User login
Dual therapy serves as well as triple for most HIV patients
based on a meta-analysis including data from more than 5,000 patients.
Although triple therapy remains the standard of care, the availability of more potent drugs has revived interest in dual and mono therapies, wrote Pisaturo Mariantonietta, MD, of the University of Campania Luigi Vanvitelli, Naples, Italy, and colleagues.
In a study published in Clinical Microbiology and Infection, the researchers identified 14 articles including 5,205 treatment-naive HIV adults. The studies were published between 2008 and 2020; 13 were randomized, controlled trials.
The dual therapies used in the studies included atazanavir/r plus maraviroc; lopinavir/r plus lamivudine; raltegravir plus darunavir/r; lopinavir/r plus tenofovir, raltegravir, efavirenz, or maraviroc; atazanavir/r plus raltegravir and darunavir/r plus maraviroc; and dolutegravir plus lamivudine.
Overall, no significant differences occurred in the primary endpoint of treatment failure across 10 studies between dual therapy and triple therapy patients based on data at 48 weeks (relative risk 1.20). “The rate of treatment failure did not differ among the two groups when stratifying the patients according to the drug used in the dual regimen,” the researchers said.
Low viral load’s link to treatment failure
Among 2,398 patients with a low HIV viral load (less than 100,000 copies/mL), dual therapy patients were significantly more likely to experience treatment failure than were triple therapy patients (RR, 1.47, P = .007). No differences were noted between dual and triple therapy failure among patients with high HIV viral loads at baseline. Patterns were similar at 96 weeks, but only three studies included 96-week data, the researchers said.
The rate of discontinuation because of adverse events was not significantly different between the groups at 48 weeks.
The study findings were limited by several factors, including the use of different regimens in the dual strategies, some of which are no longer in use, as well as there being insufficient data to fully compare outcomes at 96 weeks, and lack of information on cerebrospinal fluid viral load, the researchers noted.
However, the results suggest that dual therapy might be considered for HIV-naive patients with a low viral load, they said.
“Further RCTs that will evaluate the efficacy of antiretroviral regimens in use today among difficult-to-treat populations, such as patients with high viral load, including both intention-to-treat and per-protocol analysis, are needed to address this topic,” they concluded.
Consider range of patient factors when choosing therapies
Conducting the study at this time was important because of the expanding options for treating HIV patients, Donna E. Sweet, MD, an HIV specialist and professor of medicine at the University of Kansas, Wichita, said in an interview.
“We now have two single tablet formulations that are dual rather than triple therapy, and as treaters we are all trying to know when to use them,” she explained.
Dr. Sweet said she was not surprised by the study findings, given that well-conducted, randomized, controlled trials allowed the combination therapies to be approved.
Some of the key challenges to identifying the optimal treatment for HIV patients include factoring in the use of concomitant medications that could lead to drug-drug interactions, noted Dr. Sweet, who serves an editorial advisory board member of Internal Medicine News.
The take-home message for clinicians, in her opinion, is that “less drugs may mean less toxicity, but we don’t want to sacrifice efficacy,” she said. “There may be patients who are better suited than others for two vs. three drugs,” Dr. Sweet emphasized.
The next steps for research on the value of dual vs. triple therapy should include longer term efficacy studies, especially in those with lower CD4 counts and higher viral loads, said Dr. Sweet. In addition to factors such as CD4 counts and viral load, the food requirements of certain ART regimens could affect adherence and therefore a clinician decision to use two drugs rather than three, she noted.
Dr. Sweet disclosed past relationships with ViiV, Gilead, Merck, and Janssen on their speakers bureaus, and current advisory roles with Gilead and ViiV.
The study received no outside funding. Lead author Dr. Mariantonietta and several coauthors disclosed relationships with companies including ViiV Healthcare, AbbVie, Janssen-Cilag and Gilead Science, and Merck Sharp & Dohme, but no conflicts in connection with this study.
SOURCE: Mariantonietta P et al. Clin Microbiol Infect. 2020 Oct 5. doi: 10.1016/j.cmi.2020.09.048.
based on a meta-analysis including data from more than 5,000 patients.
Although triple therapy remains the standard of care, the availability of more potent drugs has revived interest in dual and mono therapies, wrote Pisaturo Mariantonietta, MD, of the University of Campania Luigi Vanvitelli, Naples, Italy, and colleagues.
In a study published in Clinical Microbiology and Infection, the researchers identified 14 articles including 5,205 treatment-naive HIV adults. The studies were published between 2008 and 2020; 13 were randomized, controlled trials.
The dual therapies used in the studies included atazanavir/r plus maraviroc; lopinavir/r plus lamivudine; raltegravir plus darunavir/r; lopinavir/r plus tenofovir, raltegravir, efavirenz, or maraviroc; atazanavir/r plus raltegravir and darunavir/r plus maraviroc; and dolutegravir plus lamivudine.
Overall, no significant differences occurred in the primary endpoint of treatment failure across 10 studies between dual therapy and triple therapy patients based on data at 48 weeks (relative risk 1.20). “The rate of treatment failure did not differ among the two groups when stratifying the patients according to the drug used in the dual regimen,” the researchers said.
Low viral load’s link to treatment failure
Among 2,398 patients with a low HIV viral load (less than 100,000 copies/mL), dual therapy patients were significantly more likely to experience treatment failure than were triple therapy patients (RR, 1.47, P = .007). No differences were noted between dual and triple therapy failure among patients with high HIV viral loads at baseline. Patterns were similar at 96 weeks, but only three studies included 96-week data, the researchers said.
The rate of discontinuation because of adverse events was not significantly different between the groups at 48 weeks.
The study findings were limited by several factors, including the use of different regimens in the dual strategies, some of which are no longer in use, as well as there being insufficient data to fully compare outcomes at 96 weeks, and lack of information on cerebrospinal fluid viral load, the researchers noted.
However, the results suggest that dual therapy might be considered for HIV-naive patients with a low viral load, they said.
“Further RCTs that will evaluate the efficacy of antiretroviral regimens in use today among difficult-to-treat populations, such as patients with high viral load, including both intention-to-treat and per-protocol analysis, are needed to address this topic,” they concluded.
Consider range of patient factors when choosing therapies
Conducting the study at this time was important because of the expanding options for treating HIV patients, Donna E. Sweet, MD, an HIV specialist and professor of medicine at the University of Kansas, Wichita, said in an interview.
“We now have two single tablet formulations that are dual rather than triple therapy, and as treaters we are all trying to know when to use them,” she explained.
Dr. Sweet said she was not surprised by the study findings, given that well-conducted, randomized, controlled trials allowed the combination therapies to be approved.
Some of the key challenges to identifying the optimal treatment for HIV patients include factoring in the use of concomitant medications that could lead to drug-drug interactions, noted Dr. Sweet, who serves an editorial advisory board member of Internal Medicine News.
The take-home message for clinicians, in her opinion, is that “less drugs may mean less toxicity, but we don’t want to sacrifice efficacy,” she said. “There may be patients who are better suited than others for two vs. three drugs,” Dr. Sweet emphasized.
The next steps for research on the value of dual vs. triple therapy should include longer term efficacy studies, especially in those with lower CD4 counts and higher viral loads, said Dr. Sweet. In addition to factors such as CD4 counts and viral load, the food requirements of certain ART regimens could affect adherence and therefore a clinician decision to use two drugs rather than three, she noted.
Dr. Sweet disclosed past relationships with ViiV, Gilead, Merck, and Janssen on their speakers bureaus, and current advisory roles with Gilead and ViiV.
The study received no outside funding. Lead author Dr. Mariantonietta and several coauthors disclosed relationships with companies including ViiV Healthcare, AbbVie, Janssen-Cilag and Gilead Science, and Merck Sharp & Dohme, but no conflicts in connection with this study.
SOURCE: Mariantonietta P et al. Clin Microbiol Infect. 2020 Oct 5. doi: 10.1016/j.cmi.2020.09.048.
based on a meta-analysis including data from more than 5,000 patients.
Although triple therapy remains the standard of care, the availability of more potent drugs has revived interest in dual and mono therapies, wrote Pisaturo Mariantonietta, MD, of the University of Campania Luigi Vanvitelli, Naples, Italy, and colleagues.
In a study published in Clinical Microbiology and Infection, the researchers identified 14 articles including 5,205 treatment-naive HIV adults. The studies were published between 2008 and 2020; 13 were randomized, controlled trials.
The dual therapies used in the studies included atazanavir/r plus maraviroc; lopinavir/r plus lamivudine; raltegravir plus darunavir/r; lopinavir/r plus tenofovir, raltegravir, efavirenz, or maraviroc; atazanavir/r plus raltegravir and darunavir/r plus maraviroc; and dolutegravir plus lamivudine.
Overall, no significant differences occurred in the primary endpoint of treatment failure across 10 studies between dual therapy and triple therapy patients based on data at 48 weeks (relative risk 1.20). “The rate of treatment failure did not differ among the two groups when stratifying the patients according to the drug used in the dual regimen,” the researchers said.
Low viral load’s link to treatment failure
Among 2,398 patients with a low HIV viral load (less than 100,000 copies/mL), dual therapy patients were significantly more likely to experience treatment failure than were triple therapy patients (RR, 1.47, P = .007). No differences were noted between dual and triple therapy failure among patients with high HIV viral loads at baseline. Patterns were similar at 96 weeks, but only three studies included 96-week data, the researchers said.
The rate of discontinuation because of adverse events was not significantly different between the groups at 48 weeks.
The study findings were limited by several factors, including the use of different regimens in the dual strategies, some of which are no longer in use, as well as there being insufficient data to fully compare outcomes at 96 weeks, and lack of information on cerebrospinal fluid viral load, the researchers noted.
However, the results suggest that dual therapy might be considered for HIV-naive patients with a low viral load, they said.
“Further RCTs that will evaluate the efficacy of antiretroviral regimens in use today among difficult-to-treat populations, such as patients with high viral load, including both intention-to-treat and per-protocol analysis, are needed to address this topic,” they concluded.
Consider range of patient factors when choosing therapies
Conducting the study at this time was important because of the expanding options for treating HIV patients, Donna E. Sweet, MD, an HIV specialist and professor of medicine at the University of Kansas, Wichita, said in an interview.
“We now have two single tablet formulations that are dual rather than triple therapy, and as treaters we are all trying to know when to use them,” she explained.
Dr. Sweet said she was not surprised by the study findings, given that well-conducted, randomized, controlled trials allowed the combination therapies to be approved.
Some of the key challenges to identifying the optimal treatment for HIV patients include factoring in the use of concomitant medications that could lead to drug-drug interactions, noted Dr. Sweet, who serves an editorial advisory board member of Internal Medicine News.
The take-home message for clinicians, in her opinion, is that “less drugs may mean less toxicity, but we don’t want to sacrifice efficacy,” she said. “There may be patients who are better suited than others for two vs. three drugs,” Dr. Sweet emphasized.
The next steps for research on the value of dual vs. triple therapy should include longer term efficacy studies, especially in those with lower CD4 counts and higher viral loads, said Dr. Sweet. In addition to factors such as CD4 counts and viral load, the food requirements of certain ART regimens could affect adherence and therefore a clinician decision to use two drugs rather than three, she noted.
Dr. Sweet disclosed past relationships with ViiV, Gilead, Merck, and Janssen on their speakers bureaus, and current advisory roles with Gilead and ViiV.
The study received no outside funding. Lead author Dr. Mariantonietta and several coauthors disclosed relationships with companies including ViiV Healthcare, AbbVie, Janssen-Cilag and Gilead Science, and Merck Sharp & Dohme, but no conflicts in connection with this study.
SOURCE: Mariantonietta P et al. Clin Microbiol Infect. 2020 Oct 5. doi: 10.1016/j.cmi.2020.09.048.
FROM CLINICAL MICROBIOLOGY AND INFECTION
T2D treatments create tension between glycemic and cardiovascular goals
It was no surprise that updated guidelines recently published by the European Society of Cardiology for managing cardiovascular disease in patients with diabetes highlighted optimized treatment from a cardiovascular disease perspective, while a nearly concurrent update from two major diabetes societies saw the same issue from a more glycemic point of view.
This difference led to divergent approaches to managing hyperglycemia in patients with type 2 diabetes (T2D). The two diabetes societies that wrote one set of recommendations, the American Diabetes Association and the European Association for the Study of Diabetes, put metformin at the pinnacle of their drug hierarchy. Patients with T2D and established atherosclerotic cardiovascular disease (CVD), chronic kidney disease, or heart failure should all receive metformin first unless contraindicated or not tolerated, their updated consensus report said.
Once metformin is on board, a clinician can then add a second diabetes agent from among the two drug classes recently proven to also reduce cardiovascular and renal events, either the SGLT2 (sodium-glucose transporter 2) inhibitors, or GLP-1 (glucagonlike peptide–1) receptor agonists, they advised.
Cardiovascular disease focus represents a ‘major paradigm shift’
In contrast, the ESC guidelines called for upfront, systematic assessment of CVD risk in patients with T2D before treatment starts, and for patients in high- or very high–risk strata, the guidelines recommended starting the patient first on an SGLT2 inhibitor or a GLP-1 receptor agonist, and only adding metformin in patients who need additional glycemic control.
The guidelines also recommended starting treatment-naive patients with moderate CVD risk on metformin. For patients already on metformin, the new ESC guidelines called for adding an agent from at least one of these two drug classes with proven CVD benefits for those at high or very high CVD risk. The guidelines also note that the CVD benefits of the two newer drug classes differ and hence require further individualization depending on the risks faced by each patient, such as the risk for heart failure hospitalizations.
It’s an approach “driven by data from the cardiovascular outcome trials,” that showed several drugs from both the SGLT2 inhibitor and GLP-1 receptor agonist classes have substantial benefit for preventing cardiovascular events, renal events, hospitalizations for heart failure, and in some studies all-cause mortality, said Francesco Cosentino, MD, during a discussion of the guideline differences at the virtual annual meeting of the European Association for the Study of Diabetes.
The ESC approach also represents “a major paradigm shift,” a “change from a glucose-centric approach to an approach driven by cardiovascular disease events,” summed up Dr. Cosentino, professor of cardiology at the Karolinska Institute in Stockholm and chair of the task force that wrote the ESC’s 2019 updated guidelines. The ESC approach advocates initiating drugs for treating patients with T2D “based on cardiovascular disease risk classification,” he highlighted. Results from some SGLT2 inhibitor cardiovascular outcome trials showed that the CVD benefit was similar regardless of whether or not patients also received metformin.
ADA, EASD call for ‘a different emphasis’
“There is a different emphasis” in the statement issued by the diabetologists of the ADA and EASD, admitted Peter J. Grant, MD, a professor of diabetes and endocrinology at the University of Leeds (England) and cochair of the ESC guidelines task force. Dr. Grant represented the EASD on the task force, and the Association collaborated with the ESC in producing its guidelines.
“The ADA and EASD recommendations “look primarily at glucose control, with cardiovascular disease management as secondary.” In contrast, the ESC guidelines “are primarily cardiovascular disease risk guidelines, with a glucose interest,” Dr. Grant declared.
Despite his involvement in writing the ESC guidelines, Dr. Grant tilted toward the ADA/EASD statement as more globally relevant.
“There is much more to vasculopathy in diabetes than just macrovascular disease. Many patients with type 2 diabetes without macrovascular complications have microvascular disease,” including the potential for retinopathy, nephropathy, and neuropathy, he said. These complications can also have a strong impact on psychological well being and treatment satisfaction.
“It’s important that we’re not glucocentric any more, but it’s equally important that we treat glucose because it has such a benefit for microvascular disease.” Dr. Grant also cited metformin’s long history of safety and good tolerance, clinician comfort prescribing it, and its low price. Heavier reliance on SGLT2 inhibitors and GLP-1 receptor agonists will be expensive for the short term while the cost of these drugs remains high, which places a higher burden on “knowing we’re doing it right,” said Dr. Grant.
Dr. Cosentino pointed out that the higher cost of the drugs in the two classes shown to exert important cardiovascular and renal effects needs to be considered in a cost-effectiveness context, not just by cost alone.
‘Clinical inertia’ could be a danger
Dr. Cosentino played down a major disagreement between the two guidelines, suggesting that “focusing on the differences leads to clinical inertia” by the practicing community when they are unsure how to reconcile the two positions.
Dr. Grant agreed that adding a second drug to metformin right away made sense in at least selected patients. “Look at each patient and decide whether they need glycemic control. If so, and if they also have cardiovascular disease, use both drugs,” metformin, plus one agent from one of the two newer classes.
Something both experts agreed on is that it’s time to generally steer clear of sulfonylurea drugs. “We have evidence for harmful effects from sulfonylureas,” Dr. Cosentino said.
“I’d dump sulfonylureas,” was Dr. Grant’s assessment, but he added that they still have a role for patients who need additional glycemic control but can’t afford the newer drugs.
Dr. Cosentino has had financial relationships with Abbott, AstraZeneca, Boehringer Ingelheim, Bristol-Myers Squibb, Eli Lilly, Merck, Mundipharma, Novo Nordisk, and Pfizer, Dr. Grant has lectured on behalf of AstraZeneca, GlaxoSmithKline, Merck, Novo Nordisk, the Medicines Company, and Takeda, and he has been an adviser to Amgen, AstraZeneca, Novartis, Novo Nordisk, and Synexus.
It was no surprise that updated guidelines recently published by the European Society of Cardiology for managing cardiovascular disease in patients with diabetes highlighted optimized treatment from a cardiovascular disease perspective, while a nearly concurrent update from two major diabetes societies saw the same issue from a more glycemic point of view.
This difference led to divergent approaches to managing hyperglycemia in patients with type 2 diabetes (T2D). The two diabetes societies that wrote one set of recommendations, the American Diabetes Association and the European Association for the Study of Diabetes, put metformin at the pinnacle of their drug hierarchy. Patients with T2D and established atherosclerotic cardiovascular disease (CVD), chronic kidney disease, or heart failure should all receive metformin first unless contraindicated or not tolerated, their updated consensus report said.
Once metformin is on board, a clinician can then add a second diabetes agent from among the two drug classes recently proven to also reduce cardiovascular and renal events, either the SGLT2 (sodium-glucose transporter 2) inhibitors, or GLP-1 (glucagonlike peptide–1) receptor agonists, they advised.
Cardiovascular disease focus represents a ‘major paradigm shift’
In contrast, the ESC guidelines called for upfront, systematic assessment of CVD risk in patients with T2D before treatment starts, and for patients in high- or very high–risk strata, the guidelines recommended starting the patient first on an SGLT2 inhibitor or a GLP-1 receptor agonist, and only adding metformin in patients who need additional glycemic control.
The guidelines also recommended starting treatment-naive patients with moderate CVD risk on metformin. For patients already on metformin, the new ESC guidelines called for adding an agent from at least one of these two drug classes with proven CVD benefits for those at high or very high CVD risk. The guidelines also note that the CVD benefits of the two newer drug classes differ and hence require further individualization depending on the risks faced by each patient, such as the risk for heart failure hospitalizations.
It’s an approach “driven by data from the cardiovascular outcome trials,” that showed several drugs from both the SGLT2 inhibitor and GLP-1 receptor agonist classes have substantial benefit for preventing cardiovascular events, renal events, hospitalizations for heart failure, and in some studies all-cause mortality, said Francesco Cosentino, MD, during a discussion of the guideline differences at the virtual annual meeting of the European Association for the Study of Diabetes.
The ESC approach also represents “a major paradigm shift,” a “change from a glucose-centric approach to an approach driven by cardiovascular disease events,” summed up Dr. Cosentino, professor of cardiology at the Karolinska Institute in Stockholm and chair of the task force that wrote the ESC’s 2019 updated guidelines. The ESC approach advocates initiating drugs for treating patients with T2D “based on cardiovascular disease risk classification,” he highlighted. Results from some SGLT2 inhibitor cardiovascular outcome trials showed that the CVD benefit was similar regardless of whether or not patients also received metformin.
ADA, EASD call for ‘a different emphasis’
“There is a different emphasis” in the statement issued by the diabetologists of the ADA and EASD, admitted Peter J. Grant, MD, a professor of diabetes and endocrinology at the University of Leeds (England) and cochair of the ESC guidelines task force. Dr. Grant represented the EASD on the task force, and the Association collaborated with the ESC in producing its guidelines.
“The ADA and EASD recommendations “look primarily at glucose control, with cardiovascular disease management as secondary.” In contrast, the ESC guidelines “are primarily cardiovascular disease risk guidelines, with a glucose interest,” Dr. Grant declared.
Despite his involvement in writing the ESC guidelines, Dr. Grant tilted toward the ADA/EASD statement as more globally relevant.
“There is much more to vasculopathy in diabetes than just macrovascular disease. Many patients with type 2 diabetes without macrovascular complications have microvascular disease,” including the potential for retinopathy, nephropathy, and neuropathy, he said. These complications can also have a strong impact on psychological well being and treatment satisfaction.
“It’s important that we’re not glucocentric any more, but it’s equally important that we treat glucose because it has such a benefit for microvascular disease.” Dr. Grant also cited metformin’s long history of safety and good tolerance, clinician comfort prescribing it, and its low price. Heavier reliance on SGLT2 inhibitors and GLP-1 receptor agonists will be expensive for the short term while the cost of these drugs remains high, which places a higher burden on “knowing we’re doing it right,” said Dr. Grant.
Dr. Cosentino pointed out that the higher cost of the drugs in the two classes shown to exert important cardiovascular and renal effects needs to be considered in a cost-effectiveness context, not just by cost alone.
‘Clinical inertia’ could be a danger
Dr. Cosentino played down a major disagreement between the two guidelines, suggesting that “focusing on the differences leads to clinical inertia” by the practicing community when they are unsure how to reconcile the two positions.
Dr. Grant agreed that adding a second drug to metformin right away made sense in at least selected patients. “Look at each patient and decide whether they need glycemic control. If so, and if they also have cardiovascular disease, use both drugs,” metformin, plus one agent from one of the two newer classes.
Something both experts agreed on is that it’s time to generally steer clear of sulfonylurea drugs. “We have evidence for harmful effects from sulfonylureas,” Dr. Cosentino said.
“I’d dump sulfonylureas,” was Dr. Grant’s assessment, but he added that they still have a role for patients who need additional glycemic control but can’t afford the newer drugs.
Dr. Cosentino has had financial relationships with Abbott, AstraZeneca, Boehringer Ingelheim, Bristol-Myers Squibb, Eli Lilly, Merck, Mundipharma, Novo Nordisk, and Pfizer, Dr. Grant has lectured on behalf of AstraZeneca, GlaxoSmithKline, Merck, Novo Nordisk, the Medicines Company, and Takeda, and he has been an adviser to Amgen, AstraZeneca, Novartis, Novo Nordisk, and Synexus.
It was no surprise that updated guidelines recently published by the European Society of Cardiology for managing cardiovascular disease in patients with diabetes highlighted optimized treatment from a cardiovascular disease perspective, while a nearly concurrent update from two major diabetes societies saw the same issue from a more glycemic point of view.
This difference led to divergent approaches to managing hyperglycemia in patients with type 2 diabetes (T2D). The two diabetes societies that wrote one set of recommendations, the American Diabetes Association and the European Association for the Study of Diabetes, put metformin at the pinnacle of their drug hierarchy. Patients with T2D and established atherosclerotic cardiovascular disease (CVD), chronic kidney disease, or heart failure should all receive metformin first unless contraindicated or not tolerated, their updated consensus report said.
Once metformin is on board, a clinician can then add a second diabetes agent from among the two drug classes recently proven to also reduce cardiovascular and renal events, either the SGLT2 (sodium-glucose transporter 2) inhibitors, or GLP-1 (glucagonlike peptide–1) receptor agonists, they advised.
Cardiovascular disease focus represents a ‘major paradigm shift’
In contrast, the ESC guidelines called for upfront, systematic assessment of CVD risk in patients with T2D before treatment starts, and for patients in high- or very high–risk strata, the guidelines recommended starting the patient first on an SGLT2 inhibitor or a GLP-1 receptor agonist, and only adding metformin in patients who need additional glycemic control.
The guidelines also recommended starting treatment-naive patients with moderate CVD risk on metformin. For patients already on metformin, the new ESC guidelines called for adding an agent from at least one of these two drug classes with proven CVD benefits for those at high or very high CVD risk. The guidelines also note that the CVD benefits of the two newer drug classes differ and hence require further individualization depending on the risks faced by each patient, such as the risk for heart failure hospitalizations.
It’s an approach “driven by data from the cardiovascular outcome trials,” that showed several drugs from both the SGLT2 inhibitor and GLP-1 receptor agonist classes have substantial benefit for preventing cardiovascular events, renal events, hospitalizations for heart failure, and in some studies all-cause mortality, said Francesco Cosentino, MD, during a discussion of the guideline differences at the virtual annual meeting of the European Association for the Study of Diabetes.
The ESC approach also represents “a major paradigm shift,” a “change from a glucose-centric approach to an approach driven by cardiovascular disease events,” summed up Dr. Cosentino, professor of cardiology at the Karolinska Institute in Stockholm and chair of the task force that wrote the ESC’s 2019 updated guidelines. The ESC approach advocates initiating drugs for treating patients with T2D “based on cardiovascular disease risk classification,” he highlighted. Results from some SGLT2 inhibitor cardiovascular outcome trials showed that the CVD benefit was similar regardless of whether or not patients also received metformin.
ADA, EASD call for ‘a different emphasis’
“There is a different emphasis” in the statement issued by the diabetologists of the ADA and EASD, admitted Peter J. Grant, MD, a professor of diabetes and endocrinology at the University of Leeds (England) and cochair of the ESC guidelines task force. Dr. Grant represented the EASD on the task force, and the Association collaborated with the ESC in producing its guidelines.
“The ADA and EASD recommendations “look primarily at glucose control, with cardiovascular disease management as secondary.” In contrast, the ESC guidelines “are primarily cardiovascular disease risk guidelines, with a glucose interest,” Dr. Grant declared.
Despite his involvement in writing the ESC guidelines, Dr. Grant tilted toward the ADA/EASD statement as more globally relevant.
“There is much more to vasculopathy in diabetes than just macrovascular disease. Many patients with type 2 diabetes without macrovascular complications have microvascular disease,” including the potential for retinopathy, nephropathy, and neuropathy, he said. These complications can also have a strong impact on psychological well being and treatment satisfaction.
“It’s important that we’re not glucocentric any more, but it’s equally important that we treat glucose because it has such a benefit for microvascular disease.” Dr. Grant also cited metformin’s long history of safety and good tolerance, clinician comfort prescribing it, and its low price. Heavier reliance on SGLT2 inhibitors and GLP-1 receptor agonists will be expensive for the short term while the cost of these drugs remains high, which places a higher burden on “knowing we’re doing it right,” said Dr. Grant.
Dr. Cosentino pointed out that the higher cost of the drugs in the two classes shown to exert important cardiovascular and renal effects needs to be considered in a cost-effectiveness context, not just by cost alone.
‘Clinical inertia’ could be a danger
Dr. Cosentino played down a major disagreement between the two guidelines, suggesting that “focusing on the differences leads to clinical inertia” by the practicing community when they are unsure how to reconcile the two positions.
Dr. Grant agreed that adding a second drug to metformin right away made sense in at least selected patients. “Look at each patient and decide whether they need glycemic control. If so, and if they also have cardiovascular disease, use both drugs,” metformin, plus one agent from one of the two newer classes.
Something both experts agreed on is that it’s time to generally steer clear of sulfonylurea drugs. “We have evidence for harmful effects from sulfonylureas,” Dr. Cosentino said.
“I’d dump sulfonylureas,” was Dr. Grant’s assessment, but he added that they still have a role for patients who need additional glycemic control but can’t afford the newer drugs.
Dr. Cosentino has had financial relationships with Abbott, AstraZeneca, Boehringer Ingelheim, Bristol-Myers Squibb, Eli Lilly, Merck, Mundipharma, Novo Nordisk, and Pfizer, Dr. Grant has lectured on behalf of AstraZeneca, GlaxoSmithKline, Merck, Novo Nordisk, the Medicines Company, and Takeda, and he has been an adviser to Amgen, AstraZeneca, Novartis, Novo Nordisk, and Synexus.
FROM EASD 2020
Remdesivir effective, well-tolerated in final trial report
Drug beats placebo across multiple endpoints in COVID-19 patients
In May 2020, remdesivir received Food and Drug Administration approval for emergency treatment of severe COVID-19 on the basis of a preliminary report on this trial. In August 2020, the FDA expanded the indication to include all hospitalized adult and pediatric patients with suspected or laboratory-confirmed COVID-19 infection irrespective of severity.
“Our findings were consistent with the findings of the preliminary report: a 10-day course of remdesivir was superior to placebo in the treatment of hospitalized patients with COVID-19,” reported a team of investigators led by John H. Beigel, MD, of the Division of Microbiology and Infectious Diseases at the National Institute of Allergy and Infectious Diseases, in the New England Journal of Medicine.
The drug’s broadened indication was not based on the ACTT-1 trial, according to Dr. Beigel. “Other data have demonstrated that remdesivir shortens recovery in patients with lower acuity. In our study, evidence of pneumonia was an enrollment requirement,” he explained in an interview.
In the newly published final ACTT-1 data, the median time to recovery was 10 days for those on active therapy versus 15 days for those randomized to placebo. With a rate ratio of 1.29 (P less than .001), this translated to a recovery that was about one third faster.
In this final report, remdesivir’s significant advantage over placebo regarding the trial’s primary endpoint was reinforced by efficacy on multiple secondary endpoints.
This benefits on multiple secondary endpoints included a 50% greater odds ratio (OR, 1.5; 95% CI, 1.2-1.9) of significant clinical improvement by day 15 after adjustment for baseline severity, a shorter initial length of hospital stay (12 vs. 17 days) and fewer days on oxygen supplementation (13 vs. 21 days) for the subgroup of patients on oxygen at enrollment.
Although the numerically lower mortality in the remdesivir arm (6.75 vs. 11.9%) did not reach statistical significance, Dr. Beigel said, “mortality was moving in the same direction as the other key endpoints.”
According to the study investigators, the types of rates of adverse events on remdesivir, which inhibits viral replication, “were generally similar in the remdesivir and placebo groups.”
In ACTT-1, 1,062 patients were randomized to remdesivir (200 mg loading dose followed by 100 mg daily for up to 9 days) or placebo. Patients were enrolled at study sites in North America, Europe, and Asia.
The data of ACTT-1 confirm a benefit from remdesivir in hospitalized COVID-19 patients with severe disease, but Dr. Beigel said he agrees with the current FDA indication that supports treatment in any hospitalized COVID-19 patient.
“We saw bigger benefits in patients with more severe infections. The benefits are not as large in patients with mild disease, but I think remdesivir should be considered in any hospitalized patient,” Dr. Beigel said.
This point of view is shared.
“I would give this drug to anyone in the hospital infected with COVID-19 assuming there was an ample supply and no need for rationing,” said Donna E. Sweet, MD, professor of internal medicine, University of Kansas, Wichita. She noted that this study has implications for hospital and hospital staff, as well as for patients.
“This type of reduction in recovery time means a reduction in potential exposures to hospital staff, a reduced need for PPE [personal protective equipment], and it will free up beds in the ICU [intensive care unit],” said Dr. Sweet, who also serves as an editorial advisory board member for Internal Medicine News.
An infectious disease specialist at the University of Minnesota also considers remdesivir to have an important role for conserving resources that deserves emphasis.
The reduction in time to recovery “is of benefit to the health system by maintaining hospital bed capacity,” said David R. Boulware, MD, professor of medicine at the University of Minnesota, Minneapolis.
According to his reading of the available data, including those from ACTT-1, the benefit appears to be greatest in those with a moderate degree of illness, which he defined as “sick enough to be hospitalized and require oxygen, yet not severely sick [and] requiring a ventilator or [extracorporeal membrane oxygenation].”
This does not preclude a benefit in those with more severe or milder disease, but patients with mild disease “are likely to recover regardless – or despite – whatever therapy they receive,” he said.
Dr. Beigel, the principal investigator of this trial, reports no potential conflicts of interest.
SOURCE: Beigel JH et al. N Engl J Med. 2020 Oct 8. doi: 10.1056/NEJMoa2007764.
Drug beats placebo across multiple endpoints in COVID-19 patients
Drug beats placebo across multiple endpoints in COVID-19 patients
In May 2020, remdesivir received Food and Drug Administration approval for emergency treatment of severe COVID-19 on the basis of a preliminary report on this trial. In August 2020, the FDA expanded the indication to include all hospitalized adult and pediatric patients with suspected or laboratory-confirmed COVID-19 infection irrespective of severity.
“Our findings were consistent with the findings of the preliminary report: a 10-day course of remdesivir was superior to placebo in the treatment of hospitalized patients with COVID-19,” reported a team of investigators led by John H. Beigel, MD, of the Division of Microbiology and Infectious Diseases at the National Institute of Allergy and Infectious Diseases, in the New England Journal of Medicine.
The drug’s broadened indication was not based on the ACTT-1 trial, according to Dr. Beigel. “Other data have demonstrated that remdesivir shortens recovery in patients with lower acuity. In our study, evidence of pneumonia was an enrollment requirement,” he explained in an interview.
In the newly published final ACTT-1 data, the median time to recovery was 10 days for those on active therapy versus 15 days for those randomized to placebo. With a rate ratio of 1.29 (P less than .001), this translated to a recovery that was about one third faster.
In this final report, remdesivir’s significant advantage over placebo regarding the trial’s primary endpoint was reinforced by efficacy on multiple secondary endpoints.
This benefits on multiple secondary endpoints included a 50% greater odds ratio (OR, 1.5; 95% CI, 1.2-1.9) of significant clinical improvement by day 15 after adjustment for baseline severity, a shorter initial length of hospital stay (12 vs. 17 days) and fewer days on oxygen supplementation (13 vs. 21 days) for the subgroup of patients on oxygen at enrollment.
Although the numerically lower mortality in the remdesivir arm (6.75 vs. 11.9%) did not reach statistical significance, Dr. Beigel said, “mortality was moving in the same direction as the other key endpoints.”
According to the study investigators, the types of rates of adverse events on remdesivir, which inhibits viral replication, “were generally similar in the remdesivir and placebo groups.”
In ACTT-1, 1,062 patients were randomized to remdesivir (200 mg loading dose followed by 100 mg daily for up to 9 days) or placebo. Patients were enrolled at study sites in North America, Europe, and Asia.
The data of ACTT-1 confirm a benefit from remdesivir in hospitalized COVID-19 patients with severe disease, but Dr. Beigel said he agrees with the current FDA indication that supports treatment in any hospitalized COVID-19 patient.
“We saw bigger benefits in patients with more severe infections. The benefits are not as large in patients with mild disease, but I think remdesivir should be considered in any hospitalized patient,” Dr. Beigel said.
This point of view is shared.
“I would give this drug to anyone in the hospital infected with COVID-19 assuming there was an ample supply and no need for rationing,” said Donna E. Sweet, MD, professor of internal medicine, University of Kansas, Wichita. She noted that this study has implications for hospital and hospital staff, as well as for patients.
“This type of reduction in recovery time means a reduction in potential exposures to hospital staff, a reduced need for PPE [personal protective equipment], and it will free up beds in the ICU [intensive care unit],” said Dr. Sweet, who also serves as an editorial advisory board member for Internal Medicine News.
An infectious disease specialist at the University of Minnesota also considers remdesivir to have an important role for conserving resources that deserves emphasis.
The reduction in time to recovery “is of benefit to the health system by maintaining hospital bed capacity,” said David R. Boulware, MD, professor of medicine at the University of Minnesota, Minneapolis.
According to his reading of the available data, including those from ACTT-1, the benefit appears to be greatest in those with a moderate degree of illness, which he defined as “sick enough to be hospitalized and require oxygen, yet not severely sick [and] requiring a ventilator or [extracorporeal membrane oxygenation].”
This does not preclude a benefit in those with more severe or milder disease, but patients with mild disease “are likely to recover regardless – or despite – whatever therapy they receive,” he said.
Dr. Beigel, the principal investigator of this trial, reports no potential conflicts of interest.
SOURCE: Beigel JH et al. N Engl J Med. 2020 Oct 8. doi: 10.1056/NEJMoa2007764.
In May 2020, remdesivir received Food and Drug Administration approval for emergency treatment of severe COVID-19 on the basis of a preliminary report on this trial. In August 2020, the FDA expanded the indication to include all hospitalized adult and pediatric patients with suspected or laboratory-confirmed COVID-19 infection irrespective of severity.
“Our findings were consistent with the findings of the preliminary report: a 10-day course of remdesivir was superior to placebo in the treatment of hospitalized patients with COVID-19,” reported a team of investigators led by John H. Beigel, MD, of the Division of Microbiology and Infectious Diseases at the National Institute of Allergy and Infectious Diseases, in the New England Journal of Medicine.
The drug’s broadened indication was not based on the ACTT-1 trial, according to Dr. Beigel. “Other data have demonstrated that remdesivir shortens recovery in patients with lower acuity. In our study, evidence of pneumonia was an enrollment requirement,” he explained in an interview.
In the newly published final ACTT-1 data, the median time to recovery was 10 days for those on active therapy versus 15 days for those randomized to placebo. With a rate ratio of 1.29 (P less than .001), this translated to a recovery that was about one third faster.
In this final report, remdesivir’s significant advantage over placebo regarding the trial’s primary endpoint was reinforced by efficacy on multiple secondary endpoints.
This benefits on multiple secondary endpoints included a 50% greater odds ratio (OR, 1.5; 95% CI, 1.2-1.9) of significant clinical improvement by day 15 after adjustment for baseline severity, a shorter initial length of hospital stay (12 vs. 17 days) and fewer days on oxygen supplementation (13 vs. 21 days) for the subgroup of patients on oxygen at enrollment.
Although the numerically lower mortality in the remdesivir arm (6.75 vs. 11.9%) did not reach statistical significance, Dr. Beigel said, “mortality was moving in the same direction as the other key endpoints.”
