months (hazard ratio [HR], 0.39; 95% CI, 0.32-0.49; P < 0.001). The median PFS was also significantly longer in the lenvatinib plus everolimus group compared with sunitinib, 14.7 vs 9.2 months (HR 0.65; 95% CI 0.53-0.80; P < 0.001). The PFS benefit favored the lenvatinib combination groups over sunitinib in all subgroups, including the MSKCC prognostic risk groups. The median OS was not reached with any treatment, with 79% of patients in the lenvatinib plus pembrolizumab group, 66% of patients in the lenvatinib plus everolimus group, and 70% in the sunitinib group still alive at 24 months. Survival was significantly longer in the lenvatinib plus pembrolizumab group compared with sunitinib (HR, 0.66; 95% CI, 0.49-0.88; P = 0.005). The OS favored lenvatinib/pembrolizumab over sunitinib in the PD-L1 positive or negative groups. The median duration of response in the lenvatinib plus pembrolizumab group was 25.8 months compared to 16.6 months and 14.6 months in the lenvatinib plus everolimus and sunitinib groups, respectively. Complete response rates were higher in the lenvatinib plus pembrolizumab group (16%) compared with lenvatinib/everolimus (9.8%) or sunitinib (4.2%). The median time to response was around 1.9 months in all 3 groups.

The most frequent adverse events seen in all groups were diarrhea, hypertension, fatigue, and nausea. Hypothyroidism was seen more frequently in the lenvatinib plus pembrolizumab group (47%). Grade 3 adverse events were seen in approximately 80% of patients in all groups. The most common grade 3 or higher adverse event was hypertension in all 3 groups. The median time for discontinuing treatment due to side effects was 8.97 months in the lenvatinib plus pembrolizumab arm, 5.49 months in the lenvatinib plus everolimus group, and 4.57 months in the sunitinib group. In the lenvatinib plus pembrolizumab group, 15 patients had grade 5 adverse events; 11 participants had fatal events not related to disease progression. In the lenvatinib plus everolimus group, there were 22 patients with grade 5 events, with 10 fatal events not related to disease progression. In the sunitinib group, 11 patients had grade 5 events, and only 2 fatal events were not linked to disease progression.

Conclusion. The combination of lenvatinib plus pembrolizumab significantly prolongs PFS and OS compared with sunitinib in patients with previously untreated and advanced ccRCC. The median OS has not yet been reached.

 

 

Commentary

The results of the current phase 3 CLEAR trial highlight the efficacy and safety of lenvatinib plus pembrolizumab as a first-line treatment in advanced ccRCC. This trial adds to the rapidly growing body of literature supporting the notion that the combination of anti-PD-1 based therapy with either CTLA-4 antibodies or VEGF receptor tyrosine kinase inhibitors (TKI) improves outcomes in previously untreated patients with advanced ccRCC. Previously presented data from Keynote-426 (pembrolizumab plus axitinib), Checkmate-214 (nivolumab plus ipilimumab), and Javelin Renal 101 (Avelumab plus axitinib) have also shown improved outcomes with combination therapy in the frontline setting.1-4 While the landscape of therapeutic options in the frontline setting continues to grow, there remains lack of clarity as to how to tailor our therapeutic decisions for specific patient populations. The exception would be nivolumab and ipilimumab, which are currently indicated for IMDC intermediate- or poor-risk patients.

The combination of VEGFR TKI therapy and PD-1 antibodies provides rapid disease control, with a median time to response in the current study of 1.9 months, and, generally speaking, a low risk of progression in the first 6 months of therapy. While cross-trial comparisons are always problematic, the PFS reported in this study and others with VEGFR TKI and PD-1 antibody combinations is quite impressive and surpasses that noted in Checkmate 214.3 While the median OS survival has not yet been reached, the long duration of PFS and complete response rate of 16% in this study certainly make this an attractive frontline option for newly diagnosed patients with advanced ccRCC. Longer follow-up is needed to confirm the survival benefit noted.

Applications for Clinical Practice

The current data support the use VEGFR TKI and anti-PD1 therapy in the frontline setting. How to choose between such combination regimens or combination immunotherapy remains unclear, and further biomarker-based assessments are needed to help guide therapeutic decisions for our patients.

References

1. Motzer, R, Alekseev B, Rha SY, et al. Lenvatinib plus pembrolizumab or everolimus for advanced renal cell carcinoma [published online ahead of print, 2021 Feb 13]. N Engl J Med. 2021;10.1056/NEJMoa2035716. doi:10.1056/NEJMoa2035716

2. Rini, BI, Plimack ER, Stus V, et al. Pembrolizumab plus axitinib versus sunitinib for advanced renal-cell carcinoma. N Engl J Med. 2019;380(12):1116-1127.

3. Motzer, RJ, Tannir NM, McDermott DF, et al. Nivolumab plus ipilimumab versus sunitinib in advanced renal-cell carcinoma. N Engl J Med. 2018;378(14):1277-1290.

4. Motzer, RJ, Penkov K, Haanen J, et al. Avelumab plus axitinib versus sunitinib for advanced renal-cell carcinoma. N Engl J Med. 2019;380(12):1103-1115.

Article PDF
Issue
Journal of Clinical Outcomes Management - 28(2)
Publications
Topics
Page Number
54-56
Sections
Article PDF
Article PDF

Study Overview

Objective. To evaluate the efficacy and safety of lenvatinib in combination with everolimus or pembrolizumab compared with sunitinib alone for the treatment of newly diagnosed advanced clear cell renal cell carcinoma (ccRCC).

Design. Global, multicenter, randomized, open-label, phase 3 trial.

Intervention. Patients were randomized in a 1:1:1 ratio to receive treatment with 1 of 3 regimens: lenvatinib 20 mg daily plus pembrolizumab 200 mg on day 1 of each 21-day cycle; lenvatinib 18 mg daily plus everolimus 5 mg once daily for each 21-day cycle; or sunitinib 50 mg daily for 4 weeks followed by 2 weeks off. Patients were stratified according to geographic region and Memorial Sloan Kettering Cancer Center (MSKCC) prognostic risk group.

Setting and participants. A total of 1417 patients were screened, and 1069 patients underwent randomization between October 2016 and July 2019: 355 patients were randomized to the lenvatinib plus pembrolizumab group, 357 were randomized to the lenvatinib plus everolimus group, and 357 were randomized to the sunitinib alone group. The patients must have had a diagnosis of previously untreated advanced renal cell carcinoma with a clear-cell component. All the patients need to have a Karnofsky performance status of at least 70, adequate renal function, and controlled blood pressure with or without antihypertensive medications.

Main outcome measures. The primary endpoint assessed the progression-free survival (PFS) as evaluated by independent review committee using RECIST, version 1.1. Imaging was performed at the time of screening and every 8 weeks thereafter. Secondary endpoints were safety, overall survival (OS), and objective response rate as well as investigator-assessed PFS. Also, they assessed the duration of response. During the treatment period, the safety and adverse events were assessed up to 30 days from the last dose of the trial drug.

Main results. The baseline characteristics were well balanced between the treatment groups. More than 70% of enrolled participants were male. Approximately 60% of participants were MSKCC intermediate risk, 27% were favorable risk, and 9% were poor risk. Patients with a PD-L1 combined positive score of 1% or more represented 30% of the population. The remainder had a PD-L1 combined positive score of <1% (30%) or such data were not available (38%). Liver metastases were present in 17% of patients at baseline in each group, and 70% of patients had a prior nephrectomy. The data cutoff occurred in August 2020 for PFS and the median follow-up for OS was 26.6 months. Around 40% of the participants in the lenvatinib plus pembrolizumab group, 18.8% in the sunitinib group, and 31% in the lenvatinib plus everolimus group were still receiving trial treatment at data cutoff. The leading cause for discontinuing therapy was disease progression. Approximately 50% of patients in the lenvatinib/everolimus group and sunitinib group received subsequent checkpoint inhibitor therapy after progression.

The median PFS in the lenvatinib plus pembrolizumab group was significantly longer than in the sunitinib group, 23.9 months vs 9.2 months (hazard ratio [HR], 0.39; 95% CI, 0.32-0.49; P < 0.001). The median PFS was also significantly longer in the lenvatinib plus everolimus group compared with sunitinib, 14.7 vs 9.2 months (HR 0.65; 95% CI 0.53-0.80; P < 0.001). The PFS benefit favored the lenvatinib combination groups over sunitinib in all subgroups, including the MSKCC prognostic risk groups. The median OS was not reached with any treatment, with 79% of patients in the lenvatinib plus pembrolizumab group, 66% of patients in the lenvatinib plus everolimus group, and 70% in the sunitinib group still alive at 24 months. Survival was significantly longer in the lenvatinib plus pembrolizumab group compared with sunitinib (HR, 0.66; 95% CI, 0.49-0.88; P = 0.005). The OS favored lenvatinib/pembrolizumab over sunitinib in the PD-L1 positive or negative groups. The median duration of response in the lenvatinib plus pembrolizumab group was 25.8 months compared to 16.6 months and 14.6 months in the lenvatinib plus everolimus and sunitinib groups, respectively. Complete response rates were higher in the lenvatinib plus pembrolizumab group (16%) compared with lenvatinib/everolimus (9.8%) or sunitinib (4.2%). The median time to response was around 1.9 months in all 3 groups.

The most frequent adverse events seen in all groups were diarrhea, hypertension, fatigue, and nausea. Hypothyroidism was seen more frequently in the lenvatinib plus pembrolizumab group (47%). Grade 3 adverse events were seen in approximately 80% of patients in all groups. The most common grade 3 or higher adverse event was hypertension in all 3 groups. The median time for discontinuing treatment due to side effects was 8.97 months in the lenvatinib plus pembrolizumab arm, 5.49 months in the lenvatinib plus everolimus group, and 4.57 months in the sunitinib group. In the lenvatinib plus pembrolizumab group, 15 patients had grade 5 adverse events; 11 participants had fatal events not related to disease progression. In the lenvatinib plus everolimus group, there were 22 patients with grade 5 events, with 10 fatal events not related to disease progression. In the sunitinib group, 11 patients had grade 5 events, and only 2 fatal events were not linked to disease progression.

Conclusion. The combination of lenvatinib plus pembrolizumab significantly prolongs PFS and OS compared with sunitinib in patients with previously untreated and advanced ccRCC. The median OS has not yet been reached.

 

 

Commentary

The results of the current phase 3 CLEAR trial highlight the efficacy and safety of lenvatinib plus pembrolizumab as a first-line treatment in advanced ccRCC. This trial adds to the rapidly growing body of literature supporting the notion that the combination of anti-PD-1 based therapy with either CTLA-4 antibodies or VEGF receptor tyrosine kinase inhibitors (TKI) improves outcomes in previously untreated patients with advanced ccRCC. Previously presented data from Keynote-426 (pembrolizumab plus axitinib), Checkmate-214 (nivolumab plus ipilimumab), and Javelin Renal 101 (Avelumab plus axitinib) have also shown improved outcomes with combination therapy in the frontline setting.1-4 While the landscape of therapeutic options in the frontline setting continues to grow, there remains lack of clarity as to how to tailor our therapeutic decisions for specific patient populations. The exception would be nivolumab and ipilimumab, which are currently indicated for IMDC intermediate- or poor-risk patients.

The combination of VEGFR TKI therapy and PD-1 antibodies provides rapid disease control, with a median time to response in the current study of 1.9 months, and, generally speaking, a low risk of progression in the first 6 months of therapy. While cross-trial comparisons are always problematic, the PFS reported in this study and others with VEGFR TKI and PD-1 antibody combinations is quite impressive and surpasses that noted in Checkmate 214.3 While the median OS survival has not yet been reached, the long duration of PFS and complete response rate of 16% in this study certainly make this an attractive frontline option for newly diagnosed patients with advanced ccRCC. Longer follow-up is needed to confirm the survival benefit noted.

Applications for Clinical Practice

The current data support the use VEGFR TKI and anti-PD1 therapy in the frontline setting. How to choose between such combination regimens or combination immunotherapy remains unclear, and further biomarker-based assessments are needed to help guide therapeutic decisions for our patients.

Study Overview

Objective. To evaluate the efficacy and safety of lenvatinib in combination with everolimus or pembrolizumab compared with sunitinib alone for the treatment of newly diagnosed advanced clear cell renal cell carcinoma (ccRCC).

Design. Global, multicenter, randomized, open-label, phase 3 trial.

Intervention. Patients were randomized in a 1:1:1 ratio to receive treatment with 1 of 3 regimens: lenvatinib 20 mg daily plus pembrolizumab 200 mg on day 1 of each 21-day cycle; lenvatinib 18 mg daily plus everolimus 5 mg once daily for each 21-day cycle; or sunitinib 50 mg daily for 4 weeks followed by 2 weeks off. Patients were stratified according to geographic region and Memorial Sloan Kettering Cancer Center (MSKCC) prognostic risk group.

Setting and participants. A total of 1417 patients were screened, and 1069 patients underwent randomization between October 2016 and July 2019: 355 patients were randomized to the lenvatinib plus pembrolizumab group, 357 were randomized to the lenvatinib plus everolimus group, and 357 were randomized to the sunitinib alone group. The patients must have had a diagnosis of previously untreated advanced renal cell carcinoma with a clear-cell component. All the patients need to have a Karnofsky performance status of at least 70, adequate renal function, and controlled blood pressure with or without antihypertensive medications.

Main outcome measures. The primary endpoint assessed the progression-free survival (PFS) as evaluated by independent review committee using RECIST, version 1.1. Imaging was performed at the time of screening and every 8 weeks thereafter. Secondary endpoints were safety, overall survival (OS), and objective response rate as well as investigator-assessed PFS. Also, they assessed the duration of response. During the treatment period, the safety and adverse events were assessed up to 30 days from the last dose of the trial drug.

Main results. The baseline characteristics were well balanced between the treatment groups. More than 70% of enrolled participants were male. Approximately 60% of participants were MSKCC intermediate risk, 27% were favorable risk, and 9% were poor risk. Patients with a PD-L1 combined positive score of 1% or more represented 30% of the population. The remainder had a PD-L1 combined positive score of <1% (30%) or such data were not available (38%). Liver metastases were present in 17% of patients at baseline in each group, and 70% of patients had a prior nephrectomy. The data cutoff occurred in August 2020 for PFS and the median follow-up for OS was 26.6 months. Around 40% of the participants in the lenvatinib plus pembrolizumab group, 18.8% in the sunitinib group, and 31% in the lenvatinib plus everolimus group were still receiving trial treatment at data cutoff. The leading cause for discontinuing therapy was disease progression. Approximately 50% of patients in the lenvatinib/everolimus group and sunitinib group received subsequent checkpoint inhibitor therapy after progression.

The median PFS in the lenvatinib plus pembrolizumab group was significantly longer than in the sunitinib group, 23.9 months vs 9.2 months (hazard ratio [HR], 0.39; 95% CI, 0.32-0.49; P < 0.001). The median PFS was also significantly longer in the lenvatinib plus everolimus group compared with sunitinib, 14.7 vs 9.2 months (HR 0.65; 95% CI 0.53-0.80; P < 0.001). The PFS benefit favored the lenvatinib combination groups over sunitinib in all subgroups, including the MSKCC prognostic risk groups. The median OS was not reached with any treatment, with 79% of patients in the lenvatinib plus pembrolizumab group, 66% of patients in the lenvatinib plus everolimus group, and 70% in the sunitinib group still alive at 24 months. Survival was significantly longer in the lenvatinib plus pembrolizumab group compared with sunitinib (HR, 0.66; 95% CI, 0.49-0.88; P = 0.005). The OS favored lenvatinib/pembrolizumab over sunitinib in the PD-L1 positive or negative groups. The median duration of response in the lenvatinib plus pembrolizumab group was 25.8 months compared to 16.6 months and 14.6 months in the lenvatinib plus everolimus and sunitinib groups, respectively. Complete response rates were higher in the lenvatinib plus pembrolizumab group (16%) compared with lenvatinib/everolimus (9.8%) or sunitinib (4.2%). The median time to response was around 1.9 months in all 3 groups.

The most frequent adverse events seen in all groups were diarrhea, hypertension, fatigue, and nausea. Hypothyroidism was seen more frequently in the lenvatinib plus pembrolizumab group (47%). Grade 3 adverse events were seen in approximately 80% of patients in all groups. The most common grade 3 or higher adverse event was hypertension in all 3 groups. The median time for discontinuing treatment due to side effects was 8.97 months in the lenvatinib plus pembrolizumab arm, 5.49 months in the lenvatinib plus everolimus group, and 4.57 months in the sunitinib group. In the lenvatinib plus pembrolizumab group, 15 patients had grade 5 adverse events; 11 participants had fatal events not related to disease progression. In the lenvatinib plus everolimus group, there were 22 patients with grade 5 events, with 10 fatal events not related to disease progression. In the sunitinib group, 11 patients had grade 5 events, and only 2 fatal events were not linked to disease progression.

Conclusion. The combination of lenvatinib plus pembrolizumab significantly prolongs PFS and OS compared with sunitinib in patients with previously untreated and advanced ccRCC. The median OS has not yet been reached.

 

 

Commentary

The results of the current phase 3 CLEAR trial highlight the efficacy and safety of lenvatinib plus pembrolizumab as a first-line treatment in advanced ccRCC. This trial adds to the rapidly growing body of literature supporting the notion that the combination of anti-PD-1 based therapy with either CTLA-4 antibodies or VEGF receptor tyrosine kinase inhibitors (TKI) improves outcomes in previously untreated patients with advanced ccRCC. Previously presented data from Keynote-426 (pembrolizumab plus axitinib), Checkmate-214 (nivolumab plus ipilimumab), and Javelin Renal 101 (Avelumab plus axitinib) have also shown improved outcomes with combination therapy in the frontline setting.1-4 While the landscape of therapeutic options in the frontline setting continues to grow, there remains lack of clarity as to how to tailor our therapeutic decisions for specific patient populations. The exception would be nivolumab and ipilimumab, which are currently indicated for IMDC intermediate- or poor-risk patients.

The combination of VEGFR TKI therapy and PD-1 antibodies provides rapid disease control, with a median time to response in the current study of 1.9 months, and, generally speaking, a low risk of progression in the first 6 months of therapy. While cross-trial comparisons are always problematic, the PFS reported in this study and others with VEGFR TKI and PD-1 antibody combinations is quite impressive and surpasses that noted in Checkmate 214.3 While the median OS survival has not yet been reached, the long duration of PFS and complete response rate of 16% in this study certainly make this an attractive frontline option for newly diagnosed patients with advanced ccRCC. Longer follow-up is needed to confirm the survival benefit noted.

Applications for Clinical Practice

The current data support the use VEGFR TKI and anti-PD1 therapy in the frontline setting. How to choose between such combination regimens or combination immunotherapy remains unclear, and further biomarker-based assessments are needed to help guide therapeutic decisions for our patients.

References

1. Motzer, R, Alekseev B, Rha SY, et al. Lenvatinib plus pembrolizumab or everolimus for advanced renal cell carcinoma [published online ahead of print, 2021 Feb 13]. N Engl J Med. 2021;10.1056/NEJMoa2035716. doi:10.1056/NEJMoa2035716

2. Rini, BI, Plimack ER, Stus V, et al. Pembrolizumab plus axitinib versus sunitinib for advanced renal-cell carcinoma. N Engl J Med. 2019;380(12):1116-1127.

3. Motzer, RJ, Tannir NM, McDermott DF, et al. Nivolumab plus ipilimumab versus sunitinib in advanced renal-cell carcinoma. N Engl J Med. 2018;378(14):1277-1290.

4. Motzer, RJ, Penkov K, Haanen J, et al. Avelumab plus axitinib versus sunitinib for advanced renal-cell carcinoma. N Engl J Med. 2019;380(12):1103-1115.

References

1. Motzer, R, Alekseev B, Rha SY, et al. Lenvatinib plus pembrolizumab or everolimus for advanced renal cell carcinoma [published online ahead of print, 2021 Feb 13]. N Engl J Med. 2021;10.1056/NEJMoa2035716. doi:10.1056/NEJMoa2035716

2. Rini, BI, Plimack ER, Stus V, et al. Pembrolizumab plus axitinib versus sunitinib for advanced renal-cell carcinoma. N Engl J Med. 2019;380(12):1116-1127.

3. Motzer, RJ, Tannir NM, McDermott DF, et al. Nivolumab plus ipilimumab versus sunitinib in advanced renal-cell carcinoma. N Engl J Med. 2018;378(14):1277-1290.

4. Motzer, RJ, Penkov K, Haanen J, et al. Avelumab plus axitinib versus sunitinib for advanced renal-cell carcinoma. N Engl J Med. 2019;380(12):1103-1115.

Issue
Journal of Clinical Outcomes Management - 28(2)
Issue
Journal of Clinical Outcomes Management - 28(2)
Page Number
54-56
Page Number
54-56
Publications
Publications
Topics
Article Type
Display Headline
Lenvatinib Plus Pembrolizumab Improves Outcomes in Previously Untreated Advanced Clear Cell Renal Cell Carcinoma
Display Headline
Lenvatinib Plus Pembrolizumab Improves Outcomes in Previously Untreated Advanced Clear Cell Renal Cell Carcinoma
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Article PDF Media

COVID-19 Monoclonal Antibody Infusions: A Multidisciplinary Initiative to Operationalize EUA Novel Treatment Options

Article Type
Changed
Thu, 08/26/2021 - 15:49
Display Headline
COVID-19 Monoclonal Antibody Infusions: A Multidisciplinary Initiative to Operationalize EUA Novel Treatment Options

From Mount Sinai Medical Center, Miami Beach, FL.

Abstract

Objective: To develop and implement a process for administering COVID-19 monoclonal antibody infusions for outpatients with mild or moderate COVID-19 at high risk for hospitalization, using multidisciplinary collaboration, US Food and Drug Administration (FDA) guidance, and infection prevention standards.

Methods: When monoclonal antibody therapy became available for mild or moderate COVID-19 outpatients via Emergency Use Authorization (EUA), our institution sought to provide this therapy option to our patients. We describe the process for planning, implementing, and maintaining a successful program for administering novel therapies based on FDA guidance and infection prevention standards. Key components of our implementation process were multidisciplinary planning involving decision makers and stakeholders; setting realistic goals in the process; team communication; and measuring and reporting quality improvement on a regular basis.

Results: A total of 790 COVID-19 monoclonal antibody infusions were administered from November 20, 2020 to March 5, 2021. Steps to minimize the likelihood of adverse drug reactions were implemented and a low incidence (< 1%) has occurred. There has been no concern from staff regarding infection during the process. Rarely, patients have raised cost-related concerns, typically due to incomplete communication regarding billing prior to the infusion. Patients, families, nursing staff, physicians, pharmacy, and hospital administration have expressed satisfaction with the program.

Conclusion: This process can provide a template for other hospitals or health care delivery facilities to provide novel therapies to patients with mild or moderate COVID-19 in a safe and effective manner.

Keywords: COVID-19; monoclonal antibody; infusion; emergency use authorization.

SARS-CoV-2 and the disease it causes, COVID-19, have transformed from scientific vernacular to common household terms. It began with a cluster of pneumonia cases of unknown etiology in December 2019 in Wuhan, China, with physicians there reporting a novel coronavirus strain (2019-nCoV), now referred to as SARS-CoV-2. Rapid spread of this virus resulted in the World Health Organization (WHO) declaring an international public health emergency. Since this time, the virus has evolved into a worldwide pandemic. COVID-19 has dramatically impacted our society, resulting in more than 2.63 million global deaths as of this writing, of which more than 527,000 deaths have occurred in the United States.1 This novel virus has resulted in a flurry of literature, research, therapies, and collaboration across multiple disciplines in an effort to prevent, treat, and mitigate cases and complications of this disease.

 

 

On November 9, 2020, and November 21, 2020, the US Food and Drug Administration (FDA) issued Emergency Use Authorizations (EUA) for 2 novel COVID-19 monoclonal therapies, bamlanivimab2-3 and casirivimab/imdevimab,3-4 respectively. The EUAs granted permission for these therapies to be administered for the treatment of mild to moderate COVID-19 in adult and pediatric patients (≥ 12 years and weighing at least 40 kg) with positive results of direct SARS-CoV-2 viral testing and who are at high risk for progressing to severe COVID-19 and/or hospitalization. The therapies work by targeting the SARS-CoV-2 spike protein and subsequent attachment to human angiotensin-converting enzyme 2 receptors. Clinical trial data leading to the EUA demonstrated a reduction in viral load, safe outcome, and most importantly, fewer hospitalization and emergency room visits, as compared to the placebo group.5-7 The use of monoclonal antibodies is not new and gained recognition during the Ebola crisis, when the monoclonal antibody to the Ebola virus showed a significant survival benefit.8 Providing monoclonal antibody therapy soon after symptom onset aligns with a shift from the onset of the pandemic to the current focus on the administration of pharmaceutical therapy early in the disease course. This shift prevents progression to severe COVID-19, with the goal of reducing patient mortality, hospitalizations, and strain on health care systems.

The availability of novel neutralizing monoclonal antibodies for COVID-19 led to discussions of how to incorporate these therapies as new options for patients. Our institution networked with colleagues from multiple disciplines to discuss processes and policies for the safe administration of the monoclonal antibody infusion therapies. Federal health leaders urge more use of monoclonal antibodies, but many hospitals have been unable to successfully implement infusions due to staff and logistical challenges.9 This article presents a viable process that hospitals can use to provide these novel therapies to outpatients with mild to moderate COVID-19.

The Mount Sinai Medical Center, Florida Experience

Mount Sinai Medical Center in Miami Beach, Florida, is the largest private, independent, not-for-profit teaching hospital in South Florida, comprising 672 licensed beds and supporting 150,000 emergency department (ED) visits annually. Per the EUA criteria for use, COVID-19 monoclonal antibody therapies are not authorized for patients who are hospitalized or who require oxygen therapy due to COVID-19. Therefore, options for outpatient administration needed to be evaluated. Directly following the first EUA press release, a task force of key stakeholders was assembled to brainstorm and develop a process to offer this therapy to the community. A multidisciplinary task force with representation from the ED, nursing, primary care, hospital medicine, pharmacy, risk management, billing, information technology, infection prevention, and senior level leadership participated (Table).

List of Key Stakeholders and Responsibilities

The task force reviewed institutional outpatient locations to determine whether offering this service would be feasible (eg, ED, ambulatory care facilities, cancer center). The ED was selected because it would offer the largest array of appointment times to meet the community needs with around-the-clock availability. While Mount Sinai Medical Center offers care in 3 emergency center locations in Aventura, Hialeah, and Miami Beach, it was determined to initiate the infusions at the main campus center in Miami Beach only. The main campus affords an onsite pharmacy with suitable staffing to prepare the anticipated volume of infusions in a timely manner, as both therapies have short stabilities following preparation. Thus, it was decided that patients from freestanding emergency centers in Aventura and Hialeah would be moved to the Miami Beach ED location to receive therapy. Operating at a single site also allowed for more rapid implementation, monitoring, and ability to make modifications more easily. Discussions for the possible expansion of COVID-19 monoclonal antibody infusions at satellite locations are underway.

Process implementation timeline

On November 20, 2020, 11 days after the formation of the multidisciplinary task force, the first COVID-19 monoclonal infusion was successfully administered. Figure 1 depicts the timeline from assessment to program implementation. Critical to implementation was the involvement of decision makers from all necessary departments early in the planning process to ensure that standard operating procedures were followed and that the patients, community, and organization had a positive experience. This allowed for simultaneous planning of electronic health record (Epic; EHR) builds, departmental workflows, and staff education, as described in the following section. Figure 2 shows the patient safety activities included in the implementation process.

Important patient safety initiatives

 

 

Key Stakeholder Involvement and Workflow

On the day of bamlanivimab EUA release, email communication was shared among hospital leadership with details of the press release. Departments were quickly involved to initiate a task force to assess if and how this therapy could be offered at Mount Sinai Medical Center. The following sections explain the role of each stakeholder and their essential role to operationalize these novel EUA treatment options. The task force was organized and led by our chief medical officer and chief nursing officer.

Information Technology

Medication Ordering and Documentation EHR and Smart Pumps. Early in the pandemic, the antimicrobial stewardship (ASP) clinical coordinator became the designated point person for pharmacy assessment of novel COVID-19 therapies. As such, this pharmacist began reviewing the bamlanivimab and, later, the casirivimab/imdevimab EUA Fact Sheet for Health Care Providers. All necessary elements for the complete and safe ordering and dispensing of the medication were developed and reviewed by pharmacy administration and ED nursing leadership for input, prior to submitting to the information technology team for implementation. Building the COVID-19 monoclonal medication records into the EHR allowed for detailed direction (ie, administration and preparation instructions) to be consistently applied. The medication records were also built into hospital smart pumps so that nurses could access prepopulated, accurate volumes and infusion rates to minimize errors.

