Continuous Glucose Monitoring vs Fingerstick Monitoring for Hemoglobin A1c Control in Veterans

Article Type
Changed
Wed, 11/06/2024 - 12:47
Display Headline
Continuous Glucose Monitoring vs Fingerstick Monitoring for Hemoglobin A1c Control in Veterans

In the United States, 1 in 4 veterans lives with type 2 diabetes mellitus (T2DM), double the rate of the general population.1 Medications are important for the treatment of T2DM and preventing complications that may develop if not properly managed. Common classes of medications for diabetes include biguanides, sodiumglucose cotransporter-2 (SGLT-2) inhibitors, glucagon-like peptide-1 (GLP-1) receptor agonists, dipeptidyl peptidase-4 inhibitors, thiazolidinediones, sulfonylureas, and insulin. The selection of treatment depends on patient-specific factors including hemoglobin A1c (HbA1c) goal, potential effects on weight, risk of hypoglycemia, and comorbidities such as atherosclerotic cardiovascular disease, heart failure, or chronic kidney disease.2

HbA1c level reflects the mean blood glucose over the previous 3 months and serves as an indication of diabetes control. In patients with diabetes, it is recommended that HbA1c is checked ≥ 2 times annually for those meeting treatment goals, or more often if the patient needs to adjust medications to reach their HbA1c goal. The goal HbA1c level for most adults with diabetes is < 7%.3 This target can be adjusted based on age, comorbidities, or other patient factors. It is generally recommended that frequent glucose monitoring is not needed for patients with T2DM who are only taking oral agents and/or noninsulin injectables. However, for those on insulin regimens, it is advised to monitor glucose closely, with even more frequent testing for those with an intensive insulin regimen.3

Most patients with diabetes use fingerstick testing to self-monitor their blood glucose. However, continuous glucose monitors (CGMs) are becoming widely available and offer a solution to those who do not have the ability to check their glucose multiple times a day and throughout the night. The American Diabetes Association recommends that the frequency and timing of blood glucose monitoring, or the consideration of CGM use, should be based on the specific needs and goals of each patient.3 Guidelines also encourage those on intensive insulin regimens to check glucose levels when fasting, before and after meals, prior to exercise, and when hypoglycemia or hyperglycemia is suspected. Frequent testing can become a burden for patients, whereas once a CGM sensor is placed, it can be worn for 10 to 14 days. CGMs are also capable of transmitting glucose readings every 1 to 15 minutes to a receiver or mobile phone, allowing for further adaptability to a patient’s lifestyle.3

CGMs work by measuring the interstitial glucose with a small filament sensor and have demonstrated accuracy when compared to blood glucose readings. The ability of a CGM to accurately reflect HbA1c levels is a potential benefit, reducing the need for frequent testing to determine whether patients have achieved glycemic control.4 Another benefit of a CGM is the ease of sharing data; patient accounts can be linked with a health care site, allowing clinicians to access glucose data even if the patient is not able to be seen in clinic. This allows health care practitioners (HCPs) to more efficiently tailor medications and optimize regimens based on patient-specific data that was not available by fingerstick testing alone.

Vigersky and colleagues provided one of the few studies on the long-term effects of CGM in patients managing T2DM through diet and exercise alone, oral medications, or basal insulin and found significant improvement in HbA1c after only 3 months of CGM use.5

An important aspect of CGM use is the ability to alert the patient to low blood glucose readings, which can be dangerous for those unaware of hypoglycemia. Many studies have investigated the association between CGM use and acute metabolic events, demonstrating the potential for CGMs to prevent these emergencies. Karter and colleagues found a reduction in emergency department visits and hospitalizations for hypoglycemia associated with the use of CGMs in patients with type 1 DM (T1DM) and T2DM.6

There have been few studies on the use of CGM in veterans. Langford and colleagues found a reduction of HbA1c among veterans with T2DM using CGMs. However, > 50% of the patients in the study were not receiving insulin therapy, which currently is a US Department of Veterans Affairs (VA) CGM criteria for use.7 While current studies provide evidence that supports improvement in HbA1c levels with the use of CGMs, data are lacking for veterans with T2DM taking insulin. There is also minimal research that indicates which patients should be offered a CGM. The objective of this study was to evaluate glycemic control in veterans with T2DM on insulin using a CGM who were previously monitoring blood glucose with fingerstick testing. Secondary endpoints were explored to identify subgroups that may benefit from a CGM and other potential advantages of CGMs.