According to the study investigators, the types of rates of adverse events on remdesivir, which inhibits viral replication, “were generally similar in the remdesivir and placebo groups.”
In ACTT-1, 1,062 patients were randomized to remdesivir (200 mg loading dose followed by 100 mg daily for up to 9 days) or placebo. Patients were enrolled at study sites in North America, Europe, and Asia.
The data of ACTT-1 confirm a benefit from remdesivir in hospitalized COVID-19 patients with severe disease, but Dr. Beigel said he agrees with the current FDA indication that supports treatment in any hospitalized COVID-19 patient.
“We saw bigger benefits in patients with more severe infections. The benefits are not as large in patients with mild disease, but I think remdesivir should be considered in any hospitalized patient,” Dr. Beigel said.
This point of view is shared.
“I would give this drug to anyone in the hospital infected with COVID-19 assuming there was an ample supply and no need for rationing,” said Donna E. Sweet, MD, professor of internal medicine, University of Kansas, Wichita. She noted that this study has implications for hospital and hospital staff, as well as for patients.
“This type of reduction in recovery time means a reduction in potential exposures to hospital staff, a reduced need for PPE [personal protective equipment], and it will free up beds in the ICU [intensive care unit],” said Dr. Sweet, who also serves as an editorial advisory board member for Internal Medicine News.
An infectious disease specialist at the University of Minnesota also considers remdesivir to have an important role for conserving resources that deserves emphasis.
The reduction in time to recovery “is of benefit to the health system by maintaining hospital bed capacity,” said David R. Boulware, MD, professor of medicine at the University of Minnesota, Minneapolis.
According to his reading of the available data, including those from ACTT-1, the benefit appears to be greatest in those with a moderate degree of illness, which he defined as “sick enough to be hospitalized and require oxygen, yet not severely sick [and] requiring a ventilator or [extracorporeal membrane oxygenation].”
This does not preclude a benefit in those with more severe or milder disease, but patients with mild disease “are likely to recover regardless – or despite – whatever therapy they receive,” he said.
Dr. Beigel, the principal investigator of this trial, reports no potential conflicts of interest.
SOURCE: Beigel JH et al. N Engl J Med. 2020 Oct 8. doi: 10.1056/NEJMoa2007764.
More data on impact of corticosteroids on COVID-19 mortality in patients with COPD
, a study of almost 1 million individuals in the United Kingdom has shown.
Patients with chronic obstructive pulmonary disease or asthma who used ICS on a regular basis were more likely to die from COVID-19 than COPD or asthma patients who were prescribed non-ICS therapies, reported co-lead author Anna Schultze, PhD, of London School of Hygiene & Tropical Medicine and colleagues.
Of note, the increased risk of death among ICS users likely stemmed from greater severity of preexisting chronic respiratory conditions, instead of directly from ICS usage, which has little apparent impact on COVID-19 mortality, the investigators wrote in Lancet Respiratory Medicine.
These findings conflict with a hypothesis proposed early in the pandemic: that ICS may protect individuals from SARS-CoV-2 infection and poor outcomes with COVID-19.
According to Megan Conroy, MD, of the department of internal medicine at the Ohio State University Wexner Medical Center, Columbus, this hypothesis was based on some unexpected epidemiological findings.
“In general, we tend to think people with underlying lung disease – like COPD or asthma – to be at higher risk for severe forms of lower respiratory tract infections,” Dr. Conroy said. “Somewhat surprisingly, early data in the pandemic showed patients with COPD and asthma [were] underrepresented [among patients with COVID] when compared to the prevalence of these diseases in the population.”
This raised the possibility of an incidental protective effect from regular ICS therapy, which “had some strong theoretic pathophysiologic basis,” Dr. Conroy said, referring to research that demonstrated ICS-mediated downregulation of SARS-CoV-2 entry receptors ACE2 and TMPRSS2.
Dr. Schultze and colleagues noted that investigators for two ongoing randomized controlled trials (NCT04331054, NCT04330586) are studying ICS as an intervention for COVID-19; but neither trial includes individuals already taking ICS for chronic respiratory disease.
The present observational study therefore aimed to assess mortality risk within this population. Data were drawn from electronic health records and a U.K. national mortality database, with follow-up ranging from March 1 to May 6, 2020. Eligibility required a relevant prescription within 4 months of first follow-up. In the COPD group, patients were prescribed a long-acting beta agonist plus a long-acting muscarinic antagonist (LABA–LAMA), LABA alone, LABA plus ICS, LABA–LAMA plus ICS, or ICS alone (if prescribed LABA within 4 months).
In the asthma group, patients received low/medium-dose ICS, high-dose ICS, or a short-acting beta agonist (SABA) alone. Patients with COPD were at least 35 years of age, while those with asthma were 18 years or older. Hazard ratios were adjusted for a variety of covariates, including respiratory disease–exacerbation history, age, sex, body mass index, hypertension, diabetes, and others.
These eligibility criteria returned 148,557 patients with COPD and 818,490 with asthma.
Patients with COPD who were prescribed ICS plus LABA-LAMA or ICS plus LABA had an increased risk of COVID-19-related death, compared with those who did not receive ICS (adjusted hazard ratio, 1.39; 95% confidence interval, 1.10-1.76). Separate analyses of patients who received a triple combination (LABA–LAMA plus ICS) versus those who took a dual combination (LABA plus ICS) showed that triple-combination therapy was significantly associated with increased COVID-19-related mortality (aHR, 1.43; 95% CI, 1.12-1.83), while dual-combination therapy was less so (aHR, 1.29; 95% CI, 0.96-1.74). Non–COVID-19–related mortality was significantly increased for all COPD patients who were prescribed ICS, with or without adjustment for covariates.
Asthma patients prescribed high-dose ICS instead of SABA alone had a slightly greater risk of COVID-19–related death, based on an adjusted hazard ratio of 1.55 (95% CI, 1.10-2.18). Those with asthma who received low/medium–dose ICS demonstrated a slight trend toward increased mortality risk, but this was not significant (aHR, 1.14; 95% CI, 0.85-1.54). ICS usage in the asthma group was not linked with a significant increase in non–COVID-19–related death.
“In summary, we found no evidence of a beneficial effect of regular ICS use among people with COPD and asthma on COVID-19–related mortality,” the investigators concluded.
In agreement with the investigators, Dr. Conroy said that the increased mortality rate among ICS users should not be misconstrued as a medication-related risk.
“While the study found that those with COPD or asthma taking ICS and high-dose ICS were at an increased risk of death, this could easily be explained by the likelihood that those are the patients who are more likely to have more severe underlying lung disease,” Dr. Conroy said. “While this observational study did attempt to control for exacerbation history, the ability to do so by electronic health records data is certainly imperfect.”
With this in mind, patients with chronic respiratory disease should be encouraged to adhere to their usual treatment regimen, Dr. Conroy added.
“There isn’t evidence to increase or decrease medications just because of the pandemic,” she said. “A patient with asthma or COPD should continue to take the medications that are needed to achieve good control of their lung disease.”
The study was funded by the U.K. Medical Research Council. The investigators reported additional relationships with the Wellcome Trust, the Good Thinking Foundation, the Laura and John Arnold Foundation, and others. Dr. Conroy reported no conflicts of interest.
SOURCE: Schultze A et al. Lancet Respir Med. 2020 Sep 24. doi: 10.1016/ S2213-2600(20)30415-X.
, a study of almost 1 million individuals in the United Kingdom has shown.
Patients with chronic obstructive pulmonary disease or asthma who used ICS on a regular basis were more likely to die from COVID-19 than COPD or asthma patients who were prescribed non-ICS therapies, reported co-lead author Anna Schultze, PhD, of London School of Hygiene & Tropical Medicine and colleagues.
Of note, the increased risk of death among ICS users likely stemmed from greater severity of preexisting chronic respiratory conditions, instead of directly from ICS usage, which has little apparent impact on COVID-19 mortality, the investigators wrote in Lancet Respiratory Medicine.
These findings conflict with a hypothesis proposed early in the pandemic: that ICS may protect individuals from SARS-CoV-2 infection and poor outcomes with COVID-19.
According to Megan Conroy, MD, of the department of internal medicine at the Ohio State University Wexner Medical Center, Columbus, this hypothesis was based on some unexpected epidemiological findings.
“In general, we tend to think people with underlying lung disease – like COPD or asthma – to be at higher risk for severe forms of lower respiratory tract infections,” Dr. Conroy said. “Somewhat surprisingly, early data in the pandemic showed patients with COPD and asthma [were] underrepresented [among patients with COVID] when compared to the prevalence of these diseases in the population.”
This raised the possibility of an incidental protective effect from regular ICS therapy, which “had some strong theoretic pathophysiologic basis,” Dr. Conroy said, referring to research that demonstrated ICS-mediated downregulation of SARS-CoV-2 entry receptors ACE2 and TMPRSS2.
Dr. Schultze and colleagues noted that investigators for two ongoing randomized controlled trials (NCT04331054, NCT04330586) are studying ICS as an intervention for COVID-19; but neither trial includes individuals already taking ICS for chronic respiratory disease.
The present observational study therefore aimed to assess mortality risk within this population. Data were drawn from electronic health records and a U.K. national mortality database, with follow-up ranging from March 1 to May 6, 2020. Eligibility required a relevant prescription within 4 months of first follow-up. In the COPD group, patients were prescribed a long-acting beta agonist plus a long-acting muscarinic antagonist (LABA–LAMA), LABA alone, LABA plus ICS, LABA–LAMA plus ICS, or ICS alone (if prescribed LABA within 4 months).
In the asthma group, patients received low/medium-dose ICS, high-dose ICS, or a short-acting beta agonist (SABA) alone. Patients with COPD were at least 35 years of age, while those with asthma were 18 years or older. Hazard ratios were adjusted for a variety of covariates, including respiratory disease–exacerbation history, age, sex, body mass index, hypertension, diabetes, and others.
These eligibility criteria returned 148,557 patients with COPD and 818,490 with asthma.
Patients with COPD who were prescribed ICS plus LABA-LAMA or ICS plus LABA had an increased risk of COVID-19-related death, compared with those who did not receive ICS (adjusted hazard ratio, 1.39; 95% confidence interval, 1.10-1.76). Separate analyses of patients who received a triple combination (LABA–LAMA plus ICS) versus those who took a dual combination (LABA plus ICS) showed that triple-combination therapy was significantly associated with increased COVID-19-related mortality (aHR, 1.43; 95% CI, 1.12-1.83), while dual-combination therapy was less so (aHR, 1.29; 95% CI, 0.96-1.74). Non–COVID-19–related mortality was significantly increased for all COPD patients who were prescribed ICS, with or without adjustment for covariates.
Asthma patients prescribed high-dose ICS instead of SABA alone had a slightly greater risk of COVID-19–related death, based on an adjusted hazard ratio of 1.55 (95% CI, 1.10-2.18). Those with asthma who received low/medium–dose ICS demonstrated a slight trend toward increased mortality risk, but this was not significant (aHR, 1.14; 95% CI, 0.85-1.54). ICS usage in the asthma group was not linked with a significant increase in non–COVID-19–related death.
“In summary, we found no evidence of a beneficial effect of regular ICS use among people with COPD and asthma on COVID-19–related mortality,” the investigators concluded.
In agreement with the investigators, Dr. Conroy said that the increased mortality rate among ICS users should not be misconstrued as a medication-related risk.
“While the study found that those with COPD or asthma taking ICS and high-dose ICS were at an increased risk of death, this could easily be explained by the likelihood that those are the patients who are more likely to have more severe underlying lung disease,” Dr. Conroy said. “While this observational study did attempt to control for exacerbation history, the ability to do so by electronic health records data is certainly imperfect.”
With this in mind, patients with chronic respiratory disease should be encouraged to adhere to their usual treatment regimen, Dr. Conroy added.
“There isn’t evidence to increase or decrease medications just because of the pandemic,” she said. “A patient with asthma or COPD should continue to take the medications that are needed to achieve good control of their lung disease.”
The study was funded by the U.K. Medical Research Council. The investigators reported additional relationships with the Wellcome Trust, the Good Thinking Foundation, the Laura and John Arnold Foundation, and others. Dr. Conroy reported no conflicts of interest.
SOURCE: Schultze A et al. Lancet Respir Med. 2020 Sep 24. doi: 10.1016/ S2213-2600(20)30415-X.
, a study of almost 1 million individuals in the United Kingdom has shown.
Patients with chronic obstructive pulmonary disease or asthma who used ICS on a regular basis were more likely to die from COVID-19 than COPD or asthma patients who were prescribed non-ICS therapies, reported co-lead author Anna Schultze, PhD, of London School of Hygiene & Tropical Medicine and colleagues.
Of note, the increased risk of death among ICS users likely stemmed from greater severity of preexisting chronic respiratory conditions, instead of directly from ICS usage, which has little apparent impact on COVID-19 mortality, the investigators wrote in Lancet Respiratory Medicine.
These findings conflict with a hypothesis proposed early in the pandemic: that ICS may protect individuals from SARS-CoV-2 infection and poor outcomes with COVID-19.
According to Megan Conroy, MD, of the department of internal medicine at the Ohio State University Wexner Medical Center, Columbus, this hypothesis was based on some unexpected epidemiological findings.
“In general, we tend to think people with underlying lung disease – like COPD or asthma – to be at higher risk for severe forms of lower respiratory tract infections,” Dr. Conroy said. “Somewhat surprisingly, early data in the pandemic showed patients with COPD and asthma [were] underrepresented [among patients with COVID] when compared to the prevalence of these diseases in the population.”
This raised the possibility of an incidental protective effect from regular ICS therapy, which “had some strong theoretic pathophysiologic basis,” Dr. Conroy said, referring to research that demonstrated ICS-mediated downregulation of SARS-CoV-2 entry receptors ACE2 and TMPRSS2.
Dr. Schultze and colleagues noted that investigators for two ongoing randomized controlled trials (NCT04331054, NCT04330586) are studying ICS as an intervention for COVID-19; but neither trial includes individuals already taking ICS for chronic respiratory disease.
The present observational study therefore aimed to assess mortality risk within this population. Data were drawn from electronic health records and a U.K. national mortality database, with follow-up ranging from March 1 to May 6, 2020. Eligibility required a relevant prescription within 4 months of first follow-up. In the COPD group, patients were prescribed a long-acting beta agonist plus a long-acting muscarinic antagonist (LABA–LAMA), LABA alone, LABA plus ICS, LABA–LAMA plus ICS, or ICS alone (if prescribed LABA within 4 months).
In the asthma group, patients received low/medium-dose ICS, high-dose ICS, or a short-acting beta agonist (SABA) alone. Patients with COPD were at least 35 years of age, while those with asthma were 18 years or older. Hazard ratios were adjusted for a variety of covariates, including respiratory disease–exacerbation history, age, sex, body mass index, hypertension, diabetes, and others.
These eligibility criteria returned 148,557 patients with COPD and 818,490 with asthma.
Patients with COPD who were prescribed ICS plus LABA-LAMA or ICS plus LABA had an increased risk of COVID-19-related death, compared with those who did not receive ICS (adjusted hazard ratio, 1.39; 95% confidence interval, 1.10-1.76). Separate analyses of patients who received a triple combination (LABA–LAMA plus ICS) versus those who took a dual combination (LABA plus ICS) showed that triple-combination therapy was significantly associated with increased COVID-19-related mortality (aHR, 1.43; 95% CI, 1.12-1.83), while dual-combination therapy was less so (aHR, 1.29; 95% CI, 0.96-1.74). Non–COVID-19–related mortality was significantly increased for all COPD patients who were prescribed ICS, with or without adjustment for covariates.
Asthma patients prescribed high-dose ICS instead of SABA alone had a slightly greater risk of COVID-19–related death, based on an adjusted hazard ratio of 1.55 (95% CI, 1.10-2.18). Those with asthma who received low/medium–dose ICS demonstrated a slight trend toward increased mortality risk, but this was not significant (aHR, 1.14; 95% CI, 0.85-1.54). ICS usage in the asthma group was not linked with a significant increase in non–COVID-19–related death.
“In summary, we found no evidence of a beneficial effect of regular ICS use among people with COPD and asthma on COVID-19–related mortality,” the investigators concluded.
In agreement with the investigators, Dr. Conroy said that the increased mortality rate among ICS users should not be misconstrued as a medication-related risk.
“While the study found that those with COPD or asthma taking ICS and high-dose ICS were at an increased risk of death, this could easily be explained by the likelihood that those are the patients who are more likely to have more severe underlying lung disease,” Dr. Conroy said. “While this observational study did attempt to control for exacerbation history, the ability to do so by electronic health records data is certainly imperfect.”
With this in mind, patients with chronic respiratory disease should be encouraged to adhere to their usual treatment regimen, Dr. Conroy added.
“There isn’t evidence to increase or decrease medications just because of the pandemic,” she said. “A patient with asthma or COPD should continue to take the medications that are needed to achieve good control of their lung disease.”
The study was funded by the U.K. Medical Research Council. The investigators reported additional relationships with the Wellcome Trust, the Good Thinking Foundation, the Laura and John Arnold Foundation, and others. Dr. Conroy reported no conflicts of interest.
SOURCE: Schultze A et al. Lancet Respir Med. 2020 Sep 24. doi: 10.1016/ S2213-2600(20)30415-X.
FROM LANCET RESPIRATORY MEDICINE
Retrospective Review on the Safety and Efficacy of Direct Oral Anticoagulants Compared With Warfarin in Patients With Cirrhosis
Coagulation in patients with cirrhosis is a complicated area of evolving research. Patients with cirrhosis were originally thought to be naturally anticoagulated due to the decreased production of clotting factors and platelets, combined with an increased international normalized ratio (INR).1 New data have shown that patients with cirrhosis are at a concomitant risk of bleeding and thrombosis due to increased platelet aggregation, decreased fibrinolysis, and decreased production of natural anticoagulants such as protein C and antithrombin.1 Traditionally, patients with cirrhosis needing anticoagulation therapy for comorbid conditions, such as nonvalvular atrial fibrillation (NVAF) or venous thromboembolism (VTE) were placed on warfarin therapy. Managing warfarin in patients with cirrhosis poses a challenge to clinicians due to the many food and drug interactions, narrow therapeutic index, and complications with maintaining a therapeutic INR.1
Direct oral anticoagulants (DOACs) have several benefits over warfarin therapy, including convenience, decreased monitoring, decreased drug and dietary restrictions, and faster onset of action.2 Conversely, DOACs undergo extensive hepatic metabolism giving rise to concerns about supratherapeutic drug levels and increased bleeding rates in patients with liver dysfunction.1 Consequently, patients with cirrhosis were excluded from the pivotal trials establishing DOACs for NVAF and VTE treatment. Exclusion of these patients in major clinical trials alongside the challenges of managing warfarin warrant an evaluation of the efficacy and safety of DOACs in patients with cirrhosis.
Recent retrospective studies have examined the use of DOACs in patients with cirrhosis and found favorable results. A retrospective chart review by Intagliata and colleagues consisting of 39 patients with cirrhosis using either a DOAC or warfarin found similar rates of all-cause bleeding and major bleeding between the 2 groups.3 A retrospective cohort study by Hum and colleagues consisting of 45 patients with cirrhosis compared the use of DOACs with warfarin or low-molecular weight heparin (LMWH).4 Hum and colleagues found patients prescribed a DOAC had significantly fewer major bleeding events than did patients using warfarin or LMWH.4 The largest retrospective cohort study consisted of 233 patients with chronic liver disease and found no differences among all-cause bleeding and major bleeding rates between patients using DOACs compared with those of patients using warfarin.5
The purpose of this research is to evaluate the safety and efficacy of DOACs in veteran patients with cirrhosis compared with patients using warfarin.
Methods
A retrospective single-center chart review was conducted at the Michael E. DeBakey Veterans Affairs Medical Center (MEDVAMC) in Houston, Texas, between October 31, 2014 and October 31, 2018. Patients included in the study were adults aged ≥ 18 years with a diagnosis of cirrhosis and prescribed any of the following oral anticoagulants: apixaban, dabigatran, edoxaban, rivaroxaban, or warfarin. Patients prescribed apixaban, dabigatran, edoxaban, or rivaroxaban were collectively grouped into the DOAC group, while patients prescribed warfarin were classified as the standard of care comparator group.
A diagnosis of cirrhosis was confirmed using a combination of the codes from the ninth and tenth editions of the International Classification of Diseases (ICD) for cirrhosis, documentation of diagnostic confirmation by clinicians from the gastroenterology or hepatology services, and positive liver biopsy result. Liver function tests, liver ultrasound results, and FibroSure biomarker assays were used to aid in confirming the diagnosis of cirrhosis but were not considered definitive. Patients were excluded from the trial if they had indications for anticoagulation other than NVAF and VTE and/or were prescribed triple antithrombotic therapy (dual antiplatelet therapy plus an anticoagulant). Patients who switched anticoagulant therapy during the trial period (ie, switched from warfarin to a DOAC) were also excluded from the analysis.
Patient demographic characteristics that were collected included weight; body mass index (BMI); etiology of cirrhosis; Child-Turcotte-Pugh, Model for End-Stage Liver Disease (MELD), and CHA2DS2-VASc score; concomitant antiplatelet, nonsteroidal anti-inflammatory drug (NSAID), proton pump inhibitor (PPI), and histamine-2 receptor antagonist
Two patient lists were used to identify patients for inclusion in the warfarin arm. The first patient list was generated using the US Department of Veterans Affairs (VA) Cirrhosis Tracker, which identified patients with an ICD-9/10 code for cirrhosis and an INR laboratory value. Patients generated from the VA Cirrhosis Tracker with an INR > 1.5 were screened for a warfarin prescription and then evaluated for full study inclusion. The second patient list was generated using the VA Advanced Liver Disease Dashboard which identified patients with ICD-9/10 codes for advanced liver disease and an active warfarin prescription. Patients with an active warfarin prescription were then evaluated for full study inclusion. A single patient list was generated to identify patients for inclusion in the DOAC arm. This patient list was generated using the VA DOAC dashboard, which identified patients with an active DOAC prescription and an ICD-9/10 code for cirrhosis. Patients with an ICD-9/10 code for cirrhosis and prescribed a DOAC were screened for full study inclusion. Patient data were collected from the MEDVAMC Computerized Patient Record System (CPRS) electronic health record (EHR). The research study was approved by the Baylor College of Medicine Institutional Review Board and the VA Office of Research and Development.
Outcomes
The primary endpoint for the study was all-cause bleeding. The secondary endpoints for the study were major bleeding and failed efficacy. Major bleeding was defined using the International Society on Thrombosis and Haemostasis (ISTH) 2005 definition: fatal bleeding, symptomatic bleeding in a critical organ area (ie, intracranial, intraspinal, intraocular, retroperitoneal, intraarticular, pericardial, or intramuscular with compartment syndrome), or bleeding causing a fall in hemoglobin level of > 2 g/dL or leading to the transfusion of ≥ 2 units of red cells.6 Failed efficacy was a combination endpoint that included development of VTE, stroke, myocardial infarction (MI), and/or death. A prespecified subgroup analysis was conducted at the end of the study period to analyze trends in the DOAC and warfarin groups with respect to all-cause bleeding. All-cause bleeding risk was stratified by weight, BMI, Child-Turcotte-Pugh score, MELD score, presence of gastric and/or esophageal varices, active malignancies, percentage of time within therapeutic INR range in the warfarin group, indications for anticoagulation, and antiplatelet, NSAID, PPI, and H2RA therapy.
Statistical Analysis
Data were analyzed using descriptive and inferential statistics. Continuous data were analyzed using the Student t test, and categorical data were analyzed using the Fisher exact test. Previous studies determined an all-cause bleeding rate of 10 to 17% for warfarin compared with 5% for DOACs.7,8 To detect a 12% difference in the all-cause bleeding rate between DOACs and warfarin, 212 patients would be needed to achieve 80% power at an α level of 0.05.
Results
A total of 170 patients were screened, and after applying inclusion and exclusion criteria, 79 patients were enrolled in the study (Figure). The DOAC group included 42 patients, and the warfarin group included 37 patients. In the DOAC group, 69.1% (n = 29) of patients were taking apixaban, 21.4% (n = 9) rivaroxaban, and 9.5% (n = 4) dabigatran. There were no patients prescribed edoxaban during the study period.
Baseline characteristics were similar between the 2 groups except for Child-Turcotte-Pugh score, MELD score, mean INR, and number of days on anticoagulation therapy (Table 1). Most of the patients were male (98.7%), and the mean age was 71 years. The most common causes of cirrhosis were viral (29.1%), nonalcoholic fatty liver disease (NAFLD) (24.1%), multiple causes (22.8%), and alcohol (21.5%). Sixty-two patients (78.5%) had a NVAF indication for anticoagulation. The average CHA2DS2-VASc score was 3.7. Aspirin was prescribed in 51.9% (n = 41) of patients, and PPIs were prescribed in 48.1% (n = 38) of patients. At inclusion, esophageal varices were present in 13 patients and active malignancies were present in 6 patients.
Statistically significant differences in baseline characteristics were found between mean INR, Child-Turcotte-Pugh scores, MELD scores, and number of days on anticoagulant therapy. The mean INR was 1.3 in the DOAC group compared with 2.1 in the warfarin group (P = .0001). Eighty-one percent (n = 34) of patients in the DOAC group had a Child-Turcotte-Pugh score of A compared with 43.2% (n = 16) of patients in the warfarin group (P = .0009). Eight patients in the DOAC group had a Child-Turcotte-Pugh score of B compared with 19 patients in the warfarin group (P = .004). The mean MELD score was 9.4 in the DOAC group compared with 16.3 in the warfarin group (P = .0001). The mean days on anticoagulant therapy was 500.4 days for the DOAC group compared with 1,652.4 days for the warfarin group (P = .0001).
Safety Outcome
The primary outcome comparing all-cause bleeding rates between patients on DOACs compared with warfarin are listed in Table 2. With respect to the primary outcome, 7 (16.7%) patients on DOACs experienced a bleeding event compared with 8 (21.6%) patients on warfarin (P = .77). No statistically significant differences were detected between the DOAC and warfarin groups with respect to all-cause bleeding. Seven bleeding events occurred in the DOAC group; 1 met the qualification for major bleeding with a suspected gastrointestinal (GI) bleed.6 The other 6 bleeding episodes in the DOAC group consisted of hematoma, epistaxis, hematuria, and hematochezia. Eight bleeding events occurred in the warfarin group; 2 met the qualification for major bleeding with an intracranial hemorrhage and upper GI bleed.6 The other 6 bleeding episodes in the warfarin group consisted of epistaxis, bleeding gums, hematuria, and hematochezia. There were no statistically significant differences between the rates of major bleeding and nonmajor bleeding between the DOAC and warfarin groups.
Efficacy Outcomes
There were 3 events in the DOAC group and 3 events in the warfarin group (P = .99). In the DOAC group, 2 patients experienced a pulmonary embolism, and 1 patient experienced a MI. In the warfarin group, 3 patients died (end-stage heart failure, unknown cause due to death at an outside hospital, and sepsis/organ failure). There were no statistically significant differences between the composite endpoint of failed efficacy or the individual endpoints of VTE, stroke, MI, and death.
Subgroup Analysis
A prespecified subgroup analysis was conducted to determine risk factors for all-cause bleeding within each treatment group (Table 3). No significant trends were observed in the following risk factors: Child-Turcotte-Pugh score, indication for anticoagulation, use of NSAIDs, PPIs or H2RAs, presence of gastric or esophageal varices, active malignancies, and time within therapeutic INR range in the warfarin group. Patients with bleeding events had slightly increased weight and BMI vs patients without bleeding events. Within the warfarin group, patients with bleeding events had slightly elevated MELD scores compared to patients without bleeding events. There was an equal balance of patients prescribed aspirin therapy between the groups with and without bleeding events. Overall, no significant risk factors were identified for all-cause bleeding.
Discussion
Initially, patients with cirrhosis were excluded from DOAC trials due to concerns for increased bleeding risk with hepatically eliminated medications. New retrospective research has concluded that in patients with cirrhosis, DOACs have similar or lower bleeding rates when compared directly to warfarin.9,10
In this study, no statistically significant differences were detected between the primary and secondary outcomes of all-cause bleeding, major bleeding, or failed efficacy. Subgroup analysis did not identify any significant risk factors with respect to all-cause bleeding among patients in the DOAC and warfarin groups. To meet 80% power, 212 patients needed to be enrolled in the study; however, only 79 patients were enrolled, and power was not met. The results of this study should be interpreted cautiously as hypothesis-generating due to the small sample size. Strengths of this study include similar baseline characteristics between the DOAC and warfarin groups, 4-year length of retrospective data review, and availability of both inpatient and outpatient EHR limiting the amount of missing data points.
Baseline characteristics were similar between the groups except for mean INR, Child-Turcotte-Pugh score, MELD score, and number of days on anticoagulation therapy. The difference in mean INR between groups is expected as patients in the warfarin group have a goal INR of 2 to 3 to maintain therapeutic efficacy and safety. INR is not used as a marker of efficacy or safety with DOACs; therefore, a consistent elevation in INR is not expected. Child- Turcotte-Pugh scores are calculated using INR levels.11 When calculating the score, patients with an INR < 1.7 receive 1 point; patients with an INR between 1.7 and 2.3 receive 2 points.11 Therefore, patients in the warfarin group will have artificially inflated Child-Turcotte-Pugh scores as this group has goal INR levels of 2 to 3. This makes Child-Turcotte-Pugh scores unreliable markers of disease severity in patients using warfarin therapy. When the INR scores for patients prescribed warfarin were replaced with values < 1.7, the statistical difference disappeared between the warfarin and DOAC groups. The same effect is seen on MELD scores for patients prescribed warfarin therapy. The MELD score is calculated using INR levels.12 MELD scores also will be artificially elevated in patients prescribed warfarin therapy due to the INR elevation to between 2 and 3. When MELD scores for patients prescribed warfarin were replaced with values similar to those in the DOAC group, the statistical difference disappeared between the warfarin and DOAC groups.
The last statistically significant difference was found in number of days on anticoagulant therapy. This difference was expected as warfarin is the standard of care for anticoagulation treatment in patients with cirrhosis. The first DOAC, dabigatran, was not approved by the US Food and Drug Administration until 2010.13 DOACs have only recently been used in patients with cirrhosis accounting for the statistically significant difference in days on anticoagulation therapy between the warfarin and DOAC groups.
Limitations
The inability to meet power or evaluate adherence and appropriate renal dose adjustments for DOACs limited this study. This study was conducted at a single center in a predominantly male veteran population and therefore may not be generalizable to other populations. A majority of patients in the DOAC group were prescribed apixaban (69.1%), which may have affected the overall rate of major bleeding in the DOAC group. Pivotal trials of apixaban have shown a consistent decreased risk of major bleeding in patients with NVAF or VTE when compared with warfarin.14,15 Therefore, the results of this study may not be generalizable to all DOACs.
An inherent limitation of this study was the inability to collect data verifying adherence in the DOAC group. However, in the warfarin group, percentage of time within the therapeutic INR range of 2 to 3 was collected. While not a direct marker of adherence, this does allow for limited evaluation of therapeutic efficacy and safety within the warfarin group. Last, proper dosing of DOACs in patients with and without adequate renal function was not evaluated in this study.
Conclusions
The results of this study are consistent with other retrospective research and literature reviews. There were no statistically significant differences identified between the rates of all-cause bleeding, major bleeding, and failed efficacy between the DOAC and warfarin groups. DOACs may be a safe alternative to warfarin in patients with cirrhosis requiring anticoagulation for NVAF or VTE, but large randomized trials are required to confirm these results.
1. Qamar A, Vaduganathan M, Greenberger NJ, Giugliano RP. Oral anticoagulation in patients with liver disease. J Am Coll Cardiol. 2018;71(19):2162-2175. doi:10.1016/j.jacc.2018.03.023
2. Priyanka P, Kupec JT, Krafft M, Shah NA, Reynolds GJ. Newer oral anticoagulants in the treatment of acute portal vein thrombosis in patients with and without cirrhosis. Int J Hepatol. 2018;2018:8432781. Published 2018 Jun 5. doi:10.1155/2018/8432781
3. Intagliata NM, Henry ZH, Maitland H, et al. Direct oral anticoagulants in cirrhosis patients pose similar risks of bleeding when compared to traditional anticoagulation. Dig Dis Sci. 2016;61(6):1721-1727. doi:10.1007/s10620-015-4012-2
4. Hum J, Shatzel JJ, Jou JH, Deloughery TG. The efficacy and safety of direct oral anticoagulants vs traditional anticoagulants in cirrhosis. Eur J Haematol. 2017;98(4):393-397. doi:10.1111/ejh.12844
5. Goriacko P, Veltri KT. Safety of direct oral anticoagulants vs warfarin in patients with chronic liver disease and atrial fibrillation. Eur J Haematol. 2018;100(5):488-493. doi:10.1111/ejh.13045
6. Schulman S, Kearon C; Subcommittee on Control of Anticoagulation of the Scientific and Standardization Committee of the International Society on Thrombosis and Haemostasis. Definition of major bleeding in clinical investigations of antihemostatic medicinal products in non-surgical patients. J Thromb Haemost. 2005;3(4):692-694. doi:10.1111/j.1538-7836.2005.01204.x
7. Rubboli A, Becattini C, Verheugt FW. Incidence, clinical impact and risk of bleeding during oral anticoagulation therapy. World J Cardiol. 2011;3(11):351-358. doi:10.4330/wjc.v3.i11.351
8. Ruff CT, Giugliano RP, Braunwald E, et al. Comparison of the efficacy and safety of new oral anticoagulants with warfarin in patients with atrial fibrillation: a meta-analysis of randomised trials. Lancet. 2014;383(9921):955-962. doi:10.1016/S0140-6736(13)62343-0
9. Hoolwerf EW, Kraaijpoel N, Büller HR, van Es N. Direct oral anticoagulants in patients with liver cirrhosis: A systematic review. Thromb Res. 2018;170:102-108. doi:10.1016/j.thromres.2018.08.011
10. Steuber TD, Howard ML, Nisly SA. Direct oral anticoagulants in chronic liver disease. Ann Pharmacother. 2019;53(10):1042-1049. doi:10.1177/1060028019841582
11. Janevska D, Chaloska-Ivanova V, Janevski V. Hepatocellular carcinoma: risk factors, diagnosis and treatment. Open Access Maced J Med Sci. 2015;3(4):732-736. doi:10.3889/oamjms.2015.111
12. Singal AK, Kamath PS. Model for End-Stage Liver Disease. J Clin Exp Hepatol. 2013;3(1):50-60. doi:10.1016/j.jceh.2012.11.002
13. Joppa SA, Salciccioli J, Adamski J, et al. A practical review of the emerging direct anticoagulants, laboratory monitoring, and reversal agents. J Clin Med. 2018;7(2):29. Published 2018 Feb 11. doi:10.3390/jcm7020029
14. Granger CB, Alexander JH, McMurray JJ, et al. Apixaban versus warfarin in patients with atrial fibrillation. N Engl J Med. 2011;365(11):981-992. doi:10.1056/NEJMoa1107039
15. Agnelli G, Buller HR, Cohen A, et al. Oral apixaban for the treatment of acute venous thromboembolism. N Engl J Med. 2013;369(9):799-808. doi:10.1056/NEJMoa1302507
Coagulation in patients with cirrhosis is a complicated area of evolving research. Patients with cirrhosis were originally thought to be naturally anticoagulated due to the decreased production of clotting factors and platelets, combined with an increased international normalized ratio (INR).1 New data have shown that patients with cirrhosis are at a concomitant risk of bleeding and thrombosis due to increased platelet aggregation, decreased fibrinolysis, and decreased production of natural anticoagulants such as protein C and antithrombin.1 Traditionally, patients with cirrhosis needing anticoagulation therapy for comorbid conditions, such as nonvalvular atrial fibrillation (NVAF) or venous thromboembolism (VTE) were placed on warfarin therapy. Managing warfarin in patients with cirrhosis poses a challenge to clinicians due to the many food and drug interactions, narrow therapeutic index, and complications with maintaining a therapeutic INR.1
Direct oral anticoagulants (DOACs) have several benefits over warfarin therapy, including convenience, decreased monitoring, decreased drug and dietary restrictions, and faster onset of action.2 Conversely, DOACs undergo extensive hepatic metabolism giving rise to concerns about supratherapeutic drug levels and increased bleeding rates in patients with liver dysfunction.1 Consequently, patients with cirrhosis were excluded from the pivotal trials establishing DOACs for NVAF and VTE treatment. Exclusion of these patients in major clinical trials alongside the challenges of managing warfarin warrant an evaluation of the efficacy and safety of DOACs in patients with cirrhosis.
Recent retrospective studies have examined the use of DOACs in patients with cirrhosis and found favorable results. A retrospective chart review by Intagliata and colleagues consisting of 39 patients with cirrhosis using either a DOAC or warfarin found similar rates of all-cause bleeding and major bleeding between the 2 groups.3 A retrospective cohort study by Hum and colleagues consisting of 45 patients with cirrhosis compared the use of DOACs with warfarin or low-molecular weight heparin (LMWH).4 Hum and colleagues found patients prescribed a DOAC had significantly fewer major bleeding events than did patients using warfarin or LMWH.4 The largest retrospective cohort study consisted of 233 patients with chronic liver disease and found no differences among all-cause bleeding and major bleeding rates between patients using DOACs compared with those of patients using warfarin.5
The purpose of this research is to evaluate the safety and efficacy of DOACs in veteran patients with cirrhosis compared with patients using warfarin.
Methods
A retrospective single-center chart review was conducted at the Michael E. DeBakey Veterans Affairs Medical Center (MEDVAMC) in Houston, Texas, between October 31, 2014 and October 31, 2018. Patients included in the study were adults aged ≥ 18 years with a diagnosis of cirrhosis and prescribed any of the following oral anticoagulants: apixaban, dabigatran, edoxaban, rivaroxaban, or warfarin. Patients prescribed apixaban, dabigatran, edoxaban, or rivaroxaban were collectively grouped into the DOAC group, while patients prescribed warfarin were classified as the standard of care comparator group.