Order Set Development. The pharmacy medication build was added to a comprehensive order set (Figure 3), which was then developed to guide prescribers and standardize the process around ordering of COVID-19 monoclonal therapies. While these therapies are new, oncology monoclonal therapies are regularly administered to outpatients at Mount Sinai Cancer Center. The cancer center was therefore consulted on their process surrounding best practices in administration of monoclonal antibody therapies. This included protocols for medications used in pretreatment and management of hypersensitivity reactions and potential adverse drug reactions of both COVID-19 monoclonal therapies. These medication orders were selected by default in the order set to ensure that all patients received premedications aimed at minimizing the risk of hypersensitivity reaction, and had as-needed medication orders, in the event a hypersensitivity reaction occurred. Reducing hypersensitivity reaction risk is important as well to increase the likelihood that the patient would receive full therapy, as management of this adverse drug reactions involves possible cessation of therapy depending on the level of severity. The pharmacy department also ensured these medications were stocked in ED automated dispensing cabinets to promote quick access. In addition to the aforementioned nursing orders, we added EUA criteria for use and hyperlinks to the Fact Sheets for Patients and Caregivers and Health Care Providers for each monoclonal therapy, and restricted ordering to ED physicians, nurse practitioners, and physician assistants.

COVID-19 monoclonal antibody order set

The order set underwent multidisciplinary review by pharmacy administration, the chair of emergency medicine, physicians, and ED nursing leadership prior to presentation and approval by the Pharmacy and Therapeutics Committee. Lastly, at time of implementation, the order set was added to the ED preference list, preventing inpatient access. Additionally, as a patient safety action, free- standing orders of COVID-19 monoclonal therapies were disabled, so providers could only order therapies via the approved, comprehensive order set.

Preliminary Assessment Tool. A provider assessment tool was developed to document patient-specific EUA criteria for use during initial assessment (Figure 4). This tool serves as a checklist and is visible to the full multidisciplinary team in the patient’s EHR. It is used as a resource at the time of pharmacist verification and ED physician assessment to ensure criteria for use are met.

Workflow for COVID-19 monoclonal antibody infusion

 

 

Outpatient Offices

Patient Referral. Patients with symptoms or concerns of COVID-19 exposure can make physician appointments via telemedicine or in person at Mount Sinai Medical Center’s primary care and specialty offices. At the time of patient encounter, physicians suspecting a COVID-19 diagnosis will refer patients for outpatient COVID-19 polymerase chain reaction (PCR) laboratory testing, which has an approximate 24-hour turnaround to results. Physicians also assess whether the patient meets EUA criteria for use, pending results of testing. In the event a patient meets EUA criteria for use, the physician provides patient counseling and requests verbal consent. Following this, the physician enters a note in the EHR describing the patient’s condition, criteria for use evaluation, and the patient’s verbal agreement to therapy. This preliminary screening is beneficial to begin planning with both the patient and ED to minimize delays. Patients are notified of the results of their test once available. If the COVID-19 PCR test returns positive, the physician will call the ED at the main campus and schedule the patient for COVID-19 monoclonal therapy. As the desired timeframe for administering COVID-19 monoclonal therapies is within less than 10 days of symptom onset, timely scheduling of appointments is crucial. Infusion appointments are typically provided the same or next day. The patients are informed that they must bring documentation of their positive COVID-19 PCR test to their ED visit. Lastly, because patients are pretreated with medication that may potentially impair driving, they are instructed that they cannot drive themselves home; ride shares also are not allowed in order to limit the spread of infection.

Emergency Department

Patient Arrival and Screening. A COVID-19 patient can be evaluated in the ED 1 of 2 ways. The first option is via outpatient office referral, as described previously. Upon arrival to the ED, a second screening is performed to ensure the patient still meets EUA criteria for use and the positive COVID-19 PCR test result is confirmed. If the patient no longer meets criteria, the patient is triaged accordingly, including evaluation for higher-level care (eg, supplemental oxygen, hospital admission). The second optoion is via new patient walk-ins without outpatient physician referral (Figure 4). In these cases, an initial screening is performed, documenting EUA criteria for use in the preliminary assessment (Figure 5). Physicians will consider an outside COVID-19 test as valid, so long as documentation is readily available confirming a positive PCR result. Otherwise, an in-house COVID-19 PCR test will be performed, which has a 2-hour turnaround time.

Electronic health record preliminary assessment

Infusion Schedule. The ED offers a total of 16 COVID-19 monoclonal infusions slots daily. These are broken up into 4 infusion time blocks (eg, 8 am, 12 pm, 4 pm, 8 pm), with each infusion time block consisting of 4 available patient appointments. A list of scheduled infusions for the day is emailed to the pharmacy department every morning, and patients are instructed to arrive 1 hour prior to their appointment time. This allows time for patient registration, assessment, and pharmacy notification in advance of order entry. For logistical purposes, and as a patient safety initiative to reduce the likelihood of medication errors, each of the available COVID-19 monoclonal antibodies is offered on a designated day. Bamlanivimab is offered on Tuesday, Thursday, Saturday, and Sunday, while casirivimab/imdevimab is offered Monday, Wednesday, and Friday. This provides flexibility to adjust should supply deviate based on Department of Health allocation or should new therapy options within this class of medication become available.

Patient Education. Prior to administration of the monoclonal therapy, physician and nursing staff obtain a formal, written patient consent for therapy and provide patients with the option of participating in the institutional review board (IRB) approved study. Details of this are discussed in the risk management and IRB sections of the article. Nursing staff also provides the medication-specific Fact Sheet for Patients and Caregivers in either Spanish or English, which is also included as a hyperlink on the COVID-19 Monoclonal Antibody Order Set for ease of access. Interpreter services are available for patients who speak other languages. An ED decentralized pharmacist is also available onsite Monday through Friday from 12 pm to 8:30 pm to supplement education and serve as a resource for any questions.

Infusion Ordering. Once the patient is ready to begin therapy, the he/she is brought to a dedicated overflow area of the ED. There are few, if any, patients in this location, and it is adjacent to the main emergency center for easy access by the patients, nurses, pharmacists, and physicians. The physician then enters orders in the EHR using the COVID-19 Monoclonal Antibody Order Set (Figure 3). Three discrete questions were built into the medication order: (1) Was patient consent obtained? (2) Was the Fact Sheet for Patient/Caregiver provided to the patient? (3) Is the patient COVID-19 PCR-positive? These questions were built as hard stops so that the medication orders cannot be placed without a response. This serves as another double-check to ensure processes are followed and helps facilitate timely verification by the pharmacist.

 

 

Medication Administration. One nurse is dedicated to administering the monoclonal therapies scheduled at 8 am and 12 pm and another at 4 pm and 8 pm. Each appointment block is 4 hours in duration to allow adequate time for patient registration, infusion, and postinfusion observation. The nurse administers the premedications and COVID-19 monoclonal therapy, and observes the patient for the required 1-hour postadministration observation period. Nursing orders detailing monitoring parameters for mild, moderate, and severe reactions, along with associated medication orders to administer in the event they occur, are detailed in the nursing orders of the COVID-19 Monoclonal Antibody Order Set (Figure 3). Prior to administration, the nurse scans each medication and the patient’s wrist identification band, and documents the time of administration within the EHR medication administration report.

Pharmacy Department

Medication Receipt Process. Inventory is currently allocated biweekly from the state department of health and will soon be transitioning to a direct order system. The pharmacy technician in charge of deliveries notifies the pharmacy Antimicrobial Stewardship Program (ASP) clinical coordinator upon receipt of the monoclonal therapies. Bamlanivimab is supplied as 1 vial per dose, whereas casirivimab/imdevimab is supplied as 4 vials or 8 vials per dose, depending how it is shipped. To reduce the likelihood of medication errors, the ASP clinical coordinator assembles each of the casirivimab/imdevimab vials into kits, where 1 kit equals 1 dose. Labels are then affixed to each kit indicating the medication name, number of vials which equal a full dose, and pharmacist signature. The kits are stored in a dedicated refrigerator, and inventory logs are affixed to the outside of the refrigerator and updated daily. This inventory is also communicated daily to ED physician, nursing, and pharmacy leadership, as well as the director of patient safety, who reports weekly usage to the state Department of Health and Human Services. These weekly reports are used to determine allocation amounts.

Medication Verification and Delivery. The Mount Sinai Medical Center pharmacist staffing model consists of centralized order entry and specialized, decentralized positions. All orders are verified by the ED pharmacist when scheduled (not a 24/7 service) and by the designated pharmacist for all other times. At the time of medication verification, the pharmacist documents patient-specific EUA criteria for use and confirms that consent was obtained and the Fact Sheet for Patients/Caregivers was provided. A pharmacist intervention was developed to assist with this documentation. Pharmacists input smart text “.COVIDmonoclonal” and a drop-down menu of EUA criteria for use appears. The pharmacist reviews the patient care notes and medication order question responses to ascertain this information, contacting the ED prescriber if further clarification is required. This verification serves as another check to ensure processes put in place are followed. Lastly, intravenous preparation and delivery are electronically recorded in the EHR, and the medications require nursing signature at the time of delivery to ensure a formal chain of custody.

Risk Management

At Mount Sinai Medical Center, all EUA and investigational therapies require patient consent. Consistent with this requirement, a COVID-19 monoclonal specific consent was developed by risk management. This is provided to every patient receiving a COVID-19 monoclonal infusion, in addition to the FDA EUA Fact Sheet for Patients and Caregivers, and documented as part of their EHR. The questions providers must answer are built into the order set to ensure this process is followed and these patient safety checks are incorporated into the workflow.

Billing and Finance Department

In alignment with Mount Sinai Medical Center’s mission to provide high-quality health care to its diverse community through teaching, research, charity care, and financial responsibility, it was determined that this therapy would be provided to all patients regardless of insurance type, including those who are uninsured. The billing and finance department was consulted prior to this service being offered, to provide patients with accurate and pertinent information. The billing and finance department provided guidance on how to document patient encounters at time of registration to facilitate appropriate billing. At this time, the medication is free of charge, but nonmedication-related ED fees apply. This is explained to patients so there is a clear understanding prior to booking their appointment.

 

 

Infection Prevention

As patients receiving COVID-19 monoclonal therapies can transmit the virus to others, measures to ensure protection for other patients and staff are vital. To minimize exposure, specific nursing and physician staff from the ED are assigned to the treatment of these patients, and patients receive infusions and postobservation monitoring in a designated wing of the ED. Additionally, all staff who interact with these patients are required to don full personal protective equipment. This includes not only physicians and nurses but all specialties such as physician assistants, nurse practitioners, pharmacists, and laboratory technicians. Moreover, patients are not permitted to go home in a ride share and are counseled on Centers for Disease Control and Prevention quarantining following infusion.

Measurement of Process and Outcomes and Reporting

IRB approval was sought and obtained early during initiation of this service, allowing study consent to be offered to patients at the time general consent was obtained, which maximized patient recruitment and streamlined workflow. The study is a prospective observational research study to determine the impact of administration of COVID-19 monoclonal antibody therapy on length of symptoms, chronic illness, and rate of hospitalization. Most patients were eager to participate and offer their assistance to the scientific community during this pandemic.

Staff Education

In order to successfully implement this multidisciplinary EUA treatment option, comprehensive staff education was paramount after the workflow was developed. Prior to the first day of infusions, nurses and pharmacists were provided education during multiple huddle announcements. The pharmacy team also provided screen captures via email to the pharmacists so they could become familiar with the order set, intervention documentation, and location of the preliminary assessment of EUA criteria for use at the time of order verification. The emergency medicine department chair and chief medical officer also provided education via several virtual meetings and email to referring physicians (specialists and primary care) and residents in the emergency centers involved in COVID-19 monoclonal therapy-related patient care.

Factors Contributing to Success

We believe the reasons for continued success of this process are multifactorial and include the following key elements. Multidisciplinary planning, which included decision makers and all stakeholders, began at the time the idea was conceived. This allowed quick implementation of this service by efficiently navigating barriers to engaging impacted staff early on. Throughout this process, the authors set realistic step-wise goals. While navigating through the many details to implementation described, we also kept in mind the big picture, which was to provide this potentially lifesaving therapy to as many qualifying members of our community as possible. This included being flexible with the process and adapting when needed to achieve this ultimate goal. A focus on safety remained a priority to minimize possible errors and enhance patient and staff satisfaction. The optimization of the EHR streamlined workflow, provided point-of-care resources, and enhanced patient safety. Additionally, the target date set for implementation allowed staff and department leads adequate time to plan for and anticipate the changes. Serving only 1 patient on the first day allowed time for staff to experience this new process hands-on and provided opportunity for focused education. This team communication was essential to implementing this project, including staff training of processes and procedures prior to go-live. Early incorporation of IRB approval allowed the experience to be assessed and considered for contribution to the scientific literature to tackle this novel virus that has impacted our communities locally, nationally, and abroad. Moreover, continued measurement and reporting on a regular basis leads to performance improvement. The process outlined here can be adapted to incorporate other new therapies in the future, such as the recent February 9, 2021, EUA of the COVID-19 monoclonal antibody combination bamlanivimab and etesevimab.10

Conclusion

We administered 790 COVID-19 monoclonal antibody infusions between November 20, 2020 and March 5, 2021. Steps to minimize the likelihood of hypersensitivity reactions were implemented, and a low incidence (< 1%) has been observed. There has been no incidence of infection, concern from staff about infection prevention, or risk of infection during the processes. There have been very infrequent cost-related concerns raised by patients, typically due to incomplete communication regarding billing prior to the infusion. To address these issues, staff education has been provided to enhance patient instruction on this topic. The program has provided patient and family satisfaction, as well nursing, physician, pharmacist, clinical staff, and hospital administration pride and gratification. Setting up a new program to provide a 4-hour patient encounter to infuse therapy to high-risk patients with COVID-19 requires commitment and effort. This article describes the experience, ideas, and formula others may consider using to set up such a program. Through networking and formal phone calls and meetings about monoclonal antibody therapy, we have heard about other institutions who have not been able to institute this program due to various barriers to implementation. We hope our experience serves as a resource for others to provide this therapy to their patients and expand access in an effort to mitigate COVID-19 consequences and cases affecting our communities.

Corresponding author: Kathleen Jodoin, PharmD, BCPS, Mount Sinai Medical Center, 4300 Alton Rd, Miami Beach, FL 33140; kathleen.jodoin@msmc.com.

Financial disclosures: None.

References

1. COVID Data Tracker. Center for Disease Control and Prevention. https://covid.cdc.gov/covid-data-tracker/#global-counts-rates. Accessed March 12, 2021.

2. Fact Sheet for Health Care Providers Emergency Use Authorization (EUA) of Bamlanivimab. US Food and Drug Administration. Updated February 2021. Accessed March 9, 2021. https://www.fda.gov/media/143603/download

3. Coronavirus (COVID-19) Update: FDA Authorizes Monoclonal Antibodies for Treatment of COVID-19 | FDA. https://www.fda.gov/news-events/press-announcements/coronavirus-covid-19-update-fda-authorizes-monoclonal-antibodies-treatment-covid-19. Accessed February 14, 2021.

4. Fact Sheet for Health Care Providers Emergency Use Authorization (EUA) of Casirivimab and Imdevimab. US Food and Drug Administration. Updated December 2020. Accessed March 9, 2021. https://www.fda.gov/media/143892/download

5. Chen P, Nirula A, Heller B, et al. SARS-CoV-2 Neutralizing antibody LY-CoV555 in outpatients with COVID-19. N Engl J Med. 2021;384(3):229-237. doi:10.1056/NEJMoa2029849

6. Gottlieb RL, Nirula A, Chen P, et al. Effect of bamlanivimab as monotherapy or in combination with etesevimab on viral load in patients with mild to moderate COVID-19: a randomized clinical trial. 10.1JAMA. 2021;325(7):632-644. doi:10.1001/jama.2021.0202

7. Weinreich DM, Sivapalasingam S, Norton T, et al. REGN-COV2, a neutralizing antibody cocktail, in outpatients with COVID-19. 10.1N Engl J Med. 2021;384:238-251. doi:10.1056/nejmoa2035002

8. Mulangu S, Dodd LE, Davey RT Jr, et al. A randomized, controlled trial of Ebola virus disease therapeutics. 10.1N Engl J Med. 2019;381:2293-2303. doi:10.1056/NEJMoa1910993

9. Boyle, P. Can an experimental treatment keep COVID-19 patients out of hospitals? Association of American Medical Colleges. January 29, 2021. Accessed March 9, 2021. https://www.aamc.org/news-insights/can-experimental-treatment-keep-covid-19-patients-out-hospitals

10. Fact Sheet for Health Care Providers Emergency Use Authorization (EUA) of Bamlanivimab and Etesevimab. US Food and Drug Administration. Updated February 2021. Accessed March 9, 2021. https://www.fda.gov/media/145802/download

Article PDF
Issue
Journal of Clinical Outcomes Management - 28(2)
Publications
Topics
Page Number
70-81
Sections
Article PDF
Article PDF

From Mount Sinai Medical Center, Miami Beach, FL.

Abstract

Objective: To develop and implement a process for administering COVID-19 monoclonal antibody infusions for outpatients with mild or moderate COVID-19 at high risk for hospitalization, using multidisciplinary collaboration, US Food and Drug Administration (FDA) guidance, and infection prevention standards.

Methods: When monoclonal antibody therapy became available for mild or moderate COVID-19 outpatients via Emergency Use Authorization (EUA), our institution sought to provide this therapy option to our patients. We describe the process for planning, implementing, and maintaining a successful program for administering novel therapies based on FDA guidance and infection prevention standards. Key components of our implementation process were multidisciplinary planning involving decision makers and stakeholders; setting realistic goals in the process; team communication; and measuring and reporting quality improvement on a regular basis.

Results: A total of 790 COVID-19 monoclonal antibody infusions were administered from November 20, 2020 to March 5, 2021. Steps to minimize the likelihood of adverse drug reactions were implemented and a low incidence (< 1%) has occurred. There has been no concern from staff regarding infection during the process. Rarely, patients have raised cost-related concerns, typically due to incomplete communication regarding billing prior to the infusion. Patients, families, nursing staff, physicians, pharmacy, and hospital administration have expressed satisfaction with the program.

Conclusion: This process can provide a template for other hospitals or health care delivery facilities to provide novel therapies to patients with mild or moderate COVID-19 in a safe and effective manner.

Keywords: COVID-19; monoclonal antibody; infusion; emergency use authorization.

SARS-CoV-2 and the disease it causes, COVID-19, have transformed from scientific vernacular to common household terms. It began with a cluster of pneumonia cases of unknown etiology in December 2019 in Wuhan, China, with physicians there reporting a novel coronavirus strain (2019-nCoV), now referred to as SARS-CoV-2. Rapid spread of this virus resulted in the World Health Organization (WHO) declaring an international public health emergency. Since this time, the virus has evolved into a worldwide pandemic. COVID-19 has dramatically impacted our society, resulting in more than 2.63 million global deaths as of this writing, of which more than 527,000 deaths have occurred in the United States.1 This novel virus has resulted in a flurry of literature, research, therapies, and collaboration across multiple disciplines in an effort to prevent, treat, and mitigate cases and complications of this disease.

 

 

On November 9, 2020, and November 21, 2020, the US Food and Drug Administration (FDA) issued Emergency Use Authorizations (EUA) for 2 novel COVID-19 monoclonal therapies, bamlanivimab2-3 and casirivimab/imdevimab,3-4 respectively. The EUAs granted permission for these therapies to be administered for the treatment of mild to moderate COVID-19 in adult and pediatric patients (≥ 12 years and weighing at least 40 kg) with positive results of direct SARS-CoV-2 viral testing and who are at high risk for progressing to severe COVID-19 and/or hospitalization. The therapies work by targeting the SARS-CoV-2 spike protein and subsequent attachment to human angiotensin-converting enzyme 2 receptors. Clinical trial data leading to the EUA demonstrated a reduction in viral load, safe outcome, and most importantly, fewer hospitalization and emergency room visits, as compared to the placebo group.5-7 The use of monoclonal antibodies is not new and gained recognition during the Ebola crisis, when the monoclonal antibody to the Ebola virus showed a significant survival benefit.8 Providing monoclonal antibody therapy soon after symptom onset aligns with a shift from the onset of the pandemic to the current focus on the administration of pharmaceutical therapy early in the disease course. This shift prevents progression to severe COVID-19, with the goal of reducing patient mortality, hospitalizations, and strain on health care systems.

The availability of novel neutralizing monoclonal antibodies for COVID-19 led to discussions of how to incorporate these therapies as new options for patients. Our institution networked with colleagues from multiple disciplines to discuss processes and policies for the safe administration of the monoclonal antibody infusion therapies. Federal health leaders urge more use of monoclonal antibodies, but many hospitals have been unable to successfully implement infusions due to staff and logistical challenges.9 This article presents a viable process that hospitals can use to provide these novel therapies to outpatients with mild to moderate COVID-19.

The Mount Sinai Medical Center, Florida Experience

Mount Sinai Medical Center in Miami Beach, Florida, is the largest private, independent, not-for-profit teaching hospital in South Florida, comprising 672 licensed beds and supporting 150,000 emergency department (ED) visits annually. Per the EUA criteria for use, COVID-19 monoclonal antibody therapies are not authorized for patients who are hospitalized or who require oxygen therapy due to COVID-19. Therefore, options for outpatient administration needed to be evaluated. Directly following the first EUA press release, a task force of key stakeholders was assembled to brainstorm and develop a process to offer this therapy to the community. A multidisciplinary task force with representation from the ED, nursing, primary care, hospital medicine, pharmacy, risk management, billing, information technology, infection prevention, and senior level leadership participated (Table).

List of Key Stakeholders and Responsibilities

The task force reviewed institutional outpatient locations to determine whether offering this service would be feasible (eg, ED, ambulatory care facilities, cancer center). The ED was selected because it would offer the largest array of appointment times to meet the community needs with around-the-clock availability. While Mount Sinai Medical Center offers care in 3 emergency center locations in Aventura, Hialeah, and Miami Beach, it was determined to initiate the infusions at the main campus center in Miami Beach only. The main campus affords an onsite pharmacy with suitable staffing to prepare the anticipated volume of infusions in a timely manner, as both therapies have short stabilities following preparation. Thus, it was decided that patients from freestanding emergency centers in Aventura and Hialeah would be moved to the Miami Beach ED location to receive therapy. Operating at a single site also allowed for more rapid implementation, monitoring, and ability to make modifications more easily. Discussions for the possible expansion of COVID-19 monoclonal antibody infusions at satellite locations are underway.

Process implementation timeline

On November 20, 2020, 11 days after the formation of the multidisciplinary task force, the first COVID-19 monoclonal infusion was successfully administered. Figure 1 depicts the timeline from assessment to program implementation. Critical to implementation was the involvement of decision makers from all necessary departments early in the planning process to ensure that standard operating procedures were followed and that the patients, community, and organization had a positive experience. This allowed for simultaneous planning of electronic health record (Epic; EHR) builds, departmental workflows, and staff education, as described in the following section. Figure 2 shows the patient safety activities included in the implementation process.

Important patient safety initiatives

 

 

Key Stakeholder Involvement and Workflow

On the day of bamlanivimab EUA release, email communication was shared among hospital leadership with details of the press release. Departments were quickly involved to initiate a task force to assess if and how this therapy could be offered at Mount Sinai Medical Center. The following sections explain the role of each stakeholder and their essential role to operationalize these novel EUA treatment options. The task force was organized and led by our chief medical officer and chief nursing officer.

Information Technology

Medication Ordering and Documentation EHR and Smart Pumps. Early in the pandemic, the antimicrobial stewardship (ASP) clinical coordinator became the designated point person for pharmacy assessment of novel COVID-19 therapies. As such, this pharmacist began reviewing the bamlanivimab and, later, the casirivimab/imdevimab EUA Fact Sheet for Health Care Providers. All necessary elements for the complete and safe ordering and dispensing of the medication were developed and reviewed by pharmacy administration and ED nursing leadership for input, prior to submitting to the information technology team for implementation. Building the COVID-19 monoclonal medication records into the EHR allowed for detailed direction (ie, administration and preparation instructions) to be consistently applied. The medication records were also built into hospital smart pumps so that nurses could access prepopulated, accurate volumes and infusion rates to minimize errors.

Order Set Development. The pharmacy medication build was added to a comprehensive order set (Figure 3), which was then developed to guide prescribers and standardize the process around ordering of COVID-19 monoclonal therapies. While these therapies are new, oncology monoclonal therapies are regularly administered to outpatients at Mount Sinai Cancer Center. The cancer center was therefore consulted on their process surrounding best practices in administration of monoclonal antibody therapies. This included protocols for medications used in pretreatment and management of hypersensitivity reactions and potential adverse drug reactions of both COVID-19 monoclonal therapies. These medication orders were selected by default in the order set to ensure that all patients received premedications aimed at minimizing the risk of hypersensitivity reaction, and had as-needed medication orders, in the event a hypersensitivity reaction occurred. Reducing hypersensitivity reaction risk is important as well to increase the likelihood that the patient would receive full therapy, as management of this adverse drug reactions involves possible cessation of therapy depending on the level of severity. The pharmacy department also ensured these medications were stocked in ED automated dispensing cabinets to promote quick access. In addition to the aforementioned nursing orders, we added EUA criteria for use and hyperlinks to the Fact Sheets for Patients and Caregivers and Health Care Providers for each monoclonal therapy, and restricted ordering to ED physicians, nurse practitioners, and physician assistants.

COVID-19 monoclonal antibody order set

The order set underwent multidisciplinary review by pharmacy administration, the chair of emergency medicine, physicians, and ED nursing leadership prior to presentation and approval by the Pharmacy and Therapeutics Committee. Lastly, at time of implementation, the order set was added to the ED preference list, preventing inpatient access. Additionally, as a patient safety action, free- standing orders of COVID-19 monoclonal therapies were disabled, so providers could only order therapies via the approved, comprehensive order set.

Preliminary Assessment Tool. A provider assessment tool was developed to document patient-specific EUA criteria for use during initial assessment (Figure 4). This tool serves as a checklist and is visible to the full multidisciplinary team in the patient’s EHR. It is used as a resource at the time of pharmacist verification and ED physician assessment to ensure criteria for use are met.

Workflow for COVID-19 monoclonal antibody infusion

 

 

Outpatient Offices

Patient Referral. Patients with symptoms or concerns of COVID-19 exposure can make physician appointments via telemedicine or in person at Mount Sinai Medical Center’s primary care and specialty offices. At the time of patient encounter, physicians suspecting a COVID-19 diagnosis will refer patients for outpatient COVID-19 polymerase chain reaction (PCR) laboratory testing, which has an approximate 24-hour turnaround to results. Physicians also assess whether the patient meets EUA criteria for use, pending results of testing. In the event a patient meets EUA criteria for use, the physician provides patient counseling and requests verbal consent. Following this, the physician enters a note in the EHR describing the patient’s condition, criteria for use evaluation, and the patient’s verbal agreement to therapy. This preliminary screening is beneficial to begin planning with both the patient and ED to minimize delays. Patients are notified of the results of their test once available. If the COVID-19 PCR test returns positive, the physician will call the ED at the main campus and schedule the patient for COVID-19 monoclonal therapy. As the desired timeframe for administering COVID-19 monoclonal therapies is within less than 10 days of symptom onset, timely scheduling of appointments is crucial. Infusion appointments are typically provided the same or next day. The patients are informed that they must bring documentation of their positive COVID-19 PCR test to their ED visit. Lastly, because patients are pretreated with medication that may potentially impair driving, they are instructed that they cannot drive themselves home; ride shares also are not allowed in order to limit the spread of infection.

Emergency Department

Patient Arrival and Screening. A COVID-19 patient can be evaluated in the ED 1 of 2 ways. The first option is via outpatient office referral, as described previously. Upon arrival to the ED, a second screening is performed to ensure the patient still meets EUA criteria for use and the positive COVID-19 PCR test result is confirmed. If the patient no longer meets criteria, the patient is triaged accordingly, including evaluation for higher-level care (eg, supplemental oxygen, hospital admission). The second optoion is via new patient walk-ins without outpatient physician referral (Figure 4). In these cases, an initial screening is performed, documenting EUA criteria for use in the preliminary assessment (Figure 5). Physicians will consider an outside COVID-19 test as valid, so long as documentation is readily available confirming a positive PCR result. Otherwise, an in-house COVID-19 PCR test will be performed, which has a 2-hour turnaround time.

Electronic health record preliminary assessment

Infusion Schedule. The ED offers a total of 16 COVID-19 monoclonal infusions slots daily. These are broken up into 4 infusion time blocks (eg, 8 am, 12 pm, 4 pm, 8 pm), with each infusion time block consisting of 4 available patient appointments. A list of scheduled infusions for the day is emailed to the pharmacy department every morning, and patients are instructed to arrive 1 hour prior to their appointment time. This allows time for patient registration, assessment, and pharmacy notification in advance of order entry. For logistical purposes, and as a patient safety initiative to reduce the likelihood of medication errors, each of the available COVID-19 monoclonal antibodies is offered on a designated day. Bamlanivimab is offered on Tuesday, Thursday, Saturday, and Sunday, while casirivimab/imdevimab is offered Monday, Wednesday, and Friday. This provides flexibility to adjust should supply deviate based on Department of Health allocation or should new therapy options within this class of medication become available.

Patient Education. Prior to administration of the monoclonal therapy, physician and nursing staff obtain a formal, written patient consent for therapy and provide patients with the option of participating in the institutional review board (IRB) approved study. Details of this are discussed in the risk management and IRB sections of the article. Nursing staff also provides the medication-specific Fact Sheet for Patients and Caregivers in either Spanish or English, which is also included as a hyperlink on the COVID-19 Monoclonal Antibody Order Set for ease of access. Interpreter services are available for patients who speak other languages. An ED decentralized pharmacist is also available onsite Monday through Friday from 12 pm to 8:30 pm to supplement education and serve as a resource for any questions.