Methods

This was a retrospective study of veterans who transitioned from fingerstick testing to CGM for glucose monitoring. Each veteran served as their own control to limit confounding variables when comparing HbA1c levels. Veterans with an active or suspended CGM order were identified by reviewing outpatient prescription data. All data collection and analysis were done within the Veterans Affairs Sioux Falls Health Care System.

The primary objective of this study was to assess glycemic control from the use of a CGM by evaluating the change in HbA1c after transitioning to a CGM compared to the change in HbA1c with standard fingerstick monitoring. Three HbA1c values were collected for each veteran: before starting CGM, at initiation, and following CGM initiation (Figure 1). CGM start date was the date the CGM prescription order was placed. The pre-CGM HbA1c level was ≥ 1 year prior to the CGM start date or the HbA1c closest to 1 year. The start CGM HbA1c level was within 3 months before or 1 month after the CGM start date. The post-CGM HbA1c level was the most recent time of data collection and at least 6 months after CGM initiation. The change in HbA1c from fingerstick glucose monitoring was the difference between the pre-CGM and start CGM values. The change in HbA1c from use of a CGM was the difference between start CGM and post-CGM values, which were compared to determine HbA1c reduction from CGM use.

Abbreviations: CGM, continuous glucose monitor; HbA1c, hemoglobin A1c.

This study also explored secondary outcomes including changes in HbA1c by prescriber type, differences in HbA1c reduction based on age, and changes in diabetes medications, including total daily insulin doses. For secondary outcomes, diabetes medication information and the total daily dose of insulin were gathered at the start of CGM use and at the time of data collection. The most recent CGM order prescribed was also collected.

Veterans were included if they were aged ≥ 18 years, had an active order for a CGM, T2DM diagnosis, an insulin prescription, and previously used test strips for glucose monitoring. Patients with T1DM, those who accessed CGMs or care in the community, and patients without HbA1c values pre-CGM, were excluded.

Statistical Analysis

The primary endpoint of change in HbA1c level before and after CGM use was compared using a paired t test. A 0.5% change in HbA1c was considered clinically significant, as suggested in other studies.8,9P < .05 was considered statistically significant. Analysis for continuous baseline characteristics, including age and total daily insulin, were reported as mean values. Nominal characteristics including sex, race, diabetes medications, and prescriber type are reported as percentages.

Results

A total of 402 veterans were identified with an active CGM at the time of initial data collection in January 2024 and 175 met inclusion criteria. Sixty patients were excluded due to diabetes managed through a community HCP, 38 had T1DM, and 129 lacked HbA1c within all specified time periods. The 175 veterans were randomized, and 150 were selected to perform a chart review for data collection. The mean age was 70 years, most were male and identified as White (Table 1). The majority of patients were managed by endocrinology (53.3%), followed by primary care (24.0%), and pharmacy (22.7%) (Table 2). The mean baseline HbA1c was 8.6%.

The difference in HbA1c before and after use of CGM was -0.97% (P = .0001). Prior to use of a CGM the change in HbA1c was minimal, with an increase of 0.003% with the use of selfmonitoring glucose. After use of a CGM, HbA1c decreased by 0.971%. This reduction in HbA1c would also be considered clinically significant as the change was > 0.5%. The mean pre-, at start, and post-CGM HbA1c levels were 8.6%, 8.6%, and 7.6%, respectively (Figure 2). Pharmacy prescribers had a 0.7% reduction in HbA1c post-CGM, the least of all prescribers. While most age groups saw a reduction in HbA1c, those aged ≥ 80 years had an increase of 0.18% (Table 3). There was an overall mean reduction in insulin of 22 units, which was similar between all prescribers.

Abbreviation: CGM, continuous glucose monitor.

Discussion

The primary endpoint of difference in change of HbA1c before and after CGM use was found to be statistically and clinically significant, with a nearly 1% reduction in HbA1c, which was similar to the reduction found by Vigersky and colleagues. 5 Across all prescribers, post-CGM HbA1c levels were similar; however, patients with CGM prescribed by pharmacists had the smallest change in HbA1c. VA pharmacists primarily assess veterans taking insulin who have HbA1c levels that are below the goal with the aim of decreasing insulin to reduce the risk of hypoglycemia, which could result in increased HbA1c levels. This may also explain the observed increase in post-CGM HbA1c levels in patients aged ≥ 80 years. Patients under the care of pharmacists also had baseline mean HbA1c levels that were lower than primary care and endocrinology prescribers and were closer to their HbA1c goal at baseline, which likely was reflected in the smaller reduction in post-CGM HbA1c level.