A diagnosis of cirrhosis was confirmed using a combination of the codes from the ninth and tenth editions of the International Classification of Diseases (ICD) for cirrhosis, documentation of diagnostic confirmation by clinicians from the gastroenterology or hepatology services, and positive liver biopsy result. Liver function tests, liver ultrasound results, and FibroSure biomarker assays were used to aid in confirming the diagnosis of cirrhosis but were not considered definitive. Patients were excluded from the trial if they had indications for anticoagulation other than NVAF and VTE and/or were prescribed triple antithrombotic therapy (dual antiplatelet therapy plus an anticoagulant). Patients who switched anticoagulant therapy during the trial period (ie, switched from warfarin to a DOAC) were also excluded from the analysis.
Patient demographic characteristics that were collected included weight; body mass index (BMI); etiology of cirrhosis; Child-Turcotte-Pugh, Model for End-Stage Liver Disease (MELD), and CHA2DS2-VASc score; concomitant antiplatelet, nonsteroidal anti-inflammatory drug (NSAID), proton pump inhibitor (PPI), and histamine-2 receptor antagonist
Two patient lists were used to identify patients for inclusion in the warfarin arm. The first patient list was generated using the US Department of Veterans Affairs (VA) Cirrhosis Tracker, which identified patients with an ICD-9/10 code for cirrhosis and an INR laboratory value. Patients generated from the VA Cirrhosis Tracker with an INR > 1.5 were screened for a warfarin prescription and then evaluated for full study inclusion. The second patient list was generated using the VA Advanced Liver Disease Dashboard which identified patients with ICD-9/10 codes for advanced liver disease and an active warfarin prescription. Patients with an active warfarin prescription were then evaluated for full study inclusion. A single patient list was generated to identify patients for inclusion in the DOAC arm. This patient list was generated using the VA DOAC dashboard, which identified patients with an active DOAC prescription and an ICD-9/10 code for cirrhosis. Patients with an ICD-9/10 code for cirrhosis and prescribed a DOAC were screened for full study inclusion. Patient data were collected from the MEDVAMC Computerized Patient Record System (CPRS) electronic health record (EHR). The research study was approved by the Baylor College of Medicine Institutional Review Board and the VA Office of Research and Development.
Outcomes
The primary endpoint for the study was all-cause bleeding. The secondary endpoints for the study were major bleeding and failed efficacy. Major bleeding was defined using the International Society on Thrombosis and Haemostasis (ISTH) 2005 definition: fatal bleeding, symptomatic bleeding in a critical organ area (ie, intracranial, intraspinal, intraocular, retroperitoneal, intraarticular, pericardial, or intramuscular with compartment syndrome), or bleeding causing a fall in hemoglobin level of > 2 g/dL or leading to the transfusion of ≥ 2 units of red cells.6 Failed efficacy was a combination endpoint that included development of VTE, stroke, myocardial infarction (MI), and/or death. A prespecified subgroup analysis was conducted at the end of the study period to analyze trends in the DOAC and warfarin groups with respect to all-cause bleeding. All-cause bleeding risk was stratified by weight, BMI, Child-Turcotte-Pugh score, MELD score, presence of gastric and/or esophageal varices, active malignancies, percentage of time within therapeutic INR range in the warfarin group, indications for anticoagulation, and antiplatelet, NSAID, PPI, and H2RA therapy.
Statistical Analysis
Data were analyzed using descriptive and inferential statistics. Continuous data were analyzed using the Student t test, and categorical data were analyzed using the Fisher exact test. Previous studies determined an all-cause bleeding rate of 10 to 17% for warfarin compared with 5% for DOACs.7,8 To detect a 12% difference in the all-cause bleeding rate between DOACs and warfarin, 212 patients would be needed to achieve 80% power at an α level of 0.05.
Results
A total of 170 patients were screened, and after applying inclusion and exclusion criteria, 79 patients were enrolled in the study (Figure). The DOAC group included 42 patients, and the warfarin group included 37 patients. In the DOAC group, 69.1% (n = 29) of patients were taking apixaban, 21.4% (n = 9) rivaroxaban, and 9.5% (n = 4) dabigatran. There were no patients prescribed edoxaban during the study period.
Baseline characteristics were similar between the 2 groups except for Child-Turcotte-Pugh score, MELD score, mean INR, and number of days on anticoagulation therapy (Table 1). Most of the patients were male (98.7%), and the mean age was 71 years. The most common causes of cirrhosis were viral (29.1%), nonalcoholic fatty liver disease (NAFLD) (24.1%), multiple causes (22.8%), and alcohol (21.5%). Sixty-two patients (78.5%) had a NVAF indication for anticoagulation. The average CHA2DS2-VASc score was 3.7. Aspirin was prescribed in 51.9% (n = 41) of patients, and PPIs were prescribed in 48.1% (n = 38) of patients. At inclusion, esophageal varices were present in 13 patients and active malignancies were present in 6 patients.
Statistically significant differences in baseline characteristics were found between mean INR, Child-Turcotte-Pugh scores, MELD scores, and number of days on anticoagulant therapy. The mean INR was 1.3 in the DOAC group compared with 2.1 in the warfarin group (P = .0001). Eighty-one percent (n = 34) of patients in the DOAC group had a Child-Turcotte-Pugh score of A compared with 43.2% (n = 16) of patients in the warfarin group (P = .0009). Eight patients in the DOAC group had a Child-Turcotte-Pugh score of B compared with 19 patients in the warfarin group (P = .004). The mean MELD score was 9.4 in the DOAC group compared with 16.3 in the warfarin group (P = .0001). The mean days on anticoagulant therapy was 500.4 days for the DOAC group compared with 1,652.4 days for the warfarin group (P = .0001).
Safety Outcome
The primary outcome comparing all-cause bleeding rates between patients on DOACs compared with warfarin are listed in Table 2. With respect to the primary outcome, 7 (16.7%) patients on DOACs experienced a bleeding event compared with 8 (21.6%) patients on warfarin (P = .77). No statistically significant differences were detected between the DOAC and warfarin groups with respect to all-cause bleeding. Seven bleeding events occurred in the DOAC group; 1 met the qualification for major bleeding with a suspected gastrointestinal (GI) bleed.6 The other 6 bleeding episodes in the DOAC group consisted of hematoma, epistaxis, hematuria, and hematochezia. Eight bleeding events occurred in the warfarin group; 2 met the qualification for major bleeding with an intracranial hemorrhage and upper GI bleed.6 The other 6 bleeding episodes in the warfarin group consisted of epistaxis, bleeding gums, hematuria, and hematochezia. There were no statistically significant differences between the rates of major bleeding and nonmajor bleeding between the DOAC and warfarin groups.
Efficacy Outcomes
There were 3 events in the DOAC group and 3 events in the warfarin group (P = .99). In the DOAC group, 2 patients experienced a pulmonary embolism, and 1 patient experienced a MI. In the warfarin group, 3 patients died (end-stage heart failure, unknown cause due to death at an outside hospital, and sepsis/organ failure). There were no statistically significant differences between the composite endpoint of failed efficacy or the individual endpoints of VTE, stroke, MI, and death.
Subgroup Analysis
A prespecified subgroup analysis was conducted to determine risk factors for all-cause bleeding within each treatment group (Table 3). No significant trends were observed in the following risk factors: Child-Turcotte-Pugh score, indication for anticoagulation, use of NSAIDs, PPIs or H2RAs, presence of gastric or esophageal varices, active malignancies, and time within therapeutic INR range in the warfarin group. Patients with bleeding events had slightly increased weight and BMI vs patients without bleeding events. Within the warfarin group, patients with bleeding events had slightly elevated MELD scores compared to patients without bleeding events. There was an equal balance of patients prescribed aspirin therapy between the groups with and without bleeding events. Overall, no significant risk factors were identified for all-cause bleeding.
Discussion
Initially, patients with cirrhosis were excluded from DOAC trials due to concerns for increased bleeding risk with hepatically eliminated medications. New retrospective research has concluded that in patients with cirrhosis, DOACs have similar or lower bleeding rates when compared directly to warfarin.9,10
In this study, no statistically significant differences were detected between the primary and secondary outcomes of all-cause bleeding, major bleeding, or failed efficacy. Subgroup analysis did not identify any significant risk factors with respect to all-cause bleeding among patients in the DOAC and warfarin groups. To meet 80% power, 212 patients needed to be enrolled in the study; however, only 79 patients were enrolled, and power was not met. The results of this study should be interpreted cautiously as hypothesis-generating due to the small sample size. Strengths of this study include similar baseline characteristics between the DOAC and warfarin groups, 4-year length of retrospective data review, and availability of both inpatient and outpatient EHR limiting the amount of missing data points.
Baseline characteristics were similar between the groups except for mean INR, Child-Turcotte-Pugh score, MELD score, and number of days on anticoagulation therapy. The difference in mean INR between groups is expected as patients in the warfarin group have a goal INR of 2 to 3 to maintain therapeutic efficacy and safety. INR is not used as a marker of efficacy or safety with DOACs; therefore, a consistent elevation in INR is not expected. Child- Turcotte-Pugh scores are calculated using INR levels.11 When calculating the score, patients with an INR < 1.7 receive 1 point; patients with an INR between 1.7 and 2.3 receive 2 points.11 Therefore, patients in the warfarin group will have artificially inflated Child-Turcotte-Pugh scores as this group has goal INR levels of 2 to 3. This makes Child-Turcotte-Pugh scores unreliable markers of disease severity in patients using warfarin therapy. When the INR scores for patients prescribed warfarin were replaced with values < 1.7, the statistical difference disappeared between the warfarin and DOAC groups. The same effect is seen on MELD scores for patients prescribed warfarin therapy. The MELD score is calculated using INR levels.12 MELD scores also will be artificially elevated in patients prescribed warfarin therapy due to the INR elevation to between 2 and 3. When MELD scores for patients prescribed warfarin were replaced with values similar to those in the DOAC group, the statistical difference disappeared between the warfarin and DOAC groups.
The last statistically significant difference was found in number of days on anticoagulant therapy. This difference was expected as warfarin is the standard of care for anticoagulation treatment in patients with cirrhosis. The first DOAC, dabigatran, was not approved by the US Food and Drug Administration until 2010.13 DOACs have only recently been used in patients with cirrhosis accounting for the statistically significant difference in days on anticoagulation therapy between the warfarin and DOAC groups.
Limitations
The inability to meet power or evaluate adherence and appropriate renal dose adjustments for DOACs limited this study. This study was conducted at a single center in a predominantly male veteran population and therefore may not be generalizable to other populations. A majority of patients in the DOAC group were prescribed apixaban (69.1%), which may have affected the overall rate of major bleeding in the DOAC group. Pivotal trials of apixaban have shown a consistent decreased risk of major bleeding in patients with NVAF or VTE when compared with warfarin.14,15 Therefore, the results of this study may not be generalizable to all DOACs.
An inherent limitation of this study was the inability to collect data verifying adherence in the DOAC group. However, in the warfarin group, percentage of time within the therapeutic INR range of 2 to 3 was collected. While not a direct marker of adherence, this does allow for limited evaluation of therapeutic efficacy and safety within the warfarin group. Last, proper dosing of DOACs in patients with and without adequate renal function was not evaluated in this study.
Conclusions
The results of this study are consistent with other retrospective research and literature reviews. There were no statistically significant differences identified between the rates of all-cause bleeding, major bleeding, and failed efficacy between the DOAC and warfarin groups. DOACs may be a safe alternative to warfarin in patients with cirrhosis requiring anticoagulation for NVAF or VTE, but large randomized trials are required to confirm these results.
Coagulation in patients with cirrhosis is a complicated area of evolving research. Patients with cirrhosis were originally thought to be naturally anticoagulated due to the decreased production of clotting factors and platelets, combined with an increased international normalized ratio (INR).1 New data have shown that patients with cirrhosis are at a concomitant risk of bleeding and thrombosis due to increased platelet aggregation, decreased fibrinolysis, and decreased production of natural anticoagulants such as protein C and antithrombin.1 Traditionally, patients with cirrhosis needing anticoagulation therapy for comorbid conditions, such as nonvalvular atrial fibrillation (NVAF) or venous thromboembolism (VTE) were placed on warfarin therapy. Managing warfarin in patients with cirrhosis poses a challenge to clinicians due to the many food and drug interactions, narrow therapeutic index, and complications with maintaining a therapeutic INR.1
Direct oral anticoagulants (DOACs) have several benefits over warfarin therapy, including convenience, decreased monitoring, decreased drug and dietary restrictions, and faster onset of action.2 Conversely, DOACs undergo extensive hepatic metabolism giving rise to concerns about supratherapeutic drug levels and increased bleeding rates in patients with liver dysfunction.1 Consequently, patients with cirrhosis were excluded from the pivotal trials establishing DOACs for NVAF and VTE treatment. Exclusion of these patients in major clinical trials alongside the challenges of managing warfarin warrant an evaluation of the efficacy and safety of DOACs in patients with cirrhosis.
Recent retrospective studies have examined the use of DOACs in patients with cirrhosis and found favorable results. A retrospective chart review by Intagliata and colleagues consisting of 39 patients with cirrhosis using either a DOAC or warfarin found similar rates of all-cause bleeding and major bleeding between the 2 groups.3 A retrospective cohort study by Hum and colleagues consisting of 45 patients with cirrhosis compared the use of DOACs with warfarin or low-molecular weight heparin (LMWH).4 Hum and colleagues found patients prescribed a DOAC had significantly fewer major bleeding events than did patients using warfarin or LMWH.4 The largest retrospective cohort study consisted of 233 patients with chronic liver disease and found no differences among all-cause bleeding and major bleeding rates between patients using DOACs compared with those of patients using warfarin.5
The purpose of this research is to evaluate the safety and efficacy of DOACs in veteran patients with cirrhosis compared with patients using warfarin.
Methods
A retrospective single-center chart review was conducted at the Michael E. DeBakey Veterans Affairs Medical Center (MEDVAMC) in Houston, Texas, between October 31, 2014 and October 31, 2018. Patients included in the study were adults aged ≥ 18 years with a diagnosis of cirrhosis and prescribed any of the following oral anticoagulants: apixaban, dabigatran, edoxaban, rivaroxaban, or warfarin. Patients prescribed apixaban, dabigatran, edoxaban, or rivaroxaban were collectively grouped into the DOAC group, while patients prescribed warfarin were classified as the standard of care comparator group.
A diagnosis of cirrhosis was confirmed using a combination of the codes from the ninth and tenth editions of the International Classification of Diseases (ICD) for cirrhosis, documentation of diagnostic confirmation by clinicians from the gastroenterology or hepatology services, and positive liver biopsy result. Liver function tests, liver ultrasound results, and FibroSure biomarker assays were used to aid in confirming the diagnosis of cirrhosis but were not considered definitive. Patients were excluded from the trial if they had indications for anticoagulation other than NVAF and VTE and/or were prescribed triple antithrombotic therapy (dual antiplatelet therapy plus an anticoagulant). Patients who switched anticoagulant therapy during the trial period (ie, switched from warfarin to a DOAC) were also excluded from the analysis.
Patient demographic characteristics that were collected included weight; body mass index (BMI); etiology of cirrhosis; Child-Turcotte-Pugh, Model for End-Stage Liver Disease (MELD), and CHA2DS2-VASc score; concomitant antiplatelet, nonsteroidal anti-inflammatory drug (NSAID), proton pump inhibitor (PPI), and histamine-2 receptor antagonist
Two patient lists were used to identify patients for inclusion in the warfarin arm. The first patient list was generated using the US Department of Veterans Affairs (VA) Cirrhosis Tracker, which identified patients with an ICD-9/10 code for cirrhosis and an INR laboratory value. Patients generated from the VA Cirrhosis Tracker with an INR > 1.5 were screened for a warfarin prescription and then evaluated for full study inclusion. The second patient list was generated using the VA Advanced Liver Disease Dashboard which identified patients with ICD-9/10 codes for advanced liver disease and an active warfarin prescription. Patients with an active warfarin prescription were then evaluated for full study inclusion. A single patient list was generated to identify patients for inclusion in the DOAC arm. This patient list was generated using the VA DOAC dashboard, which identified patients with an active DOAC prescription and an ICD-9/10 code for cirrhosis. Patients with an ICD-9/10 code for cirrhosis and prescribed a DOAC were screened for full study inclusion. Patient data were collected from the MEDVAMC Computerized Patient Record System (CPRS) electronic health record (EHR). The research study was approved by the Baylor College of Medicine Institutional Review Board and the VA Office of Research and Development.
Outcomes
The primary endpoint for the study was all-cause bleeding. The secondary endpoints for the study were major bleeding and failed efficacy. Major bleeding was defined using the International Society on Thrombosis and Haemostasis (ISTH) 2005 definition: fatal bleeding, symptomatic bleeding in a critical organ area (ie, intracranial, intraspinal, intraocular, retroperitoneal, intraarticular, pericardial, or intramuscular with compartment syndrome), or bleeding causing a fall in hemoglobin level of > 2 g/dL or leading to the transfusion of ≥ 2 units of red cells.6 Failed efficacy was a combination endpoint that included development of VTE, stroke, myocardial infarction (MI), and/or death. A prespecified subgroup analysis was conducted at the end of the study period to analyze trends in the DOAC and warfarin groups with respect to all-cause bleeding. All-cause bleeding risk was stratified by weight, BMI, Child-Turcotte-Pugh score, MELD score, presence of gastric and/or esophageal varices, active malignancies, percentage of time within therapeutic INR range in the warfarin group, indications for anticoagulation, and antiplatelet, NSAID, PPI, and H2RA therapy.
Statistical Analysis
Data were analyzed using descriptive and inferential statistics. Continuous data were analyzed using the Student t test, and categorical data were analyzed using the Fisher exact test. Previous studies determined an all-cause bleeding rate of 10 to 17% for warfarin compared with 5% for DOACs.7,8 To detect a 12% difference in the all-cause bleeding rate between DOACs and warfarin, 212 patients would be needed to achieve 80% power at an α level of 0.05.
Results
A total of 170 patients were screened, and after applying inclusion and exclusion criteria, 79 patients were enrolled in the study (Figure). The DOAC group included 42 patients, and the warfarin group included 37 patients. In the DOAC group, 69.1% (n = 29) of patients were taking apixaban, 21.4% (n = 9) rivaroxaban, and 9.5% (n = 4) dabigatran. There were no patients prescribed edoxaban during the study period.
Baseline characteristics were similar between the 2 groups except for Child-Turcotte-Pugh score, MELD score, mean INR, and number of days on anticoagulation therapy (Table 1). Most of the patients were male (98.7%), and the mean age was 71 years. The most common causes of cirrhosis were viral (29.1%), nonalcoholic fatty liver disease (NAFLD) (24.1%), multiple causes (22.8%), and alcohol (21.5%). Sixty-two patients (78.5%) had a NVAF indication for anticoagulation. The average CHA2DS2-VASc score was 3.7. Aspirin was prescribed in 51.9% (n = 41) of patients, and PPIs were prescribed in 48.1% (n = 38) of patients. At inclusion, esophageal varices were present in 13 patients and active malignancies were present in 6 patients.
Statistically significant differences in baseline characteristics were found between mean INR, Child-Turcotte-Pugh scores, MELD scores, and number of days on anticoagulant therapy. The mean INR was 1.3 in the DOAC group compared with 2.1 in the warfarin group (P = .0001). Eighty-one percent (n = 34) of patients in the DOAC group had a Child-Turcotte-Pugh score of A compared with 43.2% (n = 16) of patients in the warfarin group (P = .0009). Eight patients in the DOAC group had a Child-Turcotte-Pugh score of B compared with 19 patients in the warfarin group (P = .004). The mean MELD score was 9.4 in the DOAC group compared with 16.3 in the warfarin group (P = .0001). The mean days on anticoagulant therapy was 500.4 days for the DOAC group compared with 1,652.4 days for the warfarin group (P = .0001).
Safety Outcome
The primary outcome comparing all-cause bleeding rates between patients on DOACs compared with warfarin are listed in Table 2. With respect to the primary outcome, 7 (16.7%) patients on DOACs experienced a bleeding event compared with 8 (21.6%) patients on warfarin (P = .77). No statistically significant differences were detected between the DOAC and warfarin groups with respect to all-cause bleeding. Seven bleeding events occurred in the DOAC group; 1 met the qualification for major bleeding with a suspected gastrointestinal (GI) bleed.6 The other 6 bleeding episodes in the DOAC group consisted of hematoma, epistaxis, hematuria, and hematochezia. Eight bleeding events occurred in the warfarin group; 2 met the qualification for major bleeding with an intracranial hemorrhage and upper GI bleed.6 The other 6 bleeding episodes in the warfarin group consisted of epistaxis, bleeding gums, hematuria, and hematochezia. There were no statistically significant differences between the rates of major bleeding and nonmajor bleeding between the DOAC and warfarin groups.
Efficacy Outcomes
There were 3 events in the DOAC group and 3 events in the warfarin group (P = .99). In the DOAC group, 2 patients experienced a pulmonary embolism, and 1 patient experienced a MI. In the warfarin group, 3 patients died (end-stage heart failure, unknown cause due to death at an outside hospital, and sepsis/organ failure). There were no statistically significant differences between the composite endpoint of failed efficacy or the individual endpoints of VTE, stroke, MI, and death.
Subgroup Analysis
A prespecified subgroup analysis was conducted to determine risk factors for all-cause bleeding within each treatment group (Table 3). No significant trends were observed in the following risk factors: Child-Turcotte-Pugh score, indication for anticoagulation, use of NSAIDs, PPIs or H2RAs, presence of gastric or esophageal varices, active malignancies, and time within therapeutic INR range in the warfarin group. Patients with bleeding events had slightly increased weight and BMI vs patients without bleeding events. Within the warfarin group, patients with bleeding events had slightly elevated MELD scores compared to patients without bleeding events. There was an equal balance of patients prescribed aspirin therapy between the groups with and without bleeding events. Overall, no significant risk factors were identified for all-cause bleeding.
Discussion
Initially, patients with cirrhosis were excluded from DOAC trials due to concerns for increased bleeding risk with hepatically eliminated medications. New retrospective research has concluded that in patients with cirrhosis, DOACs have similar or lower bleeding rates when compared directly to warfarin.9,10
In this study, no statistically significant differences were detected between the primary and secondary outcomes of all-cause bleeding, major bleeding, or failed efficacy. Subgroup analysis did not identify any significant risk factors with respect to all-cause bleeding among patients in the DOAC and warfarin groups. To meet 80% power, 212 patients needed to be enrolled in the study; however, only 79 patients were enrolled, and power was not met. The results of this study should be interpreted cautiously as hypothesis-generating due to the small sample size. Strengths of this study include similar baseline characteristics between the DOAC and warfarin groups, 4-year length of retrospective data review, and availability of both inpatient and outpatient EHR limiting the amount of missing data points.
Baseline characteristics were similar between the groups except for mean INR, Child-Turcotte-Pugh score, MELD score, and number of days on anticoagulation therapy. The difference in mean INR between groups is expected as patients in the warfarin group have a goal INR of 2 to 3 to maintain therapeutic efficacy and safety. INR is not used as a marker of efficacy or safety with DOACs; therefore, a consistent elevation in INR is not expected. Child- Turcotte-Pugh scores are calculated using INR levels.11 When calculating the score, patients with an INR < 1.7 receive 1 point; patients with an INR between 1.7 and 2.3 receive 2 points.11 Therefore, patients in the warfarin group will have artificially inflated Child-Turcotte-Pugh scores as this group has goal INR levels of 2 to 3. This makes Child-Turcotte-Pugh scores unreliable markers of disease severity in patients using warfarin therapy. When the INR scores for patients prescribed warfarin were replaced with values < 1.7, the statistical difference disappeared between the warfarin and DOAC groups. The same effect is seen on MELD scores for patients prescribed warfarin therapy. The MELD score is calculated using INR levels.12 MELD scores also will be artificially elevated in patients prescribed warfarin therapy due to the INR elevation to between 2 and 3. When MELD scores for patients prescribed warfarin were replaced with values similar to those in the DOAC group, the statistical difference disappeared between the warfarin and DOAC groups.
The last statistically significant difference was found in number of days on anticoagulant therapy. This difference was expected as warfarin is the standard of care for anticoagulation treatment in patients with cirrhosis. The first DOAC, dabigatran, was not approved by the US Food and Drug Administration until 2010.13 DOACs have only recently been used in patients with cirrhosis accounting for the statistically significant difference in days on anticoagulation therapy between the warfarin and DOAC groups.
Limitations
The inability to meet power or evaluate adherence and appropriate renal dose adjustments for DOACs limited this study. This study was conducted at a single center in a predominantly male veteran population and therefore may not be generalizable to other populations. A majority of patients in the DOAC group were prescribed apixaban (69.1%), which may have affected the overall rate of major bleeding in the DOAC group. Pivotal trials of apixaban have shown a consistent decreased risk of major bleeding in patients with NVAF or VTE when compared with warfarin.14,15 Therefore, the results of this study may not be generalizable to all DOACs.
An inherent limitation of this study was the inability to collect data verifying adherence in the DOAC group. However, in the warfarin group, percentage of time within the therapeutic INR range of 2 to 3 was collected. While not a direct marker of adherence, this does allow for limited evaluation of therapeutic efficacy and safety within the warfarin group. Last, proper dosing of DOACs in patients with and without adequate renal function was not evaluated in this study.
Conclusions
The results of this study are consistent with other retrospective research and literature reviews. There were no statistically significant differences identified between the rates of all-cause bleeding, major bleeding, and failed efficacy between the DOAC and warfarin groups. DOACs may be a safe alternative to warfarin in patients with cirrhosis requiring anticoagulation for NVAF or VTE, but large randomized trials are required to confirm these results.
1. Qamar A, Vaduganathan M, Greenberger NJ, Giugliano RP. Oral anticoagulation in patients with liver disease. J Am Coll Cardiol. 2018;71(19):2162-2175. doi:10.1016/j.jacc.2018.03.023
2. Priyanka P, Kupec JT, Krafft M, Shah NA, Reynolds GJ. Newer oral anticoagulants in the treatment of acute portal vein thrombosis in patients with and without cirrhosis. Int J Hepatol. 2018;2018:8432781. Published 2018 Jun 5. doi:10.1155/2018/8432781
3. Intagliata NM, Henry ZH, Maitland H, et al. Direct oral anticoagulants in cirrhosis patients pose similar risks of bleeding when compared to traditional anticoagulation. Dig Dis Sci. 2016;61(6):1721-1727. doi:10.1007/s10620-015-4012-2
4. Hum J, Shatzel JJ, Jou JH, Deloughery TG. The efficacy and safety of direct oral anticoagulants vs traditional anticoagulants in cirrhosis. Eur J Haematol. 2017;98(4):393-397. doi:10.1111/ejh.12844
5. Goriacko P, Veltri KT. Safety of direct oral anticoagulants vs warfarin in patients with chronic liver disease and atrial fibrillation. Eur J Haematol. 2018;100(5):488-493. doi:10.1111/ejh.13045
6. Schulman S, Kearon C; Subcommittee on Control of Anticoagulation of the Scientific and Standardization Committee of the International Society on Thrombosis and Haemostasis. Definition of major bleeding in clinical investigations of antihemostatic medicinal products in non-surgical patients. J Thromb Haemost. 2005;3(4):692-694. doi:10.1111/j.1538-7836.2005.01204.x
7. Rubboli A, Becattini C, Verheugt FW. Incidence, clinical impact and risk of bleeding during oral anticoagulation therapy. World J Cardiol. 2011;3(11):351-358. doi:10.4330/wjc.v3.i11.351
8. Ruff CT, Giugliano RP, Braunwald E, et al. Comparison of the efficacy and safety of new oral anticoagulants with warfarin in patients with atrial fibrillation: a meta-analysis of randomised trials. Lancet. 2014;383(9921):955-962. doi:10.1016/S0140-6736(13)62343-0
9. Hoolwerf EW, Kraaijpoel N, Büller HR, van Es N. Direct oral anticoagulants in patients with liver cirrhosis: A systematic review. Thromb Res. 2018;170:102-108. doi:10.1016/j.thromres.2018.08.011
10. Steuber TD, Howard ML, Nisly SA. Direct oral anticoagulants in chronic liver disease. Ann Pharmacother. 2019;53(10):1042-1049. doi:10.1177/1060028019841582
11. Janevska D, Chaloska-Ivanova V, Janevski V. Hepatocellular carcinoma: risk factors, diagnosis and treatment. Open Access Maced J Med Sci. 2015;3(4):732-736. doi:10.3889/oamjms.2015.111
12. Singal AK, Kamath PS. Model for End-Stage Liver Disease. J Clin Exp Hepatol. 2013;3(1):50-60. doi:10.1016/j.jceh.2012.11.002
13. Joppa SA, Salciccioli J, Adamski J, et al. A practical review of the emerging direct anticoagulants, laboratory monitoring, and reversal agents. J Clin Med. 2018;7(2):29. Published 2018 Feb 11. doi:10.3390/jcm7020029
14. Granger CB, Alexander JH, McMurray JJ, et al. Apixaban versus warfarin in patients with atrial fibrillation. N Engl J Med. 2011;365(11):981-992. doi:10.1056/NEJMoa1107039
15. Agnelli G, Buller HR, Cohen A, et al. Oral apixaban for the treatment of acute venous thromboembolism. N Engl J Med. 2013;369(9):799-808. doi:10.1056/NEJMoa1302507
1. Qamar A, Vaduganathan M, Greenberger NJ, Giugliano RP. Oral anticoagulation in patients with liver disease. J Am Coll Cardiol. 2018;71(19):2162-2175. doi:10.1016/j.jacc.2018.03.023
2. Priyanka P, Kupec JT, Krafft M, Shah NA, Reynolds GJ. Newer oral anticoagulants in the treatment of acute portal vein thrombosis in patients with and without cirrhosis. Int J Hepatol. 2018;2018:8432781. Published 2018 Jun 5. doi:10.1155/2018/8432781
3. Intagliata NM, Henry ZH, Maitland H, et al. Direct oral anticoagulants in cirrhosis patients pose similar risks of bleeding when compared to traditional anticoagulation. Dig Dis Sci. 2016;61(6):1721-1727. doi:10.1007/s10620-015-4012-2
4. Hum J, Shatzel JJ, Jou JH, Deloughery TG. The efficacy and safety of direct oral anticoagulants vs traditional anticoagulants in cirrhosis. Eur J Haematol. 2017;98(4):393-397. doi:10.1111/ejh.12844
5. Goriacko P, Veltri KT. Safety of direct oral anticoagulants vs warfarin in patients with chronic liver disease and atrial fibrillation. Eur J Haematol. 2018;100(5):488-493. doi:10.1111/ejh.13045
6. Schulman S, Kearon C; Subcommittee on Control of Anticoagulation of the Scientific and Standardization Committee of the International Society on Thrombosis and Haemostasis. Definition of major bleeding in clinical investigations of antihemostatic medicinal products in non-surgical patients. J Thromb Haemost. 2005;3(4):692-694. doi:10.1111/j.1538-7836.2005.01204.x
7. Rubboli A, Becattini C, Verheugt FW. Incidence, clinical impact and risk of bleeding during oral anticoagulation therapy. World J Cardiol. 2011;3(11):351-358. doi:10.4330/wjc.v3.i11.351
8. Ruff CT, Giugliano RP, Braunwald E, et al. Comparison of the efficacy and safety of new oral anticoagulants with warfarin in patients with atrial fibrillation: a meta-analysis of randomised trials. Lancet. 2014;383(9921):955-962. doi:10.1016/S0140-6736(13)62343-0
9. Hoolwerf EW, Kraaijpoel N, Büller HR, van Es N. Direct oral anticoagulants in patients with liver cirrhosis: A systematic review. Thromb Res. 2018;170:102-108. doi:10.1016/j.thromres.2018.08.011
10. Steuber TD, Howard ML, Nisly SA. Direct oral anticoagulants in chronic liver disease. Ann Pharmacother. 2019;53(10):1042-1049. doi:10.1177/1060028019841582
11. Janevska D, Chaloska-Ivanova V, Janevski V. Hepatocellular carcinoma: risk factors, diagnosis and treatment. Open Access Maced J Med Sci. 2015;3(4):732-736. doi:10.3889/oamjms.2015.111
12. Singal AK, Kamath PS. Model for End-Stage Liver Disease. J Clin Exp Hepatol. 2013;3(1):50-60. doi:10.1016/j.jceh.2012.11.002
13. Joppa SA, Salciccioli J, Adamski J, et al. A practical review of the emerging direct anticoagulants, laboratory monitoring, and reversal agents. J Clin Med. 2018;7(2):29. Published 2018 Feb 11. doi:10.3390/jcm7020029
14. Granger CB, Alexander JH, McMurray JJ, et al. Apixaban versus warfarin in patients with atrial fibrillation. N Engl J Med. 2011;365(11):981-992. doi:10.1056/NEJMoa1107039
15. Agnelli G, Buller HR, Cohen A, et al. Oral apixaban for the treatment of acute venous thromboembolism. N Engl J Med. 2013;369(9):799-808. doi:10.1056/NEJMoa1302507
Multidisciplinary Transitional Pain Service for the Veteran Population
Despite advancements in techniques, postsurgical pain continues to be a prominent part of the patient experience. Often this experience can lead to developing chronic postsurgical pain that interferes with quality of life after the expected time to recovery.1-3 As many as 14% of patients who undergo surgery without any history of opioid use develop chronic opioid use that persists after recovery from their operation.4-8 For patients with existing chronic opioid use or a history of substance use disorder (SUD), surgeons, primary care providers, or addiction providers often do not provide sufficient presurgical planning or postsurgical coordination of care. This lack of pain care coordination can increase the risk of inadequate pain control, opioid use escalation, or SUD relapse after surgery.
Convincing arguments have been made that a perioperative surgical home can improve significantly the quality of perioperative care.9-14 This report describes our experience implementing a perioperative surgical home at the US Department of Veterans Affairs (VA) Salt Lake City VA Medical Center (SLCVAMC), focusing on pain management extending from the preoperative period until 6 months or more after surgery. This type of Transitional Pain Service (TPS) has been described previously.15-17 Our service differs from those described previously by enrolling all patients before surgery rather than select postsurgical enrollment of only patients with a history of opioid use or SUD or patients who struggle with persistent postsurgical pain.
Methods
In January 2018, we developed and implemented a new TPS at the SLCVAMC. The transitional pain team consisted of an anesthesiologist with specialization in acute pain management, a nurse practitioner (NP) with experience in both acute and chronic pain management, 2 nurse care coordinators, and a psychologist (Figure 1). Before implementation, a needs assessment took place with these key stakeholders and others at SLCVAMC to identify the following specific goals of the TPS: (1) reduce pain through pharmacologic and nonpharmacologic interventions; (2) eliminate new chronic opioid use in previously nonopioid user (NOU) patients; (3) address chronic opioid use in previous chronic opioid users (COUs) by providing support for opioid taper and alternative analgesic therapies for their chronic pain conditions; and (4) improve continuity of care by close coordination with the surgical team, primary care providers (PCPs), and mental health or chronic pain providers as needed.
Once these TPS goals were defined, the Consolidated Framework for Implementation Research (CFIR) guided the implementation. CFIR is a theory-based implementation framework consisting of 5 domains: intervention characteristics, inner setting, outer setting, characteristics of individuals, and process. These domains were used to identify barriers and facilitators during the early implementation process and helped refine TPS as it was put into clinical practice.
Patient Selection
During the initial implementation of TPS, enrollment was limited to patients scheduled for elective primary or revision knee, hip, or shoulder replacement as well as rotator cuff repair surgery. But as the TPS workflow became established after iterative refinement, we expanded the program to enroll patients with established risk factors for OUD having other types of surgery (Table 1). The diagnosis of risk factors, such as history of SUD, chronic opioid use, or significant mental health disorders (ie, history of suicidal ideation or attempt, posttraumatic stress disorder, and inpatient psychiatric care) were confirmed through both in-person interviews and electronic health record (EHR) documentation. The overall goal was to identify all at-risk patients as soon as they were indicated for surgery, to allow time for evaluation, education, developing an individualized pain plan, and opioid taper prior to surgery if indicated.
Preoperative Procedures
Once identified, patients were contacted by a TPS team member and invited to attend a onetime 90-minute presurgical expectations class held at SLCVAMC. The education curriculum was developed by the whole team, and classes were taught primarily by the TPS psychologist. The class included education about expectations for postoperative pain, available analgesic therapies, opioid education, appropriate use of opioids, and the effect of psychological factors on pain. Pain coping strategies were introduced using a mindfulness-based intervention (MBI) and the Acceptance and Commitment Therapy (ACT) matrix. Classes were offered multiple times a week to help maximize convenience for patients and were separate from the anesthesia preoperative evaluation. Patients attended class only once. High-risk patients (patients with chronic opioid therapy, recent history of or current SUDs, significant comorbid mental health issues) were encouraged to attend this class one-on-one with the TPS psychologist rather than in the group setting, so individual attention to mental health and SUD issues could be addressed directly.
Baseline history, morphine equivalent daily dose (MEDD), and patient-reported outcomes using measures from the Patient-Reported Outcome Measurement System (PROMIS) for pain intensity (PROMIS 3a), pain interference (PROMIS 6b), and physical function (PROMIS 8b), and a pain-catastrophizing scale (PCS) score were obtained on all patients.18 PROMIS measures are validated questionnaires developed with the National Institutes of Health to standardize and quantify patient-reported outcomes in many domains.19 Patients with a history of SUD or COU met with the anesthesiologist and/or NP, and a personalized pain plan was developed that included preoperative opioid taper, buprenorphine use strategy, or opioid-free strategies.