Infusion Ordering. Once the patient is ready to begin therapy, the he/she is brought to a dedicated overflow area of the ED. There are few, if any, patients in this location, and it is adjacent to the main emergency center for easy access by the patients, nurses, pharmacists, and physicians. The physician then enters orders in the EHR using the COVID-19 Monoclonal Antibody Order Set (Figure 3). Three discrete questions were built into the medication order: (1) Was patient consent obtained? (2) Was the Fact Sheet for Patient/Caregiver provided to the patient? (3) Is the patient COVID-19 PCR-positive? These questions were built as hard stops so that the medication orders cannot be placed without a response. This serves as another double-check to ensure processes are followed and helps facilitate timely verification by the pharmacist.

 

 

Medication Administration. One nurse is dedicated to administering the monoclonal therapies scheduled at 8 am and 12 pm and another at 4 pm and 8 pm. Each appointment block is 4 hours in duration to allow adequate time for patient registration, infusion, and postinfusion observation. The nurse administers the premedications and COVID-19 monoclonal therapy, and observes the patient for the required 1-hour postadministration observation period. Nursing orders detailing monitoring parameters for mild, moderate, and severe reactions, along with associated medication orders to administer in the event they occur, are detailed in the nursing orders of the COVID-19 Monoclonal Antibody Order Set (Figure 3). Prior to administration, the nurse scans each medication and the patient’s wrist identification band, and documents the time of administration within the EHR medication administration report.

Pharmacy Department

Medication Receipt Process. Inventory is currently allocated biweekly from the state department of health and will soon be transitioning to a direct order system. The pharmacy technician in charge of deliveries notifies the pharmacy Antimicrobial Stewardship Program (ASP) clinical coordinator upon receipt of the monoclonal therapies. Bamlanivimab is supplied as 1 vial per dose, whereas casirivimab/imdevimab is supplied as 4 vials or 8 vials per dose, depending how it is shipped. To reduce the likelihood of medication errors, the ASP clinical coordinator assembles each of the casirivimab/imdevimab vials into kits, where 1 kit equals 1 dose. Labels are then affixed to each kit indicating the medication name, number of vials which equal a full dose, and pharmacist signature. The kits are stored in a dedicated refrigerator, and inventory logs are affixed to the outside of the refrigerator and updated daily. This inventory is also communicated daily to ED physician, nursing, and pharmacy leadership, as well as the director of patient safety, who reports weekly usage to the state Department of Health and Human Services. These weekly reports are used to determine allocation amounts.

Medication Verification and Delivery. The Mount Sinai Medical Center pharmacist staffing model consists of centralized order entry and specialized, decentralized positions. All orders are verified by the ED pharmacist when scheduled (not a 24/7 service) and by the designated pharmacist for all other times. At the time of medication verification, the pharmacist documents patient-specific EUA criteria for use and confirms that consent was obtained and the Fact Sheet for Patients/Caregivers was provided. A pharmacist intervention was developed to assist with this documentation. Pharmacists input smart text “.COVIDmonoclonal” and a drop-down menu of EUA criteria for use appears. The pharmacist reviews the patient care notes and medication order question responses to ascertain this information, contacting the ED prescriber if further clarification is required. This verification serves as another check to ensure processes put in place are followed. Lastly, intravenous preparation and delivery are electronically recorded in the EHR, and the medications require nursing signature at the time of delivery to ensure a formal chain of custody.

Risk Management

At Mount Sinai Medical Center, all EUA and investigational therapies require patient consent. Consistent with this requirement, a COVID-19 monoclonal specific consent was developed by risk management. This is provided to every patient receiving a COVID-19 monoclonal infusion, in addition to the FDA EUA Fact Sheet for Patients and Caregivers, and documented as part of their EHR. The questions providers must answer are built into the order set to ensure this process is followed and these patient safety checks are incorporated into the workflow.

Billing and Finance Department

In alignment with Mount Sinai Medical Center’s mission to provide high-quality health care to its diverse community through teaching, research, charity care, and financial responsibility, it was determined that this therapy would be provided to all patients regardless of insurance type, including those who are uninsured. The billing and finance department was consulted prior to this service being offered, to provide patients with accurate and pertinent information. The billing and finance department provided guidance on how to document patient encounters at time of registration to facilitate appropriate billing. At this time, the medication is free of charge, but nonmedication-related ED fees apply. This is explained to patients so there is a clear understanding prior to booking their appointment.

 

 

Infection Prevention

As patients receiving COVID-19 monoclonal therapies can transmit the virus to others, measures to ensure protection for other patients and staff are vital. To minimize exposure, specific nursing and physician staff from the ED are assigned to the treatment of these patients, and patients receive infusions and postobservation monitoring in a designated wing of the ED. Additionally, all staff who interact with these patients are required to don full personal protective equipment. This includes not only physicians and nurses but all specialties such as physician assistants, nurse practitioners, pharmacists, and laboratory technicians. Moreover, patients are not permitted to go home in a ride share and are counseled on Centers for Disease Control and Prevention quarantining following infusion.

Measurement of Process and Outcomes and Reporting

IRB approval was sought and obtained early during initiation of this service, allowing study consent to be offered to patients at the time general consent was obtained, which maximized patient recruitment and streamlined workflow. The study is a prospective observational research study to determine the impact of administration of COVID-19 monoclonal antibody therapy on length of symptoms, chronic illness, and rate of hospitalization. Most patients were eager to participate and offer their assistance to the scientific community during this pandemic.

Staff Education

In order to successfully implement this multidisciplinary EUA treatment option, comprehensive staff education was paramount after the workflow was developed. Prior to the first day of infusions, nurses and pharmacists were provided education during multiple huddle announcements. The pharmacy team also provided screen captures via email to the pharmacists so they could become familiar with the order set, intervention documentation, and location of the preliminary assessment of EUA criteria for use at the time of order verification. The emergency medicine department chair and chief medical officer also provided education via several virtual meetings and email to referring physicians (specialists and primary care) and residents in the emergency centers involved in COVID-19 monoclonal therapy-related patient care.

Factors Contributing to Success

We believe the reasons for continued success of this process are multifactorial and include the following key elements. Multidisciplinary planning, which included decision makers and all stakeholders, began at the time the idea was conceived. This allowed quick implementation of this service by efficiently navigating barriers to engaging impacted staff early on. Throughout this process, the authors set realistic step-wise goals. While navigating through the many details to implementation described, we also kept in mind the big picture, which was to provide this potentially lifesaving therapy to as many qualifying members of our community as possible. This included being flexible with the process and adapting when needed to achieve this ultimate goal. A focus on safety remained a priority to minimize possible errors and enhance patient and staff satisfaction. The optimization of the EHR streamlined workflow, provided point-of-care resources, and enhanced patient safety. Additionally, the target date set for implementation allowed staff and department leads adequate time to plan for and anticipate the changes. Serving only 1 patient on the first day allowed time for staff to experience this new process hands-on and provided opportunity for focused education. This team communication was essential to implementing this project, including staff training of processes and procedures prior to go-live. Early incorporation of IRB approval allowed the experience to be assessed and considered for contribution to the scientific literature to tackle this novel virus that has impacted our communities locally, nationally, and abroad. Moreover, continued measurement and reporting on a regular basis leads to performance improvement. The process outlined here can be adapted to incorporate other new therapies in the future, such as the recent February 9, 2021, EUA of the COVID-19 monoclonal antibody combination bamlanivimab and etesevimab.10

Conclusion

We administered 790 COVID-19 monoclonal antibody infusions between November 20, 2020 and March 5, 2021. Steps to minimize the likelihood of hypersensitivity reactions were implemented, and a low incidence (< 1%) has been observed. There has been no incidence of infection, concern from staff about infection prevention, or risk of infection during the processes. There have been very infrequent cost-related concerns raised by patients, typically due to incomplete communication regarding billing prior to the infusion. To address these issues, staff education has been provided to enhance patient instruction on this topic. The program has provided patient and family satisfaction, as well nursing, physician, pharmacist, clinical staff, and hospital administration pride and gratification. Setting up a new program to provide a 4-hour patient encounter to infuse therapy to high-risk patients with COVID-19 requires commitment and effort. This article describes the experience, ideas, and formula others may consider using to set up such a program. Through networking and formal phone calls and meetings about monoclonal antibody therapy, we have heard about other institutions who have not been able to institute this program due to various barriers to implementation. We hope our experience serves as a resource for others to provide this therapy to their patients and expand access in an effort to mitigate COVID-19 consequences and cases affecting our communities.

Corresponding author: Kathleen Jodoin, PharmD, BCPS, Mount Sinai Medical Center, 4300 Alton Rd, Miami Beach, FL 33140; kathleen.jodoin@msmc.com.

Financial disclosures: None.

From Mount Sinai Medical Center, Miami Beach, FL.

Abstract

Objective: To develop and implement a process for administering COVID-19 monoclonal antibody infusions for outpatients with mild or moderate COVID-19 at high risk for hospitalization, using multidisciplinary collaboration, US Food and Drug Administration (FDA) guidance, and infection prevention standards.

Methods: When monoclonal antibody therapy became available for mild or moderate COVID-19 outpatients via Emergency Use Authorization (EUA), our institution sought to provide this therapy option to our patients. We describe the process for planning, implementing, and maintaining a successful program for administering novel therapies based on FDA guidance and infection prevention standards. Key components of our implementation process were multidisciplinary planning involving decision makers and stakeholders; setting realistic goals in the process; team communication; and measuring and reporting quality improvement on a regular basis.

Results: A total of 790 COVID-19 monoclonal antibody infusions were administered from November 20, 2020 to March 5, 2021. Steps to minimize the likelihood of adverse drug reactions were implemented and a low incidence (< 1%) has occurred. There has been no concern from staff regarding infection during the process. Rarely, patients have raised cost-related concerns, typically due to incomplete communication regarding billing prior to the infusion. Patients, families, nursing staff, physicians, pharmacy, and hospital administration have expressed satisfaction with the program.

Conclusion: This process can provide a template for other hospitals or health care delivery facilities to provide novel therapies to patients with mild or moderate COVID-19 in a safe and effective manner.

Keywords: COVID-19; monoclonal antibody; infusion; emergency use authorization.

SARS-CoV-2 and the disease it causes, COVID-19, have transformed from scientific vernacular to common household terms. It began with a cluster of pneumonia cases of unknown etiology in December 2019 in Wuhan, China, with physicians there reporting a novel coronavirus strain (2019-nCoV), now referred to as SARS-CoV-2. Rapid spread of this virus resulted in the World Health Organization (WHO) declaring an international public health emergency. Since this time, the virus has evolved into a worldwide pandemic. COVID-19 has dramatically impacted our society, resulting in more than 2.63 million global deaths as of this writing, of which more than 527,000 deaths have occurred in the United States.1 This novel virus has resulted in a flurry of literature, research, therapies, and collaboration across multiple disciplines in an effort to prevent, treat, and mitigate cases and complications of this disease.

 

 

On November 9, 2020, and November 21, 2020, the US Food and Drug Administration (FDA) issued Emergency Use Authorizations (EUA) for 2 novel COVID-19 monoclonal therapies, bamlanivimab2-3 and casirivimab/imdevimab,3-4 respectively. The EUAs granted permission for these therapies to be administered for the treatment of mild to moderate COVID-19 in adult and pediatric patients (≥ 12 years and weighing at least 40 kg) with positive results of direct SARS-CoV-2 viral testing and who are at high risk for progressing to severe COVID-19 and/or hospitalization. The therapies work by targeting the SARS-CoV-2 spike protein and subsequent attachment to human angiotensin-converting enzyme 2 receptors. Clinical trial data leading to the EUA demonstrated a reduction in viral load, safe outcome, and most importantly, fewer hospitalization and emergency room visits, as compared to the placebo group.5-7 The use of monoclonal antibodies is not new and gained recognition during the Ebola crisis, when the monoclonal antibody to the Ebola virus showed a significant survival benefit.8 Providing monoclonal antibody therapy soon after symptom onset aligns with a shift from the onset of the pandemic to the current focus on the administration of pharmaceutical therapy early in the disease course. This shift prevents progression to severe COVID-19, with the goal of reducing patient mortality, hospitalizations, and strain on health care systems.

The availability of novel neutralizing monoclonal antibodies for COVID-19 led to discussions of how to incorporate these therapies as new options for patients. Our institution networked with colleagues from multiple disciplines to discuss processes and policies for the safe administration of the monoclonal antibody infusion therapies. Federal health leaders urge more use of monoclonal antibodies, but many hospitals have been unable to successfully implement infusions due to staff and logistical challenges.9 This article presents a viable process that hospitals can use to provide these novel therapies to outpatients with mild to moderate COVID-19.

The Mount Sinai Medical Center, Florida Experience

Mount Sinai Medical Center in Miami Beach, Florida, is the largest private, independent, not-for-profit teaching hospital in South Florida, comprising 672 licensed beds and supporting 150,000 emergency department (ED) visits annually. Per the EUA criteria for use, COVID-19 monoclonal antibody therapies are not authorized for patients who are hospitalized or who require oxygen therapy due to COVID-19. Therefore, options for outpatient administration needed to be evaluated. Directly following the first EUA press release, a task force of key stakeholders was assembled to brainstorm and develop a process to offer this therapy to the community. A multidisciplinary task force with representation from the ED, nursing, primary care, hospital medicine, pharmacy, risk management, billing, information technology, infection prevention, and senior level leadership participated (Table).

List of Key Stakeholders and Responsibilities

The task force reviewed institutional outpatient locations to determine whether offering this service would be feasible (eg, ED, ambulatory care facilities, cancer center). The ED was selected because it would offer the largest array of appointment times to meet the community needs with around-the-clock availability. While Mount Sinai Medical Center offers care in 3 emergency center locations in Aventura, Hialeah, and Miami Beach, it was determined to initiate the infusions at the main campus center in Miami Beach only. The main campus affords an onsite pharmacy with suitable staffing to prepare the anticipated volume of infusions in a timely manner, as both therapies have short stabilities following preparation. Thus, it was decided that patients from freestanding emergency centers in Aventura and Hialeah would be moved to the Miami Beach ED location to receive therapy. Operating at a single site also allowed for more rapid implementation, monitoring, and ability to make modifications more easily. Discussions for the possible expansion of COVID-19 monoclonal antibody infusions at satellite locations are underway.

Process implementation timeline

On November 20, 2020, 11 days after the formation of the multidisciplinary task force, the first COVID-19 monoclonal infusion was successfully administered. Figure 1 depicts the timeline from assessment to program implementation. Critical to implementation was the involvement of decision makers from all necessary departments early in the planning process to ensure that standard operating procedures were followed and that the patients, community, and organization had a positive experience. This allowed for simultaneous planning of electronic health record (Epic; EHR) builds, departmental workflows, and staff education, as described in the following section. Figure 2 shows the patient safety activities included in the implementation process.

Important patient safety initiatives

 

 

Key Stakeholder Involvement and Workflow

On the day of bamlanivimab EUA release, email communication was shared among hospital leadership with details of the press release. Departments were quickly involved to initiate a task force to assess if and how this therapy could be offered at Mount Sinai Medical Center. The following sections explain the role of each stakeholder and their essential role to operationalize these novel EUA treatment options. The task force was organized and led by our chief medical officer and chief nursing officer.

Information Technology

Medication Ordering and Documentation EHR and Smart Pumps. Early in the pandemic, the antimicrobial stewardship (ASP) clinical coordinator became the designated point person for pharmacy assessment of novel COVID-19 therapies. As such, this pharmacist began reviewing the bamlanivimab and, later, the casirivimab/imdevimab EUA Fact Sheet for Health Care Providers. All necessary elements for the complete and safe ordering and dispensing of the medication were developed and reviewed by pharmacy administration and ED nursing leadership for input, prior to submitting to the information technology team for implementation. Building the COVID-19 monoclonal medication records into the EHR allowed for detailed direction (ie, administration and preparation instructions) to be consistently applied. The medication records were also built into hospital smart pumps so that nurses could access prepopulated, accurate volumes and infusion rates to minimize errors.

Order Set Development. The pharmacy medication build was added to a comprehensive order set (Figure 3), which was then developed to guide prescribers and standardize the process around ordering of COVID-19 monoclonal therapies. While these therapies are new, oncology monoclonal therapies are regularly administered to outpatients at Mount Sinai Cancer Center. The cancer center was therefore consulted on their process surrounding best practices in administration of monoclonal antibody therapies. This included protocols for medications used in pretreatment and management of hypersensitivity reactions and potential adverse drug reactions of both COVID-19 monoclonal therapies. These medication orders were selected by default in the order set to ensure that all patients received premedications aimed at minimizing the risk of hypersensitivity reaction, and had as-needed medication orders, in the event a hypersensitivity reaction occurred. Reducing hypersensitivity reaction risk is important as well to increase the likelihood that the patient would receive full therapy, as management of this adverse drug reactions involves possible cessation of therapy depending on the level of severity. The pharmacy department also ensured these medications were stocked in ED automated dispensing cabinets to promote quick access. In addition to the aforementioned nursing orders, we added EUA criteria for use and hyperlinks to the Fact Sheets for Patients and Caregivers and Health Care Providers for each monoclonal therapy, and restricted ordering to ED physicians, nurse practitioners, and physician assistants.

COVID-19 monoclonal antibody order set

The order set underwent multidisciplinary review by pharmacy administration, the chair of emergency medicine, physicians, and ED nursing leadership prior to presentation and approval by the Pharmacy and Therapeutics Committee. Lastly, at time of implementation, the order set was added to the ED preference list, preventing inpatient access. Additionally, as a patient safety action, free- standing orders of COVID-19 monoclonal therapies were disabled, so providers could only order therapies via the approved, comprehensive order set.

Preliminary Assessment Tool. A provider assessment tool was developed to document patient-specific EUA criteria for use during initial assessment (Figure 4). This tool serves as a checklist and is visible to the full multidisciplinary team in the patient’s EHR. It is used as a resource at the time of pharmacist verification and ED physician assessment to ensure criteria for use are met.

Workflow for COVID-19 monoclonal antibody infusion

 

 

Outpatient Offices

Patient Referral. Patients with symptoms or concerns of COVID-19 exposure can make physician appointments via telemedicine or in person at Mount Sinai Medical Center’s primary care and specialty offices. At the time of patient encounter, physicians suspecting a COVID-19 diagnosis will refer patients for outpatient COVID-19 polymerase chain reaction (PCR) laboratory testing, which has an approximate 24-hour turnaround to results. Physicians also assess whether the patient meets EUA criteria for use, pending results of testing. In the event a patient meets EUA criteria for use, the physician provides patient counseling and requests verbal consent. Following this, the physician enters a note in the EHR describing the patient’s condition, criteria for use evaluation, and the patient’s verbal agreement to therapy. This preliminary screening is beneficial to begin planning with both the patient and ED to minimize delays. Patients are notified of the results of their test once available. If the COVID-19 PCR test returns positive, the physician will call the ED at the main campus and schedule the patient for COVID-19 monoclonal therapy. As the desired timeframe for administering COVID-19 monoclonal therapies is within less than 10 days of symptom onset, timely scheduling of appointments is crucial. Infusion appointments are typically provided the same or next day. The patients are informed that they must bring documentation of their positive COVID-19 PCR test to their ED visit. Lastly, because patients are pretreated with medication that may potentially impair driving, they are instructed that they cannot drive themselves home; ride shares also are not allowed in order to limit the spread of infection.

Emergency Department

Patient Arrival and Screening. A COVID-19 patient can be evaluated in the ED 1 of 2 ways. The first option is via outpatient office referral, as described previously. Upon arrival to the ED, a second screening is performed to ensure the patient still meets EUA criteria for use and the positive COVID-19 PCR test result is confirmed. If the patient no longer meets criteria, the patient is triaged accordingly, including evaluation for higher-level care (eg, supplemental oxygen, hospital admission). The second optoion is via new patient walk-ins without outpatient physician referral (Figure 4). In these cases, an initial screening is performed, documenting EUA criteria for use in the preliminary assessment (Figure 5). Physicians will consider an outside COVID-19 test as valid, so long as documentation is readily available confirming a positive PCR result. Otherwise, an in-house COVID-19 PCR test will be performed, which has a 2-hour turnaround time.

Electronic health record preliminary assessment

Infusion Schedule. The ED offers a total of 16 COVID-19 monoclonal infusions slots daily. These are broken up into 4 infusion time blocks (eg, 8 am, 12 pm, 4 pm, 8 pm), with each infusion time block consisting of 4 available patient appointments. A list of scheduled infusions for the day is emailed to the pharmacy department every morning, and patients are instructed to arrive 1 hour prior to their appointment time. This allows time for patient registration, assessment, and pharmacy notification in advance of order entry. For logistical purposes, and as a patient safety initiative to reduce the likelihood of medication errors, each of the available COVID-19 monoclonal antibodies is offered on a designated day. Bamlanivimab is offered on Tuesday, Thursday, Saturday, and Sunday, while casirivimab/imdevimab is offered Monday, Wednesday, and Friday. This provides flexibility to adjust should supply deviate based on Department of Health allocation or should new therapy options within this class of medication become available.

Patient Education. Prior to administration of the monoclonal therapy, physician and nursing staff obtain a formal, written patient consent for therapy and provide patients with the option of participating in the institutional review board (IRB) approved study. Details of this are discussed in the risk management and IRB sections of the article. Nursing staff also provides the medication-specific Fact Sheet for Patients and Caregivers in either Spanish or English, which is also included as a hyperlink on the COVID-19 Monoclonal Antibody Order Set for ease of access. Interpreter services are available for patients who speak other languages. An ED decentralized pharmacist is also available onsite Monday through Friday from 12 pm to 8:30 pm to supplement education and serve as a resource for any questions.

Infusion Ordering. Once the patient is ready to begin therapy, the he/she is brought to a dedicated overflow area of the ED. There are few, if any, patients in this location, and it is adjacent to the main emergency center for easy access by the patients, nurses, pharmacists, and physicians. The physician then enters orders in the EHR using the COVID-19 Monoclonal Antibody Order Set (Figure 3). Three discrete questions were built into the medication order: (1) Was patient consent obtained? (2) Was the Fact Sheet for Patient/Caregiver provided to the patient? (3) Is the patient COVID-19 PCR-positive? These questions were built as hard stops so that the medication orders cannot be placed without a response. This serves as another double-check to ensure processes are followed and helps facilitate timely verification by the pharmacist.

 

 

Medication Administration. One nurse is dedicated to administering the monoclonal therapies scheduled at 8 am and 12 pm and another at 4 pm and 8 pm. Each appointment block is 4 hours in duration to allow adequate time for patient registration, infusion, and postinfusion observation. The nurse administers the premedications and COVID-19 monoclonal therapy, and observes the patient for the required 1-hour postadministration observation period. Nursing orders detailing monitoring parameters for mild, moderate, and severe reactions, along with associated medication orders to administer in the event they occur, are detailed in the nursing orders of the COVID-19 Monoclonal Antibody Order Set (Figure 3). Prior to administration, the nurse scans each medication and the patient’s wrist identification band, and documents the time of administration within the EHR medication administration report.

Pharmacy Department

Medication Receipt Process. Inventory is currently allocated biweekly from the state department of health and will soon be transitioning to a direct order system. The pharmacy technician in charge of deliveries notifies the pharmacy Antimicrobial Stewardship Program (ASP) clinical coordinator upon receipt of the monoclonal therapies. Bamlanivimab is supplied as 1 vial per dose, whereas casirivimab/imdevimab is supplied as 4 vials or 8 vials per dose, depending how it is shipped. To reduce the likelihood of medication errors, the ASP clinical coordinator assembles each of the casirivimab/imdevimab vials into kits, where 1 kit equals 1 dose. Labels are then affixed to each kit indicating the medication name, number of vials which equal a full dose, and pharmacist signature. The kits are stored in a dedicated refrigerator, and inventory logs are affixed to the outside of the refrigerator and updated daily. This inventory is also communicated daily to ED physician, nursing, and pharmacy leadership, as well as the director of patient safety, who reports weekly usage to the state Department of Health and Human Services. These weekly reports are used to determine allocation amounts.

Medication Verification and Delivery. The Mount Sinai Medical Center pharmacist staffing model consists of centralized order entry and specialized, decentralized positions. All orders are verified by the ED pharmacist when scheduled (not a 24/7 service) and by the designated pharmacist for all other times. At the time of medication verification, the pharmacist documents patient-specific EUA criteria for use and confirms that consent was obtained and the Fact Sheet for Patients/Caregivers was provided. A pharmacist intervention was developed to assist with this documentation. Pharmacists input smart text “.COVIDmonoclonal” and a drop-down menu of EUA criteria for use appears. The pharmacist reviews the patient care notes and medication order question responses to ascertain this information, contacting the ED prescriber if further clarification is required. This verification serves as another check to ensure processes put in place are followed. Lastly, intravenous preparation and delivery are electronically recorded in the EHR, and the medications require nursing signature at the time of delivery to ensure a formal chain of custody.

Risk Management

At Mount Sinai Medical Center, all EUA and investigational therapies require patient consent. Consistent with this requirement, a COVID-19 monoclonal specific consent was developed by risk management. This is provided to every patient receiving a COVID-19 monoclonal infusion, in addition to the FDA EUA Fact Sheet for Patients and Caregivers, and documented as part of their EHR. The questions providers must answer are built into the order set to ensure this process is followed and these patient safety checks are incorporated into the workflow.

Billing and Finance Department

In alignment with Mount Sinai Medical Center’s mission to provide high-quality health care to its diverse community through teaching, research, charity care, and financial responsibility, it was determined that this therapy would be provided to all patients regardless of insurance type, including those who are uninsured. The billing and finance department was consulted prior to this service being offered, to provide patients with accurate and pertinent information. The billing and finance department provided guidance on how to document patient encounters at time of registration to facilitate appropriate billing. At this time, the medication is free of charge, but nonmedication-related ED fees apply. This is explained to patients so there is a clear understanding prior to booking their appointment.

 

 

Infection Prevention

As patients receiving COVID-19 monoclonal therapies can transmit the virus to others, measures to ensure protection for other patients and staff are vital. To minimize exposure, specific nursing and physician staff from the ED are assigned to the treatment of these patients, and patients receive infusions and postobservation monitoring in a designated wing of the ED. Additionally, all staff who interact with these patients are required to don full personal protective equipment. This includes not only physicians and nurses but all specialties such as physician assistants, nurse practitioners, pharmacists, and laboratory technicians. Moreover, patients are not permitted to go home in a ride share and are counseled on Centers for Disease Control and Prevention quarantining following infusion.

Measurement of Process and Outcomes and Reporting

IRB approval was sought and obtained early during initiation of this service, allowing study consent to be offered to patients at the time general consent was obtained, which maximized patient recruitment and streamlined workflow. The study is a prospective observational research study to determine the impact of administration of COVID-19 monoclonal antibody therapy on length of symptoms, chronic illness, and rate of hospitalization. Most patients were eager to participate and offer their assistance to the scientific community during this pandemic.

Staff Education

In order to successfully implement this multidisciplinary EUA treatment option, comprehensive staff education was paramount after the workflow was developed. Prior to the first day of infusions, nurses and pharmacists were provided education during multiple huddle announcements. The pharmacy team also provided screen captures via email to the pharmacists so they could become familiar with the order set, intervention documentation, and location of the preliminary assessment of EUA criteria for use at the time of order verification. The emergency medicine department chair and chief medical officer also provided education via several virtual meetings and email to referring physicians (specialists and primary care) and residents in the emergency centers involved in COVID-19 monoclonal therapy-related patient care.

Factors Contributing to Success

We believe the reasons for continued success of this process are multifactorial and include the following key elements. Multidisciplinary planning, which included decision makers and all stakeholders, began at the time the idea was conceived. This allowed quick implementation of this service by efficiently navigating barriers to engaging impacted staff early on. Throughout this process, the authors set realistic step-wise goals. While navigating through the many details to implementation described, we also kept in mind the big picture, which was to provide this potentially lifesaving therapy to as many qualifying members of our community as possible. This included being flexible with the process and adapting when needed to achieve this ultimate goal. A focus on safety remained a priority to minimize possible errors and enhance patient and staff satisfaction. The optimization of the EHR streamlined workflow, provided point-of-care resources, and enhanced patient safety. Additionally, the target date set for implementation allowed staff and department leads adequate time to plan for and anticipate the changes. Serving only 1 patient on the first day allowed time for staff to experience this new process hands-on and provided opportunity for focused education. This team communication was essential to implementing this project, including staff training of processes and procedures prior to go-live. Early incorporation of IRB approval allowed the experience to be assessed and considered for contribution to the scientific literature to tackle this novel virus that has impacted our communities locally, nationally, and abroad. Moreover, continued measurement and reporting on a regular basis leads to performance improvement. The process outlined here can be adapted to incorporate other new therapies in the future, such as the recent February 9, 2021, EUA of the COVID-19 monoclonal antibody combination bamlanivimab and etesevimab.10

Conclusion

We administered 790 COVID-19 monoclonal antibody infusions between November 20, 2020 and March 5, 2021. Steps to minimize the likelihood of hypersensitivity reactions were implemented, and a low incidence (< 1%) has been observed. There has been no incidence of infection, concern from staff about infection prevention, or risk of infection during the processes. There have been very infrequent cost-related concerns raised by patients, typically due to incomplete communication regarding billing prior to the infusion. To address these issues, staff education has been provided to enhance patient instruction on this topic. The program has provided patient and family satisfaction, as well nursing, physician, pharmacist, clinical staff, and hospital administration pride and gratification. Setting up a new program to provide a 4-hour patient encounter to infuse therapy to high-risk patients with COVID-19 requires commitment and effort. This article describes the experience, ideas, and formula others may consider using to set up such a program. Through networking and formal phone calls and meetings about monoclonal antibody therapy, we have heard about other institutions who have not been able to institute this program due to various barriers to implementation. We hope our experience serves as a resource for others to provide this therapy to their patients and expand access in an effort to mitigate COVID-19 consequences and cases affecting our communities.