Hospital Procedures
On the day of surgery, the TPS team met with the patient preoperatively and implemented an individualized pain plan that included multimodal analgesic techniques with nonsteroidal anti-inflammatory drugs, acetaminophen, gabapentinoids, and regional anesthesia, where appropriate (Table 2). Enhanced recovery after surgery protocols were developed in conjunction with the surgeons to include local infiltration analgesia by the surgeon, postoperative multimodal analgesic strategies, and intensive physical therapy starting the day of surgery for inpatient procedures.
After surgery, the TPS team followed up with patients daily and provided recommendations for analgesic therapies. Patients were offered daily sessions with the psychologist to reinforce and practice nonpharmacologic pain-coping strategies, such as meditation and relaxation. Prior to patient discharge, the TPS team provided recommendations for discharge medications and an opioid taper plan. For some patients taking buprenorphine before surgery who had stopped this therapy prior to or during their hospital stay, TPS providers transitioned them back to buprenorphine before discharge.
Postoperative Procedures
Patients were called by the nurse care coordinators at postdischarge days 2, 7, 10, 14, 21, 28, and then monthly for ≥ 6 months. For patients who had not stopped opioid use or returned to their preoperative baseline opioid dose, weekly calls were made until opioid taper goals were achieved. At each call, nurses collected PROMIS scores for the previous 24 hours, the most recent 24-hour MEDD, the date of last opioid use, and the number of remaining opioid tablets after opioid cessation. In addition, nurses provided active listening and supportive care and encouragement as well as care coordination for issues related to rehabilitation facilities, physical therapy, transportation, medication questions, and wound questions. Nurses notified the anesthesiologist or NP when patients were unable to taper opioid use or had poor pain control as indicated by their PROMIS scores, opioid use, or directly expressed by the patient.
The TPS team prescribed alternative analgesic therapies, opioid taper plans, and communicated with surgeons and primary care providers if limited continued opioid therapy was recommended. Individual sessions with the psychologist were available to patients after discharge with a focus on ACT-matrix therapy and consultation with long-term mental health and/or substance abuse providers as indicated. Frequent communication and care coordination were maintained with the surgical team, the PCP, and other providers on the mental health or chronic pain services. This care coordination often included postsurgical joint clinic appointments in which TPS providers and nurses would be present with the surgeon or the PCP.
For patients with inadequately treated chronic pain conditions or who required long-term opioid tapers, we developed a combined clinic with the TPS and Anesthesia Chronic Pain group. This clinic allows patients to be seen by both services in the same setting, allowing a warm handoff by TPS to the chronic pain team.
Heath and Decision Support Tools
An electronic dashboard registry of surgical episodes managed by TPS was developed to achieve clinical, administrative, and quality improvement goals. The dashboard registry consists of surgical episode data, opioid doses, patient-reported outcomes, and clinical decision-making processes. Custom-built note templates capture pertinent data through embedded data labels, called health factors. Data are captured as part of routine clinical care, recorded in Computerized Patient Record System as health factors. They are available in the VA Corporate Data Warehouse as structured data. Workflows are executed daily to keep the dashboard registry current, clean, and able to process new data. Information displays direct daily clinical workflow and support point-of-care clinical decision making (Figures 2, 3, and 4). Data are aggregated across patient-care encounters and allow nurse care coordinators to concisely review pertinent patient data prior to delivering care. These data include surgical history, comorbidities, timeline of opioid use, and PROMIS scores during their course of recovery. This system allows TPS to optimize care delivery by providing longitudinal data across the surgical episode, thereby reducing the time needed to review records. Secondary purposes of captured data include measuring clinic performance and quality improvement to improve care delivery.
Results
The TPS intervention was implemented January 1, 2018. Two-hundred thirteen patients were enrolled between January and December 2018, which included 60 (28%) patients with a history of chronic opioid use and 153 (72%) patients who were considered opioid naïve. A total of 99% of patients had ≥ 1 successful follow-up within 14 days after discharge, 96% had ≥ 1 follow-up between 14 and 30 days after surgery, and 72% had completed personal follow-up 90 days after discharge (Table 3). For patients who TPS was unable to contact in person or by phone, 90-day MEDD was obtained using prescription and Controlled Substance Database reviews. The protocol for this retrospective analysis was approved by the University of Utah Institutional Review Board and the VA Research Review Committee.
By 90 days after surgery, 26 (43.3%) COUs were off opioids completely, 17 (28.3%) had decreased their opioid dose from their preoperative baseline MEDD (120 [SD, 108] vs 55 [SD, 45]), 14 (23.3%) returned to their baseline dose, and 3 (5%) increased from their baseline dose. Of the 153 patients who were NOUs before surgery, only 1 (0.7%) was taking opioids after 90 days. TPS continued to work closely with the patient and their PCP and that patient was finally able to stop opioid use 262 days after discharge. Ten patients had an additional surgery within 90 days of the initial surgery. Of these, 6 were COU, of whom 3 stopped all opioids by 90 days from their original surgery, 2 had no change in MEDD at 90 days, and 1 had a lower MEDD at 90 days. Of the 4 NOU who had additional surgery, all were off opioids by 90 days from the original surgery.
Although difficult to quantify, a meaningful outcome of TPS has been to improve satisfaction substantially among health care providers caring for complex patients at risk for chronic opioid abuse. This group includes the many members of the surgical team, PCPs, and addiction specialists who appreciate the close care coordination and assistance in caring for patients with difficult issues, especially with opioid tapers or SUDs. We also have noticed changes in prescribing practices among surgeons and PCPs for their patients who are not part of TPS.
Discussion
With any new clinical service, there are obstacles and challenges. TPS requires a considerable investment in personnel, and currently no mechanism is in place for obtaining payment for many of the provided services. We were fortunate the VA Whole Health Initiative, the VA Office of Rural Health, and the VA Centers of Innovation provided support for the development, implementation, and pilot evaluation of TPS. After we presented our initial results to hospital leadership, we also received hospital support to expand TPS service to include a total of 4 nurse care coordinators and 2 psychologists. We are currently performing a cost analysis of the service but recognize that this model may be difficult to reproduce at other institutions without a change in reimbursement standards.
Developing a working relationship with the surgical and primary care services required a concerted effort from the TPS team and a number of months to become effective. As most veterans receive primary care, mental health care, and surgical care within the VA system, this model lends itself to close care coordination. Initially there was skepticism about TPS recommendations to reduce opioid use, especially from PCPs who had cared for complex patients over many years. But this uncertainty went away as we showed evidence of close patient follow-up and detailed communication. TPS soon became the designated service for both primary care and surgical providers who were otherwise uncomfortable with how to approach opioid tapers and nonopioid pain strategies. In fact, a substantial portion of our referrals now come directly from the PCP who is referring a high-risk patient for evaluation for surgery rather than from the surgeons, and joint visits with TPS and primary care have become commonplace.
Challenges abound when working with patients with substance abuse history, opioid use history, high anxiety, significant pain catastrophizing, and those who have had previous negative experiences with surgery. We have found that the most important facet of our service comes from the amount of time and effort team members, especially the nurses, spend helping patients. Much of the nurses' work focuses on nonpain-related issues, such as assisting patients with finding transportation, housing issues, questions about medications, help scheduling appointments, etc. Through this concerted effort, patients gain trust in TPS providers and are willing to listen to and experiment with our recommendations. Many patients who were initially extremely unreceptive to the presurgery education asked for our support weeks after surgery to help with postsurgery pain.
Another challenge we continue to experience comes from the success of the program.
Conclusions
The multidisciplinary TPS supports greater preoperative to postoperative longitudinal care for surgical patients. This endeavor has resulted in better patient preparation before surgery and improved care coordination after surgery, with specific improvements in appropriate use of opioid medications and smooth transitions of care for patients with ongoing and complex needs. Development of sophisticated note templates and customized health information technology allows for accurate follow-through and data gathering for quality improvement, facilitating data-driven improvements and proving value to the facility.
Given that TPS is a multidisciplinary program with multiple interventions, it is difficult to pinpoint which specific aspects of TPS are most effective in achieving success. For example, although we have little doubt that the work our psychologists do with our patients is beneficial and even essential for the success we have had with some of our most difficult patients, it is less clear whether it matters if they use mindfulness, ACT matrix, or cognitive behavioral therapy. We think that an important part of TPS is the frequent human interaction with a caring individual. Therefore, as TPS continues to grow, maintaining the ability to provide frequent personal interaction is a priority.
The role of opioids in acute pain deserves further scrutiny. In 2018, with TPS use of opioids after orthopedic surgery decreased by > 40% from the previous year. Despite this more restricted use of opioids, pain interference and physical function scores indicated that surgical patients do not seem to experience increased pain or reduced physical function. In addition, stopping opioid use for COUs did not seem to affect the quality of recovery, pain, or physical function. Future prospective controlled studies of TPS are needed to confirm these findings and identify which aspects of TPS are most effective in improving functional recovery of patients. Also, more evidence is needed to determine the appropriateness or need for opioids in acute postsurgical pain.
TPS has expanded to include all surgical specialties. Given the high burden and limited resources, we have chosen to focus on patients at higher risk for chronic postsurgical pain by type of surgery (eg, thoracotomy, open abdominal, limb amputation, major joint surgery) and/or history of substance abuse or chronic opioid use. To better direct scarce resources where it would be of most benefit, we are now enrolling only NOUs without other risk factors postoperatively if they request a refill of opioids or are otherwise struggling with pain control after surgery. Whether this approach affects the success we had in the first year in preventing new COUs after surgery remains to be seen.
It is unlikely that any single model of a perioperative surgical home will fit the needs of the many different types of medical systems that exist. The TPS model fits well in large hospital systems, like the VA, where patients receive most of their care within the same system. However, it seems to us that the optimal TPS program in any health system will provide education, support, and care coordination beginning preoperatively to prepare the patient for surgery and then to facilitate care coordination to transition patients back to their PCPs or on to specialized chronic care.
Acknowledgments
We would like to acknowledge the contributions of Candice Harmon, RN; David Merrill, RN; Amy Beckstead, RN, who have provided invaluable assistance with establishing the TPS program at the VA Salt Lake City and helping with the evaluation process.
Funding for the implementation and evaluation of the TPS was received from the VA Whole Health Initiative, the VA Center of Innovation, the VA Office of Rural Health, and National Institutes of Health Grant UL1TR002538.
1. Ilfeld BM, Madison SJ, Suresh PJ. Persistent postmastectomy pain and pain-related physical and emotional functioning with and without a continuous paravertebral nerve block: a prospective 1-year follow-up assessment of a randomized, triple-masked, placebo-controlled study. Ann Surg Oncol. 2015;22(6):2017-2025. doi:10.1245/s10434-014-4248-7
2. Richebé P, Capdevila X, Rivat C. Persistent postsurgical pain. Anesthesiology. 2018;129(3):590-607. doi:10.1097/aln.0000000000002238
3. Glare P, Aubrey KR, Myles PS. Transition from acute to chronic pain after surgery. Lancet. 2019;393(10180):1537-1546. doi:10.1016/s0140-6736(19)30352-6
4. Brummett CM, Waljee JF, Goesling J, et al. New persistent opioid use after minor and major surgical procedures in US adults. JAMA Surgery. 2017;152(6):e170504-e170504. doi:10.1001/jamasurg.2017.0504
5. Swenson CW, Kamdar NS, Seiler K, Morgan DM, Lin P, As-Sanie S. Definition development and prevalence of new persistent opioid use following hysterectomy. Am J Obstet Gynecol. 2018;219(5):486.e1-486.e7. doi:10.1016/j.ajog.2018.06.010
6. Bartels K, Fernandez-Bustamante A, McWilliams SK, Hopfer CJ, Mikulich-Gilbertson SK. Long-term opioid use after inpatient surgery - a retrospective cohort study. Drug Alcohol Depend. 2018;187:61-65. doi:10.1016/j.drugalcdep.2018.02.013
7. Bedard N, DeMik D, Dowdle S, Callaghan J. Trends and risk factors for prolonged opioid use after unicompartmental knee arthroplasty. Bone Joint J. 2018;100-B(1)(suppl A):62-67. doi:10.1302/0301-620x.100b1.bjj-2017-0547.r1
8. Politzer CS, Kildow BJ, Goltz DE, Green CL, Bolognesi MP, Seyler T. Trends in opioid utilization before and after total knee arthroplasty. J Arthroplasty. 2018;33(7S):S147-S153.e1. doi:10.1016/j.arth.2017.10.060
9. Mariano ER, Walters TL, Kim ET, Kain ZN. Why the perioperative surgical home makes sense for Veterans Affairs health care. Anesth Analg. 2015;120(5):1163-1166. doi:10.1213/ane.0000000000000712
10. Walters TL, Howard SK, Kou A, et al. Design and implementation of a perioperative surgical home at a Veterans Affairs hospital. Semin Cardiothorac Vasc Anesth. 2016;20(2):133-140. doi:10.1177/1089253215607066
11. Walters TL, Mariano ER, Clark DJ. Perioperative surgical home and the integral role of pain medicine. Pain Med. 2015;16(9):1666-1672. doi:10.1111/pme.12796
12. Vetter TR, Kain ZN. Role of the perioperative surgical home in optimizing the perioperative use of opioids. Anesth Analg. 2017;125(5):1653-1657. doi:10.1213/ane.0000000000002280
13. Shafer SL. Anesthesia & Analgesia’s 2015 collection on the perioperative surgical home. Anesth Analg. 2015;120(5):966-967. doi:10.1213/ane.0000000000000696
14. Wenzel JT, Schwenk ES, Baratta JL, Viscusi ER. Managing opioid-tolerant patients in the perioperative surgical home. Anesthesiol Clin. 2016;34(2):287-301. doi:10.1016/j.anclin.2016.01.005
15. Katz J, Weinrib A, Fashler SR, et al. The Toronto General Hospital Transitional Pain Service: development and implementation of a multidisciplinary program to prevent chronic postsurgical pain. J Pain Res. 2015;8:695-702. doi:10.2147/jpr.s91924
16. Tiippana E, Hamunen K, Heiskanen T, Nieminen T, Kalso E, Kontinen VK. New approach for treatment of prolonged postoperative pain: APS Out-Patient Clinic. Scand J Pain. 2016;12(1):19-24. doi:10.1016/j.sjpain.2016.02.008
17. Katz J, Weinrib AZ, Clarke H. Chronic postsurgical pain: from risk factor identification to multidisciplinary management at the Toronto General Hospital Transitional Pain Service. Can J Pain. 2019;3(2):49-58. doi:10.1080/24740527.2019.1574537
18. Sullivan MJ, Bishop SR, Pivik J. The Pain Catastrophizing Scale: development and validation. Psychol Assess. 1995;7(4):524-532. doi:10.1037/1040-3590.7.4.524
19. HealthMeasures. Intro to PROMIS. https://www.healthmeasures.net/explore-measurement-systems/promis. Accessed September 28, 2020.
Despite advancements in techniques, postsurgical pain continues to be a prominent part of the patient experience. Often this experience can lead to developing chronic postsurgical pain that interferes with quality of life after the expected time to recovery.1-3 As many as 14% of patients who undergo surgery without any history of opioid use develop chronic opioid use that persists after recovery from their operation.4-8 For patients with existing chronic opioid use or a history of substance use disorder (SUD), surgeons, primary care providers, or addiction providers often do not provide sufficient presurgical planning or postsurgical coordination of care. This lack of pain care coordination can increase the risk of inadequate pain control, opioid use escalation, or SUD relapse after surgery.
Convincing arguments have been made that a perioperative surgical home can improve significantly the quality of perioperative care.9-14 This report describes our experience implementing a perioperative surgical home at the US Department of Veterans Affairs (VA) Salt Lake City VA Medical Center (SLCVAMC), focusing on pain management extending from the preoperative period until 6 months or more after surgery. This type of Transitional Pain Service (TPS) has been described previously.15-17 Our service differs from those described previously by enrolling all patients before surgery rather than select postsurgical enrollment of only patients with a history of opioid use or SUD or patients who struggle with persistent postsurgical pain.
Methods
In January 2018, we developed and implemented a new TPS at the SLCVAMC. The transitional pain team consisted of an anesthesiologist with specialization in acute pain management, a nurse practitioner (NP) with experience in both acute and chronic pain management, 2 nurse care coordinators, and a psychologist (Figure 1). Before implementation, a needs assessment took place with these key stakeholders and others at SLCVAMC to identify the following specific goals of the TPS: (1) reduce pain through pharmacologic and nonpharmacologic interventions; (2) eliminate new chronic opioid use in previously nonopioid user (NOU) patients; (3) address chronic opioid use in previous chronic opioid users (COUs) by providing support for opioid taper and alternative analgesic therapies for their chronic pain conditions; and (4) improve continuity of care by close coordination with the surgical team, primary care providers (PCPs), and mental health or chronic pain providers as needed.
Once these TPS goals were defined, the Consolidated Framework for Implementation Research (CFIR) guided the implementation. CFIR is a theory-based implementation framework consisting of 5 domains: intervention characteristics, inner setting, outer setting, characteristics of individuals, and process. These domains were used to identify barriers and facilitators during the early implementation process and helped refine TPS as it was put into clinical practice.
Patient Selection
During the initial implementation of TPS, enrollment was limited to patients scheduled for elective primary or revision knee, hip, or shoulder replacement as well as rotator cuff repair surgery. But as the TPS workflow became established after iterative refinement, we expanded the program to enroll patients with established risk factors for OUD having other types of surgery (Table 1). The diagnosis of risk factors, such as history of SUD, chronic opioid use, or significant mental health disorders (ie, history of suicidal ideation or attempt, posttraumatic stress disorder, and inpatient psychiatric care) were confirmed through both in-person interviews and electronic health record (EHR) documentation. The overall goal was to identify all at-risk patients as soon as they were indicated for surgery, to allow time for evaluation, education, developing an individualized pain plan, and opioid taper prior to surgery if indicated.
Preoperative Procedures
Once identified, patients were contacted by a TPS team member and invited to attend a onetime 90-minute presurgical expectations class held at SLCVAMC. The education curriculum was developed by the whole team, and classes were taught primarily by the TPS psychologist. The class included education about expectations for postoperative pain, available analgesic therapies, opioid education, appropriate use of opioids, and the effect of psychological factors on pain. Pain coping strategies were introduced using a mindfulness-based intervention (MBI) and the Acceptance and Commitment Therapy (ACT) matrix. Classes were offered multiple times a week to help maximize convenience for patients and were separate from the anesthesia preoperative evaluation. Patients attended class only once. High-risk patients (patients with chronic opioid therapy, recent history of or current SUDs, significant comorbid mental health issues) were encouraged to attend this class one-on-one with the TPS psychologist rather than in the group setting, so individual attention to mental health and SUD issues could be addressed directly.
Baseline history, morphine equivalent daily dose (MEDD), and patient-reported outcomes using measures from the Patient-Reported Outcome Measurement System (PROMIS) for pain intensity (PROMIS 3a), pain interference (PROMIS 6b), and physical function (PROMIS 8b), and a pain-catastrophizing scale (PCS) score were obtained on all patients.18 PROMIS measures are validated questionnaires developed with the National Institutes of Health to standardize and quantify patient-reported outcomes in many domains.19 Patients with a history of SUD or COU met with the anesthesiologist and/or NP, and a personalized pain plan was developed that included preoperative opioid taper, buprenorphine use strategy, or opioid-free strategies.
Hospital Procedures
On the day of surgery, the TPS team met with the patient preoperatively and implemented an individualized pain plan that included multimodal analgesic techniques with nonsteroidal anti-inflammatory drugs, acetaminophen, gabapentinoids, and regional anesthesia, where appropriate (Table 2). Enhanced recovery after surgery protocols were developed in conjunction with the surgeons to include local infiltration analgesia by the surgeon, postoperative multimodal analgesic strategies, and intensive physical therapy starting the day of surgery for inpatient procedures.
After surgery, the TPS team followed up with patients daily and provided recommendations for analgesic therapies. Patients were offered daily sessions with the psychologist to reinforce and practice nonpharmacologic pain-coping strategies, such as meditation and relaxation. Prior to patient discharge, the TPS team provided recommendations for discharge medications and an opioid taper plan. For some patients taking buprenorphine before surgery who had stopped this therapy prior to or during their hospital stay, TPS providers transitioned them back to buprenorphine before discharge.
Postoperative Procedures
Patients were called by the nurse care coordinators at postdischarge days 2, 7, 10, 14, 21, 28, and then monthly for ≥ 6 months. For patients who had not stopped opioid use or returned to their preoperative baseline opioid dose, weekly calls were made until opioid taper goals were achieved. At each call, nurses collected PROMIS scores for the previous 24 hours, the most recent 24-hour MEDD, the date of last opioid use, and the number of remaining opioid tablets after opioid cessation. In addition, nurses provided active listening and supportive care and encouragement as well as care coordination for issues related to rehabilitation facilities, physical therapy, transportation, medication questions, and wound questions. Nurses notified the anesthesiologist or NP when patients were unable to taper opioid use or had poor pain control as indicated by their PROMIS scores, opioid use, or directly expressed by the patient.
The TPS team prescribed alternative analgesic therapies, opioid taper plans, and communicated with surgeons and primary care providers if limited continued opioid therapy was recommended. Individual sessions with the psychologist were available to patients after discharge with a focus on ACT-matrix therapy and consultation with long-term mental health and/or substance abuse providers as indicated. Frequent communication and care coordination were maintained with the surgical team, the PCP, and other providers on the mental health or chronic pain services. This care coordination often included postsurgical joint clinic appointments in which TPS providers and nurses would be present with the surgeon or the PCP.
For patients with inadequately treated chronic pain conditions or who required long-term opioid tapers, we developed a combined clinic with the TPS and Anesthesia Chronic Pain group. This clinic allows patients to be seen by both services in the same setting, allowing a warm handoff by TPS to the chronic pain team.
Heath and Decision Support Tools
An electronic dashboard registry of surgical episodes managed by TPS was developed to achieve clinical, administrative, and quality improvement goals. The dashboard registry consists of surgical episode data, opioid doses, patient-reported outcomes, and clinical decision-making processes. Custom-built note templates capture pertinent data through embedded data labels, called health factors. Data are captured as part of routine clinical care, recorded in Computerized Patient Record System as health factors. They are available in the VA Corporate Data Warehouse as structured data. Workflows are executed daily to keep the dashboard registry current, clean, and able to process new data. Information displays direct daily clinical workflow and support point-of-care clinical decision making (Figures 2, 3, and 4). Data are aggregated across patient-care encounters and allow nurse care coordinators to concisely review pertinent patient data prior to delivering care. These data include surgical history, comorbidities, timeline of opioid use, and PROMIS scores during their course of recovery. This system allows TPS to optimize care delivery by providing longitudinal data across the surgical episode, thereby reducing the time needed to review records. Secondary purposes of captured data include measuring clinic performance and quality improvement to improve care delivery.
Results
The TPS intervention was implemented January 1, 2018. Two-hundred thirteen patients were enrolled between January and December 2018, which included 60 (28%) patients with a history of chronic opioid use and 153 (72%) patients who were considered opioid naïve. A total of 99% of patients had ≥ 1 successful follow-up within 14 days after discharge, 96% had ≥ 1 follow-up between 14 and 30 days after surgery, and 72% had completed personal follow-up 90 days after discharge (Table 3). For patients who TPS was unable to contact in person or by phone, 90-day MEDD was obtained using prescription and Controlled Substance Database reviews. The protocol for this retrospective analysis was approved by the University of Utah Institutional Review Board and the VA Research Review Committee.
By 90 days after surgery, 26 (43.3%) COUs were off opioids completely, 17 (28.3%) had decreased their opioid dose from their preoperative baseline MEDD (120 [SD, 108] vs 55 [SD, 45]), 14 (23.3%) returned to their baseline dose, and 3 (5%) increased from their baseline dose. Of the 153 patients who were NOUs before surgery, only 1 (0.7%) was taking opioids after 90 days. TPS continued to work closely with the patient and their PCP and that patient was finally able to stop opioid use 262 days after discharge. Ten patients had an additional surgery within 90 days of the initial surgery. Of these, 6 were COU, of whom 3 stopped all opioids by 90 days from their original surgery, 2 had no change in MEDD at 90 days, and 1 had a lower MEDD at 90 days. Of the 4 NOU who had additional surgery, all were off opioids by 90 days from the original surgery.
Although difficult to quantify, a meaningful outcome of TPS has been to improve satisfaction substantially among health care providers caring for complex patients at risk for chronic opioid abuse. This group includes the many members of the surgical team, PCPs, and addiction specialists who appreciate the close care coordination and assistance in caring for patients with difficult issues, especially with opioid tapers or SUDs. We also have noticed changes in prescribing practices among surgeons and PCPs for their patients who are not part of TPS.
Discussion
With any new clinical service, there are obstacles and challenges. TPS requires a considerable investment in personnel, and currently no mechanism is in place for obtaining payment for many of the provided services. We were fortunate the VA Whole Health Initiative, the VA Office of Rural Health, and the VA Centers of Innovation provided support for the development, implementation, and pilot evaluation of TPS. After we presented our initial results to hospital leadership, we also received hospital support to expand TPS service to include a total of 4 nurse care coordinators and 2 psychologists. We are currently performing a cost analysis of the service but recognize that this model may be difficult to reproduce at other institutions without a change in reimbursement standards.
Developing a working relationship with the surgical and primary care services required a concerted effort from the TPS team and a number of months to become effective. As most veterans receive primary care, mental health care, and surgical care within the VA system, this model lends itself to close care coordination. Initially there was skepticism about TPS recommendations to reduce opioid use, especially from PCPs who had cared for complex patients over many years. But this uncertainty went away as we showed evidence of close patient follow-up and detailed communication. TPS soon became the designated service for both primary care and surgical providers who were otherwise uncomfortable with how to approach opioid tapers and nonopioid pain strategies. In fact, a substantial portion of our referrals now come directly from the PCP who is referring a high-risk patient for evaluation for surgery rather than from the surgeons, and joint visits with TPS and primary care have become commonplace.
Challenges abound when working with patients with substance abuse history, opioid use history, high anxiety, significant pain catastrophizing, and those who have had previous negative experiences with surgery. We have found that the most important facet of our service comes from the amount of time and effort team members, especially the nurses, spend helping patients. Much of the nurses' work focuses on nonpain-related issues, such as assisting patients with finding transportation, housing issues, questions about medications, help scheduling appointments, etc. Through this concerted effort, patients gain trust in TPS providers and are willing to listen to and experiment with our recommendations. Many patients who were initially extremely unreceptive to the presurgery education asked for our support weeks after surgery to help with postsurgery pain.
Another challenge we continue to experience comes from the success of the program.
Conclusions
The multidisciplinary TPS supports greater preoperative to postoperative longitudinal care for surgical patients. This endeavor has resulted in better patient preparation before surgery and improved care coordination after surgery, with specific improvements in appropriate use of opioid medications and smooth transitions of care for patients with ongoing and complex needs. Development of sophisticated note templates and customized health information technology allows for accurate follow-through and data gathering for quality improvement, facilitating data-driven improvements and proving value to the facility.
Given that TPS is a multidisciplinary program with multiple interventions, it is difficult to pinpoint which specific aspects of TPS are most effective in achieving success. For example, although we have little doubt that the work our psychologists do with our patients is beneficial and even essential for the success we have had with some of our most difficult patients, it is less clear whether it matters if they use mindfulness, ACT matrix, or cognitive behavioral therapy. We think that an important part of TPS is the frequent human interaction with a caring individual. Therefore, as TPS continues to grow, maintaining the ability to provide frequent personal interaction is a priority.
The role of opioids in acute pain deserves further scrutiny. In 2018, with TPS use of opioids after orthopedic surgery decreased by > 40% from the previous year. Despite this more restricted use of opioids, pain interference and physical function scores indicated that surgical patients do not seem to experience increased pain or reduced physical function. In addition, stopping opioid use for COUs did not seem to affect the quality of recovery, pain, or physical function. Future prospective controlled studies of TPS are needed to confirm these findings and identify which aspects of TPS are most effective in improving functional recovery of patients. Also, more evidence is needed to determine the appropriateness or need for opioids in acute postsurgical pain.
TPS has expanded to include all surgical specialties. Given the high burden and limited resources, we have chosen to focus on patients at higher risk for chronic postsurgical pain by type of surgery (eg, thoracotomy, open abdominal, limb amputation, major joint surgery) and/or history of substance abuse or chronic opioid use. To better direct scarce resources where it would be of most benefit, we are now enrolling only NOUs without other risk factors postoperatively if they request a refill of opioids or are otherwise struggling with pain control after surgery. Whether this approach affects the success we had in the first year in preventing new COUs after surgery remains to be seen.
It is unlikely that any single model of a perioperative surgical home will fit the needs of the many different types of medical systems that exist. The TPS model fits well in large hospital systems, like the VA, where patients receive most of their care within the same system. However, it seems to us that the optimal TPS program in any health system will provide education, support, and care coordination beginning preoperatively to prepare the patient for surgery and then to facilitate care coordination to transition patients back to their PCPs or on to specialized chronic care.
Acknowledgments
We would like to acknowledge the contributions of Candice Harmon, RN; David Merrill, RN; Amy Beckstead, RN, who have provided invaluable assistance with establishing the TPS program at the VA Salt Lake City and helping with the evaluation process.
Funding for the implementation and evaluation of the TPS was received from the VA Whole Health Initiative, the VA Center of Innovation, the VA Office of Rural Health, and National Institutes of Health Grant UL1TR002538.
Despite advancements in techniques, postsurgical pain continues to be a prominent part of the patient experience. Often this experience can lead to developing chronic postsurgical pain that interferes with quality of life after the expected time to recovery.1-3 As many as 14% of patients who undergo surgery without any history of opioid use develop chronic opioid use that persists after recovery from their operation.4-8 For patients with existing chronic opioid use or a history of substance use disorder (SUD), surgeons, primary care providers, or addiction providers often do not provide sufficient presurgical planning or postsurgical coordination of care. This lack of pain care coordination can increase the risk of inadequate pain control, opioid use escalation, or SUD relapse after surgery.
Convincing arguments have been made that a perioperative surgical home can improve significantly the quality of perioperative care.9-14 This report describes our experience implementing a perioperative surgical home at the US Department of Veterans Affairs (VA) Salt Lake City VA Medical Center (SLCVAMC), focusing on pain management extending from the preoperative period until 6 months or more after surgery. This type of Transitional Pain Service (TPS) has been described previously.15-17 Our service differs from those described previously by enrolling all patients before surgery rather than select postsurgical enrollment of only patients with a history of opioid use or SUD or patients who struggle with persistent postsurgical pain.
Methods
In January 2018, we developed and implemented a new TPS at the SLCVAMC. The transitional pain team consisted of an anesthesiologist with specialization in acute pain management, a nurse practitioner (NP) with experience in both acute and chronic pain management, 2 nurse care coordinators, and a psychologist (Figure 1). Before implementation, a needs assessment took place with these key stakeholders and others at SLCVAMC to identify the following specific goals of the TPS: (1) reduce pain through pharmacologic and nonpharmacologic interventions; (2) eliminate new chronic opioid use in previously nonopioid user (NOU) patients; (3) address chronic opioid use in previous chronic opioid users (COUs) by providing support for opioid taper and alternative analgesic therapies for their chronic pain conditions; and (4) improve continuity of care by close coordination with the surgical team, primary care providers (PCPs), and mental health or chronic pain providers as needed.
Once these TPS goals were defined, the Consolidated Framework for Implementation Research (CFIR) guided the implementation. CFIR is a theory-based implementation framework consisting of 5 domains: intervention characteristics, inner setting, outer setting, characteristics of individuals, and process. These domains were used to identify barriers and facilitators during the early implementation process and helped refine TPS as it was put into clinical practice.
Patient Selection
During the initial implementation of TPS, enrollment was limited to patients scheduled for elective primary or revision knee, hip, or shoulder replacement as well as rotator cuff repair surgery. But as the TPS workflow became established after iterative refinement, we expanded the program to enroll patients with established risk factors for OUD having other types of surgery (Table 1). The diagnosis of risk factors, such as history of SUD, chronic opioid use, or significant mental health disorders (ie, history of suicidal ideation or attempt, posttraumatic stress disorder, and inpatient psychiatric care) were confirmed through both in-person interviews and electronic health record (EHR) documentation. The overall goal was to identify all at-risk patients as soon as they were indicated for surgery, to allow time for evaluation, education, developing an individualized pain plan, and opioid taper prior to surgery if indicated.
Preoperative Procedures
Once identified, patients were contacted by a TPS team member and invited to attend a onetime 90-minute presurgical expectations class held at SLCVAMC. The education curriculum was developed by the whole team, and classes were taught primarily by the TPS psychologist. The class included education about expectations for postoperative pain, available analgesic therapies, opioid education, appropriate use of opioids, and the effect of psychological factors on pain. Pain coping strategies were introduced using a mindfulness-based intervention (MBI) and the Acceptance and Commitment Therapy (ACT) matrix. Classes were offered multiple times a week to help maximize convenience for patients and were separate from the anesthesia preoperative evaluation. Patients attended class only once. High-risk patients (patients with chronic opioid therapy, recent history of or current SUDs, significant comorbid mental health issues) were encouraged to attend this class one-on-one with the TPS psychologist rather than in the group setting, so individual attention to mental health and SUD issues could be addressed directly.
Baseline history, morphine equivalent daily dose (MEDD), and patient-reported outcomes using measures from the Patient-Reported Outcome Measurement System (PROMIS) for pain intensity (PROMIS 3a), pain interference (PROMIS 6b), and physical function (PROMIS 8b), and a pain-catastrophizing scale (PCS) score were obtained on all patients.18 PROMIS measures are validated questionnaires developed with the National Institutes of Health to standardize and quantify patient-reported outcomes in many domains.19 Patients with a history of SUD or COU met with the anesthesiologist and/or NP, and a personalized pain plan was developed that included preoperative opioid taper, buprenorphine use strategy, or opioid-free strategies.
Hospital Procedures
On the day of surgery, the TPS team met with the patient preoperatively and implemented an individualized pain plan that included multimodal analgesic techniques with nonsteroidal anti-inflammatory drugs, acetaminophen, gabapentinoids, and regional anesthesia, where appropriate (Table 2). Enhanced recovery after surgery protocols were developed in conjunction with the surgeons to include local infiltration analgesia by the surgeon, postoperative multimodal analgesic strategies, and intensive physical therapy starting the day of surgery for inpatient procedures.
After surgery, the TPS team followed up with patients daily and provided recommendations for analgesic therapies. Patients were offered daily sessions with the psychologist to reinforce and practice nonpharmacologic pain-coping strategies, such as meditation and relaxation. Prior to patient discharge, the TPS team provided recommendations for discharge medications and an opioid taper plan. For some patients taking buprenorphine before surgery who had stopped this therapy prior to or during their hospital stay, TPS providers transitioned them back to buprenorphine before discharge.
Postoperative Procedures
Patients were called by the nurse care coordinators at postdischarge days 2, 7, 10, 14, 21, 28, and then monthly for ≥ 6 months. For patients who had not stopped opioid use or returned to their preoperative baseline opioid dose, weekly calls were made until opioid taper goals were achieved. At each call, nurses collected PROMIS scores for the previous 24 hours, the most recent 24-hour MEDD, the date of last opioid use, and the number of remaining opioid tablets after opioid cessation. In addition, nurses provided active listening and supportive care and encouragement as well as care coordination for issues related to rehabilitation facilities, physical therapy, transportation, medication questions, and wound questions. Nurses notified the anesthesiologist or NP when patients were unable to taper opioid use or had poor pain control as indicated by their PROMIS scores, opioid use, or directly expressed by the patient.
The TPS team prescribed alternative analgesic therapies, opioid taper plans, and communicated with surgeons and primary care providers if limited continued opioid therapy was recommended. Individual sessions with the psychologist were available to patients after discharge with a focus on ACT-matrix therapy and consultation with long-term mental health and/or substance abuse providers as indicated. Frequent communication and care coordination were maintained with the surgical team, the PCP, and other providers on the mental health or chronic pain services. This care coordination often included postsurgical joint clinic appointments in which TPS providers and nurses would be present with the surgeon or the PCP.
For patients with inadequately treated chronic pain conditions or who required long-term opioid tapers, we developed a combined clinic with the TPS and Anesthesia Chronic Pain group. This clinic allows patients to be seen by both services in the same setting, allowing a warm handoff by TPS to the chronic pain team.
Heath and Decision Support Tools
An electronic dashboard registry of surgical episodes managed by TPS was developed to achieve clinical, administrative, and quality improvement goals. The dashboard registry consists of surgical episode data, opioid doses, patient-reported outcomes, and clinical decision-making processes. Custom-built note templates capture pertinent data through embedded data labels, called health factors. Data are captured as part of routine clinical care, recorded in Computerized Patient Record System as health factors. They are available in the VA Corporate Data Warehouse as structured data. Workflows are executed daily to keep the dashboard registry current, clean, and able to process new data. Information displays direct daily clinical workflow and support point-of-care clinical decision making (Figures 2, 3, and 4). Data are aggregated across patient-care encounters and allow nurse care coordinators to concisely review pertinent patient data prior to delivering care. These data include surgical history, comorbidities, timeline of opioid use, and PROMIS scores during their course of recovery. This system allows TPS to optimize care delivery by providing longitudinal data across the surgical episode, thereby reducing the time needed to review records. Secondary purposes of captured data include measuring clinic performance and quality improvement to improve care delivery.
Results
The TPS intervention was implemented January 1, 2018. Two-hundred thirteen patients were enrolled between January and December 2018, which included 60 (28%) patients with a history of chronic opioid use and 153 (72%) patients who were considered opioid naïve. A total of 99% of patients had ≥ 1 successful follow-up within 14 days after discharge, 96% had ≥ 1 follow-up between 14 and 30 days after surgery, and 72% had completed personal follow-up 90 days after discharge (Table 3). For patients who TPS was unable to contact in person or by phone, 90-day MEDD was obtained using prescription and Controlled Substance Database reviews. The protocol for this retrospective analysis was approved by the University of Utah Institutional Review Board and the VA Research Review Committee.
By 90 days after surgery, 26 (43.3%) COUs were off opioids completely, 17 (28.3%) had decreased their opioid dose from their preoperative baseline MEDD (120 [SD, 108] vs 55 [SD, 45]), 14 (23.3%) returned to their baseline dose, and 3 (5%) increased from their baseline dose. Of the 153 patients who were NOUs before surgery, only 1 (0.7%) was taking opioids after 90 days. TPS continued to work closely with the patient and their PCP and that patient was finally able to stop opioid use 262 days after discharge. Ten patients had an additional surgery within 90 days of the initial surgery. Of these, 6 were COU, of whom 3 stopped all opioids by 90 days from their original surgery, 2 had no change in MEDD at 90 days, and 1 had a lower MEDD at 90 days. Of the 4 NOU who had additional surgery, all were off opioids by 90 days from the original surgery.
Although difficult to quantify, a meaningful outcome of TPS has been to improve satisfaction substantially among health care providers caring for complex patients at risk for chronic opioid abuse. This group includes the many members of the surgical team, PCPs, and addiction specialists who appreciate the close care coordination and assistance in caring for patients with difficult issues, especially with opioid tapers or SUDs. We also have noticed changes in prescribing practices among surgeons and PCPs for their patients who are not part of TPS.
Discussion
With any new clinical service, there are obstacles and challenges. TPS requires a considerable investment in personnel, and currently no mechanism is in place for obtaining payment for many of the provided services. We were fortunate the VA Whole Health Initiative, the VA Office of Rural Health, and the VA Centers of Innovation provided support for the development, implementation, and pilot evaluation of TPS. After we presented our initial results to hospital leadership, we also received hospital support to expand TPS service to include a total of 4 nurse care coordinators and 2 psychologists. We are currently performing a cost analysis of the service but recognize that this model may be difficult to reproduce at other institutions without a change in reimbursement standards.
Developing a working relationship with the surgical and primary care services required a concerted effort from the TPS team and a number of months to become effective. As most veterans receive primary care, mental health care, and surgical care within the VA system, this model lends itself to close care coordination. Initially there was skepticism about TPS recommendations to reduce opioid use, especially from PCPs who had cared for complex patients over many years. But this uncertainty went away as we showed evidence of close patient follow-up and detailed communication. TPS soon became the designated service for both primary care and surgical providers who were otherwise uncomfortable with how to approach opioid tapers and nonopioid pain strategies. In fact, a substantial portion of our referrals now come directly from the PCP who is referring a high-risk patient for evaluation for surgery rather than from the surgeons, and joint visits with TPS and primary care have become commonplace.
Challenges abound when working with patients with substance abuse history, opioid use history, high anxiety, significant pain catastrophizing, and those who have had previous negative experiences with surgery. We have found that the most important facet of our service comes from the amount of time and effort team members, especially the nurses, spend helping patients. Much of the nurses' work focuses on nonpain-related issues, such as assisting patients with finding transportation, housing issues, questions about medications, help scheduling appointments, etc. Through this concerted effort, patients gain trust in TPS providers and are willing to listen to and experiment with our recommendations. Many patients who were initially extremely unreceptive to the presurgery education asked for our support weeks after surgery to help with postsurgery pain.
Another challenge we continue to experience comes from the success of the program.
Conclusions
The multidisciplinary TPS supports greater preoperative to postoperative longitudinal care for surgical patients. This endeavor has resulted in better patient preparation before surgery and improved care coordination after surgery, with specific improvements in appropriate use of opioid medications and smooth transitions of care for patients with ongoing and complex needs. Development of sophisticated note templates and customized health information technology allows for accurate follow-through and data gathering for quality improvement, facilitating data-driven improvements and proving value to the facility.
Given that TPS is a multidisciplinary program with multiple interventions, it is difficult to pinpoint which specific aspects of TPS are most effective in achieving success. For example, although we have little doubt that the work our psychologists do with our patients is beneficial and even essential for the success we have had with some of our most difficult patients, it is less clear whether it matters if they use mindfulness, ACT matrix, or cognitive behavioral therapy. We think that an important part of TPS is the frequent human interaction with a caring individual. Therefore, as TPS continues to grow, maintaining the ability to provide frequent personal interaction is a priority.
The role of opioids in acute pain deserves further scrutiny. In 2018, with TPS use of opioids after orthopedic surgery decreased by > 40% from the previous year. Despite this more restricted use of opioids, pain interference and physical function scores indicated that surgical patients do not seem to experience increased pain or reduced physical function. In addition, stopping opioid use for COUs did not seem to affect the quality of recovery, pain, or physical function. Future prospective controlled studies of TPS are needed to confirm these findings and identify which aspects of TPS are most effective in improving functional recovery of patients. Also, more evidence is needed to determine the appropriateness or need for opioids in acute postsurgical pain.
TPS has expanded to include all surgical specialties. Given the high burden and limited resources, we have chosen to focus on patients at higher risk for chronic postsurgical pain by type of surgery (eg, thoracotomy, open abdominal, limb amputation, major joint surgery) and/or history of substance abuse or chronic opioid use. To better direct scarce resources where it would be of most benefit, we are now enrolling only NOUs without other risk factors postoperatively if they request a refill of opioids or are otherwise struggling with pain control after surgery. Whether this approach affects the success we had in the first year in preventing new COUs after surgery remains to be seen.
It is unlikely that any single model of a perioperative surgical home will fit the needs of the many different types of medical systems that exist. The TPS model fits well in large hospital systems, like the VA, where patients receive most of their care within the same system. However, it seems to us that the optimal TPS program in any health system will provide education, support, and care coordination beginning preoperatively to prepare the patient for surgery and then to facilitate care coordination to transition patients back to their PCPs or on to specialized chronic care.
Acknowledgments
We would like to acknowledge the contributions of Candice Harmon, RN; David Merrill, RN; Amy Beckstead, RN, who have provided invaluable assistance with establishing the TPS program at the VA Salt Lake City and helping with the evaluation process.
Funding for the implementation and evaluation of the TPS was received from the VA Whole Health Initiative, the VA Center of Innovation, the VA Office of Rural Health, and National Institutes of Health Grant UL1TR002538.
1. Ilfeld BM, Madison SJ, Suresh PJ. Persistent postmastectomy pain and pain-related physical and emotional functioning with and without a continuous paravertebral nerve block: a prospective 1-year follow-up assessment of a randomized, triple-masked, placebo-controlled study. Ann Surg Oncol. 2015;22(6):2017-2025. doi:10.1245/s10434-014-4248-7
2. Richebé P, Capdevila X, Rivat C. Persistent postsurgical pain. Anesthesiology. 2018;129(3):590-607. doi:10.1097/aln.0000000000002238
3. Glare P, Aubrey KR, Myles PS. Transition from acute to chronic pain after surgery. Lancet. 2019;393(10180):1537-1546. doi:10.1016/s0140-6736(19)30352-6
4. Brummett CM, Waljee JF, Goesling J, et al. New persistent opioid use after minor and major surgical procedures in US adults. JAMA Surgery. 2017;152(6):e170504-e170504. doi:10.1001/jamasurg.2017.0504
5. Swenson CW, Kamdar NS, Seiler K, Morgan DM, Lin P, As-Sanie S. Definition development and prevalence of new persistent opioid use following hysterectomy. Am J Obstet Gynecol. 2018;219(5):486.e1-486.e7. doi:10.1016/j.ajog.2018.06.010
6. Bartels K, Fernandez-Bustamante A, McWilliams SK, Hopfer CJ, Mikulich-Gilbertson SK. Long-term opioid use after inpatient surgery - a retrospective cohort study. Drug Alcohol Depend. 2018;187:61-65. doi:10.1016/j.drugalcdep.2018.02.013
7. Bedard N, DeMik D, Dowdle S, Callaghan J. Trends and risk factors for prolonged opioid use after unicompartmental knee arthroplasty. Bone Joint J. 2018;100-B(1)(suppl A):62-67. doi:10.1302/0301-620x.100b1.bjj-2017-0547.r1
8. Politzer CS, Kildow BJ, Goltz DE, Green CL, Bolognesi MP, Seyler T. Trends in opioid utilization before and after total knee arthroplasty. J Arthroplasty. 2018;33(7S):S147-S153.e1. doi:10.1016/j.arth.2017.10.060
9. Mariano ER, Walters TL, Kim ET, Kain ZN. Why the perioperative surgical home makes sense for Veterans Affairs health care. Anesth Analg. 2015;120(5):1163-1166. doi:10.1213/ane.0000000000000712
10. Walters TL, Howard SK, Kou A, et al. Design and implementation of a perioperative surgical home at a Veterans Affairs hospital. Semin Cardiothorac Vasc Anesth. 2016;20(2):133-140. doi:10.1177/1089253215607066
11. Walters TL, Mariano ER, Clark DJ. Perioperative surgical home and the integral role of pain medicine. Pain Med. 2015;16(9):1666-1672. doi:10.1111/pme.12796
12. Vetter TR, Kain ZN. Role of the perioperative surgical home in optimizing the perioperative use of opioids. Anesth Analg. 2017;125(5):1653-1657. doi:10.1213/ane.0000000000002280
13. Shafer SL. Anesthesia & Analgesia’s 2015 collection on the perioperative surgical home. Anesth Analg. 2015;120(5):966-967. doi:10.1213/ane.0000000000000696
14. Wenzel JT, Schwenk ES, Baratta JL, Viscusi ER. Managing opioid-tolerant patients in the perioperative surgical home. Anesthesiol Clin. 2016;34(2):287-301. doi:10.1016/j.anclin.2016.01.005
15. Katz J, Weinrib A, Fashler SR, et al. The Toronto General Hospital Transitional Pain Service: development and implementation of a multidisciplinary program to prevent chronic postsurgical pain. J Pain Res. 2015;8:695-702. doi:10.2147/jpr.s91924
16. Tiippana E, Hamunen K, Heiskanen T, Nieminen T, Kalso E, Kontinen VK. New approach for treatment of prolonged postoperative pain: APS Out-Patient Clinic. Scand J Pain. 2016;12(1):19-24. doi:10.1016/j.sjpain.2016.02.008
17. Katz J, Weinrib AZ, Clarke H. Chronic postsurgical pain: from risk factor identification to multidisciplinary management at the Toronto General Hospital Transitional Pain Service. Can J Pain. 2019;3(2):49-58. doi:10.1080/24740527.2019.1574537
18. Sullivan MJ, Bishop SR, Pivik J. The Pain Catastrophizing Scale: development and validation. Psychol Assess. 1995;7(4):524-532. doi:10.1037/1040-3590.7.4.524
19. HealthMeasures. Intro to PROMIS. https://www.healthmeasures.net/explore-measurement-systems/promis. Accessed September 28, 2020.
1. Ilfeld BM, Madison SJ, Suresh PJ. Persistent postmastectomy pain and pain-related physical and emotional functioning with and without a continuous paravertebral nerve block: a prospective 1-year follow-up assessment of a randomized, triple-masked, placebo-controlled study. Ann Surg Oncol. 2015;22(6):2017-2025. doi:10.1245/s10434-014-4248-7
2. Richebé P, Capdevila X, Rivat C. Persistent postsurgical pain. Anesthesiology. 2018;129(3):590-607. doi:10.1097/aln.0000000000002238
3. Glare P, Aubrey KR, Myles PS. Transition from acute to chronic pain after surgery. Lancet. 2019;393(10180):1537-1546. doi:10.1016/s0140-6736(19)30352-6
4. Brummett CM, Waljee JF, Goesling J, et al. New persistent opioid use after minor and major surgical procedures in US adults. JAMA Surgery. 2017;152(6):e170504-e170504. doi:10.1001/jamasurg.2017.0504
5. Swenson CW, Kamdar NS, Seiler K, Morgan DM, Lin P, As-Sanie S. Definition development and prevalence of new persistent opioid use following hysterectomy. Am J Obstet Gynecol. 2018;219(5):486.e1-486.e7. doi:10.1016/j.ajog.2018.06.010
6. Bartels K, Fernandez-Bustamante A, McWilliams SK, Hopfer CJ, Mikulich-Gilbertson SK. Long-term opioid use after inpatient surgery - a retrospective cohort study. Drug Alcohol Depend. 2018;187:61-65. doi:10.1016/j.drugalcdep.2018.02.013
7. Bedard N, DeMik D, Dowdle S, Callaghan J. Trends and risk factors for prolonged opioid use after unicompartmental knee arthroplasty. Bone Joint J. 2018;100-B(1)(suppl A):62-67. doi:10.1302/0301-620x.100b1.bjj-2017-0547.r1
8. Politzer CS, Kildow BJ, Goltz DE, Green CL, Bolognesi MP, Seyler T. Trends in opioid utilization before and after total knee arthroplasty. J Arthroplasty. 2018;33(7S):S147-S153.e1. doi:10.1016/j.arth.2017.10.060
9. Mariano ER, Walters TL, Kim ET, Kain ZN. Why the perioperative surgical home makes sense for Veterans Affairs health care. Anesth Analg. 2015;120(5):1163-1166. doi:10.1213/ane.0000000000000712
10. Walters TL, Howard SK, Kou A, et al. Design and implementation of a perioperative surgical home at a Veterans Affairs hospital. Semin Cardiothorac Vasc Anesth. 2016;20(2):133-140. doi:10.1177/1089253215607066
11. Walters TL, Mariano ER, Clark DJ. Perioperative surgical home and the integral role of pain medicine. Pain Med. 2015;16(9):1666-1672. doi:10.1111/pme.12796
12. Vetter TR, Kain ZN. Role of the perioperative surgical home in optimizing the perioperative use of opioids. Anesth Analg. 2017;125(5):1653-1657. doi:10.1213/ane.0000000000002280
13. Shafer SL. Anesthesia & Analgesia’s 2015 collection on the perioperative surgical home. Anesth Analg. 2015;120(5):966-967. doi:10.1213/ane.0000000000000696
14. Wenzel JT, Schwenk ES, Baratta JL, Viscusi ER. Managing opioid-tolerant patients in the perioperative surgical home. Anesthesiol Clin. 2016;34(2):287-301. doi:10.1016/j.anclin.2016.01.005
15. Katz J, Weinrib A, Fashler SR, et al. The Toronto General Hospital Transitional Pain Service: development and implementation of a multidisciplinary program to prevent chronic postsurgical pain. J Pain Res. 2015;8:695-702. doi:10.2147/jpr.s91924
16. Tiippana E, Hamunen K, Heiskanen T, Nieminen T, Kalso E, Kontinen VK. New approach for treatment of prolonged postoperative pain: APS Out-Patient Clinic. Scand J Pain. 2016;12(1):19-24. doi:10.1016/j.sjpain.2016.02.008
17. Katz J, Weinrib AZ, Clarke H. Chronic postsurgical pain: from risk factor identification to multidisciplinary management at the Toronto General Hospital Transitional Pain Service. Can J Pain. 2019;3(2):49-58. doi:10.1080/24740527.2019.1574537
18. Sullivan MJ, Bishop SR, Pivik J. The Pain Catastrophizing Scale: development and validation. Psychol Assess. 1995;7(4):524-532. doi:10.1037/1040-3590.7.4.524
19. HealthMeasures. Intro to PROMIS. https://www.healthmeasures.net/explore-measurement-systems/promis. Accessed September 28, 2020.
Experts assess infection risks for patients on biologics
In a new review, a group of infectious disease experts have summarized and made recommendations about recent findings regarding infections that can occur during treatment with an evolving set of targeted and biologic therapies for rheumatoid arthritis and psoriatic arthritis.
“We claim for the need for multicenter registries and multidisciplinary approaches, for new vaccines trials in RA and PsA, and for better defining when and how biologics can be restarted after severe infections,” lead author Olivier Lortholary, MD, of the Institut Pasteur in Paris, and his coauthors wrote in Annals of the Rheumatic Diseases.
“The take-home message is that different DMARDs [disease-modifying antirheumatic drugs], in many ways, are very similar,” said coauthor Kevin L. Winthrop, MD, MPH, professor of public health and ophthalmology at Oregon Health & Science University, Portland, in an interview. “They all have fairly similar risks when it comes to ‘classical’ or routine bacterial infections. But when you talk about opportunistic infections, you start seeing the differences between these drugs.”
The experts began by addressing the current view of the infectious risk of biologic therapies, citing a recent meta-analysis in which standard (odds ratio, 1.31; 95% confidence interval, 1.09-1.58) and high (OR, 1.90; 95% CI, 1.50-2.39) doses of biologics were associated with increased risk of serious infection. They also noted that the ‘healthy drug survivor effect’ tends to confound long-term extensions of randomized clinical trials involving biologics.
“That is largely because people who are more likely to do well or have proven themselves to do well with that infection, they tend to stay in [trials] and stay on drugs,” Dr. Winthrop said. “The ones who develop infections are more likely to drop out. You see this survival of the fittest-type situation, where healthy users dominate a cohort over time. That’s why you see incidence rates decreasing.”
In response, Arthur Kavanaugh, MD, professor of medicine in the division of rheumatology, allergy, and immunology at the University of California, San Diego, and the director of the Center for Innovative Therapy there, backed the idea of a general ‘depletion of the susceptibles’ but warned doctors to evaluate each patient and situation accordingly. “Providers need to be vigilant throughout for common infections, rarer infections, and infections at greatest risk for the individual patient based on factors like comorbidities and concomitant medications,” he said in an interview.
When considering restarting a biologic in a patient who recently suffered a serious infection, the experts prescribed no general rule and noted that it will “depend on the type of infection, on the mechanism of action of the drug, on the other available drugs for the considered disease and, of course, on the willingness of the patients to restart a drug possibly having [given] him/her a side effect.”
Assessing infection risk related to various inhibitors
Regarding infections caused by TNF-alpha inhibitors (TNFIs), the experts acknowledged a broad increase in risk for mycobacterial and fungal infections, especially tuberculosis and histoplasmosis. They added that patients on TNFIs are more prone to developing pneumonia and soft tissue infections, while smaller studies have indicated a higher risk of listeriosis, legionellosis, herpes zoster (HZ), and reactivation of chronic hepatitis B virus infection.
As for recommendations, they endorsed discontinuing TNFIs when a serious infection occurs and not restarting until after treatment and clinical response. Patients should be screened for latent tuberculosis infection (LTBI) before starting the drug, and anti-TB drugs should be presented to patients with LTBI so they do not progress to active TB.
Regarding other biologics, they cited several studies indicating that IL-6 inhibitors can increase infection risks in RA patients at a rate similar to TNFIs. Among the most common infections were pneumonia and cellulitis. In addition, although PsA patients on IL-17 inhibitors have a dose-dependent risk of mild to moderate mucocutaneous candidiasis, there was no increased risk of serious opportunistic infections like TB.
In assessing JAK inhibitors, they cited a pooled analysis that indicated pneumonia and skin and soft-tissue infections as the most common and noted the high incidence of HZ, compared with other infections. They added that abatacept (Orencia) did not appear to increase risk of infections in RA patients, such as HZ, dermatomycosis, candidiasis, or endemic mycoses. Those same patients did not see an increased overall infection risk after treatment with rituximab (Rituxan), and clinical trials containing treatment with apremilast (Otezla) reported a rare occurrence of serious infections.
Recommendation-wise, they endorsed screening for LTBI before starting IL-6 inhibitors and antiviral prophylaxis with acyclovir in particularly at-risk patients on JAK inhibitors. Age-appropriate influenza vaccinations were also recommended for rituximab, because of the development of rituximab-induced hypogammaglobulinemia.
Prediction and prevention
When it comes to predicting infections in patients on biologics, the experts wrote that it “remains a challenge.” The potential effects of pretreatment underlying disease, the lack of validated biomarkers, and the relatively low rate of infections all combine to stymie prediction. That said, they acknowledged ongoing efforts in monitoring lymphocyte subpopulation counts and immunoglobin levels, as well as a clinical score called the RABBIT Risk Score for Infections, which was validated in two separate cohorts.
“As Yogi Berra said, predictions are hard, especially about the future,” Dr. Kavanaugh said. “Discussions with your patient are always important.”
In regard to overall prevention, they acknowledged that most of their recommendations are of low evidence, except for antiviral prophylaxis for hepatitis B patients on rituximab and the aforementioned LTBI therapy in patients on TNFIs. Broadly, they advocated for all RA and PsA patients to receive a full infectious disease evaluation before the start of targeted and biologic therapies.
They also addressed vaccinations, recommending an evaluation of the patient’s immunization history and potentially planning a catch-up schedule for those in need of the influenza vaccine, a diphtheria-tetanus-pertussis booster, or the pneumococcal vaccine. More broadly, they stated that “a better response is expected if [non-live] vaccination is performed before the introduction of immunosuppressive drugs.” They added that live vaccines should be administered as soon as possible.
What rheumatologists can do
“So how do you mitigate risk?” Dr. Winthrop asked. “You have to be able to predict the risk, see what’s modifiable, and try to act on it. A lot of the risk of infection has more to do with the patient than the therapy.
“You try to minimize what you’re doing to the patient, particularly around steroids,” he said. “And then you think about screening and vaccinations. Rheumatologists need to be involved in those conversations because they’re the ones who know how these drugs interact with vaccines. A lot of the drugs might dumb down vaccine responses. Be sure to consider that and give the vaccines at times that will optimize their immunogenicity and likely efficacy.”
“Thankfully, infections are not that common,” Dr. Kavanaugh said. “Rheumatologists depend on data from trials, but more safety data comes from registry data and personal and shared experience.”
The authors declared no potential conflicts of interest.
SOURCE: Lortholary O et al. Ann Rheum Dis. 2020 Sep 22. doi: 10.1136/annrheumdis-2020-217092.
In a new review, a group of infectious disease experts have summarized and made recommendations about recent findings regarding infections that can occur during treatment with an evolving set of targeted and biologic therapies for rheumatoid arthritis and psoriatic arthritis.
“We claim for the need for multicenter registries and multidisciplinary approaches, for new vaccines trials in RA and PsA, and for better defining when and how biologics can be restarted after severe infections,” lead author Olivier Lortholary, MD, of the Institut Pasteur in Paris, and his coauthors wrote in Annals of the Rheumatic Diseases.
“The take-home message is that different DMARDs [disease-modifying antirheumatic drugs], in many ways, are very similar,” said coauthor Kevin L. Winthrop, MD, MPH, professor of public health and ophthalmology at Oregon Health & Science University, Portland, in an interview. “They all have fairly similar risks when it comes to ‘classical’ or routine bacterial infections. But when you talk about opportunistic infections, you start seeing the differences between these drugs.”
The experts began by addressing the current view of the infectious risk of biologic therapies, citing a recent meta-analysis in which standard (odds ratio, 1.31; 95% confidence interval, 1.09-1.58) and high (OR, 1.90; 95% CI, 1.50-2.39) doses of biologics were associated with increased risk of serious infection. They also noted that the ‘healthy drug survivor effect’ tends to confound long-term extensions of randomized clinical trials involving biologics.
“That is largely because people who are more likely to do well or have proven themselves to do well with that infection, they tend to stay in [trials] and stay on drugs,” Dr. Winthrop said. “The ones who develop infections are more likely to drop out. You see this survival of the fittest-type situation, where healthy users dominate a cohort over time. That’s why you see incidence rates decreasing.”
In response, Arthur Kavanaugh, MD, professor of medicine in the division of rheumatology, allergy, and immunology at the University of California, San Diego, and the director of the Center for Innovative Therapy there, backed the idea of a general ‘depletion of the susceptibles’ but warned doctors to evaluate each patient and situation accordingly. “Providers need to be vigilant throughout for common infections, rarer infections, and infections at greatest risk for the individual patient based on factors like comorbidities and concomitant medications,” he said in an interview.
When considering restarting a biologic in a patient who recently suffered a serious infection, the experts prescribed no general rule and noted that it will “depend on the type of infection, on the mechanism of action of the drug, on the other available drugs for the considered disease and, of course, on the willingness of the patients to restart a drug possibly having [given] him/her a side effect.”
Assessing infection risk related to various inhibitors
Regarding infections caused by TNF-alpha inhibitors (TNFIs), the experts acknowledged a broad increase in risk for mycobacterial and fungal infections, especially tuberculosis and histoplasmosis. They added that patients on TNFIs are more prone to developing pneumonia and soft tissue infections, while smaller studies have indicated a higher risk of listeriosis, legionellosis, herpes zoster (HZ), and reactivation of chronic hepatitis B virus infection.
As for recommendations, they endorsed discontinuing TNFIs when a serious infection occurs and not restarting until after treatment and clinical response. Patients should be screened for latent tuberculosis infection (LTBI) before starting the drug, and anti-TB drugs should be presented to patients with LTBI so they do not progress to active TB.
Regarding other biologics, they cited several studies indicating that IL-6 inhibitors can increase infection risks in RA patients at a rate similar to TNFIs. Among the most common infections were pneumonia and cellulitis. In addition, although PsA patients on IL-17 inhibitors have a dose-dependent risk of mild to moderate mucocutaneous candidiasis, there was no increased risk of serious opportunistic infections like TB.
In assessing JAK inhibitors, they cited a pooled analysis that indicated pneumonia and skin and soft-tissue infections as the most common and noted the high incidence of HZ, compared with other infections. They added that abatacept (Orencia) did not appear to increase risk of infections in RA patients, such as HZ, dermatomycosis, candidiasis, or endemic mycoses. Those same patients did not see an increased overall infection risk after treatment with rituximab (Rituxan), and clinical trials containing treatment with apremilast (Otezla) reported a rare occurrence of serious infections.
Recommendation-wise, they endorsed screening for LTBI before starting IL-6 inhibitors and antiviral prophylaxis with acyclovir in particularly at-risk patients on JAK inhibitors. Age-appropriate influenza vaccinations were also recommended for rituximab, because of the development of rituximab-induced hypogammaglobulinemia.
Prediction and prevention
When it comes to predicting infections in patients on biologics, the experts wrote that it “remains a challenge.” The potential effects of pretreatment underlying disease, the lack of validated biomarkers, and the relatively low rate of infections all combine to stymie prediction. That said, they acknowledged ongoing efforts in monitoring lymphocyte subpopulation counts and immunoglobin levels, as well as a clinical score called the RABBIT Risk Score for Infections, which was validated in two separate cohorts.
“As Yogi Berra said, predictions are hard, especially about the future,” Dr. Kavanaugh said. “Discussions with your patient are always important.”
In regard to overall prevention, they acknowledged that most of their recommendations are of low evidence, except for antiviral prophylaxis for hepatitis B patients on rituximab and the aforementioned LTBI therapy in patients on TNFIs. Broadly, they advocated for all RA and PsA patients to receive a full infectious disease evaluation before the start of targeted and biologic therapies.
They also addressed vaccinations, recommending an evaluation of the patient’s immunization history and potentially planning a catch-up schedule for those in need of the influenza vaccine, a diphtheria-tetanus-pertussis booster, or the pneumococcal vaccine. More broadly, they stated that “a better response is expected if [non-live] vaccination is performed before the introduction of immunosuppressive drugs.” They added that live vaccines should be administered as soon as possible.
What rheumatologists can do
“So how do you mitigate risk?” Dr. Winthrop asked. “You have to be able to predict the risk, see what’s modifiable, and try to act on it. A lot of the risk of infection has more to do with the patient than the therapy.
“You try to minimize what you’re doing to the patient, particularly around steroids,” he said. “And then you think about screening and vaccinations. Rheumatologists need to be involved in those conversations because they’re the ones who know how these drugs interact with vaccines. A lot of the drugs might dumb down vaccine responses. Be sure to consider that and give the vaccines at times that will optimize their immunogenicity and likely efficacy.”
“Thankfully, infections are not that common,” Dr. Kavanaugh said. “Rheumatologists depend on data from trials, but more safety data comes from registry data and personal and shared experience.”
The authors declared no potential conflicts of interest.
SOURCE: Lortholary O et al. Ann Rheum Dis. 2020 Sep 22. doi: 10.1136/annrheumdis-2020-217092.
In a new review, a group of infectious disease experts have summarized and made recommendations about recent findings regarding infections that can occur during treatment with an evolving set of targeted and biologic therapies for rheumatoid arthritis and psoriatic arthritis.
“We claim for the need for multicenter registries and multidisciplinary approaches, for new vaccines trials in RA and PsA, and for better defining when and how biologics can be restarted after severe infections,” lead author Olivier Lortholary, MD, of the Institut Pasteur in Paris, and his coauthors wrote in Annals of the Rheumatic Diseases.
“The take-home message is that different DMARDs [disease-modifying antirheumatic drugs], in many ways, are very similar,” said coauthor Kevin L. Winthrop, MD, MPH, professor of public health and ophthalmology at Oregon Health & Science University, Portland, in an interview. “They all have fairly similar risks when it comes to ‘classical’ or routine bacterial infections. But when you talk about opportunistic infections, you start seeing the differences between these drugs.”
The experts began by addressing the current view of the infectious risk of biologic therapies, citing a recent meta-analysis in which standard (odds ratio, 1.31; 95% confidence interval, 1.09-1.58) and high (OR, 1.90; 95% CI, 1.50-2.39) doses of biologics were associated with increased risk of serious infection. They also noted that the ‘healthy drug survivor effect’ tends to confound long-term extensions of randomized clinical trials involving biologics.
“That is largely because people who are more likely to do well or have proven themselves to do well with that infection, they tend to stay in [trials] and stay on drugs,” Dr. Winthrop said. “The ones who develop infections are more likely to drop out. You see this survival of the fittest-type situation, where healthy users dominate a cohort over time. That’s why you see incidence rates decreasing.”
In response, Arthur Kavanaugh, MD, professor of medicine in the division of rheumatology, allergy, and immunology at the University of California, San Diego, and the director of the Center for Innovative Therapy there, backed the idea of a general ‘depletion of the susceptibles’ but warned doctors to evaluate each patient and situation accordingly. “Providers need to be vigilant throughout for common infections, rarer infections, and infections at greatest risk for the individual patient based on factors like comorbidities and concomitant medications,” he said in an interview.
When considering restarting a biologic in a patient who recently suffered a serious infection, the experts prescribed no general rule and noted that it will “depend on the type of infection, on the mechanism of action of the drug, on the other available drugs for the considered disease and, of course, on the willingness of the patients to restart a drug possibly having [given] him/her a side effect.”
Assessing infection risk related to various inhibitors
Regarding infections caused by TNF-alpha inhibitors (TNFIs), the experts acknowledged a broad increase in risk for mycobacterial and fungal infections, especially tuberculosis and histoplasmosis. They added that patients on TNFIs are more prone to developing pneumonia and soft tissue infections, while smaller studies have indicated a higher risk of listeriosis, legionellosis, herpes zoster (HZ), and reactivation of chronic hepatitis B virus infection.
As for recommendations, they endorsed discontinuing TNFIs when a serious infection occurs and not restarting until after treatment and clinical response. Patients should be screened for latent tuberculosis infection (LTBI) before starting the drug, and anti-TB drugs should be presented to patients with LTBI so they do not progress to active TB.
Regarding other biologics, they cited several studies indicating that IL-6 inhibitors can increase infection risks in RA patients at a rate similar to TNFIs. Among the most common infections were pneumonia and cellulitis. In addition, although PsA patients on IL-17 inhibitors have a dose-dependent risk of mild to moderate mucocutaneous candidiasis, there was no increased risk of serious opportunistic infections like TB.
In assessing JAK inhibitors, they cited a pooled analysis that indicated pneumonia and skin and soft-tissue infections as the most common and noted the high incidence of HZ, compared with other infections. They added that abatacept (Orencia) did not appear to increase risk of infections in RA patients, such as HZ, dermatomycosis, candidiasis, or endemic mycoses. Those same patients did not see an increased overall infection risk after treatment with rituximab (Rituxan), and clinical trials containing treatment with apremilast (Otezla) reported a rare occurrence of serious infections.
Recommendation-wise, they endorsed screening for LTBI before starting IL-6 inhibitors and antiviral prophylaxis with acyclovir in particularly at-risk patients on JAK inhibitors. Age-appropriate influenza vaccinations were also recommended for rituximab, because of the development of rituximab-induced hypogammaglobulinemia.
Prediction and prevention
When it comes to predicting infections in patients on biologics, the experts wrote that it “remains a challenge.” The potential effects of pretreatment underlying disease, the lack of validated biomarkers, and the relatively low rate of infections all combine to stymie prediction. That said, they acknowledged ongoing efforts in monitoring lymphocyte subpopulation counts and immunoglobin levels, as well as a clinical score called the RABBIT Risk Score for Infections, which was validated in two separate cohorts.
“As Yogi Berra said, predictions are hard, especially about the future,” Dr. Kavanaugh said. “Discussions with your patient are always important.”
In regard to overall prevention, they acknowledged that most of their recommendations are of low evidence, except for antiviral prophylaxis for hepatitis B patients on rituximab and the aforementioned LTBI therapy in patients on TNFIs. Broadly, they advocated for all RA and PsA patients to receive a full infectious disease evaluation before the start of targeted and biologic therapies.
They also addressed vaccinations, recommending an evaluation of the patient’s immunization history and potentially planning a catch-up schedule for those in need of the influenza vaccine, a diphtheria-tetanus-pertussis booster, or the pneumococcal vaccine. More broadly, they stated that “a better response is expected if [non-live] vaccination is performed before the introduction of immunosuppressive drugs.” They added that live vaccines should be administered as soon as possible.
What rheumatologists can do
“So how do you mitigate risk?” Dr. Winthrop asked. “You have to be able to predict the risk, see what’s modifiable, and try to act on it. A lot of the risk of infection has more to do with the patient than the therapy.
“You try to minimize what you’re doing to the patient, particularly around steroids,” he said. “And then you think about screening and vaccinations. Rheumatologists need to be involved in those conversations because they’re the ones who know how these drugs interact with vaccines. A lot of the drugs might dumb down vaccine responses. Be sure to consider that and give the vaccines at times that will optimize their immunogenicity and likely efficacy.”
“Thankfully, infections are not that common,” Dr. Kavanaugh said. “Rheumatologists depend on data from trials, but more safety data comes from registry data and personal and shared experience.”
The authors declared no potential conflicts of interest.
SOURCE: Lortholary O et al. Ann Rheum Dis. 2020 Sep 22. doi: 10.1136/annrheumdis-2020-217092.
FROM ANNALS OF THE RHEUMATIC DISEASES
Dapagliflozin’s CKD performance sends heart failure messages
The DAPA-CKD trial results, which proved dapagliflozin’s efficacy for slowing chronic kidney disease progression in patients selected for signs of worsening renal function, also have important messages for cardiologists, especially heart failure physicians.
Those messages include findings that were “consistent” with the results of the earlier DAPA-HF trial, which tested the same sodium-glucose transporter 2 (SGLT2) inhibitor in patients selected for having heart failure with reduced ejection fraction (HFrEF). In addition, a specific action of dapagliflozin (Farxiga) on the patients in DAPA-CKD, which enrolled patients based on markers of chronic kidney disease (CKD), was prevention of first and recurrent heart failure hospitalizations, John J.V. McMurray, MD, said at the virtual annual scientific meeting of the Heart Failure Society of America, further highlighting the role that dapagliflozin has in reducing both heart failure and renal events.
What DAPA-CKD means for heart failure
The main findings from the DAPA-CKD trial, published in September in the New England Journal of Medicine, included as a secondary outcome the combined rate of death from cardiovascular causes or hospitalization for heart failure (HHF). Treatment with dapagliflozin linked with a significant 29% relative reduction in this endpoint, compared with placebo-treated patients. At the HFSA meeting, Dr. McMurray reported for the first time the specific HHF numbers, a prespecified secondary endpoint for the study.
Patients on dapagliflozin had 37 total HHF events (1.7%), including both first-time and subsequent hospitalizations, while patients in the placebo arm had a total of 71 HHF events (3.3%) during the study’s median 2.4 years of follow-up, an absolute reduction of 1.6% that translated into a relative risk reduction of 49%.
The HHF findings from DAPA-CKD importantly showed that SGLT2 inhibition in patients with signs of renal dysfunction “will not only slow progression of kidney disease but will also reduce the risk of developing heart failure, crucially in patients with or without type 2 diabetes,” explained Dr. McMurray in an interview. “Cardiologists often consult in the kidney wards and advise on management of patients with chronic kidney disease, even those without heart failure.”
The DAPA-CKD findings carry another important message for heart failure management regarding the minimum level of renal function a patient can have and still safely receive dapagliflozin or possibly another agent from the same SGLT2 inhibitor class. In DAPA-CKD, patients safely received dapagliflozin with an estimated glomerular filtration rate (eGFR) as low as 25 mL/min per 1.73 m2; 14% of enrolled patients had an eGFR of 25-29 mL/min per 1.73 m2.
“Typically, about 40%-50% of patients with heart failure have chronic kidney disease,” which makes this safety finding important to clinicians who care for heart failure patients, but it’s also important for any patient who might be a candidate for dapagliflozin or another drug from its class. “We had no strong evidence before this trial that SGLT2 inhibition could reduce hard renal endpoints,” specifically need for chronic dialysis, renal transplant, or renal death, “in patients with or without diabetes,” Dr. McMurray said.
DAPA-CKD grows the pool of eligible heart failure patients
A further consequence of the DAPA-CKD findings is that when, as expected, regulatory bodies give dapagliflozin an indication for treating the types of CKD patients enrolled in the trial, it will functionally expand this treatment to an even larger swath of heart failure patients who currently don’t qualify for this treatment, specifically patients with CKD who also have heart failure with preserved ejection fraction (HFpEF). On Oct. 2, 2020, the Food and Drug Administration fast-tracked dapagliflozin for the CKD indication by granting it Breakthrough Therapy Designation based on the DAPA-CKD results.