Corresponding author: Kathleen Jodoin, PharmD, BCPS, Mount Sinai Medical Center, 4300 Alton Rd, Miami Beach, FL 33140; kathleen.jodoin@msmc.com.

Financial disclosures: None.

References

1. COVID Data Tracker. Center for Disease Control and Prevention. https://covid.cdc.gov/covid-data-tracker/#global-counts-rates. Accessed March 12, 2021.

2. Fact Sheet for Health Care Providers Emergency Use Authorization (EUA) of Bamlanivimab. US Food and Drug Administration. Updated February 2021. Accessed March 9, 2021. https://www.fda.gov/media/143603/download

3. Coronavirus (COVID-19) Update: FDA Authorizes Monoclonal Antibodies for Treatment of COVID-19 | FDA. https://www.fda.gov/news-events/press-announcements/coronavirus-covid-19-update-fda-authorizes-monoclonal-antibodies-treatment-covid-19. Accessed February 14, 2021.

4. Fact Sheet for Health Care Providers Emergency Use Authorization (EUA) of Casirivimab and Imdevimab. US Food and Drug Administration. Updated December 2020. Accessed March 9, 2021. https://www.fda.gov/media/143892/download

5. Chen P, Nirula A, Heller B, et al. SARS-CoV-2 Neutralizing antibody LY-CoV555 in outpatients with COVID-19. N Engl J Med. 2021;384(3):229-237. doi:10.1056/NEJMoa2029849

6. Gottlieb RL, Nirula A, Chen P, et al. Effect of bamlanivimab as monotherapy or in combination with etesevimab on viral load in patients with mild to moderate COVID-19: a randomized clinical trial. 10.1JAMA. 2021;325(7):632-644. doi:10.1001/jama.2021.0202

7. Weinreich DM, Sivapalasingam S, Norton T, et al. REGN-COV2, a neutralizing antibody cocktail, in outpatients with COVID-19. 10.1N Engl J Med. 2021;384:238-251. doi:10.1056/nejmoa2035002

8. Mulangu S, Dodd LE, Davey RT Jr, et al. A randomized, controlled trial of Ebola virus disease therapeutics. 10.1N Engl J Med. 2019;381:2293-2303. doi:10.1056/NEJMoa1910993

9. Boyle, P. Can an experimental treatment keep COVID-19 patients out of hospitals? Association of American Medical Colleges. January 29, 2021. Accessed March 9, 2021. https://www.aamc.org/news-insights/can-experimental-treatment-keep-covid-19-patients-out-hospitals

10. Fact Sheet for Health Care Providers Emergency Use Authorization (EUA) of Bamlanivimab and Etesevimab. US Food and Drug Administration. Updated February 2021. Accessed March 9, 2021. https://www.fda.gov/media/145802/download

References

1. COVID Data Tracker. Center for Disease Control and Prevention. https://covid.cdc.gov/covid-data-tracker/#global-counts-rates. Accessed March 12, 2021.

2. Fact Sheet for Health Care Providers Emergency Use Authorization (EUA) of Bamlanivimab. US Food and Drug Administration. Updated February 2021. Accessed March 9, 2021. https://www.fda.gov/media/143603/download

3. Coronavirus (COVID-19) Update: FDA Authorizes Monoclonal Antibodies for Treatment of COVID-19 | FDA. https://www.fda.gov/news-events/press-announcements/coronavirus-covid-19-update-fda-authorizes-monoclonal-antibodies-treatment-covid-19. Accessed February 14, 2021.

4. Fact Sheet for Health Care Providers Emergency Use Authorization (EUA) of Casirivimab and Imdevimab. US Food and Drug Administration. Updated December 2020. Accessed March 9, 2021. https://www.fda.gov/media/143892/download

5. Chen P, Nirula A, Heller B, et al. SARS-CoV-2 Neutralizing antibody LY-CoV555 in outpatients with COVID-19. N Engl J Med. 2021;384(3):229-237. doi:10.1056/NEJMoa2029849

6. Gottlieb RL, Nirula A, Chen P, et al. Effect of bamlanivimab as monotherapy or in combination with etesevimab on viral load in patients with mild to moderate COVID-19: a randomized clinical trial. 10.1JAMA. 2021;325(7):632-644. doi:10.1001/jama.2021.0202

7. Weinreich DM, Sivapalasingam S, Norton T, et al. REGN-COV2, a neutralizing antibody cocktail, in outpatients with COVID-19. 10.1N Engl J Med. 2021;384:238-251. doi:10.1056/nejmoa2035002

8. Mulangu S, Dodd LE, Davey RT Jr, et al. A randomized, controlled trial of Ebola virus disease therapeutics. 10.1N Engl J Med. 2019;381:2293-2303. doi:10.1056/NEJMoa1910993

9. Boyle, P. Can an experimental treatment keep COVID-19 patients out of hospitals? Association of American Medical Colleges. January 29, 2021. Accessed March 9, 2021. https://www.aamc.org/news-insights/can-experimental-treatment-keep-covid-19-patients-out-hospitals

10. Fact Sheet for Health Care Providers Emergency Use Authorization (EUA) of Bamlanivimab and Etesevimab. US Food and Drug Administration. Updated February 2021. Accessed March 9, 2021. https://www.fda.gov/media/145802/download

Issue
Journal of Clinical Outcomes Management - 28(2)
Issue
Journal of Clinical Outcomes Management - 28(2)
Page Number
70-81
Page Number
70-81
Publications
Publications
Topics
Article Type
Display Headline
COVID-19 Monoclonal Antibody Infusions: A Multidisciplinary Initiative to Operationalize EUA Novel Treatment Options
Display Headline
COVID-19 Monoclonal Antibody Infusions: A Multidisciplinary Initiative to Operationalize EUA Novel Treatment Options
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
Article PDF Media

Pharmacists’ Bleed Risk Tool and Treatment Preferences Prior to Initiating Anticoagulation in Patients With Nonvalvular Atrial Fibrillation: A Cross-Sectional Survey

Article Type
Changed
Mon, 02/01/2021 - 07:52
Display Headline
Pharmacists’ Bleed Risk Tool and Treatment Preferences Prior to Initiating Anticoagulation in Patients With Nonvalvular Atrial Fibrillation: A Cross-Sectional Survey

From Nova Southeastern University College of Pharmacy, Fort Lauderdale, FL.

Abstract

  • Objective: To determine pharmacists’ preferences in bleed risk tool (BRT) usage and gastroprotection when bleed risk was lower than or equal to stroke risk in patients with nonvalvular atrial fibrillation and who were candidates for oral anticoagulation therapy (warfarin or direct oral anticoagulants [DOACs]).
  • Methods: A survey consisting of 4 domains (demographics, clinical experience, BRT usage, and treatment preferences based on cases where bleed risk was lower than or equal to stroke risk) was developed. The anonymous survey was disseminated via REDCap software to members of the American College of Clinical Pharmacy ambulatory care and cardiology Practice-based Research Networks. Descriptive statistics were calculated for all study variables and inferential statistics were employed as necessary.
  • Results: Of 165 BRT users, 97% preferred HAS-BLED. When bleed risk was lower than stroke risk, 151 respondents chose either DOACs (65%) or warfarin (35%); 15% added gastroprotection. When bleed risk was equal to stroke risk, 141 respondents chose DOACs (50%), warfarin (45%), or aspirin (5%); 40% added gastroprotection.
  • Conclusion: In addition to BRT usage, pharmacists were judicious in their recommendation to add gastroprotection and would consider doing so if there was a specific indication. As more than 80% of extracranial bleeds are gastrointestinal bleeds and most BRTs are nonspecific for predicting these bleeds, randomized, prospective studies stratified by HAS-BLED and stroke risk scores are needed to provide further guidance on the efficacy and safety of oral anticoagulation therapy with or without gastroprotection.

Keywords: NVAF; gastroprotection; proton pump inhibitors; warfarin; oral anticoagulants.

Management of patients with nonvalvular atrial fibrillation (NVAF) with oral anticoagulation therapy (OACT) requires constant attention to maintain a balance between preventing strokes and minimizing bleeds. Several validated bleed risk tools (BRTs) available for use in NVAF patients include HAS-BLED, HEMORR2HAGES, ATRIA, and mOBRI.1,2 A high bleed risk score is not a contraindication to OACT, but, prior to and throughout therapy, bleed risk should be assessed and modifiable risk factors addressed.3 While intraluminal gastrointestinal (GI) bleeds are not considered a critical bleed site, they are a common complication of chronic OACT and can result in hemodynamic compromise and permanent discontinuation of therapy.4,5 In 3233 patients with nonvariceal upper GI bleeds (2005-2016), the adjusted odds ratio of hospital admission, transfusion, and re-bleeding while on OACT (warfarin, heparin, or apixaban) was 3.48, 2.53, and 2.26, respectively.6 Addition of acid-suppressive therapy with a proton pump inhibitor (PPI) or histamine-2 receptor antagonist (H2RA) in NVAF patients at increased risk for upper GI bleeds and receiving OACT may result in fewer bleeds.7,8

Pharmacists play an integral part in managing patients on warfarin,9-11 and data on their role in managing patients receiving direct oral anticoagulants (DOACs) are increasing.12-16 Inpatient pharmacists actively participate in multidisciplinary collaborative teams and use clinical decision-support systems or enhanced monitoring to ensure safe prescribing of high-risk medications.12,15,16 Pharmacist-managed, outpatient-based anticoagulation services in patients on warfarin were associated with lower rates of bleeding and thromboembolic events and lower health care utilization versus routine care.17 However, it is unclear how pharmacists manage patients who are candidates for OACT but who may be at increased risk for upper GI bleeds. Using a US-based survey, the investigators sought to determine pharmacists’ preferences in BRT usage and gastroprotection when bleed risk was lower than or equal to stroke risk.

 

 

Methods

This cross-sectional study was conducted after receiving approval by Nova Southeastern University’s Institutional Review Board. The survey consisted of 16 items divided into 4 domains: demographics, clinical experience, use of BRTs, and treatment preferences based on cases where bleed risk was lower than or equal to stroke risk (Figure 1). Queries were multiple choice and allowed for free-text input when “Other” was selected. Licensed pharmacists ≥ 18 years of age who routinely provided care to patients with NVAF were eligible to participate in the study. Participants who reported using a BRT (users) completed all study domains, while participants who reported not using a BRT (nonusers) completed domains 1 through 3 only.

Survey tool

An invitation containing the survey link was sent to the American College of Clinical Pharmacy ambulatory care (n = 2237) and cardiology (n = 1318) pharmacists listed in the organization’s Practice-based Research Networks. The survey was administered in the United States between April and June 2016 via Research Electronic Data Capture (REDCap) software, a secure Web application for building and managing online surveys designed to support data collection for research studies.18

Survey responses were downloaded, and data were analyzed using NCSS 2019 Statistical Software, LLC (Kaysville, UT). Descriptive statistics were calculated for all study variables. Demographic and clinical experience data for the group that used a BRT versus the group that did not were compared using Pearson’s chi-square, ANOVA, or the Cochran-Armitage test for trends. Logistic regression with hierarchical forward selection with switching was used to identify predictors of drug selection and use of gastroprotection.

Results

Of 230 respondents who completed the survey (response rate 6.5%), 165 (72%) used a BRT and 65 (28%) did not. No significant differences were found for age, gender, duration in clinical practice, the percentage of time spent in patient care, or practice specialty between users and nonusers (Table). The median age of users was 32 years; 68% were females; the median duration in clinical practice was 6 years; 75% of their time was spent in clinical practice; and clinical settings included ambulatory care, cardiology, and internal medicine. A significant difference was found for practice region between users versus nonusers (P = 0.014). Respondents who managed more than 200 NVAF patients per year used a BRT more often than those who managed fewer than 100 NVAF patients per year (P = 0.001).

Respondent Demographics

Of those who used a BRT, 97% utilized the HAS-BLED tool (n = 160). The remainder used HEMORR2HAGES (n = 3), ATRIA (n = 1), and mOBRI (n = 1). Reasons for choosing HAS-BLED included “familiarity/ease-of-use,” “preference by institution/clinical team,” and the fact that it was a “validated tool for NVAF.”

When bleed risk was lower than stroke risk, 151 of 165 users (92%) chose a treatment option (Figure 2). Of those, 65% chose a DOAC and 35% chose warfarin. Fourteen respondents chose “other” and explained that they “would initiate OACT after weighing patient factors and preferences.” When a DOAC was selected, 9% (n = 9) chose PPI co-therapy and 4% (n = 4) chose a H2RA. When warfarin was selected, 13% (n = 7) chose PPI co-therapy and 4% (n = 2) chose a H2RA. Respondents who chose gastroprotection did not provide reasons for doing so, but those who did not add it explained that they “would add gastroprotection only if patient is also on an NSAID or has a history of GI bleed” or cited “patient preference.” Specific to warfarin, some respondents would not add gastroprotection, as anticoagulation with warfarin is “easily reversed.”

Pharmacists’ treatment preferences if bleed risk is less than stroke risk (n = 151)

When bleed risk was equal to stroke risk, 141 of 165 users (85%) chose a treatment option (Figure 3). Fifty percent chose DOACs, 45% chose warfarin, and 5% chose aspirin. Logistic regression analysis (outcome DOAC versus warfarin area-under-ROC curve, 0.67) showed that as the number of NVAF patients seen in 12 months increased, respondents were more likely to select a DOAC over warfarin (odds ratio, 1.7; 95% CI, 1.1-2.5). Therefore, for every 50-patient increase per year, the probability of recommending a DOAC increased 1.7-fold.

 Pharmacists’ treatment preferences if bleed risk is equal to stroke risk (n = 141)

Of respondents who selected either a DOAC or warfarin, 38% (n = 50) also added gastroprotection (Figure 3). When a DOAC was selected, 34% (n = 24) favored PPI co-therapy and 7% (n = 5) chose a H2RA. When warfarin was selected, 19% (n = 12) favored PPI co-therapy, while 13% (n = 8) chose a H2RA. Rationale for choosing gastroprotection, regardless of OACT selection, included “stroke is more devastating, so if patient wants to continue treatment, but knew risks of bleeding were similar, would recommend gastroprotection to help minimize bleeding risk” and “patient-specific consideration.” Rationales for not choosing gastroprotection included “would add gastroprotection only if patient is on dual antiplatelet therapy or has another indication”; “in most patients, stroke risk outweighs bleed risk so no need for gastroprotection unless there is a stated reason”; “would use apixaban as has lowest bleeding rate of all DOACs in clinical trials”; and “gastroprotection has not been shown to be beneficial in large scale trials.” 

Eight respondents chose aspirin because it was “easy and relatively low cost.” Twenty-four respondents chose “other” and explained that the choice of OACT depended on patient preference after they had discussed stroke and bleed risk with the patient and/or determined the etiology driving bleed risk.

 

 

Discussion

This is the first national survey exploring US pharmacists’ preferences in BRT usage and treatment based on bleed risk. Pharmacists preferred the HAS-BLED tool and considered patient-specific factors and evidence-based data when weighing the risk-benefit of OACT with or without gastroprotective therapy.

Similar to our findings, where three-quarters of pharmacists used a BRT, a recent Medscape/American College of Cardiology (ACC) survey reported that 74% of cardiologists used a BRT (eg, HAS-BLED) always/most of the time or sometimes to assess a patient’s overall risk of bleeding prior to initiating DOAC therapy; 27% never or rarely used a bleed risk score before prescribing DOACs.19 Although reasons for BRT preference were not provided, they may be similar to those reported by our respondents (ie, familiarity/ease-of-use). In both surveys, rationales for not using a BRT were not obtained, but possible reasons include lack of confidence with bleed risk calculators,20 inconsistent implementation of comprehensive assessments (stroke risk, bleed risk, and medication-related issues prior to decision-making),21 and nonspecific guideline recommendations.22

More recently, a network meta-analysis found that HAS-BLED and HEMORR2HAGES had modest but balanced sensitivity (defined as the ratio between the number of major bleeding events in high-risk stratification and the total number of bleeding events) and specificity (defined as the ratio between the number of nonmajor bleeding events in the low-risk population and total nonbleeding events) for predicting major bleeding events.2,3 Several respondents did comment that, although HAS-BLED was imprecise and only studied with warfarin, it was necessary to identify bleed risk in a patient starting a high-risk medication, and that the ACC anticoagulation application uses HAS-BLED with CHA2DS2VASc along with clinical trial data to estimate stroke risk and bleed risk, with projected risk reduction (strokes) and risk increases (bleeds) expected with each treatment (www.acc.org/tools-and-practice-support/mobile-resources/features/anticoag-evaluator). The 2019 AHA/ACC/HRS atrial fibrillation guideline recommends that HAS-BLED scores be used to assess bleed risk in patients for whom anticoagulation is being considered, and that the need for and choice of OACT should be periodically reevaluated to reassess stroke and bleed risks.23

Although more than 80% of extracranial bleeds are GI bleeds,24 most BRTs are nonspecific for predicting GI bleeds. Indeed, one respondent used a spreadsheet with several BRTs to maximize treatment guidance for patients with multiple risk factors for strokes and bleeds. A comprehensive approach to determining factors that increase bleed risk should be adopted. These factors include age (HAS-BLED, HEMORR2HAGES, mOBRI, ATRIA); anemia (mOBRI, HEMORR2HAGES, ATRIA); hepatic/renal disease (HAS-BLED, HEMORR2HAGES, ATRIA, mOBRI); concomitant medications/alcohol use, including NSAIDs, corticosteroids, and antiplatelet therapy (HAS-BLED, HEMORR2HAGES); bleed history/rebleeding risk (HEMORR2HAGES, HAS-BLED, ATRIA); and GI bleeds (mOBRI).1,2 Additional risk factors for GI bleeds include being a tobacco smoker and/or being infected with Helicobacter pylori. A prospective cohort study that analyzed data from questionnaires completed by 99,359 individuals from the Copenhagen General Population Study reported that the multivariable adjusted hazard ratio for current smokers versus never smokers was 2.20 (95% CI, 1.84-2.62) for GI bleeds.25 Presence of H pylori should be investigated, with a subsequent eradication regimen implemented, as patients with warfarin-associated upper GI bleeds who were H pylori-positive had lower HAS-BLED scores versus those who were negative.26

When bleed risk was lower than stroke risk (eg, HAS-BLED < 3, CHA2DS2VASc ≥ 1), respondents appropriately initiated therapy with an OAC (predominantly apixaban); a small proportion also added gastroprotection. If the patient did not have any other GI bleed risk factors (eg, a previous GI bleed or on chronic antiplatelet or NSAID therapy), the choice of OACT depended on the attributes of each OAC and patient preference.27 Selection of warfarin was appropriate if cost, formulary restrictions, and availability of an inexpensive reversal agent were important concerns to patients and/or their health care providers. Rivaroxaban was selected because of its once-daily dosing and low risk for GI bleeding.

 

 

The recently published ARISTOPHANES study provides evidence that apixaban is an appropriate choice in patients with a HAS-BLED score < 3. In this retrospective observational study, more than 70% of patients received standard doses of DOACs (apixaban 5 mg, dabigatran 150 mg, or rivaroxaban 20 mg) and about 20% had a bleeding history, about 30% were on PPIs, less than 25% were on NSAIDs, and about 40% had a HAS-BLED score < 3. The study found that apixaban was more effective (reduced rates of ischemic or hemorrhagic strokes/systemic embolism) and safer (reduced rates of major GI bleed or intracranial bleed) than warfarin.28 Dabigatran and rivaroxaban were also more effective than warfarin for stroke prevention and had a lower risk for major intracranial bleed risk; while the risk of major GI bleed was similar between dabigatran and warfarin, major GI bleed risk was higher for rivaroxaban. When compared with each other, the 3 DOACs were effective at stroke prevention, with apixaban more effective than dabigatran and rivaroxaban; similar efficacy was noted for dabigatran versus rivaroxaban. Apixaban was associated with fewer GI bleeds versus dabigatran and rivaroxaban, but with similar intracranial bleed risks; dabigatran was associated with fewer GI bleeds but similar intracranial bleed risks versus rivaroxaban.28 Efficacy and safety findings from a subgroup analysis based on HAS-BLED scores < 3 and ≥ 3 were generally consistent with the main results.

When bleed risk was equal to stroke risk, the difficulty was determining how OACT in a patient at high stroke risk (CHA2DS2VASc score ≥ 2) and high bleed risk (HAS-BLED score ≥ 3) should be managed. Eight respondents chose aspirin and added gastroprotection with either a PPI or H2RA; however, currently, aspirin is not recommended as the sole antithrombotic for patients with NVAF.23 With the OACT, an interesting finding was that as the number of patients seen in 12 months increased, pharmacists were almost twice as likely to select a DOAC over warfarin. Moreover, pharmacists were judicious in their recommendation to add gastroprotection, and would consider doing so if there was a specific indication. At the time of our survey, several studies described DOAC-associated GI bleeds,29-31 but data on the effectiveness of acid-suppressive therapy, specifically with PPIs, in the prevention of upper GI bleeds were sparse.4,7,32 Respondents most likely were familiar with GI bleed risk factors and prevention strategies from various guidelines published between 2009 and 2010, which did not include DOACs.33-35

Another important finding was pharmacists’ uncertainty as to the effectiveness of PPIs in preventing GI bleeds in combination with DOACs. The data are conflicting. A meta-analysis of older studies (2007-2015) showed that PPIs (but not H2RAs) reduced the risk of upper GI bleeds in patients on warfarin but not for dabigatran.36 A retrospective cohort study of Medicare beneficiaries on OACTs (2011-2015) showed the adjusted incidence of hospitalization for upper GI bleeds in patients on PPI co-therapy was significantly lower compared with patients not on PPI co-therapy (76 versus 115 per 10,000 person-years, respectively).8 Apixaban without PPI co-therapy was associated with the lowest risk of upper GI bleed hospitalizations (73/10,000 person-years), and PPI co-therapy further reduced this risk (49/10,000 person-years). Warfarin without PPI co-therapy was associated with the next lowest risk (113/10,000 person-years), followed by dabigatran (120/10,000 person-years) and rivaroxaban (144/10,000 person-years). PPI co-therapy significantly reduced the risk of upper GI bleed hospitalizations with all OACTs, but the incidence of upper GI bleed hospitalizations with rivaroxaban was significantly greater than with the other OACTs.8 Therefore, if there are concerns about the safety of PPIs,37-39 or the patient is unable to tolerate a PPI, then apixaban may be the most appropriate DOAC for a patient with high bleed risk. Notably, a 2020 review of data from the PINNACLE registry (average age, 75-77 years; 31% on PPIs) found that the relative GI bleed safety advantage of apixaban and dabigatran versus warfarin was attenuated in patients ≥ 75 years.40 Last, since the risk for lower GI bleeds is not reduced by PPIs,41 consideration of their use should be accompanied by an assessment to detect bleeds (eg, low hemoglobin/hematocrit, presence of bright red blood, hematochezia/melena, fecal occult testing), with prompt management as necessary.5

Limitations

Limitations of our survey included an overall low response rate, which can generate a biased sample if respondents are systematically different from nonrespondents. In addition, to maintain simplicity and reduce respondents’ time commitment, our survey did not include actual CHA2DS2VASc stroke risk scores, HAS-BLED bleed risk scores, or specific GI bleed risk factors when querying pharmacists about treatment options based on bleed risk. The addition of these variables would have improved the robustness of the data.

Conclusion

In addition to applying BRTs in the management of NVAF patients, pharmacists considered patient-specific variables, prescriber preferences, and evidence-based guidance when recommending OACT with or without gastroprotection. To avoid suboptimal patient management, busy pharmacists should be granted time to attend continuing education programs describing optimal OACT selection and formulation of individualized, evidence-based plans to address modifiable risk factors for bleeding, including the appropriate use of gastroprotection. Randomized, prospective, long-term studies stratified by HAS-BLED and CHA2DS2VASc scores are needed to further clarify efficacy, safety, and cost-effectiveness of OACT, with and without PPIs, in patients who may be at risk for upper GI bleeds.

Acknowledgments: The authors thank Robin J. Jacobs, PhD, MSW, MS, MPH, Patrick C. Hardigan, PhD, Steven Brettler, PharmD, MPH, Maria-Isabel A. Cabral, PharmD, and Reginald Gyapong, PharmD, for their participation in this project. The authors also sincerely thank Fabio Franco, BS Computer Science, who organized the database to enable efficient data management.

Corresponding author: Devada Singh-Franco, PharmD, CDE, Nova Southeastern University College of Pharmacy, 3200 S University Drive, Fort Lauderdale, FL 33328; singh@nova.edu

Disclosures: None.

Funding: The study was supported by Nova Southeastern University’s Health Professions Division Internal Research Grant.

References

1. Apostolakis S, Lane DA, Guo Y, et al. Performance of the HEMORR2HAGES, ATRIA, and HAS-BLED bleeding risk–prediction scores in patients with atrial fibrillation undergoing anticoagulation. J Am Coll Cardiol. 2012;60:861-867.

2. Chang G, Xie Q, Ma L, et al. Accuracy of HAS-BLED and other bleeding risk assessment tools in predicting major bleeding events in atrial fibrillation: A network meta-analysis. J Thromb Haemost. 2020;18:791-801.

3. Ding WY, Harrison SL, Lane DA, Lip GYH. Considerations when choosing an appropriate bleeding risk assessment tool for patients with atrial fibrillation. J Thromb Haemost. 2020;18:788-790.

4. Lauffenburger JC, Rhoney DH, Farley JF, et al. Predictors of gastrointestinal bleeding among patients with atrial fibrillation after initiating dabigatran therapy. Pharmacotherapy. 2015;35:560-568.

5. Tomaselli GF, Mahaffey KW, Cuker A, et al. 2020 ACC Expert Consensus Decision Pathway on Management of Bleeding in Patients on Oral Anticoagulants: A Report of the American College of Cardiology Solution Set Oversight Committee. J Am Coll Cardiol. 2020;76:594-622.

6. Taha A, McCloskey C, Craigen T, Angerson W. Antiplatelet versus anticoagulant effects in non-variceal upper gastrointestinal bleeding. Gut. 2019;68(suppl 2):A152.

7. Chan EW, Lau WC, Leung WK, et al. Prevention of dabigatran-related gastrointestinal bleeding with gastroprotective agents: A population-based study. Gastroenterology. 2015;149:586-595.

8. Ray WA, Chung CP, Murray KT, et al. Association of oral anticoagulants and proton pump inhibitor cotherapy with hospitalization for upper gastrointestinal tract bleeding. JAMA. 2018;320:2221-2230.

9. Brunetti L, Lee S-M, Doherty N, et al. Impact of warfarin discharge education program on hospital readmission and treatment costs. Int J Clin Pharm. 2018;40:721-729.

10. Hasan SS, Kow CS, Curley LE, et al. Economic evaluation of prescribing conventional and newer oral anticoagulants in older adults. Expert Rev Pharmacoecon Outcomes Res. 2018;18:371-377.

11. Phelps E, Delate T, Witt DM, et al. Effect of increased time in the therapeutic range on atrial fibrillation outcomes within a centralized anticoagulation service. Thromb Res. 2018;163:54-59.

12. Ahuja T, Raco V, Papadopoulos J, Green D. Antithrombotic stewardship: Assessing use of computerized clinical decision support tools to enhance safe prescribing of direct oral anticoagulants in hospitalized patients. J Patient Saf. 2018 Sep 25. [Epub ahead of print]

13. Leef GC, Perino AC, Askari M, et al. Appropriateness of direct oral anticoagulant dosing in patients with atrial fibrillation: Insights from the Veterans Health Administration. J Pharm Pract. 2020;33:647-653.

14. Papastergiou J, Kheir N, Ladova K, et al. Pharmacists’ confidence when providing pharmaceutical care on anticoagulants, a multinational survey. Int J Clin Pharm. 2017;39:1282-1290.

15. Perlman A, Horwitz E, Hirsh-Raccah B, et al. Clinical pharmacist led hospital-wide direct oral anticoagulant stewardship program. Isr J Health Policy Res. 2019;8:19.

16. Uppuluri EM, McComb MN, Shapiro NL. Implementation of a direct oral anticoagulation screening service at a large academic medical center provided by a pharmacist-managed antithrombosis clinic as a method to expand antithrombotic stewardship efforts. J Pharm Pract. 2020;33:271-275.

17. Manzoor BS, Cheng W-H, Lee JC, et al. Quality of pharmacist-managed anticoagulation therapy in long-term ambulatory settings: A systematic review. Ann Pharmacother. 2017;51:1122-1137.