Results first reported in 2019 from the DAPA-HF trial led to dapagliflozin receiving a labeled indication for treating HFrEF, the types of heart failure patients enrolled in the trial. Direct evidence on the efficacy of SGLT2 inhibitors for patients with HFpEF will not be available until results from a few trials now in progress become available during the next 12 months.
In the meantime, nearly half of patients with HFpEF also have CKD, noted Dr. McMurray, and another large portion of HFpEF patients have type 2 diabetes and hence qualify for SGLT2 inhibitor treatment that way. “Obviously, we would like to know specifically about heart failure outcomes in patients with HFpEF” on SGLT2 inhibitor treatment, he acknowledged. But the recent approval of dapagliflozin for patients with HFrEF and the likely indication coming soon for treating CKD means that the number of patients with heart failure who are not eligible for SGLT2 inhibitor treatment is dwindling down to some extent.
New DAPA-HF results show no drug, device interactions
In a separate session at the HFSA virtual meeting, Dr. McMurray and several collaborators on the DAPA-HF trial presented results from some new analyses. Dr. McMurray looked at the impact of dapagliflozin treatment on the primary endpoint when patients were stratified by the diuretic dosage they received at study entry. The results showed that “the benefits from dapagliflozin were irrespective of the use of background diuretic therapy or the diuretic dose,” he reported. Study findings also showed that roughly three-quarters of patients in the study had no change in their diuretic dosage during the course of the trial, that the fraction of patients who had an increase in their dosage was about the same as those whose diuretic dosage decreased, and that this pattern was similar in both the patients on dapagliflozin and in those randomized to placebo.
Another set of new analyses from DAPA-HF looked at the impact on dapagliflozin efficacy of background medical and device therapies for heart failure, as well as background diabetes therapies. The findings showed no signal of an interaction with background therapies. “The effects of dapagliflozin are incremental and complimentary to conventional therapies for HFrEF,” concluded Lars Kober, MD, a professor and heart failure physician at Copenhagen University Hospital.
DAPA-CKD was funded by AstraZeneca, the company that markets dapagliflozin (Farxiga). Dr. McMurray’s employer, Glasgow University, has received payments from AstraZeneca and several other companies to compensate for his time overseeing various clinical trials. Dr. Kober has received honoraria for speaking on behalf of several companies including AstraZeneca.
The DAPA-CKD trial results, which proved dapagliflozin’s efficacy for slowing chronic kidney disease progression in patients selected for signs of worsening renal function, also have important messages for cardiologists, especially heart failure physicians.
Those messages include findings that were “consistent” with the results of the earlier DAPA-HF trial, which tested the same sodium-glucose transporter 2 (SGLT2) inhibitor in patients selected for having heart failure with reduced ejection fraction (HFrEF). In addition, a specific action of dapagliflozin (Farxiga) on the patients in DAPA-CKD, which enrolled patients based on markers of chronic kidney disease (CKD), was prevention of first and recurrent heart failure hospitalizations, John J.V. McMurray, MD, said at the virtual annual scientific meeting of the Heart Failure Society of America, further highlighting the role that dapagliflozin has in reducing both heart failure and renal events.
What DAPA-CKD means for heart failure
The main findings from the DAPA-CKD trial, published in September in the New England Journal of Medicine, included as a secondary outcome the combined rate of death from cardiovascular causes or hospitalization for heart failure (HHF). Treatment with dapagliflozin linked with a significant 29% relative reduction in this endpoint, compared with placebo-treated patients. At the HFSA meeting, Dr. McMurray reported for the first time the specific HHF numbers, a prespecified secondary endpoint for the study.
Patients on dapagliflozin had 37 total HHF events (1.7%), including both first-time and subsequent hospitalizations, while patients in the placebo arm had a total of 71 HHF events (3.3%) during the study’s median 2.4 years of follow-up, an absolute reduction of 1.6% that translated into a relative risk reduction of 49%.
The HHF findings from DAPA-CKD importantly showed that SGLT2 inhibition in patients with signs of renal dysfunction “will not only slow progression of kidney disease but will also reduce the risk of developing heart failure, crucially in patients with or without type 2 diabetes,” explained Dr. McMurray in an interview. “Cardiologists often consult in the kidney wards and advise on management of patients with chronic kidney disease, even those without heart failure.”
The DAPA-CKD findings carry another important message for heart failure management regarding the minimum level of renal function a patient can have and still safely receive dapagliflozin or possibly another agent from the same SGLT2 inhibitor class. In DAPA-CKD, patients safely received dapagliflozin with an estimated glomerular filtration rate (eGFR) as low as 25 mL/min per 1.73 m2; 14% of enrolled patients had an eGFR of 25-29 mL/min per 1.73 m2.
“Typically, about 40%-50% of patients with heart failure have chronic kidney disease,” which makes this safety finding important to clinicians who care for heart failure patients, but it’s also important for any patient who might be a candidate for dapagliflozin or another drug from its class. “We had no strong evidence before this trial that SGLT2 inhibition could reduce hard renal endpoints,” specifically need for chronic dialysis, renal transplant, or renal death, “in patients with or without diabetes,” Dr. McMurray said.
DAPA-CKD grows the pool of eligible heart failure patients
A further consequence of the DAPA-CKD findings is that when, as expected, regulatory bodies give dapagliflozin an indication for treating the types of CKD patients enrolled in the trial, it will functionally expand this treatment to an even larger swath of heart failure patients who currently don’t qualify for this treatment, specifically patients with CKD who also have heart failure with preserved ejection fraction (HFpEF). On Oct. 2, 2020, the Food and Drug Administration fast-tracked dapagliflozin for the CKD indication by granting it Breakthrough Therapy Designation based on the DAPA-CKD results.
Results first reported in 2019 from the DAPA-HF trial led to dapagliflozin receiving a labeled indication for treating HFrEF, the types of heart failure patients enrolled in the trial. Direct evidence on the efficacy of SGLT2 inhibitors for patients with HFpEF will not be available until results from a few trials now in progress become available during the next 12 months.
In the meantime, nearly half of patients with HFpEF also have CKD, noted Dr. McMurray, and another large portion of HFpEF patients have type 2 diabetes and hence qualify for SGLT2 inhibitor treatment that way. “Obviously, we would like to know specifically about heart failure outcomes in patients with HFpEF” on SGLT2 inhibitor treatment, he acknowledged. But the recent approval of dapagliflozin for patients with HFrEF and the likely indication coming soon for treating CKD means that the number of patients with heart failure who are not eligible for SGLT2 inhibitor treatment is dwindling down to some extent.
New DAPA-HF results show no drug, device interactions
In a separate session at the HFSA virtual meeting, Dr. McMurray and several collaborators on the DAPA-HF trial presented results from some new analyses. Dr. McMurray looked at the impact of dapagliflozin treatment on the primary endpoint when patients were stratified by the diuretic dosage they received at study entry. The results showed that “the benefits from dapagliflozin were irrespective of the use of background diuretic therapy or the diuretic dose,” he reported. Study findings also showed that roughly three-quarters of patients in the study had no change in their diuretic dosage during the course of the trial, that the fraction of patients who had an increase in their dosage was about the same as those whose diuretic dosage decreased, and that this pattern was similar in both the patients on dapagliflozin and in those randomized to placebo.
Another set of new analyses from DAPA-HF looked at the impact on dapagliflozin efficacy of background medical and device therapies for heart failure, as well as background diabetes therapies. The findings showed no signal of an interaction with background therapies. “The effects of dapagliflozin are incremental and complimentary to conventional therapies for HFrEF,” concluded Lars Kober, MD, a professor and heart failure physician at Copenhagen University Hospital.
DAPA-CKD was funded by AstraZeneca, the company that markets dapagliflozin (Farxiga). Dr. McMurray’s employer, Glasgow University, has received payments from AstraZeneca and several other companies to compensate for his time overseeing various clinical trials. Dr. Kober has received honoraria for speaking on behalf of several companies including AstraZeneca.
The DAPA-CKD trial results, which proved dapagliflozin’s efficacy for slowing chronic kidney disease progression in patients selected for signs of worsening renal function, also have important messages for cardiologists, especially heart failure physicians.
Those messages include findings that were “consistent” with the results of the earlier DAPA-HF trial, which tested the same sodium-glucose transporter 2 (SGLT2) inhibitor in patients selected for having heart failure with reduced ejection fraction (HFrEF). In addition, a specific action of dapagliflozin (Farxiga) on the patients in DAPA-CKD, which enrolled patients based on markers of chronic kidney disease (CKD), was prevention of first and recurrent heart failure hospitalizations, John J.V. McMurray, MD, said at the virtual annual scientific meeting of the Heart Failure Society of America, further highlighting the role that dapagliflozin has in reducing both heart failure and renal events.
What DAPA-CKD means for heart failure
The main findings from the DAPA-CKD trial, published in September in the New England Journal of Medicine, included as a secondary outcome the combined rate of death from cardiovascular causes or hospitalization for heart failure (HHF). Treatment with dapagliflozin linked with a significant 29% relative reduction in this endpoint, compared with placebo-treated patients. At the HFSA meeting, Dr. McMurray reported for the first time the specific HHF numbers, a prespecified secondary endpoint for the study.
Patients on dapagliflozin had 37 total HHF events (1.7%), including both first-time and subsequent hospitalizations, while patients in the placebo arm had a total of 71 HHF events (3.3%) during the study’s median 2.4 years of follow-up, an absolute reduction of 1.6% that translated into a relative risk reduction of 49%.
The HHF findings from DAPA-CKD importantly showed that SGLT2 inhibition in patients with signs of renal dysfunction “will not only slow progression of kidney disease but will also reduce the risk of developing heart failure, crucially in patients with or without type 2 diabetes,” explained Dr. McMurray in an interview. “Cardiologists often consult in the kidney wards and advise on management of patients with chronic kidney disease, even those without heart failure.”
The DAPA-CKD findings carry another important message for heart failure management regarding the minimum level of renal function a patient can have and still safely receive dapagliflozin or possibly another agent from the same SGLT2 inhibitor class. In DAPA-CKD, patients safely received dapagliflozin with an estimated glomerular filtration rate (eGFR) as low as 25 mL/min per 1.73 m2; 14% of enrolled patients had an eGFR of 25-29 mL/min per 1.73 m2.
“Typically, about 40%-50% of patients with heart failure have chronic kidney disease,” which makes this safety finding important to clinicians who care for heart failure patients, but it’s also important for any patient who might be a candidate for dapagliflozin or another drug from its class. “We had no strong evidence before this trial that SGLT2 inhibition could reduce hard renal endpoints,” specifically need for chronic dialysis, renal transplant, or renal death, “in patients with or without diabetes,” Dr. McMurray said.
DAPA-CKD grows the pool of eligible heart failure patients
A further consequence of the DAPA-CKD findings is that when, as expected, regulatory bodies give dapagliflozin an indication for treating the types of CKD patients enrolled in the trial, it will functionally expand this treatment to an even larger swath of heart failure patients who currently don’t qualify for this treatment, specifically patients with CKD who also have heart failure with preserved ejection fraction (HFpEF). On Oct. 2, 2020, the Food and Drug Administration fast-tracked dapagliflozin for the CKD indication by granting it Breakthrough Therapy Designation based on the DAPA-CKD results.
Results first reported in 2019 from the DAPA-HF trial led to dapagliflozin receiving a labeled indication for treating HFrEF, the types of heart failure patients enrolled in the trial. Direct evidence on the efficacy of SGLT2 inhibitors for patients with HFpEF will not be available until results from a few trials now in progress become available during the next 12 months.
In the meantime, nearly half of patients with HFpEF also have CKD, noted Dr. McMurray, and another large portion of HFpEF patients have type 2 diabetes and hence qualify for SGLT2 inhibitor treatment that way. “Obviously, we would like to know specifically about heart failure outcomes in patients with HFpEF” on SGLT2 inhibitor treatment, he acknowledged. But the recent approval of dapagliflozin for patients with HFrEF and the likely indication coming soon for treating CKD means that the number of patients with heart failure who are not eligible for SGLT2 inhibitor treatment is dwindling down to some extent.
New DAPA-HF results show no drug, device interactions
In a separate session at the HFSA virtual meeting, Dr. McMurray and several collaborators on the DAPA-HF trial presented results from some new analyses. Dr. McMurray looked at the impact of dapagliflozin treatment on the primary endpoint when patients were stratified by the diuretic dosage they received at study entry. The results showed that “the benefits from dapagliflozin were irrespective of the use of background diuretic therapy or the diuretic dose,” he reported. Study findings also showed that roughly three-quarters of patients in the study had no change in their diuretic dosage during the course of the trial, that the fraction of patients who had an increase in their dosage was about the same as those whose diuretic dosage decreased, and that this pattern was similar in both the patients on dapagliflozin and in those randomized to placebo.
Another set of new analyses from DAPA-HF looked at the impact on dapagliflozin efficacy of background medical and device therapies for heart failure, as well as background diabetes therapies. The findings showed no signal of an interaction with background therapies. “The effects of dapagliflozin are incremental and complimentary to conventional therapies for HFrEF,” concluded Lars Kober, MD, a professor and heart failure physician at Copenhagen University Hospital.
DAPA-CKD was funded by AstraZeneca, the company that markets dapagliflozin (Farxiga). Dr. McMurray’s employer, Glasgow University, has received payments from AstraZeneca and several other companies to compensate for his time overseeing various clinical trials. Dr. Kober has received honoraria for speaking on behalf of several companies including AstraZeneca.
FROM HFSA 2020
Perceived Barriers and Facilitators of Clozapine Use: A National Survey of Veterans Affairs Prescribers (FULL)
Clozapine is an atypical antipsychotic that the US Food and Drug Administration (FDA) approved for use in schizophrenia and suicidality associated with schizophrenia or schizoaffective disorder. Clozapine has been shown to be superior to other antipsychotic treatment for treatment resistant schizophrenia (TRS), which is defined as failure of 2 adequate trials of antipsychotic therapy.1 Up to 30% of patients with schizophrenia are classified as treatment resistant.2
Clozapine is considered the drug of choice for patients with TRS in both the US Department of Veterans Affairs (VA) policies and other evidence-based guidelines and remains the only antipsychotic with FDA approval for TRS.2-5 Patients treated with clozapine have fewer psychiatric hospitalizations, fewer suicide attempts, lower rates of nonadherence, and less antipsychotic polypharmacy compared with patients who are treated with other antipsychotic therapy.6,7 A 2016 study by Gören and colleagues found that in addition to the clinical benefits, there is the potential for cost savings of $22,000 for each veteran switched to and treated with clozapine for 1 year even when accounting for the cost of monitoring and potential adverse event management.8 This translates to a total savings of > $80 million if current utilization were doubled and half of those patients continued treatment for 1 year within the Veterans Health Administration (VHA). However, despite evidence supporting use, < 10% of Medicaid-eligible patients and only 4% of patients with schizophrenia in the VHA are prescribed clozapine.8,9
Clozapine is underutilized for a variety of reasons, including intensive monitoring requirements, potential for severe adverse drug reactions, and concern for patient adherence.8 Common adverse effects (AEs) can range from mild to severe and include weight gain, constipation, sedation, orthostatic hypotension, and excessive salivation. Clozapine also carries a boxed warning for agranulocytosis, seizures, myocarditis, other cardiovascular and respiratory AEs (including orthostatic hypotension), and increased mortality in elderly patients with dementia.
Severe agranulocytosis occurs in between 0.05% and 0.86% of patients, which led the FDA to implement a Risk Evaluation and Mitigation Strategy (REMS) program for clozapine prescribing in 2015. Prior to the REMS program, each of the 6 clozapine manufacturers were required to maintain a registry to monitor for agranulocytosis. Per the REMS program requirements, health care providers (HCPs), dispensing pharmacies, and patients must be enrolled in the program and provide an updated absolute neutrophil count (ANC) prior to prescribing or dispensing clozapine. This is potentially time consuming, particularly during the first 6 months of treatment when the ANC must be monitored weekly and prescriptions are restricted to a 7-day supply. With recent changes to the REMS program, pharmacists are no longer permitted to enroll patients in the REMS system. This adds to the administrative burden on HCPs and may decrease further the likelihood of prescribing clozapine due to lack of time for these tasks. Within the VHA, a separate entity, the VA National Clozapine Coordinating Center (NCCC), reduces the administrative burden on HCPs by monitoring laboratory values, controlling dispensing, and communicating data electronically to the FDA REMS program.10
Despite the various administrative and clinical barriers and facilitators to prescribing that exist, previous studies have found that certain organizational characteristics also may influence clozapine prescribing rates. Gören and colleagues found that utilization at VHA facilities ranged from < 5% to about 20% of patients with schizophrenia. In this study, facilities with higher utilization of clozapine were more likely to have integrated nonphysician psychiatric providers in clinics and to have clear organizational structure and processes for the treatment of severe mental illness, while facilities with lower utilization rates were less likely to have a point person for clozapine management.11
Although many national efforts have been made to increase clozapine use in recent years, no study has examined HCP perception of barriers and facilitators of clozapine use in the VHA. The objective of this study is to identify barriers and facilitators of clozapine use within the VHA as perceived by HCPs so that these may be addressed to increase appropriate utilization of clozapine in veterans with TRS.
Methods
This study was conducted as a national survey of mental health providers within the VHA who had a scope of practice that allowed clozapine prescribing. Any HCP in a solely administrative role was excluded. The survey tool was reviewed by clinical pharmacy specialists at the Lexington VA Health Care System for content and ease of administration. Following appropriate institutional review board approval, the survey was submitted to the organizational assessment subcommittee and the 5 national VA unions for approval per VA policy. The survey tool was built and administered through REDCap (Nashville, Tennessee) software. An electronic link was sent out to the national VA psychiatric pharmacist and national psychiatry chief listservs for dissemination to the psychiatric providers at each facility with weekly reminders sent out during the 4-week study period to maximize participation. The 29-item survey was developed to assess demographic information, HCP characteristics, perceived barriers and facilitators of clozapine use, and general clozapine knowledge. Knowledge-based questions included appropriate indications, starting dose, baseline ANC requirement, ANC monitoring requirements, and possible AEs.
Primary outcomes assessed were perceived barriers to clozapine prescribing, opinions of potential interventions to facilitate clozapine prescribing, knowledge regarding clozapine, and the impact of medication management clinics on clozapine prescribing. For the purposes of this study, a clozapine clinic was defined as an interdisciplinary team dedicated to clozapine prescribing and monitoring.
Secondary outcomes included a comparison of clozapine prescribing rates among different subgroups of HCPs. Subgroups included HCP discipline, geographic region, presence of academic affiliation, level of comfort or familiarity with clozapine, and percentage of time spent in direct patient care. The regional Veterans Integrated Service Networks (VISN) were used to evaluate the effect of geographic region on prescribing practices.
Results of the survey were analyzed using descriptive statistics. The Mann-Whitney U test was utilized to compare ordinal data from questions that were scored on a Likert scale, and nominal data was compared utilizing the χ2 test. For all objectives, an α of < .05 was considered significant.
Results
Ninety-eight HCPs from 17 VISNs responded during the 4-week survey period. One participant was excluded due to a solely administrative role. HCP characteristics and demographics are described in Table 1. The majority of respondents practice in an outpatient mental health setting either at the main VA campus or at a community-based outpatient clinic (CBOC).
Primary Outcomes
Perceived Barriers to Prescribing
The majority of survey respondents rated all factors listed as at least somewhat of a barrier to prescribing. Table 2 describes the perception of these various factors as barriers to clozapine prescribing. Along with prespecified variables, a free text box was available to participants to identify other perceived barriers not listed. Among other concerns listed in this text box were patient buy-in (11.3%), process/coordination of prescribing (8.2%), time restrictions (7.2%), prescriber restrictions (7.2%), access (3.1%), credentialing problems (2.1%), and lack of clear education materials (1%).
Perceived Facilitators to Prescribing
When asked to consider the potential for increased prescribing with various interventions, most participants reported that all identified facilitators would be at least somewhat likely to increase their clozapine utilization. Table 3 describes the perception of these various factors as facilitators to clozapine prescribing. Other identified facilitators included nursing or pharmacy support for follow-ups (4.1%), advanced practice registered nurse credentialing for VHA prescribing (3.1%), utilization of national REMS program without the NCCC (3.1%), outside pharmacy use during titration phase (2.1%), prespecified coverage for HCPs while on leave (1%), and increased access to specialty consults for AEs (1%).
Clozapine Knowledge Assessment
Overall, the average score on the clozapine knowledge assessment portion of the survey was 85.6%. The most commonly missed questions concerned the minimum ANC required to initiate clozapine and the appropriate starting dose for clozapine (Table 4). No significant difference was seen in clozapine utilization based on the clozapine knowledge assessment score when HCPs who scored≤ 60% were compared with those who scored ≥ 80% (P = .29).
Clozapine Clinic
No statistically significant difference was found (P = .35) when rates of prescribing between facilities with or without a dedicated clozapine clinic were compared (Table 5). Additionally, the involvement of a pharmacist in clozapine management clinics did not lead to a statistically significant difference in utilization rates (P = .45).
Secondary Outcomes
Self-rated level of comfort with clozapine prescribing was significantly associated with rates of clozapine prescribing (P < .01). HCPs who rated themselves as somewhat or very comfortable were significantly more likely to prescribe clozapine (Table 6). Providers who rated themselves as very familiar with clozapine monitoring requirements (Table 7) were significantly more likely to prescribe clozapine (P < .01). This significance remained when comparing HCPs who rated themselves as very familiar to those who ranked themselves as somewhat familiar (P = .01). There was no statistically significant difference in clozapine prescribing based on academic medical center affiliation, time spent in direct patient care, or geographic location.
Discussion
This survey targeted VHA HCPs who were licensed to prescribe clozapine to identify barriers and facilitators of use, along with HCP characteristics that may impact clozapine utilization. The findings of this study indicate that even though HCPs may perceive many legitimate barriers to clozapine prescribing, such as the frequent laboratory monitoring requirements, some factors may increase their willingness to prescribe clozapine. Many of these facilitators involve addressing logistical concerns and the administrative burden that accompanies clozapine use. These findings echo previous studies done within and outside the VHA.8,9
While some identified barriers would require national policy changes to address, others could be addressed at VHA facilities. It may be prudent for each VA facility to identify a HCP who is familiar with clozapine to serve as a subject matter expert. This would be beneficial to those HCPs who feel their patients may benefit from clozapine, but who lack experience in prescribing, or for those with concerns about appropriateness of a specific patient. Additionally, this point of contact could be a valuable resource for concerns regarding administrative issues that may arise with the laboratory reporting system. In some facilities, it may be beneficial to set aside dedicated prescriber time in a clinic designed for clozapine management. Many HCPs in this survey identified the establishment of a clozapine clinic as an intervention that would increase their likelihood of prescribing clozapine. This type of clinic may alleviate some of the concerns regarding appointment availability for weekly or bimonthly appointments early in therapy by having additional staff and time dedicated to accommodating the need for frequent visits.
The majority of respondents to this survey were concerned about the logistics of clozapine monitoring and prescribing; however, this is largely dictated by FDA and VHA policies and regulations. Per national guidance, patients within the VHA should only receive prescriptions for clozapine from their local VA facility pharmacy. It takes many veterans ≥ 1 hour to travel to the closest VA hospital or CBOC. This is especially true for facilities with largely rural catchments. These patients often lack many resources that may be present in more urban areas, such as reliable public transportation. This creates challenges for both weekly laboratory monitoring and dispensing of weekly clozapine prescriptions early in therapy. The option to get clozapine from a local non-VA pharmacy and complete laboratory monitoring at a non-VA laboratory facility could make a clozapine trial more feasible for these veterans. Another consideration is increasing the availability of VA-funded transportation for these patients to assist them in getting to their appointments. Serious mental illness case workers or mental health intensive case management services also may prove useful in arranging for transportation for laboratory monitoring.
Providers with higher self-rated comfort and familiarity with monitoring requirements had a significantly increased likelihood of clozapine utilization. Lack of experience was commonly identified as a barrier to prescribing. Subsequently, the majority of respondents felt that educational sessions would increase their likelihood to prescribe clozapine. This could be addressed at both a facility and national level. As discussed above, a subject matter expert at each facility could provide some of this education and guidance for prescribers who have little or no experience with clozapine. Additionally, national educational presentations and academic detailing campaigns may be an efficient way to provide standardized education across the VHA. Dissemination of required education via the VA Talent Management System is another potential route that would ensure all providers received adequate training regarding the specific challenges of prescribing clozapine within the VA.
Strengths and Limitations
The strengths of this study lie in directly assessing HCP perceptions of barriers and facilitators. It is ultimately up to each individual HCP to decide to use clozapine. Addressing the concerns of these HCPs will be advantageous in efforts to increase clozapine utilization. Additionally, to the authors’ knowledge this is the first study to assess provider characteristics and knowledge of clozapine in relation to utilization rates.
The method of distribution was a major limitation of this study. This survey was distributed via national e-mail listservs; however, no listserv exists within the VA that targets all psychiatric providers. This study relied on the psychiatry chiefs and psychiatric pharmacists within each facility to further disseminate the survey, which could have led to lower response rates than what may be gathered via more direct contact methods. In addition, targeting psychiatric section chiefs and pharmacists may have introduced response bias. Another limitation to this study was the small number of responses. It is possible that this study was not adequately powered to detect significant differences in clozapine prescribing based on HCP characteristics or clozapine clinic availability. Further studies investigating the impact of provider characteristics on clozapine utilization are warranted.
Conclusion
Even though clozapine is an effective medication for TRS, providers underutilize it for a variety of reasons. Commonly identified barriers to prescribing in this study included frequent monitoring requirements, logistics of prescribing (including the REMS program and transportation for laboratory monitoring), pharmacotherapy preferences, and concern about the potential AEs. Facilitators identified in this study included implementation of clozapine clinics, having a specified contact point within the facility to assist with administrative responsibility, educational sessions, and the ability to utilize outside laboratories.
While some of these barriers and facilitators cannot be fully addressed without national policy change, individual facilities should make every effort to identify institution-specific concerns and address these. Clozapine clinic implementation and educational sessions appear to be reasonable considerations. This study did not identify any HCP characteristics that significantly impacted the likelihood of prescribing clozapine aside from self-rated comfort and familiarity with clozapine. However, further studies are needed to fully assess the impact of provider characteristics on clozapine utilization.
1. Siskind D, Mccartney L, Goldschlager R, Kisely S. Clozapine v. first- and second-generation antipsychotics in treatment-refractory schizophrenia: systematic review and meta-analysis. Br J Psychiatry. 2016;209(5):385-392.
2. Lehman A, Lieberman JA, Dixon LB, et al; American Psychiatric Association; Steering Committee on Practice Guidelines. Practice guidelines for the treatment of patients with schizophrenia, second edition. Am J Psychiatry. 2004;161(2 suppl):1-56.
3. US Department of Veterans Affairs. Recommendations for antipsychotic selection in schizophrenia and schizoaffective disorders. https://www.pbm.va.gov/PBM/clinicalguidance/clinicalrecommendations/AntipsychoticSelectionAlgorithmSchizophreniaJune2012.doc. Published June 2012. Accessed September 12, 2019.
4. Dixon L, Perkins D, Calmes C. Guidelines watch (September 2009): practice guidelines for the treatment of patients with schizophrenia. https://psychiatryonline.org/pb/assets/raw/sitewide/practice_guidelines/guidelines/schizophrenia-watch.pdf. Published September 2009. Accessed September 12, 2019.
5. National Institute for Health and Care Excellence. Psychosis and schizophrenia in adults: prevention and management. https://www.nice.org.uk/guidance/cg178. Updated March 2014. Accessed September 12, 2019.
6. Meltzer HY, Alphs L, Green AI, et al; International Suicide Prevention Trial Study Group. Clozapine treatment for suicidality in schizophrenia: International Suicide Prevention Trial (InterSePT). Arch Gen Psychiatry. 2003;60(1):82-91.
7. Stroup TS, Gerhard T, Crystal S, Huang C, Olfson M. Comparative effectiveness of clozapine and standard antipsychotic treatment in adults with schizophrenia. Am J Psychiatry. 2016;173(2):166-173.
8. Gören JL, Rose AJ, Smith EG, Ney JP. The business case for expanded clozapine utilization. Psychiatr Serv. 2016;67(11):1197-1205.
9. Kelly DL, Freudenreich O, Sayer MA, Love RC. Addressing barriers to clozapine underutilization: a national effort. Psychiatr Serv. 2018;69(2):224-227.
10. US Department of Veterans Affairs. Clozapine patient management protocol (CPMP). https://www.va.gov/vhapublications/ViewPublication.asp?pub_ID=1818. Published December 23, 2008. Accessed September 12, 2019.
11. Gören JL, Rose AJ, Engle RL, et al. Organizational characteristics of Veterans Affairs clinics with high and low utilization of clozapine. Psychiatr Serv. 2016;67(11):1189-1196.
Clozapine is an atypical antipsychotic that the US Food and Drug Administration (FDA) approved for use in schizophrenia and suicidality associated with schizophrenia or schizoaffective disorder. Clozapine has been shown to be superior to other antipsychotic treatment for treatment resistant schizophrenia (TRS), which is defined as failure of 2 adequate trials of antipsychotic therapy.1 Up to 30% of patients with schizophrenia are classified as treatment resistant.2
Clozapine is considered the drug of choice for patients with TRS in both the US Department of Veterans Affairs (VA) policies and other evidence-based guidelines and remains the only antipsychotic with FDA approval for TRS.2-5 Patients treated with clozapine have fewer psychiatric hospitalizations, fewer suicide attempts, lower rates of nonadherence, and less antipsychotic polypharmacy compared with patients who are treated with other antipsychotic therapy.6,7 A 2016 study by Gören and colleagues found that in addition to the clinical benefits, there is the potential for cost savings of $22,000 for each veteran switched to and treated with clozapine for 1 year even when accounting for the cost of monitoring and potential adverse event management.8 This translates to a total savings of > $80 million if current utilization were doubled and half of those patients continued treatment for 1 year within the Veterans Health Administration (VHA). However, despite evidence supporting use, < 10% of Medicaid-eligible patients and only 4% of patients with schizophrenia in the VHA are prescribed clozapine.8,9
Clozapine is underutilized for a variety of reasons, including intensive monitoring requirements, potential for severe adverse drug reactions, and concern for patient adherence.8 Common adverse effects (AEs) can range from mild to severe and include weight gain, constipation, sedation, orthostatic hypotension, and excessive salivation. Clozapine also carries a boxed warning for agranulocytosis, seizures, myocarditis, other cardiovascular and respiratory AEs (including orthostatic hypotension), and increased mortality in elderly patients with dementia.
Severe agranulocytosis occurs in between 0.05% and 0.86% of patients, which led the FDA to implement a Risk Evaluation and Mitigation Strategy (REMS) program for clozapine prescribing in 2015. Prior to the REMS program, each of the 6 clozapine manufacturers were required to maintain a registry to monitor for agranulocytosis. Per the REMS program requirements, health care providers (HCPs), dispensing pharmacies, and patients must be enrolled in the program and provide an updated absolute neutrophil count (ANC) prior to prescribing or dispensing clozapine. This is potentially time consuming, particularly during the first 6 months of treatment when the ANC must be monitored weekly and prescriptions are restricted to a 7-day supply. With recent changes to the REMS program, pharmacists are no longer permitted to enroll patients in the REMS system. This adds to the administrative burden on HCPs and may decrease further the likelihood of prescribing clozapine due to lack of time for these tasks. Within the VHA, a separate entity, the VA National Clozapine Coordinating Center (NCCC), reduces the administrative burden on HCPs by monitoring laboratory values, controlling dispensing, and communicating data electronically to the FDA REMS program.10
Despite the various administrative and clinical barriers and facilitators to prescribing that exist, previous studies have found that certain organizational characteristics also may influence clozapine prescribing rates. Gören and colleagues found that utilization at VHA facilities ranged from < 5% to about 20% of patients with schizophrenia. In this study, facilities with higher utilization of clozapine were more likely to have integrated nonphysician psychiatric providers in clinics and to have clear organizational structure and processes for the treatment of severe mental illness, while facilities with lower utilization rates were less likely to have a point person for clozapine management.11
Although many national efforts have been made to increase clozapine use in recent years, no study has examined HCP perception of barriers and facilitators of clozapine use in the VHA. The objective of this study is to identify barriers and facilitators of clozapine use within the VHA as perceived by HCPs so that these may be addressed to increase appropriate utilization of clozapine in veterans with TRS.
Methods
This study was conducted as a national survey of mental health providers within the VHA who had a scope of practice that allowed clozapine prescribing. Any HCP in a solely administrative role was excluded. The survey tool was reviewed by clinical pharmacy specialists at the Lexington VA Health Care System for content and ease of administration. Following appropriate institutional review board approval, the survey was submitted to the organizational assessment subcommittee and the 5 national VA unions for approval per VA policy. The survey tool was built and administered through REDCap (Nashville, Tennessee) software. An electronic link was sent out to the national VA psychiatric pharmacist and national psychiatry chief listservs for dissemination to the psychiatric providers at each facility with weekly reminders sent out during the 4-week study period to maximize participation. The 29-item survey was developed to assess demographic information, HCP characteristics, perceived barriers and facilitators of clozapine use, and general clozapine knowledge. Knowledge-based questions included appropriate indications, starting dose, baseline ANC requirement, ANC monitoring requirements, and possible AEs.
Primary outcomes assessed were perceived barriers to clozapine prescribing, opinions of potential interventions to facilitate clozapine prescribing, knowledge regarding clozapine, and the impact of medication management clinics on clozapine prescribing. For the purposes of this study, a clozapine clinic was defined as an interdisciplinary team dedicated to clozapine prescribing and monitoring.
Secondary outcomes included a comparison of clozapine prescribing rates among different subgroups of HCPs. Subgroups included HCP discipline, geographic region, presence of academic affiliation, level of comfort or familiarity with clozapine, and percentage of time spent in direct patient care. The regional Veterans Integrated Service Networks (VISN) were used to evaluate the effect of geographic region on prescribing practices.
Results of the survey were analyzed using descriptive statistics. The Mann-Whitney U test was utilized to compare ordinal data from questions that were scored on a Likert scale, and nominal data was compared utilizing the χ2 test. For all objectives, an α of < .05 was considered significant.
Results
Ninety-eight HCPs from 17 VISNs responded during the 4-week survey period. One participant was excluded due to a solely administrative role. HCP characteristics and demographics are described in Table 1. The majority of respondents practice in an outpatient mental health setting either at the main VA campus or at a community-based outpatient clinic (CBOC).
Primary Outcomes
Perceived Barriers to Prescribing
The majority of survey respondents rated all factors listed as at least somewhat of a barrier to prescribing. Table 2 describes the perception of these various factors as barriers to clozapine prescribing. Along with prespecified variables, a free text box was available to participants to identify other perceived barriers not listed. Among other concerns listed in this text box were patient buy-in (11.3%), process/coordination of prescribing (8.2%), time restrictions (7.2%), prescriber restrictions (7.2%), access (3.1%), credentialing problems (2.1%), and lack of clear education materials (1%).
Perceived Facilitators to Prescribing
When asked to consider the potential for increased prescribing with various interventions, most participants reported that all identified facilitators would be at least somewhat likely to increase their clozapine utilization. Table 3 describes the perception of these various factors as facilitators to clozapine prescribing. Other identified facilitators included nursing or pharmacy support for follow-ups (4.1%), advanced practice registered nurse credentialing for VHA prescribing (3.1%), utilization of national REMS program without the NCCC (3.1%), outside pharmacy use during titration phase (2.1%), prespecified coverage for HCPs while on leave (1%), and increased access to specialty consults for AEs (1%).
Clozapine Knowledge Assessment
Overall, the average score on the clozapine knowledge assessment portion of the survey was 85.6%. The most commonly missed questions concerned the minimum ANC required to initiate clozapine and the appropriate starting dose for clozapine (Table 4). No significant difference was seen in clozapine utilization based on the clozapine knowledge assessment score when HCPs who scored≤ 60% were compared with those who scored ≥ 80% (P = .29).
Clozapine Clinic
No statistically significant difference was found (P = .35) when rates of prescribing between facilities with or without a dedicated clozapine clinic were compared (Table 5). Additionally, the involvement of a pharmacist in clozapine management clinics did not lead to a statistically significant difference in utilization rates (P = .45).
Secondary Outcomes
Self-rated level of comfort with clozapine prescribing was significantly associated with rates of clozapine prescribing (P < .01). HCPs who rated themselves as somewhat or very comfortable were significantly more likely to prescribe clozapine (Table 6). Providers who rated themselves as very familiar with clozapine monitoring requirements (Table 7) were significantly more likely to prescribe clozapine (P < .01). This significance remained when comparing HCPs who rated themselves as very familiar to those who ranked themselves as somewhat familiar (P = .01). There was no statistically significant difference in clozapine prescribing based on academic medical center affiliation, time spent in direct patient care, or geographic location.
Discussion
This survey targeted VHA HCPs who were licensed to prescribe clozapine to identify barriers and facilitators of use, along with HCP characteristics that may impact clozapine utilization. The findings of this study indicate that even though HCPs may perceive many legitimate barriers to clozapine prescribing, such as the frequent laboratory monitoring requirements, some factors may increase their willingness to prescribe clozapine. Many of these facilitators involve addressing logistical concerns and the administrative burden that accompanies clozapine use. These findings echo previous studies done within and outside the VHA.8,9
While some identified barriers would require national policy changes to address, others could be addressed at VHA facilities. It may be prudent for each VA facility to identify a HCP who is familiar with clozapine to serve as a subject matter expert. This would be beneficial to those HCPs who feel their patients may benefit from clozapine, but who lack experience in prescribing, or for those with concerns about appropriateness of a specific patient. Additionally, this point of contact could be a valuable resource for concerns regarding administrative issues that may arise with the laboratory reporting system. In some facilities, it may be beneficial to set aside dedicated prescriber time in a clinic designed for clozapine management. Many HCPs in this survey identified the establishment of a clozapine clinic as an intervention that would increase their likelihood of prescribing clozapine. This type of clinic may alleviate some of the concerns regarding appointment availability for weekly or bimonthly appointments early in therapy by having additional staff and time dedicated to accommodating the need for frequent visits.