18. Harris PA, Taylor R, Thielke R, et al. Research Electronic Data Capture (REDCap)—A metadata-driven methodology and workflow process for providing translational research informatics support. J Biomed Inform. 2009;42:377-381.

19. Brooks M. AF management: Are clinicians in agreement? Medscape. May 30, 2019. Accessed December 29, 2020. https://www.medscape.com/viewarticle/913386

20. Amroze A, Mazor K, Crawford S, et al. Survey of confidence in use of stroke and bleeding risk calculators, knowledge of anticoagulants, and comfort with prescription of anticoagulation in challenging scenarios: SUPPORT-AF II study. J Thromb Thrombolysis. 2019;48:629-637.

21. Wang Y, Bajorek B. Decision-making around antithrombotics for stroke prevention in atrial fibrillation: the health professionals’ views. Int J Clin Pharm. 2016;38:985-995.

22. January CT, Wann LS, Alpert JS, et al. 2014 AHA/ACC/HRS guideline for the management of patients with atrial fibrillation: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines and the Heart Rhythm Society. Circulation. 2014;130:e199-e267.

23. January CT, Wann LS, Calkins H, et al. 2019 AHA/ACC/HRS Focused Update of the 2014 AHA/ACC/HRS Guideline for the Management of Patients With Atrial Fibrillation: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines and the Heart Rhythm Society. J Am Coll Cardiol. 2019;74:104-132.

24. Anghel L, Sascu R, Trifan A, et al. Non-vitamin K antagonist oral anticoagulants and the gastrointestinal bleeding risk in real-world studies. J Clin Med. 2020;9:1398.

25. Langsted A, Nordestgaard BG. Smoking is associated with increased risk of major bleeding: a prospective cohort study. Thromb Haemost. 2019;119:39-47.

26. Faye AS, Hung KW, Cheng K, et al. HAS-BLED scores underestimate gastrointestinal bleeding risk among those with H. pylori. Am J Gastroenterol. 2019;114:S364.

27. Fawzy AM, Yang W-Y, Lip GY. Safety of direct oral anticoagulants in real-world clinical practice: translating the trials to everyday clinical management. Expert Opin Drug Saf. 2019;18:187-209.

28. Lip GYH, Keshishian A, Li X, et al. Effectiveness and safety of oral anticoagulants among nonvalvular atrial fibrillation patients. Stroke. 2018;49:2933-2944.

29. Abraham NS, Singh S, Alexander GC, et al. Comparative risk of gastrointestinal bleeding with dabigatran, rivaroxaban, and warfarin: population based cohort study. BMJ. 2015;350:h1857.

30. Holster IL, Valkhoff VE, Kuipers EJ, Tjwa E. New oral anticoagulants increase risk for gastrointestinal bleeding: a systematic review and meta-analysis. Gastroenterology. 2013;145:105-112.

31. Sherwood MW, Nessel CC, Hellkamp AS, et al. Gastrointestinal bleeding in patients with atrial fibrillation treated with rivaroxaban or warfarin: ROCKET AF Trial. J Am Coll Cardiol. 2015;66:2271-2281.

32. Di Minno A, Spadarella G, Spadarella E, et al. Gastrointestinal bleeding in patients receiving oral anticoagulation: Current treatment and pharmacological perspectives. Thromb Res. 2015;136:1074-1081.

33. Abraham NS, Hlatky MA, Antman EM, et al. ACCF/ACG/AHA 2010 Expert Consensus Document on the Concomitant Use of Proton Pump Inhibitors and Thienopyridines: A Focused Update of the ACCF/ACG/AHA 2008 Expert Consensus Document on Reducing the Gastrointestinal Risks of Antiplatelet Therapy and NSAID Use. Circulation. 2010;122:2619-2633.

34. Bhatt DL, Scheiman J, Abraham NS, et al. ACCF/ACG/AHA 2008 expert consensus document on reducing the gastrointestinal risks of antiplatelet therapy and NSAID use: a report of the American College of Cardiology Foundation Task Force on Clinical Expert Consensus Documents. J Am Coll Cardiol. 2008;52:1502-1517.

35. Lanza FL, Chan FK, Quigley EM. Guidelines for prevention of NSAID-related ulcer complications. Am J Gastroenterol. 2009;104:728-738.

36. Bang CS, Joo MK, Kim BW, et al. The role of acid suppressants in the prevention of anticoagulant-related gastrointestinal bleeding: a systematic review and meta-analysis. Gut Liver. 2020;14:57-66.

37. Farrell B, Pottie K, Thompson W, et al. Deprescribing proton pump inhibitors: Evidence-based clinical practice guideline. Can Fam Physician. 2017;63:354-364.

38. Fossmark R, Martinsen TC, Waldum HL. Adverse effects of proton pump inhibitors—evidence and plausibility. Int J Mol Sci. 2019;20:5203.

39. Haastrup PF, Thompson W, Sondergaard J, Jarbol DE. Side effects of long-term proton pump inhibitor use: A review. Basic Clin Pharmacol Toxicol. 2018;123:114-121.

40. Wong JM, Maddox TM, Kennedy K, Shaw RE. Comparing major bleeding risk in outpatients with atrial fibrillation or flutter by oral anticoagulant type (from the National Cardiovascular Disease Registry’s Practice Innovation and Clinical Excellence Registry). Am J Cardiol. 2020;125:1500-1507.

41. Nagata N, Niikura R, Aoki T, et al. Effect of proton-pump inhibitors on the risk of lower gastrointestinal bleeding associated with NSAIDs, aspirin, clopidogrel, and warfarin. J Gastroenterol. 2015;50:1079-1086.

Article PDF
Issue
Journal of Clinical Outcomes Management - 28(1)
Publications
Topics
Page Number
9-16
Sections
Article PDF
Article PDF

From Nova Southeastern University College of Pharmacy, Fort Lauderdale, FL.

Abstract

  • Objective: To determine pharmacists’ preferences in bleed risk tool (BRT) usage and gastroprotection when bleed risk was lower than or equal to stroke risk in patients with nonvalvular atrial fibrillation and who were candidates for oral anticoagulation therapy (warfarin or direct oral anticoagulants [DOACs]).
  • Methods: A survey consisting of 4 domains (demographics, clinical experience, BRT usage, and treatment preferences based on cases where bleed risk was lower than or equal to stroke risk) was developed. The anonymous survey was disseminated via REDCap software to members of the American College of Clinical Pharmacy ambulatory care and cardiology Practice-based Research Networks. Descriptive statistics were calculated for all study variables and inferential statistics were employed as necessary.
  • Results: Of 165 BRT users, 97% preferred HAS-BLED. When bleed risk was lower than stroke risk, 151 respondents chose either DOACs (65%) or warfarin (35%); 15% added gastroprotection. When bleed risk was equal to stroke risk, 141 respondents chose DOACs (50%), warfarin (45%), or aspirin (5%); 40% added gastroprotection.
  • Conclusion: In addition to BRT usage, pharmacists were judicious in their recommendation to add gastroprotection and would consider doing so if there was a specific indication. As more than 80% of extracranial bleeds are gastrointestinal bleeds and most BRTs are nonspecific for predicting these bleeds, randomized, prospective studies stratified by HAS-BLED and stroke risk scores are needed to provide further guidance on the efficacy and safety of oral anticoagulation therapy with or without gastroprotection.

Keywords: NVAF; gastroprotection; proton pump inhibitors; warfarin; oral anticoagulants.

Management of patients with nonvalvular atrial fibrillation (NVAF) with oral anticoagulation therapy (OACT) requires constant attention to maintain a balance between preventing strokes and minimizing bleeds. Several validated bleed risk tools (BRTs) available for use in NVAF patients include HAS-BLED, HEMORR2HAGES, ATRIA, and mOBRI.1,2 A high bleed risk score is not a contraindication to OACT, but, prior to and throughout therapy, bleed risk should be assessed and modifiable risk factors addressed.3 While intraluminal gastrointestinal (GI) bleeds are not considered a critical bleed site, they are a common complication of chronic OACT and can result in hemodynamic compromise and permanent discontinuation of therapy.4,5 In 3233 patients with nonvariceal upper GI bleeds (2005-2016), the adjusted odds ratio of hospital admission, transfusion, and re-bleeding while on OACT (warfarin, heparin, or apixaban) was 3.48, 2.53, and 2.26, respectively.6 Addition of acid-suppressive therapy with a proton pump inhibitor (PPI) or histamine-2 receptor antagonist (H2RA) in NVAF patients at increased risk for upper GI bleeds and receiving OACT may result in fewer bleeds.7,8

Pharmacists play an integral part in managing patients on warfarin,9-11 and data on their role in managing patients receiving direct oral anticoagulants (DOACs) are increasing.12-16 Inpatient pharmacists actively participate in multidisciplinary collaborative teams and use clinical decision-support systems or enhanced monitoring to ensure safe prescribing of high-risk medications.12,15,16 Pharmacist-managed, outpatient-based anticoagulation services in patients on warfarin were associated with lower rates of bleeding and thromboembolic events and lower health care utilization versus routine care.17 However, it is unclear how pharmacists manage patients who are candidates for OACT but who may be at increased risk for upper GI bleeds. Using a US-based survey, the investigators sought to determine pharmacists’ preferences in BRT usage and gastroprotection when bleed risk was lower than or equal to stroke risk.

 

 

Methods

This cross-sectional study was conducted after receiving approval by Nova Southeastern University’s Institutional Review Board. The survey consisted of 16 items divided into 4 domains: demographics, clinical experience, use of BRTs, and treatment preferences based on cases where bleed risk was lower than or equal to stroke risk (Figure 1). Queries were multiple choice and allowed for free-text input when “Other” was selected. Licensed pharmacists ≥ 18 years of age who routinely provided care to patients with NVAF were eligible to participate in the study. Participants who reported using a BRT (users) completed all study domains, while participants who reported not using a BRT (nonusers) completed domains 1 through 3 only.

Survey tool

An invitation containing the survey link was sent to the American College of Clinical Pharmacy ambulatory care (n = 2237) and cardiology (n = 1318) pharmacists listed in the organization’s Practice-based Research Networks. The survey was administered in the United States between April and June 2016 via Research Electronic Data Capture (REDCap) software, a secure Web application for building and managing online surveys designed to support data collection for research studies.18

Survey responses were downloaded, and data were analyzed using NCSS 2019 Statistical Software, LLC (Kaysville, UT). Descriptive statistics were calculated for all study variables. Demographic and clinical experience data for the group that used a BRT versus the group that did not were compared using Pearson’s chi-square, ANOVA, or the Cochran-Armitage test for trends. Logistic regression with hierarchical forward selection with switching was used to identify predictors of drug selection and use of gastroprotection.

Results

Of 230 respondents who completed the survey (response rate 6.5%), 165 (72%) used a BRT and 65 (28%) did not. No significant differences were found for age, gender, duration in clinical practice, the percentage of time spent in patient care, or practice specialty between users and nonusers (Table). The median age of users was 32 years; 68% were females; the median duration in clinical practice was 6 years; 75% of their time was spent in clinical practice; and clinical settings included ambulatory care, cardiology, and internal medicine. A significant difference was found for practice region between users versus nonusers (P = 0.014). Respondents who managed more than 200 NVAF patients per year used a BRT more often than those who managed fewer than 100 NVAF patients per year (P = 0.001).

Respondent Demographics

Of those who used a BRT, 97% utilized the HAS-BLED tool (n = 160). The remainder used HEMORR2HAGES (n = 3), ATRIA (n = 1), and mOBRI (n = 1). Reasons for choosing HAS-BLED included “familiarity/ease-of-use,” “preference by institution/clinical team,” and the fact that it was a “validated tool for NVAF.”

When bleed risk was lower than stroke risk, 151 of 165 users (92%) chose a treatment option (Figure 2). Of those, 65% chose a DOAC and 35% chose warfarin. Fourteen respondents chose “other” and explained that they “would initiate OACT after weighing patient factors and preferences.” When a DOAC was selected, 9% (n = 9) chose PPI co-therapy and 4% (n = 4) chose a H2RA. When warfarin was selected, 13% (n = 7) chose PPI co-therapy and 4% (n = 2) chose a H2RA. Respondents who chose gastroprotection did not provide reasons for doing so, but those who did not add it explained that they “would add gastroprotection only if patient is also on an NSAID or has a history of GI bleed” or cited “patient preference.” Specific to warfarin, some respondents would not add gastroprotection, as anticoagulation with warfarin is “easily reversed.”

Pharmacists’ treatment preferences if bleed risk is less than stroke risk (n = 151)

When bleed risk was equal to stroke risk, 141 of 165 users (85%) chose a treatment option (Figure 3). Fifty percent chose DOACs, 45% chose warfarin, and 5% chose aspirin. Logistic regression analysis (outcome DOAC versus warfarin area-under-ROC curve, 0.67) showed that as the number of NVAF patients seen in 12 months increased, respondents were more likely to select a DOAC over warfarin (odds ratio, 1.7; 95% CI, 1.1-2.5). Therefore, for every 50-patient increase per year, the probability of recommending a DOAC increased 1.7-fold.

 Pharmacists’ treatment preferences if bleed risk is equal to stroke risk (n = 141)

Of respondents who selected either a DOAC or warfarin, 38% (n = 50) also added gastroprotection (Figure 3). When a DOAC was selected, 34% (n = 24) favored PPI co-therapy and 7% (n = 5) chose a H2RA. When warfarin was selected, 19% (n = 12) favored PPI co-therapy, while 13% (n = 8) chose a H2RA. Rationale for choosing gastroprotection, regardless of OACT selection, included “stroke is more devastating, so if patient wants to continue treatment, but knew risks of bleeding were similar, would recommend gastroprotection to help minimize bleeding risk” and “patient-specific consideration.” Rationales for not choosing gastroprotection included “would add gastroprotection only if patient is on dual antiplatelet therapy or has another indication”; “in most patients, stroke risk outweighs bleed risk so no need for gastroprotection unless there is a stated reason”; “would use apixaban as has lowest bleeding rate of all DOACs in clinical trials”; and “gastroprotection has not been shown to be beneficial in large scale trials.” 

Eight respondents chose aspirin because it was “easy and relatively low cost.” Twenty-four respondents chose “other” and explained that the choice of OACT depended on patient preference after they had discussed stroke and bleed risk with the patient and/or determined the etiology driving bleed risk.

 

 

Discussion

This is the first national survey exploring US pharmacists’ preferences in BRT usage and treatment based on bleed risk. Pharmacists preferred the HAS-BLED tool and considered patient-specific factors and evidence-based data when weighing the risk-benefit of OACT with or without gastroprotective therapy.

Similar to our findings, where three-quarters of pharmacists used a BRT, a recent Medscape/American College of Cardiology (ACC) survey reported that 74% of cardiologists used a BRT (eg, HAS-BLED) always/most of the time or sometimes to assess a patient’s overall risk of bleeding prior to initiating DOAC therapy; 27% never or rarely used a bleed risk score before prescribing DOACs.19 Although reasons for BRT preference were not provided, they may be similar to those reported by our respondents (ie, familiarity/ease-of-use). In both surveys, rationales for not using a BRT were not obtained, but possible reasons include lack of confidence with bleed risk calculators,20 inconsistent implementation of comprehensive assessments (stroke risk, bleed risk, and medication-related issues prior to decision-making),21 and nonspecific guideline recommendations.22

More recently, a network meta-analysis found that HAS-BLED and HEMORR2HAGES had modest but balanced sensitivity (defined as the ratio between the number of major bleeding events in high-risk stratification and the total number of bleeding events) and specificity (defined as the ratio between the number of nonmajor bleeding events in the low-risk population and total nonbleeding events) for predicting major bleeding events.2,3 Several respondents did comment that, although HAS-BLED was imprecise and only studied with warfarin, it was necessary to identify bleed risk in a patient starting a high-risk medication, and that the ACC anticoagulation application uses HAS-BLED with CHA2DS2VASc along with clinical trial data to estimate stroke risk and bleed risk, with projected risk reduction (strokes) and risk increases (bleeds) expected with each treatment (www.acc.org/tools-and-practice-support/mobile-resources/features/anticoag-evaluator). The 2019 AHA/ACC/HRS atrial fibrillation guideline recommends that HAS-BLED scores be used to assess bleed risk in patients for whom anticoagulation is being considered, and that the need for and choice of OACT should be periodically reevaluated to reassess stroke and bleed risks.23

Although more than 80% of extracranial bleeds are GI bleeds,24 most BRTs are nonspecific for predicting GI bleeds. Indeed, one respondent used a spreadsheet with several BRTs to maximize treatment guidance for patients with multiple risk factors for strokes and bleeds. A comprehensive approach to determining factors that increase bleed risk should be adopted. These factors include age (HAS-BLED, HEMORR2HAGES, mOBRI, ATRIA); anemia (mOBRI, HEMORR2HAGES, ATRIA); hepatic/renal disease (HAS-BLED, HEMORR2HAGES, ATRIA, mOBRI); concomitant medications/alcohol use, including NSAIDs, corticosteroids, and antiplatelet therapy (HAS-BLED, HEMORR2HAGES); bleed history/rebleeding risk (HEMORR2HAGES, HAS-BLED, ATRIA); and GI bleeds (mOBRI).1,2 Additional risk factors for GI bleeds include being a tobacco smoker and/or being infected with Helicobacter pylori. A prospective cohort study that analyzed data from questionnaires completed by 99,359 individuals from the Copenhagen General Population Study reported that the multivariable adjusted hazard ratio for current smokers versus never smokers was 2.20 (95% CI, 1.84-2.62) for GI bleeds.25 Presence of H pylori should be investigated, with a subsequent eradication regimen implemented, as patients with warfarin-associated upper GI bleeds who were H pylori-positive had lower HAS-BLED scores versus those who were negative.26

When bleed risk was lower than stroke risk (eg, HAS-BLED < 3, CHA2DS2VASc ≥ 1), respondents appropriately initiated therapy with an OAC (predominantly apixaban); a small proportion also added gastroprotection. If the patient did not have any other GI bleed risk factors (eg, a previous GI bleed or on chronic antiplatelet or NSAID therapy), the choice of OACT depended on the attributes of each OAC and patient preference.27 Selection of warfarin was appropriate if cost, formulary restrictions, and availability of an inexpensive reversal agent were important concerns to patients and/or their health care providers. Rivaroxaban was selected because of its once-daily dosing and low risk for GI bleeding.

 

 

The recently published ARISTOPHANES study provides evidence that apixaban is an appropriate choice in patients with a HAS-BLED score < 3. In this retrospective observational study, more than 70% of patients received standard doses of DOACs (apixaban 5 mg, dabigatran 150 mg, or rivaroxaban 20 mg) and about 20% had a bleeding history, about 30% were on PPIs, less than 25% were on NSAIDs, and about 40% had a HAS-BLED score < 3. The study found that apixaban was more effective (reduced rates of ischemic or hemorrhagic strokes/systemic embolism) and safer (reduced rates of major GI bleed or intracranial bleed) than warfarin.28 Dabigatran and rivaroxaban were also more effective than warfarin for stroke prevention and had a lower risk for major intracranial bleed risk; while the risk of major GI bleed was similar between dabigatran and warfarin, major GI bleed risk was higher for rivaroxaban. When compared with each other, the 3 DOACs were effective at stroke prevention, with apixaban more effective than dabigatran and rivaroxaban; similar efficacy was noted for dabigatran versus rivaroxaban. Apixaban was associated with fewer GI bleeds versus dabigatran and rivaroxaban, but with similar intracranial bleed risks; dabigatran was associated with fewer GI bleeds but similar intracranial bleed risks versus rivaroxaban.28 Efficacy and safety findings from a subgroup analysis based on HAS-BLED scores < 3 and ≥ 3 were generally consistent with the main results.

When bleed risk was equal to stroke risk, the difficulty was determining how OACT in a patient at high stroke risk (CHA2DS2VASc score ≥ 2) and high bleed risk (HAS-BLED score ≥ 3) should be managed. Eight respondents chose aspirin and added gastroprotection with either a PPI or H2RA; however, currently, aspirin is not recommended as the sole antithrombotic for patients with NVAF.23 With the OACT, an interesting finding was that as the number of patients seen in 12 months increased, pharmacists were almost twice as likely to select a DOAC over warfarin. Moreover, pharmacists were judicious in their recommendation to add gastroprotection, and would consider doing so if there was a specific indication. At the time of our survey, several studies described DOAC-associated GI bleeds,29-31 but data on the effectiveness of acid-suppressive therapy, specifically with PPIs, in the prevention of upper GI bleeds were sparse.4,7,32 Respondents most likely were familiar with GI bleed risk factors and prevention strategies from various guidelines published between 2009 and 2010, which did not include DOACs.33-35

Another important finding was pharmacists’ uncertainty as to the effectiveness of PPIs in preventing GI bleeds in combination with DOACs. The data are conflicting. A meta-analysis of older studies (2007-2015) showed that PPIs (but not H2RAs) reduced the risk of upper GI bleeds in patients on warfarin but not for dabigatran.36 A retrospective cohort study of Medicare beneficiaries on OACTs (2011-2015) showed the adjusted incidence of hospitalization for upper GI bleeds in patients on PPI co-therapy was significantly lower compared with patients not on PPI co-therapy (76 versus 115 per 10,000 person-years, respectively).8 Apixaban without PPI co-therapy was associated with the lowest risk of upper GI bleed hospitalizations (73/10,000 person-years), and PPI co-therapy further reduced this risk (49/10,000 person-years). Warfarin without PPI co-therapy was associated with the next lowest risk (113/10,000 person-years), followed by dabigatran (120/10,000 person-years) and rivaroxaban (144/10,000 person-years). PPI co-therapy significantly reduced the risk of upper GI bleed hospitalizations with all OACTs, but the incidence of upper GI bleed hospitalizations with rivaroxaban was significantly greater than with the other OACTs.8 Therefore, if there are concerns about the safety of PPIs,37-39 or the patient is unable to tolerate a PPI, then apixaban may be the most appropriate DOAC for a patient with high bleed risk. Notably, a 2020 review of data from the PINNACLE registry (average age, 75-77 years; 31% on PPIs) found that the relative GI bleed safety advantage of apixaban and dabigatran versus warfarin was attenuated in patients ≥ 75 years.40 Last, since the risk for lower GI bleeds is not reduced by PPIs,41 consideration of their use should be accompanied by an assessment to detect bleeds (eg, low hemoglobin/hematocrit, presence of bright red blood, hematochezia/melena, fecal occult testing), with prompt management as necessary.5

Limitations

Limitations of our survey included an overall low response rate, which can generate a biased sample if respondents are systematically different from nonrespondents. In addition, to maintain simplicity and reduce respondents’ time commitment, our survey did not include actual CHA2DS2VASc stroke risk scores, HAS-BLED bleed risk scores, or specific GI bleed risk factors when querying pharmacists about treatment options based on bleed risk. The addition of these variables would have improved the robustness of the data.

Conclusion

In addition to applying BRTs in the management of NVAF patients, pharmacists considered patient-specific variables, prescriber preferences, and evidence-based guidance when recommending OACT with or without gastroprotection. To avoid suboptimal patient management, busy pharmacists should be granted time to attend continuing education programs describing optimal OACT selection and formulation of individualized, evidence-based plans to address modifiable risk factors for bleeding, including the appropriate use of gastroprotection. Randomized, prospective, long-term studies stratified by HAS-BLED and CHA2DS2VASc scores are needed to further clarify efficacy, safety, and cost-effectiveness of OACT, with and without PPIs, in patients who may be at risk for upper GI bleeds.

Acknowledgments: The authors thank Robin J. Jacobs, PhD, MSW, MS, MPH, Patrick C. Hardigan, PhD, Steven Brettler, PharmD, MPH, Maria-Isabel A. Cabral, PharmD, and Reginald Gyapong, PharmD, for their participation in this project. The authors also sincerely thank Fabio Franco, BS Computer Science, who organized the database to enable efficient data management.

Corresponding author: Devada Singh-Franco, PharmD, CDE, Nova Southeastern University College of Pharmacy, 3200 S University Drive, Fort Lauderdale, FL 33328; singh@nova.edu

Disclosures: None.

Funding: The study was supported by Nova Southeastern University’s Health Professions Division Internal Research Grant.

From Nova Southeastern University College of Pharmacy, Fort Lauderdale, FL.

Abstract

  • Objective: To determine pharmacists’ preferences in bleed risk tool (BRT) usage and gastroprotection when bleed risk was lower than or equal to stroke risk in patients with nonvalvular atrial fibrillation and who were candidates for oral anticoagulation therapy (warfarin or direct oral anticoagulants [DOACs]).
  • Methods: A survey consisting of 4 domains (demographics, clinical experience, BRT usage, and treatment preferences based on cases where bleed risk was lower than or equal to stroke risk) was developed. The anonymous survey was disseminated via REDCap software to members of the American College of Clinical Pharmacy ambulatory care and cardiology Practice-based Research Networks. Descriptive statistics were calculated for all study variables and inferential statistics were employed as necessary.
  • Results: Of 165 BRT users, 97% preferred HAS-BLED. When bleed risk was lower than stroke risk, 151 respondents chose either DOACs (65%) or warfarin (35%); 15% added gastroprotection. When bleed risk was equal to stroke risk, 141 respondents chose DOACs (50%), warfarin (45%), or aspirin (5%); 40% added gastroprotection.
  • Conclusion: In addition to BRT usage, pharmacists were judicious in their recommendation to add gastroprotection and would consider doing so if there was a specific indication. As more than 80% of extracranial bleeds are gastrointestinal bleeds and most BRTs are nonspecific for predicting these bleeds, randomized, prospective studies stratified by HAS-BLED and stroke risk scores are needed to provide further guidance on the efficacy and safety of oral anticoagulation therapy with or without gastroprotection.

Keywords: NVAF; gastroprotection; proton pump inhibitors; warfarin; oral anticoagulants.

Management of patients with nonvalvular atrial fibrillation (NVAF) with oral anticoagulation therapy (OACT) requires constant attention to maintain a balance between preventing strokes and minimizing bleeds. Several validated bleed risk tools (BRTs) available for use in NVAF patients include HAS-BLED, HEMORR2HAGES, ATRIA, and mOBRI.1,2 A high bleed risk score is not a contraindication to OACT, but, prior to and throughout therapy, bleed risk should be assessed and modifiable risk factors addressed.3 While intraluminal gastrointestinal (GI) bleeds are not considered a critical bleed site, they are a common complication of chronic OACT and can result in hemodynamic compromise and permanent discontinuation of therapy.4,5 In 3233 patients with nonvariceal upper GI bleeds (2005-2016), the adjusted odds ratio of hospital admission, transfusion, and re-bleeding while on OACT (warfarin, heparin, or apixaban) was 3.48, 2.53, and 2.26, respectively.6 Addition of acid-suppressive therapy with a proton pump inhibitor (PPI) or histamine-2 receptor antagonist (H2RA) in NVAF patients at increased risk for upper GI bleeds and receiving OACT may result in fewer bleeds.7,8

Pharmacists play an integral part in managing patients on warfarin,9-11 and data on their role in managing patients receiving direct oral anticoagulants (DOACs) are increasing.12-16 Inpatient pharmacists actively participate in multidisciplinary collaborative teams and use clinical decision-support systems or enhanced monitoring to ensure safe prescribing of high-risk medications.12,15,16 Pharmacist-managed, outpatient-based anticoagulation services in patients on warfarin were associated with lower rates of bleeding and thromboembolic events and lower health care utilization versus routine care.17 However, it is unclear how pharmacists manage patients who are candidates for OACT but who may be at increased risk for upper GI bleeds. Using a US-based survey, the investigators sought to determine pharmacists’ preferences in BRT usage and gastroprotection when bleed risk was lower than or equal to stroke risk.

 

 

Methods

This cross-sectional study was conducted after receiving approval by Nova Southeastern University’s Institutional Review Board. The survey consisted of 16 items divided into 4 domains: demographics, clinical experience, use of BRTs, and treatment preferences based on cases where bleed risk was lower than or equal to stroke risk (Figure 1). Queries were multiple choice and allowed for free-text input when “Other” was selected. Licensed pharmacists ≥ 18 years of age who routinely provided care to patients with NVAF were eligible to participate in the study. Participants who reported using a BRT (users) completed all study domains, while participants who reported not using a BRT (nonusers) completed domains 1 through 3 only.

Survey tool

An invitation containing the survey link was sent to the American College of Clinical Pharmacy ambulatory care (n = 2237) and cardiology (n = 1318) pharmacists listed in the organization’s Practice-based Research Networks. The survey was administered in the United States between April and June 2016 via Research Electronic Data Capture (REDCap) software, a secure Web application for building and managing online surveys designed to support data collection for research studies.18

Survey responses were downloaded, and data were analyzed using NCSS 2019 Statistical Software, LLC (Kaysville, UT). Descriptive statistics were calculated for all study variables. Demographic and clinical experience data for the group that used a BRT versus the group that did not were compared using Pearson’s chi-square, ANOVA, or the Cochran-Armitage test for trends. Logistic regression with hierarchical forward selection with switching was used to identify predictors of drug selection and use of gastroprotection.

Results

Of 230 respondents who completed the survey (response rate 6.5%), 165 (72%) used a BRT and 65 (28%) did not. No significant differences were found for age, gender, duration in clinical practice, the percentage of time spent in patient care, or practice specialty between users and nonusers (Table). The median age of users was 32 years; 68% were females; the median duration in clinical practice was 6 years; 75% of their time was spent in clinical practice; and clinical settings included ambulatory care, cardiology, and internal medicine. A significant difference was found for practice region between users versus nonusers (P = 0.014). Respondents who managed more than 200 NVAF patients per year used a BRT more often than those who managed fewer than 100 NVAF patients per year (P = 0.001).

Respondent Demographics

Of those who used a BRT, 97% utilized the HAS-BLED tool (n = 160). The remainder used HEMORR2HAGES (n = 3), ATRIA (n = 1), and mOBRI (n = 1). Reasons for choosing HAS-BLED included “familiarity/ease-of-use,” “preference by institution/clinical team,” and the fact that it was a “validated tool for NVAF.”

When bleed risk was lower than stroke risk, 151 of 165 users (92%) chose a treatment option (Figure 2). Of those, 65% chose a DOAC and 35% chose warfarin. Fourteen respondents chose “other” and explained that they “would initiate OACT after weighing patient factors and preferences.” When a DOAC was selected, 9% (n = 9) chose PPI co-therapy and 4% (n = 4) chose a H2RA. When warfarin was selected, 13% (n = 7) chose PPI co-therapy and 4% (n = 2) chose a H2RA. Respondents who chose gastroprotection did not provide reasons for doing so, but those who did not add it explained that they “would add gastroprotection only if patient is also on an NSAID or has a history of GI bleed” or cited “patient preference.” Specific to warfarin, some respondents would not add gastroprotection, as anticoagulation with warfarin is “easily reversed.”

Pharmacists’ treatment preferences if bleed risk is less than stroke risk (n = 151)

When bleed risk was equal to stroke risk, 141 of 165 users (85%) chose a treatment option (Figure 3). Fifty percent chose DOACs, 45% chose warfarin, and 5% chose aspirin. Logistic regression analysis (outcome DOAC versus warfarin area-under-ROC curve, 0.67) showed that as the number of NVAF patients seen in 12 months increased, respondents were more likely to select a DOAC over warfarin (odds ratio, 1.7; 95% CI, 1.1-2.5). Therefore, for every 50-patient increase per year, the probability of recommending a DOAC increased 1.7-fold.

 Pharmacists’ treatment preferences if bleed risk is equal to stroke risk (n = 141)

Of respondents who selected either a DOAC or warfarin, 38% (n = 50) also added gastroprotection (Figure 3). When a DOAC was selected, 34% (n = 24) favored PPI co-therapy and 7% (n = 5) chose a H2RA. When warfarin was selected, 19% (n = 12) favored PPI co-therapy, while 13% (n = 8) chose a H2RA. Rationale for choosing gastroprotection, regardless of OACT selection, included “stroke is more devastating, so if patient wants to continue treatment, but knew risks of bleeding were similar, would recommend gastroprotection to help minimize bleeding risk” and “patient-specific consideration.” Rationales for not choosing gastroprotection included “would add gastroprotection only if patient is on dual antiplatelet therapy or has another indication”; “in most patients, stroke risk outweighs bleed risk so no need for gastroprotection unless there is a stated reason”; “would use apixaban as has lowest bleeding rate of all DOACs in clinical trials”; and “gastroprotection has not been shown to be beneficial in large scale trials.” 

Eight respondents chose aspirin because it was “easy and relatively low cost.” Twenty-four respondents chose “other” and explained that the choice of OACT depended on patient preference after they had discussed stroke and bleed risk with the patient and/or determined the etiology driving bleed risk.

 

 

Discussion

This is the first national survey exploring US pharmacists’ preferences in BRT usage and treatment based on bleed risk. Pharmacists preferred the HAS-BLED tool and considered patient-specific factors and evidence-based data when weighing the risk-benefit of OACT with or without gastroprotective therapy.

Similar to our findings, where three-quarters of pharmacists used a BRT, a recent Medscape/American College of Cardiology (ACC) survey reported that 74% of cardiologists used a BRT (eg, HAS-BLED) always/most of the time or sometimes to assess a patient’s overall risk of bleeding prior to initiating DOAC therapy; 27% never or rarely used a bleed risk score before prescribing DOACs.19 Although reasons for BRT preference were not provided, they may be similar to those reported by our respondents (ie, familiarity/ease-of-use). In both surveys, rationales for not using a BRT were not obtained, but possible reasons include lack of confidence with bleed risk calculators,20 inconsistent implementation of comprehensive assessments (stroke risk, bleed risk, and medication-related issues prior to decision-making),21 and nonspecific guideline recommendations.22

More recently, a network meta-analysis found that HAS-BLED and HEMORR2HAGES had modest but balanced sensitivity (defined as the ratio between the number of major bleeding events in high-risk stratification and the total number of bleeding events) and specificity (defined as the ratio between the number of nonmajor bleeding events in the low-risk population and total nonbleeding events) for predicting major bleeding events.2,3 Several respondents did comment that, although HAS-BLED was imprecise and only studied with warfarin, it was necessary to identify bleed risk in a patient starting a high-risk medication, and that the ACC anticoagulation application uses HAS-BLED with CHA2DS2VASc along with clinical trial data to estimate stroke risk and bleed risk, with projected risk reduction (strokes) and risk increases (bleeds) expected with each treatment (www.acc.org/tools-and-practice-support/mobile-resources/features/anticoag-evaluator). The 2019 AHA/ACC/HRS atrial fibrillation guideline recommends that HAS-BLED scores be used to assess bleed risk in patients for whom anticoagulation is being considered, and that the need for and choice of OACT should be periodically reevaluated to reassess stroke and bleed risks.23

Although more than 80% of extracranial bleeds are GI bleeds,24 most BRTs are nonspecific for predicting GI bleeds. Indeed, one respondent used a spreadsheet with several BRTs to maximize treatment guidance for patients with multiple risk factors for strokes and bleeds. A comprehensive approach to determining factors that increase bleed risk should be adopted. These factors include age (HAS-BLED, HEMORR2HAGES, mOBRI, ATRIA); anemia (mOBRI, HEMORR2HAGES, ATRIA); hepatic/renal disease (HAS-BLED, HEMORR2HAGES, ATRIA, mOBRI); concomitant medications/alcohol use, including NSAIDs, corticosteroids, and antiplatelet therapy (HAS-BLED, HEMORR2HAGES); bleed history/rebleeding risk (HEMORR2HAGES, HAS-BLED, ATRIA); and GI bleeds (mOBRI).1,2 Additional risk factors for GI bleeds include being a tobacco smoker and/or being infected with Helicobacter pylori. A prospective cohort study that analyzed data from questionnaires completed by 99,359 individuals from the Copenhagen General Population Study reported that the multivariable adjusted hazard ratio for current smokers versus never smokers was 2.20 (95% CI, 1.84-2.62) for GI bleeds.25 Presence of H pylori should be investigated, with a subsequent eradication regimen implemented, as patients with warfarin-associated upper GI bleeds who were H pylori-positive had lower HAS-BLED scores versus those who were negative.26

When bleed risk was lower than stroke risk (eg, HAS-BLED < 3, CHA2DS2VASc ≥ 1), respondents appropriately initiated therapy with an OAC (predominantly apixaban); a small proportion also added gastroprotection. If the patient did not have any other GI bleed risk factors (eg, a previous GI bleed or on chronic antiplatelet or NSAID therapy), the choice of OACT depended on the attributes of each OAC and patient preference.27 Selection of warfarin was appropriate if cost, formulary restrictions, and availability of an inexpensive reversal agent were important concerns to patients and/or their health care providers. Rivaroxaban was selected because of its once-daily dosing and low risk for GI bleeding.

 

 

The recently published ARISTOPHANES study provides evidence that apixaban is an appropriate choice in patients with a HAS-BLED score < 3. In this retrospective observational study, more than 70% of patients received standard doses of DOACs (apixaban 5 mg, dabigatran 150 mg, or rivaroxaban 20 mg) and about 20% had a bleeding history, about 30% were on PPIs, less than 25% were on NSAIDs, and about 40% had a HAS-BLED score < 3. The study found that apixaban was more effective (reduced rates of ischemic or hemorrhagic strokes/systemic embolism) and safer (reduced rates of major GI bleed or intracranial bleed) than warfarin.28 Dabigatran and rivaroxaban were also more effective than warfarin for stroke prevention and had a lower risk for major intracranial bleed risk; while the risk of major GI bleed was similar between dabigatran and warfarin, major GI bleed risk was higher for rivaroxaban. When compared with each other, the 3 DOACs were effective at stroke prevention, with apixaban more effective than dabigatran and rivaroxaban; similar efficacy was noted for dabigatran versus rivaroxaban. Apixaban was associated with fewer GI bleeds versus dabigatran and rivaroxaban, but with similar intracranial bleed risks; dabigatran was associated with fewer GI bleeds but similar intracranial bleed risks versus rivaroxaban.28 Efficacy and safety findings from a subgroup analysis based on HAS-BLED scores < 3 and ≥ 3 were generally consistent with the main results.

When bleed risk was equal to stroke risk, the difficulty was determining how OACT in a patient at high stroke risk (CHA2DS2VASc score ≥ 2) and high bleed risk (HAS-BLED score ≥ 3) should be managed. Eight respondents chose aspirin and added gastroprotection with either a PPI or H2RA; however, currently, aspirin is not recommended as the sole antithrombotic for patients with NVAF.23 With the OACT, an interesting finding was that as the number of patients seen in 12 months increased, pharmacists were almost twice as likely to select a DOAC over warfarin. Moreover, pharmacists were judicious in their recommendation to add gastroprotection, and would consider doing so if there was a specific indication. At the time of our survey, several studies described DOAC-associated GI bleeds,29-31 but data on the effectiveness of acid-suppressive therapy, specifically with PPIs, in the prevention of upper GI bleeds were sparse.4,7,32 Respondents most likely were familiar with GI bleed risk factors and prevention strategies from various guidelines published between 2009 and 2010, which did not include DOACs.33-35

Another important finding was pharmacists’ uncertainty as to the effectiveness of PPIs in preventing GI bleeds in combination with DOACs. The data are conflicting. A meta-analysis of older studies (2007-2015) showed that PPIs (but not H2RAs) reduced the risk of upper GI bleeds in patients on warfarin but not for dabigatran.36 A retrospective cohort study of Medicare beneficiaries on OACTs (2011-2015) showed the adjusted incidence of hospitalization for upper GI bleeds in patients on PPI co-therapy was significantly lower compared with patients not on PPI co-therapy (76 versus 115 per 10,000 person-years, respectively).8 Apixaban without PPI co-therapy was associated with the lowest risk of upper GI bleed hospitalizations (73/10,000 person-years), and PPI co-therapy further reduced this risk (49/10,000 person-years). Warfarin without PPI co-therapy was associated with the next lowest risk (113/10,000 person-years), followed by dabigatran (120/10,000 person-years) and rivaroxaban (144/10,000 person-years). PPI co-therapy significantly reduced the risk of upper GI bleed hospitalizations with all OACTs, but the incidence of upper GI bleed hospitalizations with rivaroxaban was significantly greater than with the other OACTs.8 Therefore, if there are concerns about the safety of PPIs,37-39 or the patient is unable to tolerate a PPI, then apixaban may be the most appropriate DOAC for a patient with high bleed risk. Notably, a 2020 review of data from the PINNACLE registry (average age, 75-77 years; 31% on PPIs) found that the relative GI bleed safety advantage of apixaban and dabigatran versus warfarin was attenuated in patients ≥ 75 years.40 Last, since the risk for lower GI bleeds is not reduced by PPIs,41 consideration of their use should be accompanied by an assessment to detect bleeds (eg, low hemoglobin/hematocrit, presence of bright red blood, hematochezia/melena, fecal occult testing), with prompt management as necessary.5

Limitations

Limitations of our survey included an overall low response rate, which can generate a biased sample if respondents are systematically different from nonrespondents. In addition, to maintain simplicity and reduce respondents’ time commitment, our survey did not include actual CHA2DS2VASc stroke risk scores, HAS-BLED bleed risk scores, or specific GI bleed risk factors when querying pharmacists about treatment options based on bleed risk. The addition of these variables would have improved the robustness of the data.

Conclusion

In addition to applying BRTs in the management of NVAF patients, pharmacists considered patient-specific variables, prescriber preferences, and evidence-based guidance when recommending OACT with or without gastroprotection. To avoid suboptimal patient management, busy pharmacists should be granted time to attend continuing education programs describing optimal OACT selection and formulation of individualized, evidence-based plans to address modifiable risk factors for bleeding, including the appropriate use of gastroprotection. Randomized, prospective, long-term studies stratified by HAS-BLED and CHA2DS2VASc scores are needed to further clarify efficacy, safety, and cost-effectiveness of OACT, with and without PPIs, in patients who may be at risk for upper GI bleeds.

Acknowledgments: The authors thank Robin J. Jacobs, PhD, MSW, MS, MPH, Patrick C. Hardigan, PhD, Steven Brettler, PharmD, MPH, Maria-Isabel A. Cabral, PharmD, and Reginald Gyapong, PharmD, for their participation in this project. The authors also sincerely thank Fabio Franco, BS Computer Science, who organized the database to enable efficient data management.

Corresponding author: Devada Singh-Franco, PharmD, CDE, Nova Southeastern University College of Pharmacy, 3200 S University Drive, Fort Lauderdale, FL 33328; singh@nova.edu

Disclosures: None.

Funding: The study was supported by Nova Southeastern University’s Health Professions Division Internal Research Grant.

References

1. Apostolakis S, Lane DA, Guo Y, et al. Performance of the HEMORR2HAGES, ATRIA, and HAS-BLED bleeding risk–prediction scores in patients with atrial fibrillation undergoing anticoagulation. J Am Coll Cardiol. 2012;60:861-867.

2. Chang G, Xie Q, Ma L, et al. Accuracy of HAS-BLED and other bleeding risk assessment tools in predicting major bleeding events in atrial fibrillation: A network meta-analysis. J Thromb Haemost. 2020;18:791-801.

3. Ding WY, Harrison SL, Lane DA, Lip GYH. Considerations when choosing an appropriate bleeding risk assessment tool for patients with atrial fibrillation. J Thromb Haemost. 2020;18:788-790.

4. Lauffenburger JC, Rhoney DH, Farley JF, et al. Predictors of gastrointestinal bleeding among patients with atrial fibrillation after initiating dabigatran therapy. Pharmacotherapy. 2015;35:560-568.

5. Tomaselli GF, Mahaffey KW, Cuker A, et al. 2020 ACC Expert Consensus Decision Pathway on Management of Bleeding in Patients on Oral Anticoagulants: A Report of the American College of Cardiology Solution Set Oversight Committee. J Am Coll Cardiol. 2020;76:594-622.

6. Taha A, McCloskey C, Craigen T, Angerson W. Antiplatelet versus anticoagulant effects in non-variceal upper gastrointestinal bleeding. Gut. 2019;68(suppl 2):A152.

7. Chan EW, Lau WC, Leung WK, et al. Prevention of dabigatran-related gastrointestinal bleeding with gastroprotective agents: A population-based study. Gastroenterology. 2015;149:586-595.

8. Ray WA, Chung CP, Murray KT, et al. Association of oral anticoagulants and proton pump inhibitor cotherapy with hospitalization for upper gastrointestinal tract bleeding. JAMA. 2018;320:2221-2230.

9. Brunetti L, Lee S-M, Doherty N, et al. Impact of warfarin discharge education program on hospital readmission and treatment costs. Int J Clin Pharm. 2018;40:721-729.

10. Hasan SS, Kow CS, Curley LE, et al. Economic evaluation of prescribing conventional and newer oral anticoagulants in older adults. Expert Rev Pharmacoecon Outcomes Res. 2018;18:371-377.

11. Phelps E, Delate T, Witt DM, et al. Effect of increased time in the therapeutic range on atrial fibrillation outcomes within a centralized anticoagulation service. Thromb Res. 2018;163:54-59.

12. Ahuja T, Raco V, Papadopoulos J, Green D. Antithrombotic stewardship: Assessing use of computerized clinical decision support tools to enhance safe prescribing of direct oral anticoagulants in hospitalized patients. J Patient Saf. 2018 Sep 25. [Epub ahead of print]

13. Leef GC, Perino AC, Askari M, et al. Appropriateness of direct oral anticoagulant dosing in patients with atrial fibrillation: Insights from the Veterans Health Administration. J Pharm Pract. 2020;33:647-653.

14. Papastergiou J, Kheir N, Ladova K, et al. Pharmacists’ confidence when providing pharmaceutical care on anticoagulants, a multinational survey. Int J Clin Pharm. 2017;39:1282-1290.

15. Perlman A, Horwitz E, Hirsh-Raccah B, et al. Clinical pharmacist led hospital-wide direct oral anticoagulant stewardship program. Isr J Health Policy Res. 2019;8:19.

16. Uppuluri EM, McComb MN, Shapiro NL. Implementation of a direct oral anticoagulation screening service at a large academic medical center provided by a pharmacist-managed antithrombosis clinic as a method to expand antithrombotic stewardship efforts. J Pharm Pract. 2020;33:271-275.

17. Manzoor BS, Cheng W-H, Lee JC, et al. Quality of pharmacist-managed anticoagulation therapy in long-term ambulatory settings: A systematic review. Ann Pharmacother. 2017;51:1122-1137.

18. Harris PA, Taylor R, Thielke R, et al. Research Electronic Data Capture (REDCap)—A metadata-driven methodology and workflow process for providing translational research informatics support. J Biomed Inform. 2009;42:377-381.

19. Brooks M. AF management: Are clinicians in agreement? Medscape. May 30, 2019. Accessed December 29, 2020. https://www.medscape.com/viewarticle/913386

20. Amroze A, Mazor K, Crawford S, et al. Survey of confidence in use of stroke and bleeding risk calculators, knowledge of anticoagulants, and comfort with prescription of anticoagulation in challenging scenarios: SUPPORT-AF II study. J Thromb Thrombolysis. 2019;48:629-637.

21. Wang Y, Bajorek B. Decision-making around antithrombotics for stroke prevention in atrial fibrillation: the health professionals’ views. Int J Clin Pharm. 2016;38:985-995.

22. January CT, Wann LS, Alpert JS, et al. 2014 AHA/ACC/HRS guideline for the management of patients with atrial fibrillation: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines and the Heart Rhythm Society. Circulation. 2014;130:e199-e267.

23. January CT, Wann LS, Calkins H, et al. 2019 AHA/ACC/HRS Focused Update of the 2014 AHA/ACC/HRS Guideline for the Management of Patients With Atrial Fibrillation: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines and the Heart Rhythm Society. J Am Coll Cardiol. 2019;74:104-132.

24. Anghel L, Sascu R, Trifan A, et al. Non-vitamin K antagonist oral anticoagulants and the gastrointestinal bleeding risk in real-world studies. J Clin Med. 2020;9:1398.

25. Langsted A, Nordestgaard BG. Smoking is associated with increased risk of major bleeding: a prospective cohort study. Thromb Haemost. 2019;119:39-47.

26. Faye AS, Hung KW, Cheng K, et al. HAS-BLED scores underestimate gastrointestinal bleeding risk among those with H. pylori. Am J Gastroenterol. 2019;114:S364.

27. Fawzy AM, Yang W-Y, Lip GY. Safety of direct oral anticoagulants in real-world clinical practice: translating the trials to everyday clinical management. Expert Opin Drug Saf. 2019;18:187-209.

28. Lip GYH, Keshishian A, Li X, et al. Effectiveness and safety of oral anticoagulants among nonvalvular atrial fibrillation patients. Stroke. 2018;49:2933-2944.

29. Abraham NS, Singh S, Alexander GC, et al. Comparative risk of gastrointestinal bleeding with dabigatran, rivaroxaban, and warfarin: population based cohort study. BMJ. 2015;350:h1857.

30. Holster IL, Valkhoff VE, Kuipers EJ, Tjwa E. New oral anticoagulants increase risk for gastrointestinal bleeding: a systematic review and meta-analysis. Gastroenterology. 2013;145:105-112.

31. Sherwood MW, Nessel CC, Hellkamp AS, et al. Gastrointestinal bleeding in patients with atrial fibrillation treated with rivaroxaban or warfarin: ROCKET AF Trial. J Am Coll Cardiol. 2015;66:2271-2281.

32. Di Minno A, Spadarella G, Spadarella E, et al. Gastrointestinal bleeding in patients receiving oral anticoagulation: Current treatment and pharmacological perspectives. Thromb Res. 2015;136:1074-1081.

33. Abraham NS, Hlatky MA, Antman EM, et al. ACCF/ACG/AHA 2010 Expert Consensus Document on the Concomitant Use of Proton Pump Inhibitors and Thienopyridines: A Focused Update of the ACCF/ACG/AHA 2008 Expert Consensus Document on Reducing the Gastrointestinal Risks of Antiplatelet Therapy and NSAID Use. Circulation. 2010;122:2619-2633.

34. Bhatt DL, Scheiman J, Abraham NS, et al. ACCF/ACG/AHA 2008 expert consensus document on reducing the gastrointestinal risks of antiplatelet therapy and NSAID use: a report of the American College of Cardiology Foundation Task Force on Clinical Expert Consensus Documents. J Am Coll Cardiol. 2008;52:1502-1517.

35. Lanza FL, Chan FK, Quigley EM. Guidelines for prevention of NSAID-related ulcer complications. Am J Gastroenterol. 2009;104:728-738.

36. Bang CS, Joo MK, Kim BW, et al. The role of acid suppressants in the prevention of anticoagulant-related gastrointestinal bleeding: a systematic review and meta-analysis. Gut Liver. 2020;14:57-66.

37. Farrell B, Pottie K, Thompson W, et al. Deprescribing proton pump inhibitors: Evidence-based clinical practice guideline. Can Fam Physician. 2017;63:354-364.

38. Fossmark R, Martinsen TC, Waldum HL. Adverse effects of proton pump inhibitors—evidence and plausibility. Int J Mol Sci. 2019;20:5203.

39. Haastrup PF, Thompson W, Sondergaard J, Jarbol DE. Side effects of long-term proton pump inhibitor use: A review. Basic Clin Pharmacol Toxicol. 2018;123:114-121.

40. Wong JM, Maddox TM, Kennedy K, Shaw RE. Comparing major bleeding risk in outpatients with atrial fibrillation or flutter by oral anticoagulant type (from the National Cardiovascular Disease Registry’s Practice Innovation and Clinical Excellence Registry). Am J Cardiol. 2020;125:1500-1507.

41. Nagata N, Niikura R, Aoki T, et al. Effect of proton-pump inhibitors on the risk of lower gastrointestinal bleeding associated with NSAIDs, aspirin, clopidogrel, and warfarin. J Gastroenterol. 2015;50:1079-1086.

References

1. Apostolakis S, Lane DA, Guo Y, et al. Performance of the HEMORR2HAGES, ATRIA, and HAS-BLED bleeding risk–prediction scores in patients with atrial fibrillation undergoing anticoagulation. J Am Coll Cardiol. 2012;60:861-867.

2. Chang G, Xie Q, Ma L, et al. Accuracy of HAS-BLED and other bleeding risk assessment tools in predicting major bleeding events in atrial fibrillation: A network meta-analysis. J Thromb Haemost. 2020;18:791-801.

3. Ding WY, Harrison SL, Lane DA, Lip GYH. Considerations when choosing an appropriate bleeding risk assessment tool for patients with atrial fibrillation. J Thromb Haemost. 2020;18:788-790.

4. Lauffenburger JC, Rhoney DH, Farley JF, et al. Predictors of gastrointestinal bleeding among patients with atrial fibrillation after initiating dabigatran therapy. Pharmacotherapy. 2015;35:560-568.

5. Tomaselli GF, Mahaffey KW, Cuker A, et al. 2020 ACC Expert Consensus Decision Pathway on Management of Bleeding in Patients on Oral Anticoagulants: A Report of the American College of Cardiology Solution Set Oversight Committee. J Am Coll Cardiol. 2020;76:594-622.

6. Taha A, McCloskey C, Craigen T, Angerson W. Antiplatelet versus anticoagulant effects in non-variceal upper gastrointestinal bleeding. Gut. 2019;68(suppl 2):A152.

7. Chan EW, Lau WC, Leung WK, et al. Prevention of dabigatran-related gastrointestinal bleeding with gastroprotective agents: A population-based study. Gastroenterology. 2015;149:586-595.

8. Ray WA, Chung CP, Murray KT, et al. Association of oral anticoagulants and proton pump inhibitor cotherapy with hospitalization for upper gastrointestinal tract bleeding. JAMA. 2018;320:2221-2230.

9. Brunetti L, Lee S-M, Doherty N, et al. Impact of warfarin discharge education program on hospital readmission and treatment costs. Int J Clin Pharm. 2018;40:721-729.

10. Hasan SS, Kow CS, Curley LE, et al. Economic evaluation of prescribing conventional and newer oral anticoagulants in older adults. Expert Rev Pharmacoecon Outcomes Res. 2018;18:371-377.

11. Phelps E, Delate T, Witt DM, et al. Effect of increased time in the therapeutic range on atrial fibrillation outcomes within a centralized anticoagulation service. Thromb Res. 2018;163:54-59.

12. Ahuja T, Raco V, Papadopoulos J, Green D. Antithrombotic stewardship: Assessing use of computerized clinical decision support tools to enhance safe prescribing of direct oral anticoagulants in hospitalized patients. J Patient Saf. 2018 Sep 25. [Epub ahead of print]

13. Leef GC, Perino AC, Askari M, et al. Appropriateness of direct oral anticoagulant dosing in patients with atrial fibrillation: Insights from the Veterans Health Administration. J Pharm Pract. 2020;33:647-653.

14. Papastergiou J, Kheir N, Ladova K, et al. Pharmacists’ confidence when providing pharmaceutical care on anticoagulants, a multinational survey. Int J Clin Pharm. 2017;39:1282-1290.

15. Perlman A, Horwitz E, Hirsh-Raccah B, et al. Clinical pharmacist led hospital-wide direct oral anticoagulant stewardship program. Isr J Health Policy Res. 2019;8:19.

16. Uppuluri EM, McComb MN, Shapiro NL. Implementation of a direct oral anticoagulation screening service at a large academic medical center provided by a pharmacist-managed antithrombosis clinic as a method to expand antithrombotic stewardship efforts. J Pharm Pract. 2020;33:271-275.

17. Manzoor BS, Cheng W-H, Lee JC, et al. Quality of pharmacist-managed anticoagulation therapy in long-term ambulatory settings: A systematic review. Ann Pharmacother. 2017;51:1122-1137.

18. Harris PA, Taylor R, Thielke R, et al. Research Electronic Data Capture (REDCap)—A metadata-driven methodology and workflow process for providing translational research informatics support. J Biomed Inform. 2009;42:377-381.

19. Brooks M. AF management: Are clinicians in agreement? Medscape. May 30, 2019. Accessed December 29, 2020. https://www.medscape.com/viewarticle/913386

20. Amroze A, Mazor K, Crawford S, et al. Survey of confidence in use of stroke and bleeding risk calculators, knowledge of anticoagulants, and comfort with prescription of anticoagulation in challenging scenarios: SUPPORT-AF II study. J Thromb Thrombolysis. 2019;48:629-637.

21. Wang Y, Bajorek B. Decision-making around antithrombotics for stroke prevention in atrial fibrillation: the health professionals’ views. Int J Clin Pharm. 2016;38:985-995.

22. January CT, Wann LS, Alpert JS, et al. 2014 AHA/ACC/HRS guideline for the management of patients with atrial fibrillation: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines and the Heart Rhythm Society. Circulation. 2014;130:e199-e267.

23. January CT, Wann LS, Calkins H, et al. 2019 AHA/ACC/HRS Focused Update of the 2014 AHA/ACC/HRS Guideline for the Management of Patients With Atrial Fibrillation: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines and the Heart Rhythm Society. J Am Coll Cardiol. 2019;74:104-132.

24. Anghel L, Sascu R, Trifan A, et al. Non-vitamin K antagonist oral anticoagulants and the gastrointestinal bleeding risk in real-world studies. J Clin Med. 2020;9:1398.

25. Langsted A, Nordestgaard BG. Smoking is associated with increased risk of major bleeding: a prospective cohort study. Thromb Haemost. 2019;119:39-47.

26. Faye AS, Hung KW, Cheng K, et al. HAS-BLED scores underestimate gastrointestinal bleeding risk among those with H. pylori. Am J Gastroenterol. 2019;114:S364.

27. Fawzy AM, Yang W-Y, Lip GY. Safety of direct oral anticoagulants in real-world clinical practice: translating the trials to everyday clinical management. Expert Opin Drug Saf. 2019;18:187-209.

28. Lip GYH, Keshishian A, Li X, et al. Effectiveness and safety of oral anticoagulants among nonvalvular atrial fibrillation patients. Stroke. 2018;49:2933-2944.

29. Abraham NS, Singh S, Alexander GC, et al. Comparative risk of gastrointestinal bleeding with dabigatran, rivaroxaban, and warfarin: population based cohort study. BMJ. 2015;350:h1857.

30. Holster IL, Valkhoff VE, Kuipers EJ, Tjwa E. New oral anticoagulants increase risk for gastrointestinal bleeding: a systematic review and meta-analysis. Gastroenterology. 2013;145:105-112.