The majority of respondents to this survey were concerned about the logistics of clozapine monitoring and prescribing; however, this is largely dictated by FDA and VHA policies and regulations. Per national guidance, patients within the VHA should only receive prescriptions for clozapine from their local VA facility pharmacy. It takes many veterans ≥ 1 hour to travel to the closest VA hospital or CBOC. This is especially true for facilities with largely rural catchments. These patients often lack many resources that may be present in more urban areas, such as reliable public transportation. This creates challenges for both weekly laboratory monitoring and dispensing of weekly clozapine prescriptions early in therapy. The option to get clozapine from a local non-VA pharmacy and complete laboratory monitoring at a non-VA laboratory facility could make a clozapine trial more feasible for these veterans. Another consideration is increasing the availability of VA-funded transportation for these patients to assist them in getting to their appointments. Serious mental illness case workers or mental health intensive case management services also may prove useful in arranging for transportation for laboratory monitoring.
Providers with higher self-rated comfort and familiarity with monitoring requirements had a significantly increased likelihood of clozapine utilization. Lack of experience was commonly identified as a barrier to prescribing. Subsequently, the majority of respondents felt that educational sessions would increase their likelihood to prescribe clozapine. This could be addressed at both a facility and national level. As discussed above, a subject matter expert at each facility could provide some of this education and guidance for prescribers who have little or no experience with clozapine. Additionally, national educational presentations and academic detailing campaigns may be an efficient way to provide standardized education across the VHA. Dissemination of required education via the VA Talent Management System is another potential route that would ensure all providers received adequate training regarding the specific challenges of prescribing clozapine within the VA.
Strengths and Limitations
The strengths of this study lie in directly assessing HCP perceptions of barriers and facilitators. It is ultimately up to each individual HCP to decide to use clozapine. Addressing the concerns of these HCPs will be advantageous in efforts to increase clozapine utilization. Additionally, to the authors’ knowledge this is the first study to assess provider characteristics and knowledge of clozapine in relation to utilization rates.
The method of distribution was a major limitation of this study. This survey was distributed via national e-mail listservs; however, no listserv exists within the VA that targets all psychiatric providers. This study relied on the psychiatry chiefs and psychiatric pharmacists within each facility to further disseminate the survey, which could have led to lower response rates than what may be gathered via more direct contact methods. In addition, targeting psychiatric section chiefs and pharmacists may have introduced response bias. Another limitation to this study was the small number of responses. It is possible that this study was not adequately powered to detect significant differences in clozapine prescribing based on HCP characteristics or clozapine clinic availability. Further studies investigating the impact of provider characteristics on clozapine utilization are warranted.
Conclusion
Even though clozapine is an effective medication for TRS, providers underutilize it for a variety of reasons. Commonly identified barriers to prescribing in this study included frequent monitoring requirements, logistics of prescribing (including the REMS program and transportation for laboratory monitoring), pharmacotherapy preferences, and concern about the potential AEs. Facilitators identified in this study included implementation of clozapine clinics, having a specified contact point within the facility to assist with administrative responsibility, educational sessions, and the ability to utilize outside laboratories.
While some of these barriers and facilitators cannot be fully addressed without national policy change, individual facilities should make every effort to identify institution-specific concerns and address these. Clozapine clinic implementation and educational sessions appear to be reasonable considerations. This study did not identify any HCP characteristics that significantly impacted the likelihood of prescribing clozapine aside from self-rated comfort and familiarity with clozapine. However, further studies are needed to fully assess the impact of provider characteristics on clozapine utilization.
Clozapine is an atypical antipsychotic that the US Food and Drug Administration (FDA) approved for use in schizophrenia and suicidality associated with schizophrenia or schizoaffective disorder. Clozapine has been shown to be superior to other antipsychotic treatment for treatment resistant schizophrenia (TRS), which is defined as failure of 2 adequate trials of antipsychotic therapy.1 Up to 30% of patients with schizophrenia are classified as treatment resistant.2
Clozapine is considered the drug of choice for patients with TRS in both the US Department of Veterans Affairs (VA) policies and other evidence-based guidelines and remains the only antipsychotic with FDA approval for TRS.2-5 Patients treated with clozapine have fewer psychiatric hospitalizations, fewer suicide attempts, lower rates of nonadherence, and less antipsychotic polypharmacy compared with patients who are treated with other antipsychotic therapy.6,7 A 2016 study by Gören and colleagues found that in addition to the clinical benefits, there is the potential for cost savings of $22,000 for each veteran switched to and treated with clozapine for 1 year even when accounting for the cost of monitoring and potential adverse event management.8 This translates to a total savings of > $80 million if current utilization were doubled and half of those patients continued treatment for 1 year within the Veterans Health Administration (VHA). However, despite evidence supporting use, < 10% of Medicaid-eligible patients and only 4% of patients with schizophrenia in the VHA are prescribed clozapine.8,9
Clozapine is underutilized for a variety of reasons, including intensive monitoring requirements, potential for severe adverse drug reactions, and concern for patient adherence.8 Common adverse effects (AEs) can range from mild to severe and include weight gain, constipation, sedation, orthostatic hypotension, and excessive salivation. Clozapine also carries a boxed warning for agranulocytosis, seizures, myocarditis, other cardiovascular and respiratory AEs (including orthostatic hypotension), and increased mortality in elderly patients with dementia.
Severe agranulocytosis occurs in between 0.05% and 0.86% of patients, which led the FDA to implement a Risk Evaluation and Mitigation Strategy (REMS) program for clozapine prescribing in 2015. Prior to the REMS program, each of the 6 clozapine manufacturers were required to maintain a registry to monitor for agranulocytosis. Per the REMS program requirements, health care providers (HCPs), dispensing pharmacies, and patients must be enrolled in the program and provide an updated absolute neutrophil count (ANC) prior to prescribing or dispensing clozapine. This is potentially time consuming, particularly during the first 6 months of treatment when the ANC must be monitored weekly and prescriptions are restricted to a 7-day supply. With recent changes to the REMS program, pharmacists are no longer permitted to enroll patients in the REMS system. This adds to the administrative burden on HCPs and may decrease further the likelihood of prescribing clozapine due to lack of time for these tasks. Within the VHA, a separate entity, the VA National Clozapine Coordinating Center (NCCC), reduces the administrative burden on HCPs by monitoring laboratory values, controlling dispensing, and communicating data electronically to the FDA REMS program.10
Despite the various administrative and clinical barriers and facilitators to prescribing that exist, previous studies have found that certain organizational characteristics also may influence clozapine prescribing rates. Gören and colleagues found that utilization at VHA facilities ranged from < 5% to about 20% of patients with schizophrenia. In this study, facilities with higher utilization of clozapine were more likely to have integrated nonphysician psychiatric providers in clinics and to have clear organizational structure and processes for the treatment of severe mental illness, while facilities with lower utilization rates were less likely to have a point person for clozapine management.11
Although many national efforts have been made to increase clozapine use in recent years, no study has examined HCP perception of barriers and facilitators of clozapine use in the VHA. The objective of this study is to identify barriers and facilitators of clozapine use within the VHA as perceived by HCPs so that these may be addressed to increase appropriate utilization of clozapine in veterans with TRS.
Methods
This study was conducted as a national survey of mental health providers within the VHA who had a scope of practice that allowed clozapine prescribing. Any HCP in a solely administrative role was excluded. The survey tool was reviewed by clinical pharmacy specialists at the Lexington VA Health Care System for content and ease of administration. Following appropriate institutional review board approval, the survey was submitted to the organizational assessment subcommittee and the 5 national VA unions for approval per VA policy. The survey tool was built and administered through REDCap (Nashville, Tennessee) software. An electronic link was sent out to the national VA psychiatric pharmacist and national psychiatry chief listservs for dissemination to the psychiatric providers at each facility with weekly reminders sent out during the 4-week study period to maximize participation. The 29-item survey was developed to assess demographic information, HCP characteristics, perceived barriers and facilitators of clozapine use, and general clozapine knowledge. Knowledge-based questions included appropriate indications, starting dose, baseline ANC requirement, ANC monitoring requirements, and possible AEs.
Primary outcomes assessed were perceived barriers to clozapine prescribing, opinions of potential interventions to facilitate clozapine prescribing, knowledge regarding clozapine, and the impact of medication management clinics on clozapine prescribing. For the purposes of this study, a clozapine clinic was defined as an interdisciplinary team dedicated to clozapine prescribing and monitoring.
Secondary outcomes included a comparison of clozapine prescribing rates among different subgroups of HCPs. Subgroups included HCP discipline, geographic region, presence of academic affiliation, level of comfort or familiarity with clozapine, and percentage of time spent in direct patient care. The regional Veterans Integrated Service Networks (VISN) were used to evaluate the effect of geographic region on prescribing practices.
Results of the survey were analyzed using descriptive statistics. The Mann-Whitney U test was utilized to compare ordinal data from questions that were scored on a Likert scale, and nominal data was compared utilizing the χ2 test. For all objectives, an α of < .05 was considered significant.
Results
Ninety-eight HCPs from 17 VISNs responded during the 4-week survey period. One participant was excluded due to a solely administrative role. HCP characteristics and demographics are described in Table 1. The majority of respondents practice in an outpatient mental health setting either at the main VA campus or at a community-based outpatient clinic (CBOC).
Primary Outcomes
Perceived Barriers to Prescribing
The majority of survey respondents rated all factors listed as at least somewhat of a barrier to prescribing. Table 2 describes the perception of these various factors as barriers to clozapine prescribing. Along with prespecified variables, a free text box was available to participants to identify other perceived barriers not listed. Among other concerns listed in this text box were patient buy-in (11.3%), process/coordination of prescribing (8.2%), time restrictions (7.2%), prescriber restrictions (7.2%), access (3.1%), credentialing problems (2.1%), and lack of clear education materials (1%).
Perceived Facilitators to Prescribing
When asked to consider the potential for increased prescribing with various interventions, most participants reported that all identified facilitators would be at least somewhat likely to increase their clozapine utilization. Table 3 describes the perception of these various factors as facilitators to clozapine prescribing. Other identified facilitators included nursing or pharmacy support for follow-ups (4.1%), advanced practice registered nurse credentialing for VHA prescribing (3.1%), utilization of national REMS program without the NCCC (3.1%), outside pharmacy use during titration phase (2.1%), prespecified coverage for HCPs while on leave (1%), and increased access to specialty consults for AEs (1%).
Clozapine Knowledge Assessment
Overall, the average score on the clozapine knowledge assessment portion of the survey was 85.6%. The most commonly missed questions concerned the minimum ANC required to initiate clozapine and the appropriate starting dose for clozapine (Table 4). No significant difference was seen in clozapine utilization based on the clozapine knowledge assessment score when HCPs who scored≤ 60% were compared with those who scored ≥ 80% (P = .29).
Clozapine Clinic
No statistically significant difference was found (P = .35) when rates of prescribing between facilities with or without a dedicated clozapine clinic were compared (Table 5). Additionally, the involvement of a pharmacist in clozapine management clinics did not lead to a statistically significant difference in utilization rates (P = .45).
Secondary Outcomes
Self-rated level of comfort with clozapine prescribing was significantly associated with rates of clozapine prescribing (P < .01). HCPs who rated themselves as somewhat or very comfortable were significantly more likely to prescribe clozapine (Table 6). Providers who rated themselves as very familiar with clozapine monitoring requirements (Table 7) were significantly more likely to prescribe clozapine (P < .01). This significance remained when comparing HCPs who rated themselves as very familiar to those who ranked themselves as somewhat familiar (P = .01). There was no statistically significant difference in clozapine prescribing based on academic medical center affiliation, time spent in direct patient care, or geographic location.
Discussion
This survey targeted VHA HCPs who were licensed to prescribe clozapine to identify barriers and facilitators of use, along with HCP characteristics that may impact clozapine utilization. The findings of this study indicate that even though HCPs may perceive many legitimate barriers to clozapine prescribing, such as the frequent laboratory monitoring requirements, some factors may increase their willingness to prescribe clozapine. Many of these facilitators involve addressing logistical concerns and the administrative burden that accompanies clozapine use. These findings echo previous studies done within and outside the VHA.8,9
While some identified barriers would require national policy changes to address, others could be addressed at VHA facilities. It may be prudent for each VA facility to identify a HCP who is familiar with clozapine to serve as a subject matter expert. This would be beneficial to those HCPs who feel their patients may benefit from clozapine, but who lack experience in prescribing, or for those with concerns about appropriateness of a specific patient. Additionally, this point of contact could be a valuable resource for concerns regarding administrative issues that may arise with the laboratory reporting system. In some facilities, it may be beneficial to set aside dedicated prescriber time in a clinic designed for clozapine management. Many HCPs in this survey identified the establishment of a clozapine clinic as an intervention that would increase their likelihood of prescribing clozapine. This type of clinic may alleviate some of the concerns regarding appointment availability for weekly or bimonthly appointments early in therapy by having additional staff and time dedicated to accommodating the need for frequent visits.
The majority of respondents to this survey were concerned about the logistics of clozapine monitoring and prescribing; however, this is largely dictated by FDA and VHA policies and regulations. Per national guidance, patients within the VHA should only receive prescriptions for clozapine from their local VA facility pharmacy. It takes many veterans ≥ 1 hour to travel to the closest VA hospital or CBOC. This is especially true for facilities with largely rural catchments. These patients often lack many resources that may be present in more urban areas, such as reliable public transportation. This creates challenges for both weekly laboratory monitoring and dispensing of weekly clozapine prescriptions early in therapy. The option to get clozapine from a local non-VA pharmacy and complete laboratory monitoring at a non-VA laboratory facility could make a clozapine trial more feasible for these veterans. Another consideration is increasing the availability of VA-funded transportation for these patients to assist them in getting to their appointments. Serious mental illness case workers or mental health intensive case management services also may prove useful in arranging for transportation for laboratory monitoring.
Providers with higher self-rated comfort and familiarity with monitoring requirements had a significantly increased likelihood of clozapine utilization. Lack of experience was commonly identified as a barrier to prescribing. Subsequently, the majority of respondents felt that educational sessions would increase their likelihood to prescribe clozapine. This could be addressed at both a facility and national level. As discussed above, a subject matter expert at each facility could provide some of this education and guidance for prescribers who have little or no experience with clozapine. Additionally, national educational presentations and academic detailing campaigns may be an efficient way to provide standardized education across the VHA. Dissemination of required education via the VA Talent Management System is another potential route that would ensure all providers received adequate training regarding the specific challenges of prescribing clozapine within the VA.
Strengths and Limitations
The strengths of this study lie in directly assessing HCP perceptions of barriers and facilitators. It is ultimately up to each individual HCP to decide to use clozapine. Addressing the concerns of these HCPs will be advantageous in efforts to increase clozapine utilization. Additionally, to the authors’ knowledge this is the first study to assess provider characteristics and knowledge of clozapine in relation to utilization rates.
The method of distribution was a major limitation of this study. This survey was distributed via national e-mail listservs; however, no listserv exists within the VA that targets all psychiatric providers. This study relied on the psychiatry chiefs and psychiatric pharmacists within each facility to further disseminate the survey, which could have led to lower response rates than what may be gathered via more direct contact methods. In addition, targeting psychiatric section chiefs and pharmacists may have introduced response bias. Another limitation to this study was the small number of responses. It is possible that this study was not adequately powered to detect significant differences in clozapine prescribing based on HCP characteristics or clozapine clinic availability. Further studies investigating the impact of provider characteristics on clozapine utilization are warranted.
Conclusion
Even though clozapine is an effective medication for TRS, providers underutilize it for a variety of reasons. Commonly identified barriers to prescribing in this study included frequent monitoring requirements, logistics of prescribing (including the REMS program and transportation for laboratory monitoring), pharmacotherapy preferences, and concern about the potential AEs. Facilitators identified in this study included implementation of clozapine clinics, having a specified contact point within the facility to assist with administrative responsibility, educational sessions, and the ability to utilize outside laboratories.
While some of these barriers and facilitators cannot be fully addressed without national policy change, individual facilities should make every effort to identify institution-specific concerns and address these. Clozapine clinic implementation and educational sessions appear to be reasonable considerations. This study did not identify any HCP characteristics that significantly impacted the likelihood of prescribing clozapine aside from self-rated comfort and familiarity with clozapine. However, further studies are needed to fully assess the impact of provider characteristics on clozapine utilization.
1. Siskind D, Mccartney L, Goldschlager R, Kisely S. Clozapine v. first- and second-generation antipsychotics in treatment-refractory schizophrenia: systematic review and meta-analysis. Br J Psychiatry. 2016;209(5):385-392.
2. Lehman A, Lieberman JA, Dixon LB, et al; American Psychiatric Association; Steering Committee on Practice Guidelines. Practice guidelines for the treatment of patients with schizophrenia, second edition. Am J Psychiatry. 2004;161(2 suppl):1-56.
3. US Department of Veterans Affairs. Recommendations for antipsychotic selection in schizophrenia and schizoaffective disorders. https://www.pbm.va.gov/PBM/clinicalguidance/clinicalrecommendations/AntipsychoticSelectionAlgorithmSchizophreniaJune2012.doc. Published June 2012. Accessed September 12, 2019.
4. Dixon L, Perkins D, Calmes C. Guidelines watch (September 2009): practice guidelines for the treatment of patients with schizophrenia. https://psychiatryonline.org/pb/assets/raw/sitewide/practice_guidelines/guidelines/schizophrenia-watch.pdf. Published September 2009. Accessed September 12, 2019.
5. National Institute for Health and Care Excellence. Psychosis and schizophrenia in adults: prevention and management. https://www.nice.org.uk/guidance/cg178. Updated March 2014. Accessed September 12, 2019.
6. Meltzer HY, Alphs L, Green AI, et al; International Suicide Prevention Trial Study Group. Clozapine treatment for suicidality in schizophrenia: International Suicide Prevention Trial (InterSePT). Arch Gen Psychiatry. 2003;60(1):82-91.
7. Stroup TS, Gerhard T, Crystal S, Huang C, Olfson M. Comparative effectiveness of clozapine and standard antipsychotic treatment in adults with schizophrenia. Am J Psychiatry. 2016;173(2):166-173.
8. Gören JL, Rose AJ, Smith EG, Ney JP. The business case for expanded clozapine utilization. Psychiatr Serv. 2016;67(11):1197-1205.
9. Kelly DL, Freudenreich O, Sayer MA, Love RC. Addressing barriers to clozapine underutilization: a national effort. Psychiatr Serv. 2018;69(2):224-227.
10. US Department of Veterans Affairs. Clozapine patient management protocol (CPMP). https://www.va.gov/vhapublications/ViewPublication.asp?pub_ID=1818. Published December 23, 2008. Accessed September 12, 2019.
11. Gören JL, Rose AJ, Engle RL, et al. Organizational characteristics of Veterans Affairs clinics with high and low utilization of clozapine. Psychiatr Serv. 2016;67(11):1189-1196.
1. Siskind D, Mccartney L, Goldschlager R, Kisely S. Clozapine v. first- and second-generation antipsychotics in treatment-refractory schizophrenia: systematic review and meta-analysis. Br J Psychiatry. 2016;209(5):385-392.
2. Lehman A, Lieberman JA, Dixon LB, et al; American Psychiatric Association; Steering Committee on Practice Guidelines. Practice guidelines for the treatment of patients with schizophrenia, second edition. Am J Psychiatry. 2004;161(2 suppl):1-56.
3. US Department of Veterans Affairs. Recommendations for antipsychotic selection in schizophrenia and schizoaffective disorders. https://www.pbm.va.gov/PBM/clinicalguidance/clinicalrecommendations/AntipsychoticSelectionAlgorithmSchizophreniaJune2012.doc. Published June 2012. Accessed September 12, 2019.
4. Dixon L, Perkins D, Calmes C. Guidelines watch (September 2009): practice guidelines for the treatment of patients with schizophrenia. https://psychiatryonline.org/pb/assets/raw/sitewide/practice_guidelines/guidelines/schizophrenia-watch.pdf. Published September 2009. Accessed September 12, 2019.
5. National Institute for Health and Care Excellence. Psychosis and schizophrenia in adults: prevention and management. https://www.nice.org.uk/guidance/cg178. Updated March 2014. Accessed September 12, 2019.
6. Meltzer HY, Alphs L, Green AI, et al; International Suicide Prevention Trial Study Group. Clozapine treatment for suicidality in schizophrenia: International Suicide Prevention Trial (InterSePT). Arch Gen Psychiatry. 2003;60(1):82-91.
7. Stroup TS, Gerhard T, Crystal S, Huang C, Olfson M. Comparative effectiveness of clozapine and standard antipsychotic treatment in adults with schizophrenia. Am J Psychiatry. 2016;173(2):166-173.
8. Gören JL, Rose AJ, Smith EG, Ney JP. The business case for expanded clozapine utilization. Psychiatr Serv. 2016;67(11):1197-1205.
9. Kelly DL, Freudenreich O, Sayer MA, Love RC. Addressing barriers to clozapine underutilization: a national effort. Psychiatr Serv. 2018;69(2):224-227.
10. US Department of Veterans Affairs. Clozapine patient management protocol (CPMP). https://www.va.gov/vhapublications/ViewPublication.asp?pub_ID=1818. Published December 23, 2008. Accessed September 12, 2019.
11. Gören JL, Rose AJ, Engle RL, et al. Organizational characteristics of Veterans Affairs clinics with high and low utilization of clozapine. Psychiatr Serv. 2016;67(11):1189-1196.
Assessing Refill Data Among Different Classes of Antidepressants (FULL)
Depression affects about 4.4% of the global population.1 Major depressive disorder (MDD) is currently the fourth highest cause of disability in the world and by 2030 MDD is expected to be third.2 Research has determined that 1 in 3 veterans seen in primary care shows depressive symptoms. Of these, 1 in 5 have symptoms severe enough to warrant further evaluation for MDD, and 1 in 10 require treatment.3 With this high rate of depression, optimized treatment strategies are needed, including antidepressants and psychotherapy. Antidepressants have grown in popularity since market entry in the 1950s; currently 1 in 10 US citizens aged ≥ 12 years are prescribed an antidepressant.4
Antidepressant Adherence
Antidepressant adherence is crucial for response and remission. Sansone and Sansone reported that, on average, < 50% of patients are adherent to their antidepressant treatment regimen 6 months after initiation (range, 5.4% - 87.6%).5 Fortney and colleagues found that, based on patient report, < 20% of veterans maintained at least 80% adherence at 6 months.6 Patients who are nonadherent are at an increased risk for relapse and recurrence and are more likely to seek care at an emergency department or to become hospitalized.2 In addition to the negative impact on patient outcomes, antidepressant nonadherence may also result in increased economic burden. In the US alone, the annual cost of treating MDD exceeds $210 billion, which will continue to increase if nonadherence is not mitigated.1
Patient-specific characteristics such as lack of knowledge about proper administration techniques, misguided beliefs, and negative attitudes towards treatment may affect adherence.5 In the veteran population, reasons for discontinuation also include lack of perceived benefit and adverse effects, specifically sexual difficulties.6 Sociodemographic and other patient characteristics also may be risk factors for nonadherence, including multiple medical comorbidities; substance use disorder (SUD) diagnosis; male gender; younger age; lack of health insurance or a higher medical cost burden; lack of or low involvement in psychotherapy; infrequent follow up visits; and high illness severity.1,7,8
Appreciating the adherence rates among the different antidepressant classes may help in antidepressant selection. To our knowledge, there have been no prior studies conducted in the veteran population that compared adherence rates among antidepressant classes. Studies in the nonveteran population report differing adherence rates among the antidepressant classes with generally higher adherence in patients prescribed serotonin norepinephrine reuptake inhibitors (SNRIs) and selective serotonin reuptake inhibitors (SSRIs). A retrospective review of commercial, Medicare, and Medicaid claims in > 5000 patients found that SNRIs had a significantly higher 3-month adherence rate based on the portion of days covered model (47%; P < .001) than other antidepressant classes (SSRIs, 42%; other antidepressants, 37%; tricyclic antidepressants [TCAs], 24%).7 Monoamine oxidase inhibitors (MAOIs) prescribed to 1% of the study population had the highest adherence rate at 48%.7 A study reviewing > 25 000 patient claims sourced from the IBM MarketScan research database (Armonk, NY) found that SSRIs (Odds ratio [OR], 1.26; P < .001) and norepinephrine dopamine reuptake inhibitors (NDRIs) (OR, 1.23; P = .007) had the highest ORs for adherence according to the portion of days covered model, while other serotonin modulators (OR, 0.65; P = .001) and tri/tetracyclic antidepressants (OR, 0.49; P < . 001) had the lowest ORs and were associated with lower adherence.1
VA Approaches to Adherence
To address antidepressant adherence, the US Department of Veteran Affairs (VA) adopted 2 measures from the Healthcare Effectiveness Data and Information Set: MDD43h and MDD47h. Measure MDD43h is defined as the proportion of patients with a depression diagnosis newly treated with an antidepressant medication who remained on the antidepressant medication for at least 84 out of 114 days (3 months). MDD47h is similar, but assesses patients remaining on an antidepressant medication for at least 180 out of 230 days (6 months).9 These constitute a SAIL (Strategic Analytics for Improvement and Learning) measure by which VA hospitals are compared. High performance on these measures aids in improving the comparative status of a VA facility.
To help improve performance on these measures, the VA Psychotropic Drug Safety Initiative developed the Antidepressant Nonadherence Report, which serves as a case finder for clinicians to identify veterans with low adherence and/or those overdue for a refill. The dashboard uses the medication possession ratio (MPR) to calculate adherence. While the optimal value is still widely debated, an MPR of ≥ 80% is generally accepted for many disease states.10 The dashboard defines low adherence as ≤ 60%.
As of September 2018, the Antidepressant Nonadherence Report for the Michael E. DeBakey VA Medical Center (MEDVAMC) in Houston, Texas, included > 5000 patients in both MEDVAMC and associated community-based outpatient clinics. About 30% of patients were categorized as overdue for a refill.
Study Objectives
To better understand the problem of antidepressant adherence within this population, we decided to study the relationship between antidepressant class and adherence rates, as well as how adherence relates to patient-specific characteristics. By highlighting predisposing risk factors to low adherence, we hope to provide better interventions.
The primary objective of this study was to determine whether 3-month adherence rates, measured by the MPR, differ between antidepressant classes in veterans newly initiated on antidepressant therapy. A secondary objective was to identify whether there are differences in patient characteristics between those with high MPR (≥ 80%) and low MPR (≤ 60%).
Methods
This study used a retrospective, cross-sectional chart review of MEDVAMC patients from the Antidepressant Nonadherence Report. Patients were: aged ≥ 18 years; newly initiated on an antidepressant with no previous use of the same medication; outpatient for the entire study period; and seen by a physician, physician assistant, nurse practitioner, or pharmacist mental health provider (MHP) within the 3-month study period. All patients’ charts showed a depression diagnosis—an inclusion criterion for the MDD43h and MDD47h measures. However, for this study, the indication(s) for the chosen antidepressant were determined by the MHP note in the patient electronic health record on the date that the medication was prescribed. Study patients may not have had a current depression diagnosis based upon the MHP assessment on the index date. We chose to determine the antidepressant indication(s) in this way because the MHP note would have the most detailed patient assessment.
Patients with previous use of the prescribed antidepressant were excluded because previous exposure may bias the patient and affect current adherence. Patients who were hospitalized at the VA for any reason during the 3-month study period were excluded because of a known risk during transitions of care for medications to be held or discontinued, which could impact refills and MPR. Some patients were excluded if they were taking the antidepressant for a nonmood-related indication (insomnia, neuropathy, migraine prophylaxis, etc). Patients also were excluded if the antidepressant was prescribed to take as-needed; if trazodone was the only antidepressant prescribed; if they were diagnosed with cognitive impairment including dementia or history of stroke; or if they were diagnosed with schizophrenia, schizoaffective disorder, or borderline personality disorder. Use of trazodone as the only antidepressant was excluded because of the relatively common practice to use it in the treatment of insomnia rather than depression.
Primary and Secondary Outcomes
Information collected for the primary outcome, including antidepressant class and MPR, was obtained from the Antidepressant Nonadherence Report. For the secondary outcome, the following data was collected for each patient: age, gender, race, housing status, Medication Regimen Complexity Index (MRCI), number and type of psychiatric diagnoses, number of previous antidepressants, psychotherapy involvement, and number of mental health visits during the 3-month study period. The MRCI is an objective, validated tool that determines relative medication regimen complexity by taking into consideration the number of medications, route and frequency of administration, splitting/multiple dosage units, and presence of any special instructions.11
The primary outcome was tested using a one-way analysis of variance (ANOVA). Nominal secondary outcomes were analyzed using the Fisher’s Exact. Continuous secondary outcomes were examined using an unpaired t-test.
Results
Of 320 charts, 212 patients were excluded and 108 were included (Figure). The most common reason for exclusion was a previously prescribed antidepressant. Of the included patients 49 had an MPR ≥ 80% and 24 had an MPR ≤ 60%. The characteristics of the study population are found in Table 1 and the antidepressant frequencies and MPRs are included in Table 2.
About 87% of study patients had a diagnosis of depression. Other concomitant psychiatric diagnoses include posttraumatic stress disorder (PTSD), anxiety, insomnia, and 2 cases of intermittent explosive disorder. There were no significant differences in mean MPR between the antidepressant classes (P = .31). Within each drug class, we identified the proportion of patients with high adherence (MPR ≥ 80%). Bupropion had the greatest percentage of highly adherent patients (50%) compared with SSRIs (42.5%), SNRIs (38.5%), and mirtazapine (31.3%).
Table 3 compares the characteristics between high MPR and low MPR patients. The low MPR group showed a significantly greater proportion of patients with an SUD than the high adherence group (41.7% vs 10.2%, respectively; P = .04). The most common type of SUD was alcohol use disorder followed by cannabis use disorder. There were no other statistically significant differences identified between high and low MPR groups. There was a trend towards significance when comparing MRCI between the 2 groups (high MPR, 15.2; low MPR, 10.8; P = .06).
Discussion
In our study, there was no significant difference in 3-month adherence rates between veterans on SSRIs, SNRIs, bupropion, and mirtazapine. This result differs from a study by Keyloun and colleagues that found that SNRIs had a significantly higher adherence rate when compared with other antidepressants.7
SSRIs were the most commonly prescribed antidepressant in our study, and also had the greatest mean 3-month MPR. The high use of SSRIs may be due to the greater number of SSRI choices to select from compared with other classes. SSRIs may also have been selected more frequently because nearly half (45.4%) of the patients had comorbid PTSD, for which 3 of the 4 first-line treatment options are SSRIs (sertraline, paroxetine, fluoxetine).
As previously stated, Keyloun and colleagues previously found that SNRIs had the highest 3-month adherence rate in a study of > 5000 patients.7 In our study, SNRIs had the second highest mean 3-month MPR at about 75%, but the difference was not considered significant when compared with other antidepressant classes.
Bupropion was prescribed least frequently, but had the largest proportion of adherent patients. Gaspar and colleagues demonstrated similar outcomes, reporting that patients prescribed bupropion had a high OR for adherence.1 Bupropion may have had relatively low prescribing rates in our study because 64% of patients were diagnosed with a comorbid anxiety disorder and/or PTSD. For these patients, bupropion avoidance may have been intentional so as to not exacerbate anxiety.
Mirtazapine had both the lowest mean MPR and the lowest proportion of adherent patients. While no significant difference between antidepressant 3-month adherence rates were found, this study’s findings were similar to previous studies that found lower adherence to mirtazapine.1,5 Adverse effects such as sedation, increased appetite, and weight gain may have contributed to low adherence with mirtazapine.4 Patients may also have been using the agent on an as needed basis to treat insomnia despite the order being written for daily use.
Substance Use Disorder Influence
A significantly greater proportion of patients had an SUD in the low MPR group, suggesting that an SUD diagnosis may be a risk factor for low adherence. This finding is consistent with previous studies that also found that an SUD was associated with poor medication adherence.1 Patients with depression and an SUD have been shown to have suboptimal outcomes compared to those without an SUD, including a lower response to antidepressant therapy and increased illness severity.11,12
In a study of 131 outpatients with dual diagnosis (26% with depression) predictors for low self-reported adherence were a medication-related variable (increased adverse effects), a cognitive variable (low self-efficacy for drug avoidance), and a social factor (low social support for recovery). This variety of predictors seems to indicate that simple memory aids may not improve adherence. “Dual focus” mutual aid groups that provide social support for patients with dual diagnosis have been shown to improve adherence.13
The MEDVAMC Substance Dependence Treatment Program (SDTP) is an outpatient program that uses group education to aid veterans, often those with comorbid psychiatric disorders, to build relapse prevention skills and provide social support. Further exploration into the relationship between involvement in SDTP groups and antidepressant adherence in patients with dual diagnosis may be warranted.
Secondary Outcomes
Trends identified in the secondary outcome were similar to outcomes of previous studies: younger age, lower therapy involvement, and more comorbid psychiatric diagnoses were associated with lower adherence.1,7,8 The presence of increased previous use of antidepressants in the low adherence group may suggest that these patients have an increased illness severity, although objective scales, such as the Patient Health Questionnaire 9 (PHQ9), were not consistently conducted and therefore not included in this analysis. It is unknown whether the previous antidepressant prescriptions were of adequate duration. These patients may have also had intolerances that led to multiple different antidepressant prescriptions and self-discontinuation.
The average MRCI of study patients was 13.5 (range 2 - 53), which was significantly lower than a previous study of geriatric patients with depression reporting an average MRCI of 25.4 (range 6 - 64).14 The positive trend between MRCI and adherence seen in this study was puzzling and counterintuitive. A more complex regimen is generally thought to be associated with poor adherence. Patients with a greater number of comorbid conditions may inherently be on more medications and thus have a more complex medication regimen. Manzano-Garcia and colleagues identified a negative relationship between adherence and the number of comorbidities (OR, 1.04-1.57; P = .021) and the MRCI (OR, 1.14-1.26; P < .001) in patients with HIV.15 Further studies are needed to clarify the relationship between medication adherence and medication regimen complexity in patients with mental health disorders. A better understanding of this relationship could possibly facilitate improved individualized prescribing practices and follow-up.
Limitations
Findings from our study should be interpreted within several limitations. Generalizability and statistical power were limited due to the small sample size, a practice site limited to 1 facility, and population type. The retrospective design of the study introduces inherent bias that would be minimized had a prospective study been conducted. The primary outcome was based upon MPR, which only accounts for refills within a specified time period and does not assess for actual or accurate use of the medication. Data collection was limited to VA and US Department of Defense records.
Geographically diverse studies with larger sample sizes need to be conducted to better understand antidepressant adherence and its barriers and facilitators in the veteran population. The exclusion of patients with previous trials of the prescribed antidepressant may have led to a possible selection bias favoring inclusion of younger patients. These patients may have a more limited period for assessment and treatment when compared with older patients, and thus may have had a smaller chance of previous exposure to the prescribed antidepressant. Neither MAOIs or TCAs were included in this study. No patients taking MAOIs were identified from the Antidepressant Nonadherence Report during the study period. Three patients on TCAs were chart reviewed, but excluded from the study because of prior use of the antidepressant or a non-mental health indication. Additionally, no newer antidepressants, including vortioxetine and vilazodone, were included, likely secondary to their nonformulary status at the VA.
Conclusion
As this study’s purpose was to improve the quality of care at our facility, we will discuss our findings with local MHPs to develop strategies to improve antidepressant adherence. While larger studies need to be conducted to confirm our findings, it is worthwhile to consider risk factors for low adherence such as SUD when prescribing antidepressant medications. Patients with SUD could be encouraged to enroll in our facility’s telephone nursing depression care management program for more frequent follow up and medication adherence counseling.
This study did not find a significant difference in 3-month adherence rates between SSRIs, SNRIs, bupropion, and mirtazapine. SUD was significantly more common in patients with low adherence than those categorized as adherent and may be a risk factor for low adherence based upon our findings and those of previous studies.
1. Gaspar FW, Zaidel CS, Dewa CS. Rates and determinants of use of pharmacotherapy and psychotherapy by patients with major depressive disorder. Psychiatr Serv. 2019;70(4):262-270.
2. Ho SC, Jacob SA, Tangiisuran B. Barriers and facilitators of adherence to antidepressants among outpatients with major depressive disorder: a qualitative study. PLoS One. 2017;12(6):e0179290.
3. US Department of Veterans Affairs, Office of Research and Development. VA research on: depression. https://www.research.va.gov/topics/depression.cfm#research1. Accessed May 30, 2019.
4. Santarsieri D, Schwartz TL. Antidepressant efficacy and side-effect burden: a quick guide for clinicians. Drugs Context. 2015;4:212290.
5. Sansone RA, Sansone LA. Antidepressant adherence: are patients taking their medications? Innov Clin Neurosci. 2012;9(5-6):41-46.
6. Fortney JC, Pyne JM, Edlund MJ, et al. Reasons for antidepressant nonadherence among veterans treated in primary care clinics. J Clin Psychiatry. 2011;72(6):827-834.
7. Keyloun KR, Hansen RN, Hepp Z, Gillard P, Thase ME, Devine EB. Adherence and persistence across antidepressant therapeutic classes: a retrospective claims analysis among insured US patients with major depressive disorder (MDD). [erratum: CNS Drugs. 2017;31(6):511.] CNS Drugs. 2017;31(5):421-432.
8. Mcinnis MG. Adherence to treatment regimens in major depression: perspectives, problems, and progress. https://www.psychiatrictimes.com/depression/adherence-treatment-regimens-major-depression-perspectives-problems-and-progress. Published September 15, 2007. Accessed September 10, 2019.