31. Sherwood MW, Nessel CC, Hellkamp AS, et al. Gastrointestinal bleeding in patients with atrial fibrillation treated with rivaroxaban or warfarin: ROCKET AF Trial. J Am Coll Cardiol. 2015;66:2271-2281.

32. Di Minno A, Spadarella G, Spadarella E, et al. Gastrointestinal bleeding in patients receiving oral anticoagulation: Current treatment and pharmacological perspectives. Thromb Res. 2015;136:1074-1081.

33. Abraham NS, Hlatky MA, Antman EM, et al. ACCF/ACG/AHA 2010 Expert Consensus Document on the Concomitant Use of Proton Pump Inhibitors and Thienopyridines: A Focused Update of the ACCF/ACG/AHA 2008 Expert Consensus Document on Reducing the Gastrointestinal Risks of Antiplatelet Therapy and NSAID Use. Circulation. 2010;122:2619-2633.

34. Bhatt DL, Scheiman J, Abraham NS, et al. ACCF/ACG/AHA 2008 expert consensus document on reducing the gastrointestinal risks of antiplatelet therapy and NSAID use: a report of the American College of Cardiology Foundation Task Force on Clinical Expert Consensus Documents. J Am Coll Cardiol. 2008;52:1502-1517.

35. Lanza FL, Chan FK, Quigley EM. Guidelines for prevention of NSAID-related ulcer complications. Am J Gastroenterol. 2009;104:728-738.

36. Bang CS, Joo MK, Kim BW, et al. The role of acid suppressants in the prevention of anticoagulant-related gastrointestinal bleeding: a systematic review and meta-analysis. Gut Liver. 2020;14:57-66.

37. Farrell B, Pottie K, Thompson W, et al. Deprescribing proton pump inhibitors: Evidence-based clinical practice guideline. Can Fam Physician. 2017;63:354-364.

38. Fossmark R, Martinsen TC, Waldum HL. Adverse effects of proton pump inhibitors—evidence and plausibility. Int J Mol Sci. 2019;20:5203.

39. Haastrup PF, Thompson W, Sondergaard J, Jarbol DE. Side effects of long-term proton pump inhibitor use: A review. Basic Clin Pharmacol Toxicol. 2018;123:114-121.

40. Wong JM, Maddox TM, Kennedy K, Shaw RE. Comparing major bleeding risk in outpatients with atrial fibrillation or flutter by oral anticoagulant type (from the National Cardiovascular Disease Registry’s Practice Innovation and Clinical Excellence Registry). Am J Cardiol. 2020;125:1500-1507.

41. Nagata N, Niikura R, Aoki T, et al. Effect of proton-pump inhibitors on the risk of lower gastrointestinal bleeding associated with NSAIDs, aspirin, clopidogrel, and warfarin. J Gastroenterol. 2015;50:1079-1086.

Issue
Journal of Clinical Outcomes Management - 28(1)
Issue
Journal of Clinical Outcomes Management - 28(1)
Page Number
9-16
Page Number
9-16
Publications
Publications
Topics
Article Type
Display Headline
Pharmacists’ Bleed Risk Tool and Treatment Preferences Prior to Initiating Anticoagulation in Patients With Nonvalvular Atrial Fibrillation: A Cross-Sectional Survey
Display Headline
Pharmacists’ Bleed Risk Tool and Treatment Preferences Prior to Initiating Anticoagulation in Patients With Nonvalvular Atrial Fibrillation: A Cross-Sectional Survey
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Article PDF Media

Addressing Maternal Mortality Through Education: The Mommies Methadone Program

Article Type
Changed
Tue, 01/12/2021 - 10:12
Display Headline
Addressing Maternal Mortality Through Education: The Mommies Methadone Program

From the UT Health Long School of Medicine San Antonio, Texas.

Abstract

Objective: To educate pregnant patients with opioid use disorder (OUD) about the effects of opioids in order to improve understanding and help achieve sustained abstinence.

Methods: The Center for Health Care Services and University Hospital System (UHS) in San Antonio, TX, jointly operate a methadone clinic, the Mommies Program, for pregnant women with OUD. As part of this program, medical students discuss methadone use in pregnancy and the health effects of OUD on mother and child and review resources for sustained peripartum abstinence at prenatal visits with pregnant patients with OUD concurrently enrolled in the program. To evaluate the impact of this education intervention, students assessed patients’ knowledge of methadone effects on mother and baby, state laws concerning heroin and methadone use in pregnancy, and motivation to quit using opioids with pre- and post-intervention surveys.

Results: Of 68 women enrolled in the program, 33 completed both the pre-survey and the post-survey (48.5%). Nearly half (48%) were very motivated to quit before pregnancy, but 85% were very motivated to quit once pregnant. All participants said learning more about the effects of opiates would increase motivation for sobriety. Prior to the educational intervention, 39% of participants knew it was safe to breastfeed on methadone, which improved to 97% in the post-survey, and 76% incorrectly thought they would be reported to authorities by their health care providers if they used illegal drugs during pregnancy, while in the post-survey, 100% knew they would not be reported for doing so.

Conclusion: Pregnancy and education about opioids increased patients’ motivation to quit. Patients also advanced their breastfeeding knowledge and learned about patient-provider confidentiality. Our greatest challenge was participant follow-up; however, this improved with the help of a full-time Mommies Program nurse. Our future aim is to increase project awareness and extend the educational research.

Keywords: pregnancy; addiction; opioids; OUD; counseling.

In 2012 more than 259 million prescriptions for opioids were written in the United States, which was a 200% increase since 1998.1 Since the early 2000s, admissions to opioid substance abuse programs and the death rate from opioids have quadrupled.2-4 Specifically, the rate of heroin use increased more than 300% from 2010 to 2014.5 Opioid use in pregnancy has also escalated in recent years, with a 3- to 4-fold increase from 2000 to 2009 and with 4 in 1000 deliveries being complicated by opioid use disorder (OUD) in 2011.6-8

Between 2000 and 2014, the maternal mortality rate in the United States increased 24%, making it the only industrialized nation with a maternal mortality rate that is rising rather than falling.9 The Texas Maternal Mortality and Morbidity Task Force found that between 2012 and 2015 drug overdose was the leading cause of maternal death in the period from delivery to 365 days postpartum, and it has increased dramatically since 2010.10,11

 

 

In addition, maternal mortality reviews in several states have identified substance use as a major risk factor for pregnancy-associated deaths.12,13 In Texas between 2012 and 2015, opioids were found in 58% of maternal drug overdoses.10 In 2007, 22.8% of women who were enrolled in Medicaid programs in 46 states filled an opioid prescription during pregnancy.14 Additionally, the rising prevalence of opioid use in pregnancy has led to a sharp increase in neonatal abstinence syndrome (NAS), rising from 1.5 cases per 1000 hospital births in 1999 to 6.0 per 1000 hospital births in 2013.15 Unsurprisingly, states with the highest rates of opioid prescribing also have the highest rates of NAS.16

Methadone combined with counseling and behavioral therapy has been the standard of care for the treatment of OUD in pregnancy since the 1970s. Methadone treatment prevents opioid withdrawal symptoms and increases adherence to prenatal care.17 One of the largest methadone treatment clinics in the San Antonio, TX, area is the Center for Health Care Services (CHCS). University Health System in San Antonio (UHS) has established a clinic called The Mommies Program, where mothers addicted to opioids can receive prenatal care by a dedicated physician, registered nurse, and a certified nurse midwife, who work in collaboration with the CHCS methadone clinic. Pregnant patients with OUD in pregnancy are concurrently enrolled in the Mommies Program and receive prenatal care through UHS and methadone treatment and counseling through CHCS. The continuity effort aims to increase prenatal care rates and adherence to methadone treatment.

Once mothers are off illicit opioids and on methadone, it is essential to discuss breastfeeding with them, as many mothers addicted to illicit opioids may have been told that they should not be breastfeeding. However, breastfeeding should be encouraged in women who are stable on methadone if they are not using illicit drugs and do not have other contraindications, regardless of maternal methadone dose, since the transfer of methadone into breast milk is minimal.18-20 Breastfeeding is beneficial in women taking methadone and has been associated with decreased severity of NAS symptoms, decreased need for pharmacotherapy, and a shorter hospital stay for the baby.21 In addition, breastfeeding contributes to the development of an attachment between mother and infant, while also providing the infant with natural immunity. Women should be counseled about the need to stop breastfeeding in the event of a relapse.22

Finally, the postpartum period represents a time of increased stressors, such as loss of sleep, child protective services involvement, and frustration with constant demands from new baby. For mothers with addiction, this is an especially sensitive time, as the stressors may be exacerbated by their new sobriety and a sudden end to the motivation they experienced from pregnancy.23 Therefore, early and frequent postpartum care with methadone dose evaluation is essential in order to decrease drug relapse and screen for postpartum depression in detail, since patients with a history of drug use are at increased risk of postpartum depression.

In 2017 medical students at UT Health Long School of Medicine in San Antonio created a project to educate women about OUD in pregnancy and provide motivational incentives for sustained abstinence; this project has continued each year since. Students provide education about methadone treatment and the dangers of using illicit opioids during and after pregnancy. Students especially focus on educating patients on the key problem areas in the literature, such as overdose, NAS, breastfeeding, postpartum substance use, and postpartum depression.

 

 

Methods

From October 2018 to February 2020, a total of 15 medical students volunteered between 1 and 20 times at the Mommies Program clinic, which was held once or twice per week from 8 am to 12 pm. Prior to attending clinic, a fourth-year medical student oriented that year’s group of first-, second-, and third-year medical students who volunteered to attend the clinic. The orientation introduced students to the Mommies Program and instructed them on how and why methadone is prescribed and distributed. Students were also taught how to begin and proceed through each patient encounter, and were given a standard handout to review with patients. This handout made it possible for students of every level to attend the Mommies Program and ensured that the education was standardized at every session (Figure 1).

Handout used by medical students during discussion with participants in the Mommies Program

The only inclusion criteria for participating in the educational intervention and survey was participants had to be 18 years of age or older and enrolled in the Mommies Program. Patients who met the inclusion criteria and agreed to participate completed a pre-survey administered by the students during the patient’s initial prenatal visit (Figure 2). This survey collected baseline information about the patient’s history with opioid use and their current knowledge of methadone treatment, NAS, legal aspects of drug use disclosure, and drug testing prior to the education portion of the encounter. After the pre-survey was administered, students spent 30 minutes reviewing the correct answers of the survey with the patients by utilizing the standardized handout to help patients understand details of methadone and opioid use in pregnancy (Figure 1). The post-survey was administered by a student once patients entered the third trimester to assess whether the education session increased patients’ knowledge of these topics.

Survey administered before and after educational intervention

At the time patients completed the post-survey, they received a Baby Bag as well as education regarding each item in the bag. The aim of distributing Baby Bags was to relieve some possible postnatal stressors and educate the patients about infant care. Items included in the bag were diapers, wipes, bottles, clothes, and swaddles. Prenatal vitamins were added in January 2020, as many patients struggle to afford vitamins if they are not currently covered by Medicaid or have other barriers. The Baby Bag items were purchased through a Community Service Learning grant through UT Health San Antonio.

Results

Of 68 women enrolled in the Mommies Program during the intervention period, 33 completed the pre-survey and the post-survey (48.5%). Even though all patients enrolled in the program met the inclusion criteria, patients were not included in the educational program for multiple reasons, including refusal to participate, poor clinic follow-up, or lack of students to collect surveys. However, all patients who completed the pre-survey did complete the post-survey. In the pre-survey, only 39% of participants knew it was safe to breastfeed while on methadone. In the post-survey, 97% knew it safe to breastfeed. Nearly half (48%) reported being very motivated to quit opioids before pregnancy, but 85% were very motivated to quit once pregnant. In the pre-survey, 76% incorrectly thought they would be reported to authorities by their health providers if they used illegal drugs during pregnancy, while in the post-survey, 100% knew they would not be reported for doing so. Also, all participants said learning more about the effects of opiates would increase motivation for sobriety.

 

 

Discussion

Questions assessed during the educational surveys revolved around patients’ knowledge of the intricacies, legally and physiologically, of methadone treatment for OUD, as well as beneficial aspects for patients and future child health, such as breastfeeding and motivation to quit and stay sober.

It was clear that there was a lack of knowledge and education about breastfeeding, as only 39% of the participants thought that it was safe to breastfeed while on methadone in the pre-survey; in the post survey, this improved to 97%. Students spent a large portion of the educational time going over the safety of breastfeeding for patients on methadone and the many benefits to mother and baby. Students also reviewed breastfeeding with patients every time patients came in for a visit and debunked any falsehoods about the negatives of breastfeeding while on methadone. This is another testament to the benefits of reinforcement around patient education.

The area of trust between provider and patient is essential in all provider-patient relationships. However, in the area of addiction, a trusting bond is especially important, as patients must feel confident and comfortable to disclose every aspect of their lives so the provider can give the best care. It was clear from our initial data that many patients did not feel this trust or understand the legal aspects regarding the provider-patient relationship in the terms of drug use, as the pre-survey shows 76% of patients originally thought they would be reported to authorities if they told their provider they used illegal drugs during pregnancy. This was an enormous issue in the clinic and something that needed to be addressed because, based on these data, we feared many patients would not be honest about using illegal drugs to supplement their methadone if they believed they would be reported to the authorities or even jailed. The medical student education team continually assured patients that their honesty about illegal drug use during pregnancy would not be revealed to the authorities, and also made it clear to patients that it was essential they were honest about illegal drug use so the optimal care could be provided by the team. These discussions were successful, as the post-survey showed that 100% of patients knew they would not be reported to the authorities if they used illegal drugs during the pregnancy. This showed an increase in knowledge, but also suggested an increase in confidence in the provider-patient relationship by patients, which we speculate allowed for a better patient experience, better patient outcomes, and less emotional stress for the patient and provider.

Last, we wanted to study and address the motivation to quit using drugs and stay sober through learning about the effects of opiates and how this motivation was related to pregnancy. A study by Mitchell et al makes clear that pregnancy is a motivation to seek treatment for drug use and to quit,24 and our survey data support these findings, with 48% of patients motivated to quit before they were pregnant and 85% motivated to quit once they knew they were pregnant. In addition, all patients attested on the pre- and post-survey that learning more about opioids would increase their motivation for sobriety. Therefore, we believe education about the use of opioids and other drugs is a strong motivation towards sobriety and should be further studied in methadone treatment and other drugs as well.

We will continue to focus on sobriety postpartum by using the education in pregnancy as a springboard to further postpartum education, as education seems to be very beneficial to future sobriety. In the future, we believe extending the educational program past pregnancy and discussing opioid use and addiction with patients at multiple follow-up visits will be beneficial to patients’ sobriety.

 

 

We faced 2 main challenges in implementing this intervention and survey: patients would often miss multiple appointments during their third trimester or would not attend their postpartum visit if they only had 1 prenatal visit; and many clinic sessions had low student attendance because students often had many other responsibilities in medical school and there were only 15 volunteers over the study time. These challenges decreased our post-survey completion rate. However, there has been improvement in follow-up as the project has continued. The Mommies Program now has a full-time registered nurse, and a larger number of medical student teachers have volunteered to attend the clinic. In the future, we aim to increase awareness of our project and the benefits of participation, expand advertising at our medical school to increase student participation, and increase follow-up education in the postpartum period.

Another future direction is to include local, free doula services, which are offered through Catholic Charities in San Antonio. Doulas provide antepartum, intrapartum, and postpartum services, which we believe will help our patients through advocacy and support for sobriety during this emotional and stressful time.

Conclusion

Counseled participants were receptive to learning about the effects of OUD and methadone on themselves and their newborn. Participants unanimously stated that learning more about OUD increased their motivation for sobriety. It was also clear that the increased motivation to be sober during pregnancy, as compared to before pregnancy, is an opportunity to help these women take steps to get sober. Patients also advanced their breastfeeding knowledge, as we helped debunk falsehoods surrounding breastfeeding while on methadone, and we anticipate this will lead to greater breastfeeding rates for our patients on methadone, although this was not specifically studied. Finally, patients learned about patient-provider confidentiality, which allowed for more open and clear communication with patients so they could be cared for to the greatest degree and trust could remain paramount.

Drug use is a common problem in the health care system, and exposure to patients with addiction is important for medical students in training. We believe that attending the Mommies Program allowed medical students to gain exposure and skills to better help patients with OUD.

Corresponding author: Nicholas Stansbury, MD, Nmstansbury85@gmail.com.

Financial disclosures: None.

References

1. Centers for Disease Control and Prevention. Opioid painkiller prescribing: where you live makes a difference. CDC website. www.cdc.gov/vitalsigns/opioid-prescribing. Accessed October 28, 2020. 

2. Substance Abuse and Mental Health Services Administration. Drug Abuse Warning Network, 2011: national estimates of drug-related emergency department visits. HHS Publication No. (SMA) 13-4760, DAWN Series D-39. Rockville (MD): SAMHSA; 2013. www.samhsa.gov/data/sites/default/files/DAWN2k11ED/DAWN2k11ED/DAWN2k11ED.pdf. Accessed October 28, 2020.

3. Compton WM, Jones CM, Baldwin GT. Relationship between nonmedical prescription-opioid use and heroin use. N Engl J Med. 2016;374:154-63.

4. National Center for Health Statistics. NCHS data on drug-poisoning deaths. NCHS Factsheet. https://www.cdc.gov/nchs/data/factsheets/factsheet-drug-poisoning-H.pdf. Accessed October 28, 2020.

5. National Institute on Drug Abuse. America’s addiction to opioids: heroin and prescription drug abuse. Bethesda (MD): NIDA; 2014. www.drugabuse.gov/about-nida/legislative-activities/testimony-to-congress/2016/americas-addiction-to-opioids-heroin-prescription-drug-abuse. Accessed October 28, 2020.

6. Substance Abuse and Mental Health Services Administration (SAMHSA). Results from the 2010 National Survey on Drug Use and Health: Summary of National Findings. Rockville, MD: SAMHSA, 2011 Contract No.: HHS Publication no. (SMA) 11–4658.

7. Maeda A, Bateman BT, Clancy CR, et al. Opioid abuse and dependence during pregnancy: temporal trends and obstetrical outcomes. Anesthesiology. 2014;121:1158-1165.

8. Whiteman VE, Salemi JL, Mogos MF, et al. Maternal opioid drug use during pregnancy and its impact on perinatal morbidity, mortality, and the costs of medical care in the United States. J Pregnancy. 2014;2014:1-8

9. Pregnancy Mortality Surveillance System. www.cdc.gov/reproductivehealth/maternal-mortality/pregnancy-mortality-surveillance-system.htm#trends. Accessed February 4, 2020.

10. Macdorman MF, Declercq E, Cabral H, Morton C. Recent increases in the U.S. maternal mortality rate. Obstet Gynecol. 2016;128:447-455.

11. Texas Health and Human Services. Maternal Mortality and Morbidity Task Force and Department of State Health Services Joint Biennial Report, September 2018. www.dshs.texas.gov/legislative/2018-Reports/MMMTFJointReport2018.pdf

12. Virginia Department of Health. Pregnancy-associated deaths from drug overdose in Virginia, 1999-2007: a report from the Virginia Maternal Mortality Review Team. Richmond, VA: VDH; 2015. www.vdh.virginia.gov/content/uploads/sites/18/2016/04/Final-Pregnancy-Associated-Deaths-Due-to-Drug-Overdose.pdf. Accessed October 28, 2020. 

13. Maryland Department of Health and Mental Hygiene. Maryland maternal mortality review 2016 annual report. Baltimore: DHMH; 2016. https://phpa.health.maryland.gov/Documents/Maryland-Maternal-Mortality-Review-2016-Report.pdf. Accessed October 28, 2020.

14. Desai RJ, Hernandez-Diaz S, Bateman BT, Huybrechts KF. Increase in prescription opioid use during pregnancy among Medicaid-enrolled women. Obstet Gynecol. 2014;123:997-1002.

15. Reddy UM, Davis JM, Ren Z, et al. Opioid use in pregnancy, neonatal abstinence syndrome, and childhood outcomes. Obstet Gynecol Survey. 2017;72:703-705.

16. Patrick SW, Davis MM, Lehmann CU, Cooper WO. Increasing incidence and geographic distribution of neonatal abstinence syndrome: United States 2009 to 2012. J Perinatol. 2015;35:650-655.

17. Center for Substance Abuse Treatment. Medication-assisted treatment for opioid addiction during pregnancy. In: Medication-assisted treatment for opioid addiction in opioid treatment programs. Treatment Improvement Protocol (TIP) Series, No. 43. Rockville, MD: Substance Abuse and Mental Health Services Administration; 2005:211-224.

18. Wojnar-Horton RE, Kristensen JH, Yapp P, et al. Methadone distribution and excretion into breast milk of clients in a methadone maintenance programme. Br J Clin Pharmacol. 1997;44:543-547.

19. Reece-Stremtan S, Marinelli KA. ABM clinical protocol #21: guidelines for breastfeeding and substance use or substance use disorder, revised 2015. Breastfeed Med. 2015;10:135-141.

20. Sachs HC. The transfer of drugs and therapeutics into human breast milk: an update on selected topics. Committee on Drugs. Pediatrics. 2013;132:e796-809.

21. Bagley SM, Wachman EM, Holland E, Brogly SB. Review of the assessment and management of neonatal abstinence syndrome. Addict Sci Clin Pract. 2014;9:19.

22. Opioid use and opioid use disorder in pregnancy. Committee Opinion No. 711. Obstet Gynecol. 2017;130:488-489.

23. Gopman S. Prenatal and postpartum care of women with substance use disorders. Obstet Gynecol Clin North Am. 2014;41:213-228.

24. Mitchell M, Severtson S, Latimer W. Pregnancy and race/ethnicity as predictors of motivation for drug treatment. Am J Drug Alcohol Abuse. 2008;34:397-404.

Article PDF
Issue
Journal of Clinical Outcomes Management - 27(6)
Publications
Topics
Page Number
e4-e9
Sections
Article PDF
Article PDF

From the UT Health Long School of Medicine San Antonio, Texas.

Abstract

Objective: To educate pregnant patients with opioid use disorder (OUD) about the effects of opioids in order to improve understanding and help achieve sustained abstinence.

Methods: The Center for Health Care Services and University Hospital System (UHS) in San Antonio, TX, jointly operate a methadone clinic, the Mommies Program, for pregnant women with OUD. As part of this program, medical students discuss methadone use in pregnancy and the health effects of OUD on mother and child and review resources for sustained peripartum abstinence at prenatal visits with pregnant patients with OUD concurrently enrolled in the program. To evaluate the impact of this education intervention, students assessed patients’ knowledge of methadone effects on mother and baby, state laws concerning heroin and methadone use in pregnancy, and motivation to quit using opioids with pre- and post-intervention surveys.

Results: Of 68 women enrolled in the program, 33 completed both the pre-survey and the post-survey (48.5%). Nearly half (48%) were very motivated to quit before pregnancy, but 85% were very motivated to quit once pregnant. All participants said learning more about the effects of opiates would increase motivation for sobriety. Prior to the educational intervention, 39% of participants knew it was safe to breastfeed on methadone, which improved to 97% in the post-survey, and 76% incorrectly thought they would be reported to authorities by their health care providers if they used illegal drugs during pregnancy, while in the post-survey, 100% knew they would not be reported for doing so.

Conclusion: Pregnancy and education about opioids increased patients’ motivation to quit. Patients also advanced their breastfeeding knowledge and learned about patient-provider confidentiality. Our greatest challenge was participant follow-up; however, this improved with the help of a full-time Mommies Program nurse. Our future aim is to increase project awareness and extend the educational research.

Keywords: pregnancy; addiction; opioids; OUD; counseling.

In 2012 more than 259 million prescriptions for opioids were written in the United States, which was a 200% increase since 1998.1 Since the early 2000s, admissions to opioid substance abuse programs and the death rate from opioids have quadrupled.2-4 Specifically, the rate of heroin use increased more than 300% from 2010 to 2014.5 Opioid use in pregnancy has also escalated in recent years, with a 3- to 4-fold increase from 2000 to 2009 and with 4 in 1000 deliveries being complicated by opioid use disorder (OUD) in 2011.6-8

Between 2000 and 2014, the maternal mortality rate in the United States increased 24%, making it the only industrialized nation with a maternal mortality rate that is rising rather than falling.9 The Texas Maternal Mortality and Morbidity Task Force found that between 2012 and 2015 drug overdose was the leading cause of maternal death in the period from delivery to 365 days postpartum, and it has increased dramatically since 2010.10,11

 

 

In addition, maternal mortality reviews in several states have identified substance use as a major risk factor for pregnancy-associated deaths.12,13 In Texas between 2012 and 2015, opioids were found in 58% of maternal drug overdoses.10 In 2007, 22.8% of women who were enrolled in Medicaid programs in 46 states filled an opioid prescription during pregnancy.14 Additionally, the rising prevalence of opioid use in pregnancy has led to a sharp increase in neonatal abstinence syndrome (NAS), rising from 1.5 cases per 1000 hospital births in 1999 to 6.0 per 1000 hospital births in 2013.15 Unsurprisingly, states with the highest rates of opioid prescribing also have the highest rates of NAS.16

Methadone combined with counseling and behavioral therapy has been the standard of care for the treatment of OUD in pregnancy since the 1970s. Methadone treatment prevents opioid withdrawal symptoms and increases adherence to prenatal care.17 One of the largest methadone treatment clinics in the San Antonio, TX, area is the Center for Health Care Services (CHCS). University Health System in San Antonio (UHS) has established a clinic called The Mommies Program, where mothers addicted to opioids can receive prenatal care by a dedicated physician, registered nurse, and a certified nurse midwife, who work in collaboration with the CHCS methadone clinic. Pregnant patients with OUD in pregnancy are concurrently enrolled in the Mommies Program and receive prenatal care through UHS and methadone treatment and counseling through CHCS. The continuity effort aims to increase prenatal care rates and adherence to methadone treatment.

Once mothers are off illicit opioids and on methadone, it is essential to discuss breastfeeding with them, as many mothers addicted to illicit opioids may have been told that they should not be breastfeeding. However, breastfeeding should be encouraged in women who are stable on methadone if they are not using illicit drugs and do not have other contraindications, regardless of maternal methadone dose, since the transfer of methadone into breast milk is minimal.18-20 Breastfeeding is beneficial in women taking methadone and has been associated with decreased severity of NAS symptoms, decreased need for pharmacotherapy, and a shorter hospital stay for the baby.21 In addition, breastfeeding contributes to the development of an attachment between mother and infant, while also providing the infant with natural immunity. Women should be counseled about the need to stop breastfeeding in the event of a relapse.22

Finally, the postpartum period represents a time of increased stressors, such as loss of sleep, child protective services involvement, and frustration with constant demands from new baby. For mothers with addiction, this is an especially sensitive time, as the stressors may be exacerbated by their new sobriety and a sudden end to the motivation they experienced from pregnancy.23 Therefore, early and frequent postpartum care with methadone dose evaluation is essential in order to decrease drug relapse and screen for postpartum depression in detail, since patients with a history of drug use are at increased risk of postpartum depression.

In 2017 medical students at UT Health Long School of Medicine in San Antonio created a project to educate women about OUD in pregnancy and provide motivational incentives for sustained abstinence; this project has continued each year since. Students provide education about methadone treatment and the dangers of using illicit opioids during and after pregnancy. Students especially focus on educating patients on the key problem areas in the literature, such as overdose, NAS, breastfeeding, postpartum substance use, and postpartum depression.

 

 

Methods

From October 2018 to February 2020, a total of 15 medical students volunteered between 1 and 20 times at the Mommies Program clinic, which was held once or twice per week from 8 am to 12 pm. Prior to attending clinic, a fourth-year medical student oriented that year’s group of first-, second-, and third-year medical students who volunteered to attend the clinic. The orientation introduced students to the Mommies Program and instructed them on how and why methadone is prescribed and distributed. Students were also taught how to begin and proceed through each patient encounter, and were given a standard handout to review with patients. This handout made it possible for students of every level to attend the Mommies Program and ensured that the education was standardized at every session (Figure 1).

Handout used by medical students during discussion with participants in the Mommies Program

The only inclusion criteria for participating in the educational intervention and survey was participants had to be 18 years of age or older and enrolled in the Mommies Program. Patients who met the inclusion criteria and agreed to participate completed a pre-survey administered by the students during the patient’s initial prenatal visit (Figure 2). This survey collected baseline information about the patient’s history with opioid use and their current knowledge of methadone treatment, NAS, legal aspects of drug use disclosure, and drug testing prior to the education portion of the encounter. After the pre-survey was administered, students spent 30 minutes reviewing the correct answers of the survey with the patients by utilizing the standardized handout to help patients understand details of methadone and opioid use in pregnancy (Figure 1). The post-survey was administered by a student once patients entered the third trimester to assess whether the education session increased patients’ knowledge of these topics.

Survey administered before and after educational intervention

At the time patients completed the post-survey, they received a Baby Bag as well as education regarding each item in the bag. The aim of distributing Baby Bags was to relieve some possible postnatal stressors and educate the patients about infant care. Items included in the bag were diapers, wipes, bottles, clothes, and swaddles. Prenatal vitamins were added in January 2020, as many patients struggle to afford vitamins if they are not currently covered by Medicaid or have other barriers. The Baby Bag items were purchased through a Community Service Learning grant through UT Health San Antonio.