9. US Department of Veterans Affairs, Office of Mental Health Operations. Clinical support portal. User Guide – antidepressant non-adherence report (MDD43h MDD47h). https://spsites.cdw.va.gov/sites/OMHO_PsychPharm/_layouts/15/WopiFrame.aspx?sourcedoc=/sites/OMHO_PsychPharm/AnalyticsReports/UserGuideMDD43H47H.pdf. Accessed July 29, 2018. [Nonpublic site]
10. Crowe M. Do you know the difference between these adherence measures? https://www.pharmacytimes.com/contributor/michael-crowe-pharmd-mba-csp-fmpa/2015/07/do-you-know-the-difference-between-these-adherence-measures. Published July 5, 2015. Accessed September 13, 2019.
11. Watkins KE, Paddock SM, Zhang L, Wells KB. Improving care for depression in patients with comorbid substance misuse. Am J Psychiatry. 2006;163(1):125-132.
12. Magura S, Rosenblum A, Fong C. Factors associated with medication adherence among psychiatric outpatients at substance abuse risk. Open Addict J. 2011;4:58-64.
13. Magura S, Rosenblum A, Villano CL, Vogel HS, Fong C, Betzler T. Dual-focus mutual aid for co-occurring disorders: a quasi-experimental outcome evaluation study. Am J Drug Alcohol Abuse. 2008;34(1):61-74.
14. Libby AM, Fish DN, Hosokawa PW, et al. Patient-level medication regimen complexity across populations with chronic disease. Clin Ther. 2013;35(4):385-398.e1.
15. Manzano-García M, Pérez-Guerrero C, Álvarez de Sotomayor Paz M, Robustillo-Cortés MLA, Almeida-González CV, Morillo-Verdugo R. Identification of the medication regimen complexity index as an associated factor of nonadherence to antiretroviral treatment in HIV positive patients. Ann Pharmacother. 2018;52(9):862-867.
Depression affects about 4.4% of the global population.1 Major depressive disorder (MDD) is currently the fourth highest cause of disability in the world and by 2030 MDD is expected to be third.2 Research has determined that 1 in 3 veterans seen in primary care shows depressive symptoms. Of these, 1 in 5 have symptoms severe enough to warrant further evaluation for MDD, and 1 in 10 require treatment.3 With this high rate of depression, optimized treatment strategies are needed, including antidepressants and psychotherapy. Antidepressants have grown in popularity since market entry in the 1950s; currently 1 in 10 US citizens aged ≥ 12 years are prescribed an antidepressant.4
Antidepressant Adherence
Antidepressant adherence is crucial for response and remission. Sansone and Sansone reported that, on average, < 50% of patients are adherent to their antidepressant treatment regimen 6 months after initiation (range, 5.4% - 87.6%).5 Fortney and colleagues found that, based on patient report, < 20% of veterans maintained at least 80% adherence at 6 months.6 Patients who are nonadherent are at an increased risk for relapse and recurrence and are more likely to seek care at an emergency department or to become hospitalized.2 In addition to the negative impact on patient outcomes, antidepressant nonadherence may also result in increased economic burden. In the US alone, the annual cost of treating MDD exceeds $210 billion, which will continue to increase if nonadherence is not mitigated.1
Patient-specific characteristics such as lack of knowledge about proper administration techniques, misguided beliefs, and negative attitudes towards treatment may affect adherence.5 In the veteran population, reasons for discontinuation also include lack of perceived benefit and adverse effects, specifically sexual difficulties.6 Sociodemographic and other patient characteristics also may be risk factors for nonadherence, including multiple medical comorbidities; substance use disorder (SUD) diagnosis; male gender; younger age; lack of health insurance or a higher medical cost burden; lack of or low involvement in psychotherapy; infrequent follow up visits; and high illness severity.1,7,8
Appreciating the adherence rates among the different antidepressant classes may help in antidepressant selection. To our knowledge, there have been no prior studies conducted in the veteran population that compared adherence rates among antidepressant classes. Studies in the nonveteran population report differing adherence rates among the antidepressant classes with generally higher adherence in patients prescribed serotonin norepinephrine reuptake inhibitors (SNRIs) and selective serotonin reuptake inhibitors (SSRIs). A retrospective review of commercial, Medicare, and Medicaid claims in > 5000 patients found that SNRIs had a significantly higher 3-month adherence rate based on the portion of days covered model (47%; P < .001) than other antidepressant classes (SSRIs, 42%; other antidepressants, 37%; tricyclic antidepressants [TCAs], 24%).7 Monoamine oxidase inhibitors (MAOIs) prescribed to 1% of the study population had the highest adherence rate at 48%.7 A study reviewing > 25 000 patient claims sourced from the IBM MarketScan research database (Armonk, NY) found that SSRIs (Odds ratio [OR], 1.26; P < .001) and norepinephrine dopamine reuptake inhibitors (NDRIs) (OR, 1.23; P = .007) had the highest ORs for adherence according to the portion of days covered model, while other serotonin modulators (OR, 0.65; P = .001) and tri/tetracyclic antidepressants (OR, 0.49; P < . 001) had the lowest ORs and were associated with lower adherence.1
VA Approaches to Adherence
To address antidepressant adherence, the US Department of Veteran Affairs (VA) adopted 2 measures from the Healthcare Effectiveness Data and Information Set: MDD43h and MDD47h. Measure MDD43h is defined as the proportion of patients with a depression diagnosis newly treated with an antidepressant medication who remained on the antidepressant medication for at least 84 out of 114 days (3 months). MDD47h is similar, but assesses patients remaining on an antidepressant medication for at least 180 out of 230 days (6 months).9 These constitute a SAIL (Strategic Analytics for Improvement and Learning) measure by which VA hospitals are compared. High performance on these measures aids in improving the comparative status of a VA facility.
To help improve performance on these measures, the VA Psychotropic Drug Safety Initiative developed the Antidepressant Nonadherence Report, which serves as a case finder for clinicians to identify veterans with low adherence and/or those overdue for a refill. The dashboard uses the medication possession ratio (MPR) to calculate adherence. While the optimal value is still widely debated, an MPR of ≥ 80% is generally accepted for many disease states.10 The dashboard defines low adherence as ≤ 60%.
As of September 2018, the Antidepressant Nonadherence Report for the Michael E. DeBakey VA Medical Center (MEDVAMC) in Houston, Texas, included > 5000 patients in both MEDVAMC and associated community-based outpatient clinics. About 30% of patients were categorized as overdue for a refill.
Study Objectives
To better understand the problem of antidepressant adherence within this population, we decided to study the relationship between antidepressant class and adherence rates, as well as how adherence relates to patient-specific characteristics. By highlighting predisposing risk factors to low adherence, we hope to provide better interventions.
The primary objective of this study was to determine whether 3-month adherence rates, measured by the MPR, differ between antidepressant classes in veterans newly initiated on antidepressant therapy. A secondary objective was to identify whether there are differences in patient characteristics between those with high MPR (≥ 80%) and low MPR (≤ 60%).
Methods
This study used a retrospective, cross-sectional chart review of MEDVAMC patients from the Antidepressant Nonadherence Report. Patients were: aged ≥ 18 years; newly initiated on an antidepressant with no previous use of the same medication; outpatient for the entire study period; and seen by a physician, physician assistant, nurse practitioner, or pharmacist mental health provider (MHP) within the 3-month study period. All patients’ charts showed a depression diagnosis—an inclusion criterion for the MDD43h and MDD47h measures. However, for this study, the indication(s) for the chosen antidepressant were determined by the MHP note in the patient electronic health record on the date that the medication was prescribed. Study patients may not have had a current depression diagnosis based upon the MHP assessment on the index date. We chose to determine the antidepressant indication(s) in this way because the MHP note would have the most detailed patient assessment.
Patients with previous use of the prescribed antidepressant were excluded because previous exposure may bias the patient and affect current adherence. Patients who were hospitalized at the VA for any reason during the 3-month study period were excluded because of a known risk during transitions of care for medications to be held or discontinued, which could impact refills and MPR. Some patients were excluded if they were taking the antidepressant for a nonmood-related indication (insomnia, neuropathy, migraine prophylaxis, etc). Patients also were excluded if the antidepressant was prescribed to take as-needed; if trazodone was the only antidepressant prescribed; if they were diagnosed with cognitive impairment including dementia or history of stroke; or if they were diagnosed with schizophrenia, schizoaffective disorder, or borderline personality disorder. Use of trazodone as the only antidepressant was excluded because of the relatively common practice to use it in the treatment of insomnia rather than depression.
Primary and Secondary Outcomes
Information collected for the primary outcome, including antidepressant class and MPR, was obtained from the Antidepressant Nonadherence Report. For the secondary outcome, the following data was collected for each patient: age, gender, race, housing status, Medication Regimen Complexity Index (MRCI), number and type of psychiatric diagnoses, number of previous antidepressants, psychotherapy involvement, and number of mental health visits during the 3-month study period. The MRCI is an objective, validated tool that determines relative medication regimen complexity by taking into consideration the number of medications, route and frequency of administration, splitting/multiple dosage units, and presence of any special instructions.11
The primary outcome was tested using a one-way analysis of variance (ANOVA). Nominal secondary outcomes were analyzed using the Fisher’s Exact. Continuous secondary outcomes were examined using an unpaired t-test.
Results
Of 320 charts, 212 patients were excluded and 108 were included (Figure). The most common reason for exclusion was a previously prescribed antidepressant. Of the included patients 49 had an MPR ≥ 80% and 24 had an MPR ≤ 60%. The characteristics of the study population are found in Table 1 and the antidepressant frequencies and MPRs are included in Table 2.
About 87% of study patients had a diagnosis of depression. Other concomitant psychiatric diagnoses include posttraumatic stress disorder (PTSD), anxiety, insomnia, and 2 cases of intermittent explosive disorder. There were no significant differences in mean MPR between the antidepressant classes (P = .31). Within each drug class, we identified the proportion of patients with high adherence (MPR ≥ 80%). Bupropion had the greatest percentage of highly adherent patients (50%) compared with SSRIs (42.5%), SNRIs (38.5%), and mirtazapine (31.3%).
Table 3 compares the characteristics between high MPR and low MPR patients. The low MPR group showed a significantly greater proportion of patients with an SUD than the high adherence group (41.7% vs 10.2%, respectively; P = .04). The most common type of SUD was alcohol use disorder followed by cannabis use disorder. There were no other statistically significant differences identified between high and low MPR groups. There was a trend towards significance when comparing MRCI between the 2 groups (high MPR, 15.2; low MPR, 10.8; P = .06).
Discussion
In our study, there was no significant difference in 3-month adherence rates between veterans on SSRIs, SNRIs, bupropion, and mirtazapine. This result differs from a study by Keyloun and colleagues that found that SNRIs had a significantly higher adherence rate when compared with other antidepressants.7
SSRIs were the most commonly prescribed antidepressant in our study, and also had the greatest mean 3-month MPR. The high use of SSRIs may be due to the greater number of SSRI choices to select from compared with other classes. SSRIs may also have been selected more frequently because nearly half (45.4%) of the patients had comorbid PTSD, for which 3 of the 4 first-line treatment options are SSRIs (sertraline, paroxetine, fluoxetine).
As previously stated, Keyloun and colleagues previously found that SNRIs had the highest 3-month adherence rate in a study of > 5000 patients.7 In our study, SNRIs had the second highest mean 3-month MPR at about 75%, but the difference was not considered significant when compared with other antidepressant classes.
Bupropion was prescribed least frequently, but had the largest proportion of adherent patients. Gaspar and colleagues demonstrated similar outcomes, reporting that patients prescribed bupropion had a high OR for adherence.1 Bupropion may have had relatively low prescribing rates in our study because 64% of patients were diagnosed with a comorbid anxiety disorder and/or PTSD. For these patients, bupropion avoidance may have been intentional so as to not exacerbate anxiety.
Mirtazapine had both the lowest mean MPR and the lowest proportion of adherent patients. While no significant difference between antidepressant 3-month adherence rates were found, this study’s findings were similar to previous studies that found lower adherence to mirtazapine.1,5 Adverse effects such as sedation, increased appetite, and weight gain may have contributed to low adherence with mirtazapine.4 Patients may also have been using the agent on an as needed basis to treat insomnia despite the order being written for daily use.
Substance Use Disorder Influence
A significantly greater proportion of patients had an SUD in the low MPR group, suggesting that an SUD diagnosis may be a risk factor for low adherence. This finding is consistent with previous studies that also found that an SUD was associated with poor medication adherence.1 Patients with depression and an SUD have been shown to have suboptimal outcomes compared to those without an SUD, including a lower response to antidepressant therapy and increased illness severity.11,12
In a study of 131 outpatients with dual diagnosis (26% with depression) predictors for low self-reported adherence were a medication-related variable (increased adverse effects), a cognitive variable (low self-efficacy for drug avoidance), and a social factor (low social support for recovery). This variety of predictors seems to indicate that simple memory aids may not improve adherence. “Dual focus” mutual aid groups that provide social support for patients with dual diagnosis have been shown to improve adherence.13
The MEDVAMC Substance Dependence Treatment Program (SDTP) is an outpatient program that uses group education to aid veterans, often those with comorbid psychiatric disorders, to build relapse prevention skills and provide social support. Further exploration into the relationship between involvement in SDTP groups and antidepressant adherence in patients with dual diagnosis may be warranted.
Secondary Outcomes
Trends identified in the secondary outcome were similar to outcomes of previous studies: younger age, lower therapy involvement, and more comorbid psychiatric diagnoses were associated with lower adherence.1,7,8 The presence of increased previous use of antidepressants in the low adherence group may suggest that these patients have an increased illness severity, although objective scales, such as the Patient Health Questionnaire 9 (PHQ9), were not consistently conducted and therefore not included in this analysis. It is unknown whether the previous antidepressant prescriptions were of adequate duration. These patients may have also had intolerances that led to multiple different antidepressant prescriptions and self-discontinuation.
The average MRCI of study patients was 13.5 (range 2 - 53), which was significantly lower than a previous study of geriatric patients with depression reporting an average MRCI of 25.4 (range 6 - 64).14 The positive trend between MRCI and adherence seen in this study was puzzling and counterintuitive. A more complex regimen is generally thought to be associated with poor adherence. Patients with a greater number of comorbid conditions may inherently be on more medications and thus have a more complex medication regimen. Manzano-Garcia and colleagues identified a negative relationship between adherence and the number of comorbidities (OR, 1.04-1.57; P = .021) and the MRCI (OR, 1.14-1.26; P < .001) in patients with HIV.15 Further studies are needed to clarify the relationship between medication adherence and medication regimen complexity in patients with mental health disorders. A better understanding of this relationship could possibly facilitate improved individualized prescribing practices and follow-up.
Limitations
Findings from our study should be interpreted within several limitations. Generalizability and statistical power were limited due to the small sample size, a practice site limited to 1 facility, and population type. The retrospective design of the study introduces inherent bias that would be minimized had a prospective study been conducted. The primary outcome was based upon MPR, which only accounts for refills within a specified time period and does not assess for actual or accurate use of the medication. Data collection was limited to VA and US Department of Defense records.
Geographically diverse studies with larger sample sizes need to be conducted to better understand antidepressant adherence and its barriers and facilitators in the veteran population. The exclusion of patients with previous trials of the prescribed antidepressant may have led to a possible selection bias favoring inclusion of younger patients. These patients may have a more limited period for assessment and treatment when compared with older patients, and thus may have had a smaller chance of previous exposure to the prescribed antidepressant. Neither MAOIs or TCAs were included in this study. No patients taking MAOIs were identified from the Antidepressant Nonadherence Report during the study period. Three patients on TCAs were chart reviewed, but excluded from the study because of prior use of the antidepressant or a non-mental health indication. Additionally, no newer antidepressants, including vortioxetine and vilazodone, were included, likely secondary to their nonformulary status at the VA.
Conclusion
As this study’s purpose was to improve the quality of care at our facility, we will discuss our findings with local MHPs to develop strategies to improve antidepressant adherence. While larger studies need to be conducted to confirm our findings, it is worthwhile to consider risk factors for low adherence such as SUD when prescribing antidepressant medications. Patients with SUD could be encouraged to enroll in our facility’s telephone nursing depression care management program for more frequent follow up and medication adherence counseling.
This study did not find a significant difference in 3-month adherence rates between SSRIs, SNRIs, bupropion, and mirtazapine. SUD was significantly more common in patients with low adherence than those categorized as adherent and may be a risk factor for low adherence based upon our findings and those of previous studies.
Depression affects about 4.4% of the global population.1 Major depressive disorder (MDD) is currently the fourth highest cause of disability in the world and by 2030 MDD is expected to be third.2 Research has determined that 1 in 3 veterans seen in primary care shows depressive symptoms. Of these, 1 in 5 have symptoms severe enough to warrant further evaluation for MDD, and 1 in 10 require treatment.3 With this high rate of depression, optimized treatment strategies are needed, including antidepressants and psychotherapy. Antidepressants have grown in popularity since market entry in the 1950s; currently 1 in 10 US citizens aged ≥ 12 years are prescribed an antidepressant.4
Antidepressant Adherence
Antidepressant adherence is crucial for response and remission. Sansone and Sansone reported that, on average, < 50% of patients are adherent to their antidepressant treatment regimen 6 months after initiation (range, 5.4% - 87.6%).5 Fortney and colleagues found that, based on patient report, < 20% of veterans maintained at least 80% adherence at 6 months.6 Patients who are nonadherent are at an increased risk for relapse and recurrence and are more likely to seek care at an emergency department or to become hospitalized.2 In addition to the negative impact on patient outcomes, antidepressant nonadherence may also result in increased economic burden. In the US alone, the annual cost of treating MDD exceeds $210 billion, which will continue to increase if nonadherence is not mitigated.1
Patient-specific characteristics such as lack of knowledge about proper administration techniques, misguided beliefs, and negative attitudes towards treatment may affect adherence.5 In the veteran population, reasons for discontinuation also include lack of perceived benefit and adverse effects, specifically sexual difficulties.6 Sociodemographic and other patient characteristics also may be risk factors for nonadherence, including multiple medical comorbidities; substance use disorder (SUD) diagnosis; male gender; younger age; lack of health insurance or a higher medical cost burden; lack of or low involvement in psychotherapy; infrequent follow up visits; and high illness severity.1,7,8
Appreciating the adherence rates among the different antidepressant classes may help in antidepressant selection. To our knowledge, there have been no prior studies conducted in the veteran population that compared adherence rates among antidepressant classes. Studies in the nonveteran population report differing adherence rates among the antidepressant classes with generally higher adherence in patients prescribed serotonin norepinephrine reuptake inhibitors (SNRIs) and selective serotonin reuptake inhibitors (SSRIs). A retrospective review of commercial, Medicare, and Medicaid claims in > 5000 patients found that SNRIs had a significantly higher 3-month adherence rate based on the portion of days covered model (47%; P < .001) than other antidepressant classes (SSRIs, 42%; other antidepressants, 37%; tricyclic antidepressants [TCAs], 24%).7 Monoamine oxidase inhibitors (MAOIs) prescribed to 1% of the study population had the highest adherence rate at 48%.7 A study reviewing > 25 000 patient claims sourced from the IBM MarketScan research database (Armonk, NY) found that SSRIs (Odds ratio [OR], 1.26; P < .001) and norepinephrine dopamine reuptake inhibitors (NDRIs) (OR, 1.23; P = .007) had the highest ORs for adherence according to the portion of days covered model, while other serotonin modulators (OR, 0.65; P = .001) and tri/tetracyclic antidepressants (OR, 0.49; P < . 001) had the lowest ORs and were associated with lower adherence.1
VA Approaches to Adherence
To address antidepressant adherence, the US Department of Veteran Affairs (VA) adopted 2 measures from the Healthcare Effectiveness Data and Information Set: MDD43h and MDD47h. Measure MDD43h is defined as the proportion of patients with a depression diagnosis newly treated with an antidepressant medication who remained on the antidepressant medication for at least 84 out of 114 days (3 months). MDD47h is similar, but assesses patients remaining on an antidepressant medication for at least 180 out of 230 days (6 months).9 These constitute a SAIL (Strategic Analytics for Improvement and Learning) measure by which VA hospitals are compared. High performance on these measures aids in improving the comparative status of a VA facility.
To help improve performance on these measures, the VA Psychotropic Drug Safety Initiative developed the Antidepressant Nonadherence Report, which serves as a case finder for clinicians to identify veterans with low adherence and/or those overdue for a refill. The dashboard uses the medication possession ratio (MPR) to calculate adherence. While the optimal value is still widely debated, an MPR of ≥ 80% is generally accepted for many disease states.10 The dashboard defines low adherence as ≤ 60%.
As of September 2018, the Antidepressant Nonadherence Report for the Michael E. DeBakey VA Medical Center (MEDVAMC) in Houston, Texas, included > 5000 patients in both MEDVAMC and associated community-based outpatient clinics. About 30% of patients were categorized as overdue for a refill.
Study Objectives
To better understand the problem of antidepressant adherence within this population, we decided to study the relationship between antidepressant class and adherence rates, as well as how adherence relates to patient-specific characteristics. By highlighting predisposing risk factors to low adherence, we hope to provide better interventions.
The primary objective of this study was to determine whether 3-month adherence rates, measured by the MPR, differ between antidepressant classes in veterans newly initiated on antidepressant therapy. A secondary objective was to identify whether there are differences in patient characteristics between those with high MPR (≥ 80%) and low MPR (≤ 60%).
Methods
This study used a retrospective, cross-sectional chart review of MEDVAMC patients from the Antidepressant Nonadherence Report. Patients were: aged ≥ 18 years; newly initiated on an antidepressant with no previous use of the same medication; outpatient for the entire study period; and seen by a physician, physician assistant, nurse practitioner, or pharmacist mental health provider (MHP) within the 3-month study period. All patients’ charts showed a depression diagnosis—an inclusion criterion for the MDD43h and MDD47h measures. However, for this study, the indication(s) for the chosen antidepressant were determined by the MHP note in the patient electronic health record on the date that the medication was prescribed. Study patients may not have had a current depression diagnosis based upon the MHP assessment on the index date. We chose to determine the antidepressant indication(s) in this way because the MHP note would have the most detailed patient assessment.
Patients with previous use of the prescribed antidepressant were excluded because previous exposure may bias the patient and affect current adherence. Patients who were hospitalized at the VA for any reason during the 3-month study period were excluded because of a known risk during transitions of care for medications to be held or discontinued, which could impact refills and MPR. Some patients were excluded if they were taking the antidepressant for a nonmood-related indication (insomnia, neuropathy, migraine prophylaxis, etc). Patients also were excluded if the antidepressant was prescribed to take as-needed; if trazodone was the only antidepressant prescribed; if they were diagnosed with cognitive impairment including dementia or history of stroke; or if they were diagnosed with schizophrenia, schizoaffective disorder, or borderline personality disorder. Use of trazodone as the only antidepressant was excluded because of the relatively common practice to use it in the treatment of insomnia rather than depression.
Primary and Secondary Outcomes
Information collected for the primary outcome, including antidepressant class and MPR, was obtained from the Antidepressant Nonadherence Report. For the secondary outcome, the following data was collected for each patient: age, gender, race, housing status, Medication Regimen Complexity Index (MRCI), number and type of psychiatric diagnoses, number of previous antidepressants, psychotherapy involvement, and number of mental health visits during the 3-month study period. The MRCI is an objective, validated tool that determines relative medication regimen complexity by taking into consideration the number of medications, route and frequency of administration, splitting/multiple dosage units, and presence of any special instructions.11
The primary outcome was tested using a one-way analysis of variance (ANOVA). Nominal secondary outcomes were analyzed using the Fisher’s Exact. Continuous secondary outcomes were examined using an unpaired t-test.
Results
Of 320 charts, 212 patients were excluded and 108 were included (Figure). The most common reason for exclusion was a previously prescribed antidepressant. Of the included patients 49 had an MPR ≥ 80% and 24 had an MPR ≤ 60%. The characteristics of the study population are found in Table 1 and the antidepressant frequencies and MPRs are included in Table 2.
About 87% of study patients had a diagnosis of depression. Other concomitant psychiatric diagnoses include posttraumatic stress disorder (PTSD), anxiety, insomnia, and 2 cases of intermittent explosive disorder. There were no significant differences in mean MPR between the antidepressant classes (P = .31). Within each drug class, we identified the proportion of patients with high adherence (MPR ≥ 80%). Bupropion had the greatest percentage of highly adherent patients (50%) compared with SSRIs (42.5%), SNRIs (38.5%), and mirtazapine (31.3%).
Table 3 compares the characteristics between high MPR and low MPR patients. The low MPR group showed a significantly greater proportion of patients with an SUD than the high adherence group (41.7% vs 10.2%, respectively; P = .04). The most common type of SUD was alcohol use disorder followed by cannabis use disorder. There were no other statistically significant differences identified between high and low MPR groups. There was a trend towards significance when comparing MRCI between the 2 groups (high MPR, 15.2; low MPR, 10.8; P = .06).
Discussion
In our study, there was no significant difference in 3-month adherence rates between veterans on SSRIs, SNRIs, bupropion, and mirtazapine. This result differs from a study by Keyloun and colleagues that found that SNRIs had a significantly higher adherence rate when compared with other antidepressants.7
SSRIs were the most commonly prescribed antidepressant in our study, and also had the greatest mean 3-month MPR. The high use of SSRIs may be due to the greater number of SSRI choices to select from compared with other classes. SSRIs may also have been selected more frequently because nearly half (45.4%) of the patients had comorbid PTSD, for which 3 of the 4 first-line treatment options are SSRIs (sertraline, paroxetine, fluoxetine).
As previously stated, Keyloun and colleagues previously found that SNRIs had the highest 3-month adherence rate in a study of > 5000 patients.7 In our study, SNRIs had the second highest mean 3-month MPR at about 75%, but the difference was not considered significant when compared with other antidepressant classes.
Bupropion was prescribed least frequently, but had the largest proportion of adherent patients. Gaspar and colleagues demonstrated similar outcomes, reporting that patients prescribed bupropion had a high OR for adherence.1 Bupropion may have had relatively low prescribing rates in our study because 64% of patients were diagnosed with a comorbid anxiety disorder and/or PTSD. For these patients, bupropion avoidance may have been intentional so as to not exacerbate anxiety.
Mirtazapine had both the lowest mean MPR and the lowest proportion of adherent patients. While no significant difference between antidepressant 3-month adherence rates were found, this study’s findings were similar to previous studies that found lower adherence to mirtazapine.1,5 Adverse effects such as sedation, increased appetite, and weight gain may have contributed to low adherence with mirtazapine.4 Patients may also have been using the agent on an as needed basis to treat insomnia despite the order being written for daily use.
Substance Use Disorder Influence
A significantly greater proportion of patients had an SUD in the low MPR group, suggesting that an SUD diagnosis may be a risk factor for low adherence. This finding is consistent with previous studies that also found that an SUD was associated with poor medication adherence.1 Patients with depression and an SUD have been shown to have suboptimal outcomes compared to those without an SUD, including a lower response to antidepressant therapy and increased illness severity.11,12
In a study of 131 outpatients with dual diagnosis (26% with depression) predictors for low self-reported adherence were a medication-related variable (increased adverse effects), a cognitive variable (low self-efficacy for drug avoidance), and a social factor (low social support for recovery). This variety of predictors seems to indicate that simple memory aids may not improve adherence. “Dual focus” mutual aid groups that provide social support for patients with dual diagnosis have been shown to improve adherence.13
The MEDVAMC Substance Dependence Treatment Program (SDTP) is an outpatient program that uses group education to aid veterans, often those with comorbid psychiatric disorders, to build relapse prevention skills and provide social support. Further exploration into the relationship between involvement in SDTP groups and antidepressant adherence in patients with dual diagnosis may be warranted.
Secondary Outcomes
Trends identified in the secondary outcome were similar to outcomes of previous studies: younger age, lower therapy involvement, and more comorbid psychiatric diagnoses were associated with lower adherence.1,7,8 The presence of increased previous use of antidepressants in the low adherence group may suggest that these patients have an increased illness severity, although objective scales, such as the Patient Health Questionnaire 9 (PHQ9), were not consistently conducted and therefore not included in this analysis. It is unknown whether the previous antidepressant prescriptions were of adequate duration. These patients may have also had intolerances that led to multiple different antidepressant prescriptions and self-discontinuation.
The average MRCI of study patients was 13.5 (range 2 - 53), which was significantly lower than a previous study of geriatric patients with depression reporting an average MRCI of 25.4 (range 6 - 64).14 The positive trend between MRCI and adherence seen in this study was puzzling and counterintuitive. A more complex regimen is generally thought to be associated with poor adherence. Patients with a greater number of comorbid conditions may inherently be on more medications and thus have a more complex medication regimen. Manzano-Garcia and colleagues identified a negative relationship between adherence and the number of comorbidities (OR, 1.04-1.57; P = .021) and the MRCI (OR, 1.14-1.26; P < .001) in patients with HIV.15 Further studies are needed to clarify the relationship between medication adherence and medication regimen complexity in patients with mental health disorders. A better understanding of this relationship could possibly facilitate improved individualized prescribing practices and follow-up.
Limitations
Findings from our study should be interpreted within several limitations. Generalizability and statistical power were limited due to the small sample size, a practice site limited to 1 facility, and population type. The retrospective design of the study introduces inherent bias that would be minimized had a prospective study been conducted. The primary outcome was based upon MPR, which only accounts for refills within a specified time period and does not assess for actual or accurate use of the medication. Data collection was limited to VA and US Department of Defense records.
Geographically diverse studies with larger sample sizes need to be conducted to better understand antidepressant adherence and its barriers and facilitators in the veteran population. The exclusion of patients with previous trials of the prescribed antidepressant may have led to a possible selection bias favoring inclusion of younger patients. These patients may have a more limited period for assessment and treatment when compared with older patients, and thus may have had a smaller chance of previous exposure to the prescribed antidepressant. Neither MAOIs or TCAs were included in this study. No patients taking MAOIs were identified from the Antidepressant Nonadherence Report during the study period. Three patients on TCAs were chart reviewed, but excluded from the study because of prior use of the antidepressant or a non-mental health indication. Additionally, no newer antidepressants, including vortioxetine and vilazodone, were included, likely secondary to their nonformulary status at the VA.
Conclusion
As this study’s purpose was to improve the quality of care at our facility, we will discuss our findings with local MHPs to develop strategies to improve antidepressant adherence. While larger studies need to be conducted to confirm our findings, it is worthwhile to consider risk factors for low adherence such as SUD when prescribing antidepressant medications. Patients with SUD could be encouraged to enroll in our facility’s telephone nursing depression care management program for more frequent follow up and medication adherence counseling.
This study did not find a significant difference in 3-month adherence rates between SSRIs, SNRIs, bupropion, and mirtazapine. SUD was significantly more common in patients with low adherence than those categorized as adherent and may be a risk factor for low adherence based upon our findings and those of previous studies.
1. Gaspar FW, Zaidel CS, Dewa CS. Rates and determinants of use of pharmacotherapy and psychotherapy by patients with major depressive disorder. Psychiatr Serv. 2019;70(4):262-270.
2. Ho SC, Jacob SA, Tangiisuran B. Barriers and facilitators of adherence to antidepressants among outpatients with major depressive disorder: a qualitative study. PLoS One. 2017;12(6):e0179290.
3. US Department of Veterans Affairs, Office of Research and Development. VA research on: depression. https://www.research.va.gov/topics/depression.cfm#research1. Accessed May 30, 2019.
4. Santarsieri D, Schwartz TL. Antidepressant efficacy and side-effect burden: a quick guide for clinicians. Drugs Context. 2015;4:212290.
5. Sansone RA, Sansone LA. Antidepressant adherence: are patients taking their medications? Innov Clin Neurosci. 2012;9(5-6):41-46.
6. Fortney JC, Pyne JM, Edlund MJ, et al. Reasons for antidepressant nonadherence among veterans treated in primary care clinics. J Clin Psychiatry. 2011;72(6):827-834.
7. Keyloun KR, Hansen RN, Hepp Z, Gillard P, Thase ME, Devine EB. Adherence and persistence across antidepressant therapeutic classes: a retrospective claims analysis among insured US patients with major depressive disorder (MDD). [erratum: CNS Drugs. 2017;31(6):511.] CNS Drugs. 2017;31(5):421-432.
8. Mcinnis MG. Adherence to treatment regimens in major depression: perspectives, problems, and progress. https://www.psychiatrictimes.com/depression/adherence-treatment-regimens-major-depression-perspectives-problems-and-progress. Published September 15, 2007. Accessed September 10, 2019.
9. US Department of Veterans Affairs, Office of Mental Health Operations. Clinical support portal. User Guide – antidepressant non-adherence report (MDD43h MDD47h). https://spsites.cdw.va.gov/sites/OMHO_PsychPharm/_layouts/15/WopiFrame.aspx?sourcedoc=/sites/OMHO_PsychPharm/AnalyticsReports/UserGuideMDD43H47H.pdf. Accessed July 29, 2018. [Nonpublic site]
10. Crowe M. Do you know the difference between these adherence measures? https://www.pharmacytimes.com/contributor/michael-crowe-pharmd-mba-csp-fmpa/2015/07/do-you-know-the-difference-between-these-adherence-measures. Published July 5, 2015. Accessed September 13, 2019.
11. Watkins KE, Paddock SM, Zhang L, Wells KB. Improving care for depression in patients with comorbid substance misuse. Am J Psychiatry. 2006;163(1):125-132.
12. Magura S, Rosenblum A, Fong C. Factors associated with medication adherence among psychiatric outpatients at substance abuse risk. Open Addict J. 2011;4:58-64.
13. Magura S, Rosenblum A, Villano CL, Vogel HS, Fong C, Betzler T. Dual-focus mutual aid for co-occurring disorders: a quasi-experimental outcome evaluation study. Am J Drug Alcohol Abuse. 2008;34(1):61-74.
14. Libby AM, Fish DN, Hosokawa PW, et al. Patient-level medication regimen complexity across populations with chronic disease. Clin Ther. 2013;35(4):385-398.e1.
15. Manzano-García M, Pérez-Guerrero C, Álvarez de Sotomayor Paz M, Robustillo-Cortés MLA, Almeida-González CV, Morillo-Verdugo R. Identification of the medication regimen complexity index as an associated factor of nonadherence to antiretroviral treatment in HIV positive patients. Ann Pharmacother. 2018;52(9):862-867.
1. Gaspar FW, Zaidel CS, Dewa CS. Rates and determinants of use of pharmacotherapy and psychotherapy by patients with major depressive disorder. Psychiatr Serv. 2019;70(4):262-270.
2. Ho SC, Jacob SA, Tangiisuran B. Barriers and facilitators of adherence to antidepressants among outpatients with major depressive disorder: a qualitative study. PLoS One. 2017;12(6):e0179290.
3. US Department of Veterans Affairs, Office of Research and Development. VA research on: depression. https://www.research.va.gov/topics/depression.cfm#research1. Accessed May 30, 2019.
4. Santarsieri D, Schwartz TL. Antidepressant efficacy and side-effect burden: a quick guide for clinicians. Drugs Context. 2015;4:212290.
5. Sansone RA, Sansone LA. Antidepressant adherence: are patients taking their medications? Innov Clin Neurosci. 2012;9(5-6):41-46.
6. Fortney JC, Pyne JM, Edlund MJ, et al. Reasons for antidepressant nonadherence among veterans treated in primary care clinics. J Clin Psychiatry. 2011;72(6):827-834.
7. Keyloun KR, Hansen RN, Hepp Z, Gillard P, Thase ME, Devine EB. Adherence and persistence across antidepressant therapeutic classes: a retrospective claims analysis among insured US patients with major depressive disorder (MDD). [erratum: CNS Drugs. 2017;31(6):511.] CNS Drugs. 2017;31(5):421-432.
8. Mcinnis MG. Adherence to treatment regimens in major depression: perspectives, problems, and progress. https://www.psychiatrictimes.com/depression/adherence-treatment-regimens-major-depression-perspectives-problems-and-progress. Published September 15, 2007. Accessed September 10, 2019.
9. US Department of Veterans Affairs, Office of Mental Health Operations. Clinical support portal. User Guide – antidepressant non-adherence report (MDD43h MDD47h). https://spsites.cdw.va.gov/sites/OMHO_PsychPharm/_layouts/15/WopiFrame.aspx?sourcedoc=/sites/OMHO_PsychPharm/AnalyticsReports/UserGuideMDD43H47H.pdf. Accessed July 29, 2018. [Nonpublic site]
10. Crowe M. Do you know the difference between these adherence measures? https://www.pharmacytimes.com/contributor/michael-crowe-pharmd-mba-csp-fmpa/2015/07/do-you-know-the-difference-between-these-adherence-measures. Published July 5, 2015. Accessed September 13, 2019.
11. Watkins KE, Paddock SM, Zhang L, Wells KB. Improving care for depression in patients with comorbid substance misuse. Am J Psychiatry. 2006;163(1):125-132.
12. Magura S, Rosenblum A, Fong C. Factors associated with medication adherence among psychiatric outpatients at substance abuse risk. Open Addict J. 2011;4:58-64.
13. Magura S, Rosenblum A, Villano CL, Vogel HS, Fong C, Betzler T. Dual-focus mutual aid for co-occurring disorders: a quasi-experimental outcome evaluation study. Am J Drug Alcohol Abuse. 2008;34(1):61-74.
14. Libby AM, Fish DN, Hosokawa PW, et al. Patient-level medication regimen complexity across populations with chronic disease. Clin Ther. 2013;35(4):385-398.e1.
15. Manzano-García M, Pérez-Guerrero C, Álvarez de Sotomayor Paz M, Robustillo-Cortés MLA, Almeida-González CV, Morillo-Verdugo R. Identification of the medication regimen complexity index as an associated factor of nonadherence to antiretroviral treatment in HIV positive patients. Ann Pharmacother. 2018;52(9):862-867.
