Results

Of 68 women enrolled in the Mommies Program during the intervention period, 33 completed the pre-survey and the post-survey (48.5%). Even though all patients enrolled in the program met the inclusion criteria, patients were not included in the educational program for multiple reasons, including refusal to participate, poor clinic follow-up, or lack of students to collect surveys. However, all patients who completed the pre-survey did complete the post-survey. In the pre-survey, only 39% of participants knew it was safe to breastfeed while on methadone. In the post-survey, 97% knew it safe to breastfeed. Nearly half (48%) reported being very motivated to quit opioids before pregnancy, but 85% were very motivated to quit once pregnant. In the pre-survey, 76% incorrectly thought they would be reported to authorities by their health providers if they used illegal drugs during pregnancy, while in the post-survey, 100% knew they would not be reported for doing so. Also, all participants said learning more about the effects of opiates would increase motivation for sobriety.

 

 

Discussion

Questions assessed during the educational surveys revolved around patients’ knowledge of the intricacies, legally and physiologically, of methadone treatment for OUD, as well as beneficial aspects for patients and future child health, such as breastfeeding and motivation to quit and stay sober.

It was clear that there was a lack of knowledge and education about breastfeeding, as only 39% of the participants thought that it was safe to breastfeed while on methadone in the pre-survey; in the post survey, this improved to 97%. Students spent a large portion of the educational time going over the safety of breastfeeding for patients on methadone and the many benefits to mother and baby. Students also reviewed breastfeeding with patients every time patients came in for a visit and debunked any falsehoods about the negatives of breastfeeding while on methadone. This is another testament to the benefits of reinforcement around patient education.

The area of trust between provider and patient is essential in all provider-patient relationships. However, in the area of addiction, a trusting bond is especially important, as patients must feel confident and comfortable to disclose every aspect of their lives so the provider can give the best care. It was clear from our initial data that many patients did not feel this trust or understand the legal aspects regarding the provider-patient relationship in the terms of drug use, as the pre-survey shows 76% of patients originally thought they would be reported to authorities if they told their provider they used illegal drugs during pregnancy. This was an enormous issue in the clinic and something that needed to be addressed because, based on these data, we feared many patients would not be honest about using illegal drugs to supplement their methadone if they believed they would be reported to the authorities or even jailed. The medical student education team continually assured patients that their honesty about illegal drug use during pregnancy would not be revealed to the authorities, and also made it clear to patients that it was essential they were honest about illegal drug use so the optimal care could be provided by the team. These discussions were successful, as the post-survey showed that 100% of patients knew they would not be reported to the authorities if they used illegal drugs during the pregnancy. This showed an increase in knowledge, but also suggested an increase in confidence in the provider-patient relationship by patients, which we speculate allowed for a better patient experience, better patient outcomes, and less emotional stress for the patient and provider.

Last, we wanted to study and address the motivation to quit using drugs and stay sober through learning about the effects of opiates and how this motivation was related to pregnancy. A study by Mitchell et al makes clear that pregnancy is a motivation to seek treatment for drug use and to quit,24 and our survey data support these findings, with 48% of patients motivated to quit before they were pregnant and 85% motivated to quit once they knew they were pregnant. In addition, all patients attested on the pre- and post-survey that learning more about opioids would increase their motivation for sobriety. Therefore, we believe education about the use of opioids and other drugs is a strong motivation towards sobriety and should be further studied in methadone treatment and other drugs as well.

We will continue to focus on sobriety postpartum by using the education in pregnancy as a springboard to further postpartum education, as education seems to be very beneficial to future sobriety. In the future, we believe extending the educational program past pregnancy and discussing opioid use and addiction with patients at multiple follow-up visits will be beneficial to patients’ sobriety.

 

 

We faced 2 main challenges in implementing this intervention and survey: patients would often miss multiple appointments during their third trimester or would not attend their postpartum visit if they only had 1 prenatal visit; and many clinic sessions had low student attendance because students often had many other responsibilities in medical school and there were only 15 volunteers over the study time. These challenges decreased our post-survey completion rate. However, there has been improvement in follow-up as the project has continued. The Mommies Program now has a full-time registered nurse, and a larger number of medical student teachers have volunteered to attend the clinic. In the future, we aim to increase awareness of our project and the benefits of participation, expand advertising at our medical school to increase student participation, and increase follow-up education in the postpartum period.

Another future direction is to include local, free doula services, which are offered through Catholic Charities in San Antonio. Doulas provide antepartum, intrapartum, and postpartum services, which we believe will help our patients through advocacy and support for sobriety during this emotional and stressful time.

Conclusion

Counseled participants were receptive to learning about the effects of OUD and methadone on themselves and their newborn. Participants unanimously stated that learning more about OUD increased their motivation for sobriety. It was also clear that the increased motivation to be sober during pregnancy, as compared to before pregnancy, is an opportunity to help these women take steps to get sober. Patients also advanced their breastfeeding knowledge, as we helped debunk falsehoods surrounding breastfeeding while on methadone, and we anticipate this will lead to greater breastfeeding rates for our patients on methadone, although this was not specifically studied. Finally, patients learned about patient-provider confidentiality, which allowed for more open and clear communication with patients so they could be cared for to the greatest degree and trust could remain paramount.

Drug use is a common problem in the health care system, and exposure to patients with addiction is important for medical students in training. We believe that attending the Mommies Program allowed medical students to gain exposure and skills to better help patients with OUD.

Corresponding author: Nicholas Stansbury, MD, Nmstansbury85@gmail.com.

Financial disclosures: None.

From the UT Health Long School of Medicine San Antonio, Texas.

Abstract

Objective: To educate pregnant patients with opioid use disorder (OUD) about the effects of opioids in order to improve understanding and help achieve sustained abstinence.

Methods: The Center for Health Care Services and University Hospital System (UHS) in San Antonio, TX, jointly operate a methadone clinic, the Mommies Program, for pregnant women with OUD. As part of this program, medical students discuss methadone use in pregnancy and the health effects of OUD on mother and child and review resources for sustained peripartum abstinence at prenatal visits with pregnant patients with OUD concurrently enrolled in the program. To evaluate the impact of this education intervention, students assessed patients’ knowledge of methadone effects on mother and baby, state laws concerning heroin and methadone use in pregnancy, and motivation to quit using opioids with pre- and post-intervention surveys.

Results: Of 68 women enrolled in the program, 33 completed both the pre-survey and the post-survey (48.5%). Nearly half (48%) were very motivated to quit before pregnancy, but 85% were very motivated to quit once pregnant. All participants said learning more about the effects of opiates would increase motivation for sobriety. Prior to the educational intervention, 39% of participants knew it was safe to breastfeed on methadone, which improved to 97% in the post-survey, and 76% incorrectly thought they would be reported to authorities by their health care providers if they used illegal drugs during pregnancy, while in the post-survey, 100% knew they would not be reported for doing so.

Conclusion: Pregnancy and education about opioids increased patients’ motivation to quit. Patients also advanced their breastfeeding knowledge and learned about patient-provider confidentiality. Our greatest challenge was participant follow-up; however, this improved with the help of a full-time Mommies Program nurse. Our future aim is to increase project awareness and extend the educational research.

Keywords: pregnancy; addiction; opioids; OUD; counseling.

In 2012 more than 259 million prescriptions for opioids were written in the United States, which was a 200% increase since 1998.1 Since the early 2000s, admissions to opioid substance abuse programs and the death rate from opioids have quadrupled.2-4 Specifically, the rate of heroin use increased more than 300% from 2010 to 2014.5 Opioid use in pregnancy has also escalated in recent years, with a 3- to 4-fold increase from 2000 to 2009 and with 4 in 1000 deliveries being complicated by opioid use disorder (OUD) in 2011.6-8

Between 2000 and 2014, the maternal mortality rate in the United States increased 24%, making it the only industrialized nation with a maternal mortality rate that is rising rather than falling.9 The Texas Maternal Mortality and Morbidity Task Force found that between 2012 and 2015 drug overdose was the leading cause of maternal death in the period from delivery to 365 days postpartum, and it has increased dramatically since 2010.10,11

 

 

In addition, maternal mortality reviews in several states have identified substance use as a major risk factor for pregnancy-associated deaths.12,13 In Texas between 2012 and 2015, opioids were found in 58% of maternal drug overdoses.10 In 2007, 22.8% of women who were enrolled in Medicaid programs in 46 states filled an opioid prescription during pregnancy.14 Additionally, the rising prevalence of opioid use in pregnancy has led to a sharp increase in neonatal abstinence syndrome (NAS), rising from 1.5 cases per 1000 hospital births in 1999 to 6.0 per 1000 hospital births in 2013.15 Unsurprisingly, states with the highest rates of opioid prescribing also have the highest rates of NAS.16

Methadone combined with counseling and behavioral therapy has been the standard of care for the treatment of OUD in pregnancy since the 1970s. Methadone treatment prevents opioid withdrawal symptoms and increases adherence to prenatal care.17 One of the largest methadone treatment clinics in the San Antonio, TX, area is the Center for Health Care Services (CHCS). University Health System in San Antonio (UHS) has established a clinic called The Mommies Program, where mothers addicted to opioids can receive prenatal care by a dedicated physician, registered nurse, and a certified nurse midwife, who work in collaboration with the CHCS methadone clinic. Pregnant patients with OUD in pregnancy are concurrently enrolled in the Mommies Program and receive prenatal care through UHS and methadone treatment and counseling through CHCS. The continuity effort aims to increase prenatal care rates and adherence to methadone treatment.

Once mothers are off illicit opioids and on methadone, it is essential to discuss breastfeeding with them, as many mothers addicted to illicit opioids may have been told that they should not be breastfeeding. However, breastfeeding should be encouraged in women who are stable on methadone if they are not using illicit drugs and do not have other contraindications, regardless of maternal methadone dose, since the transfer of methadone into breast milk is minimal.18-20 Breastfeeding is beneficial in women taking methadone and has been associated with decreased severity of NAS symptoms, decreased need for pharmacotherapy, and a shorter hospital stay for the baby.21 In addition, breastfeeding contributes to the development of an attachment between mother and infant, while also providing the infant with natural immunity. Women should be counseled about the need to stop breastfeeding in the event of a relapse.22

Finally, the postpartum period represents a time of increased stressors, such as loss of sleep, child protective services involvement, and frustration with constant demands from new baby. For mothers with addiction, this is an especially sensitive time, as the stressors may be exacerbated by their new sobriety and a sudden end to the motivation they experienced from pregnancy.23 Therefore, early and frequent postpartum care with methadone dose evaluation is essential in order to decrease drug relapse and screen for postpartum depression in detail, since patients with a history of drug use are at increased risk of postpartum depression.

In 2017 medical students at UT Health Long School of Medicine in San Antonio created a project to educate women about OUD in pregnancy and provide motivational incentives for sustained abstinence; this project has continued each year since. Students provide education about methadone treatment and the dangers of using illicit opioids during and after pregnancy. Students especially focus on educating patients on the key problem areas in the literature, such as overdose, NAS, breastfeeding, postpartum substance use, and postpartum depression.

 

 

Methods

From October 2018 to February 2020, a total of 15 medical students volunteered between 1 and 20 times at the Mommies Program clinic, which was held once or twice per week from 8 am to 12 pm. Prior to attending clinic, a fourth-year medical student oriented that year’s group of first-, second-, and third-year medical students who volunteered to attend the clinic. The orientation introduced students to the Mommies Program and instructed them on how and why methadone is prescribed and distributed. Students were also taught how to begin and proceed through each patient encounter, and were given a standard handout to review with patients. This handout made it possible for students of every level to attend the Mommies Program and ensured that the education was standardized at every session (Figure 1).

Handout used by medical students during discussion with participants in the Mommies Program

The only inclusion criteria for participating in the educational intervention and survey was participants had to be 18 years of age or older and enrolled in the Mommies Program. Patients who met the inclusion criteria and agreed to participate completed a pre-survey administered by the students during the patient’s initial prenatal visit (Figure 2). This survey collected baseline information about the patient’s history with opioid use and their current knowledge of methadone treatment, NAS, legal aspects of drug use disclosure, and drug testing prior to the education portion of the encounter. After the pre-survey was administered, students spent 30 minutes reviewing the correct answers of the survey with the patients by utilizing the standardized handout to help patients understand details of methadone and opioid use in pregnancy (Figure 1). The post-survey was administered by a student once patients entered the third trimester to assess whether the education session increased patients’ knowledge of these topics.

Survey administered before and after educational intervention

At the time patients completed the post-survey, they received a Baby Bag as well as education regarding each item in the bag. The aim of distributing Baby Bags was to relieve some possible postnatal stressors and educate the patients about infant care. Items included in the bag were diapers, wipes, bottles, clothes, and swaddles. Prenatal vitamins were added in January 2020, as many patients struggle to afford vitamins if they are not currently covered by Medicaid or have other barriers. The Baby Bag items were purchased through a Community Service Learning grant through UT Health San Antonio.

Results

Of 68 women enrolled in the Mommies Program during the intervention period, 33 completed the pre-survey and the post-survey (48.5%). Even though all patients enrolled in the program met the inclusion criteria, patients were not included in the educational program for multiple reasons, including refusal to participate, poor clinic follow-up, or lack of students to collect surveys. However, all patients who completed the pre-survey did complete the post-survey. In the pre-survey, only 39% of participants knew it was safe to breastfeed while on methadone. In the post-survey, 97% knew it safe to breastfeed. Nearly half (48%) reported being very motivated to quit opioids before pregnancy, but 85% were very motivated to quit once pregnant. In the pre-survey, 76% incorrectly thought they would be reported to authorities by their health providers if they used illegal drugs during pregnancy, while in the post-survey, 100% knew they would not be reported for doing so. Also, all participants said learning more about the effects of opiates would increase motivation for sobriety.

 

 

Discussion

Questions assessed during the educational surveys revolved around patients’ knowledge of the intricacies, legally and physiologically, of methadone treatment for OUD, as well as beneficial aspects for patients and future child health, such as breastfeeding and motivation to quit and stay sober.

It was clear that there was a lack of knowledge and education about breastfeeding, as only 39% of the participants thought that it was safe to breastfeed while on methadone in the pre-survey; in the post survey, this improved to 97%. Students spent a large portion of the educational time going over the safety of breastfeeding for patients on methadone and the many benefits to mother and baby. Students also reviewed breastfeeding with patients every time patients came in for a visit and debunked any falsehoods about the negatives of breastfeeding while on methadone. This is another testament to the benefits of reinforcement around patient education.

The area of trust between provider and patient is essential in all provider-patient relationships. However, in the area of addiction, a trusting bond is especially important, as patients must feel confident and comfortable to disclose every aspect of their lives so the provider can give the best care. It was clear from our initial data that many patients did not feel this trust or understand the legal aspects regarding the provider-patient relationship in the terms of drug use, as the pre-survey shows 76% of patients originally thought they would be reported to authorities if they told their provider they used illegal drugs during pregnancy. This was an enormous issue in the clinic and something that needed to be addressed because, based on these data, we feared many patients would not be honest about using illegal drugs to supplement their methadone if they believed they would be reported to the authorities or even jailed. The medical student education team continually assured patients that their honesty about illegal drug use during pregnancy would not be revealed to the authorities, and also made it clear to patients that it was essential they were honest about illegal drug use so the optimal care could be provided by the team. These discussions were successful, as the post-survey showed that 100% of patients knew they would not be reported to the authorities if they used illegal drugs during the pregnancy. This showed an increase in knowledge, but also suggested an increase in confidence in the provider-patient relationship by patients, which we speculate allowed for a better patient experience, better patient outcomes, and less emotional stress for the patient and provider.

Last, we wanted to study and address the motivation to quit using drugs and stay sober through learning about the effects of opiates and how this motivation was related to pregnancy. A study by Mitchell et al makes clear that pregnancy is a motivation to seek treatment for drug use and to quit,24 and our survey data support these findings, with 48% of patients motivated to quit before they were pregnant and 85% motivated to quit once they knew they were pregnant. In addition, all patients attested on the pre- and post-survey that learning more about opioids would increase their motivation for sobriety. Therefore, we believe education about the use of opioids and other drugs is a strong motivation towards sobriety and should be further studied in methadone treatment and other drugs as well.

We will continue to focus on sobriety postpartum by using the education in pregnancy as a springboard to further postpartum education, as education seems to be very beneficial to future sobriety. In the future, we believe extending the educational program past pregnancy and discussing opioid use and addiction with patients at multiple follow-up visits will be beneficial to patients’ sobriety.

 

 

We faced 2 main challenges in implementing this intervention and survey: patients would often miss multiple appointments during their third trimester or would not attend their postpartum visit if they only had 1 prenatal visit; and many clinic sessions had low student attendance because students often had many other responsibilities in medical school and there were only 15 volunteers over the study time. These challenges decreased our post-survey completion rate. However, there has been improvement in follow-up as the project has continued. The Mommies Program now has a full-time registered nurse, and a larger number of medical student teachers have volunteered to attend the clinic. In the future, we aim to increase awareness of our project and the benefits of participation, expand advertising at our medical school to increase student participation, and increase follow-up education in the postpartum period.

Another future direction is to include local, free doula services, which are offered through Catholic Charities in San Antonio. Doulas provide antepartum, intrapartum, and postpartum services, which we believe will help our patients through advocacy and support for sobriety during this emotional and stressful time.

Conclusion

Counseled participants were receptive to learning about the effects of OUD and methadone on themselves and their newborn. Participants unanimously stated that learning more about OUD increased their motivation for sobriety. It was also clear that the increased motivation to be sober during pregnancy, as compared to before pregnancy, is an opportunity to help these women take steps to get sober. Patients also advanced their breastfeeding knowledge, as we helped debunk falsehoods surrounding breastfeeding while on methadone, and we anticipate this will lead to greater breastfeeding rates for our patients on methadone, although this was not specifically studied. Finally, patients learned about patient-provider confidentiality, which allowed for more open and clear communication with patients so they could be cared for to the greatest degree and trust could remain paramount.

Drug use is a common problem in the health care system, and exposure to patients with addiction is important for medical students in training. We believe that attending the Mommies Program allowed medical students to gain exposure and skills to better help patients with OUD.

Corresponding author: Nicholas Stansbury, MD, Nmstansbury85@gmail.com.

Financial disclosures: None.

References

1. Centers for Disease Control and Prevention. Opioid painkiller prescribing: where you live makes a difference. CDC website. www.cdc.gov/vitalsigns/opioid-prescribing. Accessed October 28, 2020. 

2. Substance Abuse and Mental Health Services Administration. Drug Abuse Warning Network, 2011: national estimates of drug-related emergency department visits. HHS Publication No. (SMA) 13-4760, DAWN Series D-39. Rockville (MD): SAMHSA; 2013. www.samhsa.gov/data/sites/default/files/DAWN2k11ED/DAWN2k11ED/DAWN2k11ED.pdf. Accessed October 28, 2020.

3. Compton WM, Jones CM, Baldwin GT. Relationship between nonmedical prescription-opioid use and heroin use. N Engl J Med. 2016;374:154-63.

4. National Center for Health Statistics. NCHS data on drug-poisoning deaths. NCHS Factsheet. https://www.cdc.gov/nchs/data/factsheets/factsheet-drug-poisoning-H.pdf. Accessed October 28, 2020.

5. National Institute on Drug Abuse. America’s addiction to opioids: heroin and prescription drug abuse. Bethesda (MD): NIDA; 2014. www.drugabuse.gov/about-nida/legislative-activities/testimony-to-congress/2016/americas-addiction-to-opioids-heroin-prescription-drug-abuse. Accessed October 28, 2020.

6. Substance Abuse and Mental Health Services Administration (SAMHSA). Results from the 2010 National Survey on Drug Use and Health: Summary of National Findings. Rockville, MD: SAMHSA, 2011 Contract No.: HHS Publication no. (SMA) 11–4658.

7. Maeda A, Bateman BT, Clancy CR, et al. Opioid abuse and dependence during pregnancy: temporal trends and obstetrical outcomes. Anesthesiology. 2014;121:1158-1165.

8. Whiteman VE, Salemi JL, Mogos MF, et al. Maternal opioid drug use during pregnancy and its impact on perinatal morbidity, mortality, and the costs of medical care in the United States. J Pregnancy. 2014;2014:1-8

9. Pregnancy Mortality Surveillance System. www.cdc.gov/reproductivehealth/maternal-mortality/pregnancy-mortality-surveillance-system.htm#trends. Accessed February 4, 2020.

10. Macdorman MF, Declercq E, Cabral H, Morton C. Recent increases in the U.S. maternal mortality rate. Obstet Gynecol. 2016;128:447-455.

11. Texas Health and Human Services. Maternal Mortality and Morbidity Task Force and Department of State Health Services Joint Biennial Report, September 2018. www.dshs.texas.gov/legislative/2018-Reports/MMMTFJointReport2018.pdf

12. Virginia Department of Health. Pregnancy-associated deaths from drug overdose in Virginia, 1999-2007: a report from the Virginia Maternal Mortality Review Team. Richmond, VA: VDH; 2015. www.vdh.virginia.gov/content/uploads/sites/18/2016/04/Final-Pregnancy-Associated-Deaths-Due-to-Drug-Overdose.pdf. Accessed October 28, 2020. 

13. Maryland Department of Health and Mental Hygiene. Maryland maternal mortality review 2016 annual report. Baltimore: DHMH; 2016. https://phpa.health.maryland.gov/Documents/Maryland-Maternal-Mortality-Review-2016-Report.pdf. Accessed October 28, 2020.

14. Desai RJ, Hernandez-Diaz S, Bateman BT, Huybrechts KF. Increase in prescription opioid use during pregnancy among Medicaid-enrolled women. Obstet Gynecol. 2014;123:997-1002.

15. Reddy UM, Davis JM, Ren Z, et al. Opioid use in pregnancy, neonatal abstinence syndrome, and childhood outcomes. Obstet Gynecol Survey. 2017;72:703-705.

16. Patrick SW, Davis MM, Lehmann CU, Cooper WO. Increasing incidence and geographic distribution of neonatal abstinence syndrome: United States 2009 to 2012. J Perinatol. 2015;35:650-655.

17. Center for Substance Abuse Treatment. Medication-assisted treatment for opioid addiction during pregnancy. In: Medication-assisted treatment for opioid addiction in opioid treatment programs. Treatment Improvement Protocol (TIP) Series, No. 43. Rockville, MD: Substance Abuse and Mental Health Services Administration; 2005:211-224.

18. Wojnar-Horton RE, Kristensen JH, Yapp P, et al. Methadone distribution and excretion into breast milk of clients in a methadone maintenance programme. Br J Clin Pharmacol. 1997;44:543-547.

19. Reece-Stremtan S, Marinelli KA. ABM clinical protocol #21: guidelines for breastfeeding and substance use or substance use disorder, revised 2015. Breastfeed Med. 2015;10:135-141.

20. Sachs HC. The transfer of drugs and therapeutics into human breast milk: an update on selected topics. Committee on Drugs. Pediatrics. 2013;132:e796-809.

21. Bagley SM, Wachman EM, Holland E, Brogly SB. Review of the assessment and management of neonatal abstinence syndrome. Addict Sci Clin Pract. 2014;9:19.

22. Opioid use and opioid use disorder in pregnancy. Committee Opinion No. 711. Obstet Gynecol. 2017;130:488-489.

23. Gopman S. Prenatal and postpartum care of women with substance use disorders. Obstet Gynecol Clin North Am. 2014;41:213-228.

24. Mitchell M, Severtson S, Latimer W. Pregnancy and race/ethnicity as predictors of motivation for drug treatment. Am J Drug Alcohol Abuse. 2008;34:397-404.

References

1. Centers for Disease Control and Prevention. Opioid painkiller prescribing: where you live makes a difference. CDC website. www.cdc.gov/vitalsigns/opioid-prescribing. Accessed October 28, 2020. 

2. Substance Abuse and Mental Health Services Administration. Drug Abuse Warning Network, 2011: national estimates of drug-related emergency department visits. HHS Publication No. (SMA) 13-4760, DAWN Series D-39. Rockville (MD): SAMHSA; 2013. www.samhsa.gov/data/sites/default/files/DAWN2k11ED/DAWN2k11ED/DAWN2k11ED.pdf. Accessed October 28, 2020.

3. Compton WM, Jones CM, Baldwin GT. Relationship between nonmedical prescription-opioid use and heroin use. N Engl J Med. 2016;374:154-63.

4. National Center for Health Statistics. NCHS data on drug-poisoning deaths. NCHS Factsheet. https://www.cdc.gov/nchs/data/factsheets/factsheet-drug-poisoning-H.pdf. Accessed October 28, 2020.

5. National Institute on Drug Abuse. America’s addiction to opioids: heroin and prescription drug abuse. Bethesda (MD): NIDA; 2014. www.drugabuse.gov/about-nida/legislative-activities/testimony-to-congress/2016/americas-addiction-to-opioids-heroin-prescription-drug-abuse. Accessed October 28, 2020.

6. Substance Abuse and Mental Health Services Administration (SAMHSA). Results from the 2010 National Survey on Drug Use and Health: Summary of National Findings. Rockville, MD: SAMHSA, 2011 Contract No.: HHS Publication no. (SMA) 11–4658.

7. Maeda A, Bateman BT, Clancy CR, et al. Opioid abuse and dependence during pregnancy: temporal trends and obstetrical outcomes. Anesthesiology. 2014;121:1158-1165.

8. Whiteman VE, Salemi JL, Mogos MF, et al. Maternal opioid drug use during pregnancy and its impact on perinatal morbidity, mortality, and the costs of medical care in the United States. J Pregnancy. 2014;2014:1-8

9. Pregnancy Mortality Surveillance System. www.cdc.gov/reproductivehealth/maternal-mortality/pregnancy-mortality-surveillance-system.htm#trends. Accessed February 4, 2020.

10. Macdorman MF, Declercq E, Cabral H, Morton C. Recent increases in the U.S. maternal mortality rate. Obstet Gynecol. 2016;128:447-455.

11. Texas Health and Human Services. Maternal Mortality and Morbidity Task Force and Department of State Health Services Joint Biennial Report, September 2018. www.dshs.texas.gov/legislative/2018-Reports/MMMTFJointReport2018.pdf

12. Virginia Department of Health. Pregnancy-associated deaths from drug overdose in Virginia, 1999-2007: a report from the Virginia Maternal Mortality Review Team. Richmond, VA: VDH; 2015. www.vdh.virginia.gov/content/uploads/sites/18/2016/04/Final-Pregnancy-Associated-Deaths-Due-to-Drug-Overdose.pdf. Accessed October 28, 2020. 

13. Maryland Department of Health and Mental Hygiene. Maryland maternal mortality review 2016 annual report. Baltimore: DHMH; 2016. https://phpa.health.maryland.gov/Documents/Maryland-Maternal-Mortality-Review-2016-Report.pdf. Accessed October 28, 2020.

14. Desai RJ, Hernandez-Diaz S, Bateman BT, Huybrechts KF. Increase in prescription opioid use during pregnancy among Medicaid-enrolled women. Obstet Gynecol. 2014;123:997-1002.

15. Reddy UM, Davis JM, Ren Z, et al. Opioid use in pregnancy, neonatal abstinence syndrome, and childhood outcomes. Obstet Gynecol Survey. 2017;72:703-705.

16. Patrick SW, Davis MM, Lehmann CU, Cooper WO. Increasing incidence and geographic distribution of neonatal abstinence syndrome: United States 2009 to 2012. J Perinatol. 2015;35:650-655.

17. Center for Substance Abuse Treatment. Medication-assisted treatment for opioid addiction during pregnancy. In: Medication-assisted treatment for opioid addiction in opioid treatment programs. Treatment Improvement Protocol (TIP) Series, No. 43. Rockville, MD: Substance Abuse and Mental Health Services Administration; 2005:211-224.

18. Wojnar-Horton RE, Kristensen JH, Yapp P, et al. Methadone distribution and excretion into breast milk of clients in a methadone maintenance programme. Br J Clin Pharmacol. 1997;44:543-547.

19. Reece-Stremtan S, Marinelli KA. ABM clinical protocol #21: guidelines for breastfeeding and substance use or substance use disorder, revised 2015. Breastfeed Med. 2015;10:135-141.

20. Sachs HC. The transfer of drugs and therapeutics into human breast milk: an update on selected topics. Committee on Drugs. Pediatrics. 2013;132:e796-809.

21. Bagley SM, Wachman EM, Holland E, Brogly SB. Review of the assessment and management of neonatal abstinence syndrome. Addict Sci Clin Pract. 2014;9:19.

22. Opioid use and opioid use disorder in pregnancy. Committee Opinion No. 711. Obstet Gynecol. 2017;130:488-489.

23. Gopman S. Prenatal and postpartum care of women with substance use disorders. Obstet Gynecol Clin North Am. 2014;41:213-228.

24. Mitchell M, Severtson S, Latimer W. Pregnancy and race/ethnicity as predictors of motivation for drug treatment. Am J Drug Alcohol Abuse. 2008;34:397-404.

Issue
Journal of Clinical Outcomes Management - 27(6)
Issue
Journal of Clinical Outcomes Management - 27(6)
Page Number
e4-e9
Page Number
e4-e9
Publications
Publications
Topics
Article Type
Display Headline
Addressing Maternal Mortality Through Education: The Mommies Methadone Program
Display Headline
Addressing Maternal Mortality Through Education: The Mommies Methadone Program
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Article PDF Media