While there was a decrease in HbA1c levels with CGM use, there were also changes to medications during this timeframe that also may have impacted HbA1c levels. The most common diabetes medications started during CGM use were GLP-1 agonists and SGLT2-inhibitors. Additionally, there was a reduction in the total daily dose of insulin in the study population. These results demonstrate the potential benefits of CGMs for prescribers who take advantage of the CGM glucose data available to assist with medication adjustments. Another consideration for differences in changes of HbA1c among prescriber types is the opportunity for more frequent follow- up visits with pharmacy or endocrinology compared with primary care. If veterans are followed more closely, it may be associated with improved HbA1c control. Further research investigating changes in HbA1c levels based on followup frequency may be useful.

Strengths and Limitations

The crossover design was a strength of this study. This design reduced confounding variables by having veterans serve as their own controls. In addition, the collection of multiple secondary outcomes adds to the knowledge base for future studies. This study focused on a unique population of veterans with T2DM who were taking insulin, an area that previously had very little data available to determine the benefits of CGM use.

Although the use of a CGM showed statistical significance in lowering HbA1c, many veterans were started on new diabetes medication during the period of CGM use, which also likely contributed to the reduction in HbA1c and may have confounded the results. The study was limited by its small population size due to time constraints of chart reviews and the limited generalizability of results outside of the VA system. The majority of patients were from a single site, male and identified as White, which may not be reflective of other VA and community health care systems. It was also noted that the time from the initiation of CGM use to the most recent HbA1c level varied from 6 months to several years. Additionally, veterans managed by community-based HCPs with complex diabetes cases were excluded.

Conclusions

This study demonstrated a clinically and statistically significant reduction in HbA1c with the use of a CGM compared to fingerstick monitoring in veterans with T2DM who were being treated with insulin. The change in post-CGM HbA1c levels across prescribers was similar. In the subgroup analysis of change in HbA1c among age groups, there was a lower HbA1c reduction in individuals aged ≥ 80 years. The results from this study support the idea that CGM use may be beneficial for patients who require a reduction in HbA1c by allowing more precise adjustments to medications and optimization of therapy, as well as the potential to reduce insulin requirements, which is especially valuable in the older adult veteran population.

References
  1. US Department of Veterans Affairs. VA supports veterans who have type 2 diabetes. VA News. Accessed September 30, 2024. https://news.va.gov/107579/va-supports-veterans-who-have-type-2-diabetes/
  2. ElSayed NA, Aleppo G, Aroda VR, et al. 9. Pharmacologic approaches to glycemic treatment: standards of care in diabetes-2023. Diabetes Care. 2023;46(Suppl 1):S140- S157. doi:10.2337/dc23-S009
  3. ElSayed NA, Aleppo G, Aroda VR, et al. 6. Glycemic targets: standards of care in diabetes-2023. Diabetes Care. 2023;46(Suppl 1):S97-S110. doi:10.2337/dc23-S006
  4. Miller E, Gavin JR, Kruger DF, Brunton SA. Continuous glucose monitoring: optimizing diabetes care: executive summary. Clin Diabetes. 2022;40(4):394-398. doi:10.2337/cd22-0043
  5. Vigersky RA, Fonda SJ, Chellappa M, Walker MS, Ehrhardt NM. Short- and long-term effects of real-time continuous glucose monitoring in patients with type 2 diabetes. Diabetes Care. 2012;35(1):32-38. doi:10.2337/dc11-1438
  6. Karter AJ, Parker MM, Moffet HH, Gilliam LK, Dlott R. Association of real-time continuous glucose monitoring with glycemic control and acute metabolic events among patients with insulin-treated diabetes. JAMA. 2021;325(22):2273-2284. doi:10.1001/JAMA.2021.6530
  7. Langford SN, Lane M, Karounos D. Continuous blood glucose monitoring outcomes in veterans with type 2 diabetes. Fed Pract. 2021;38(Suppl 4):S14-S17. doi:10.12788/fp.0189
  8. Radin MS. Pitfalls in hemoglobin A1c measurement: when results may be misleading. J Gen Intern Med. 2014;29(2):388-394. doi:10.1007/s11606-013-2595-x.
  9. Little RR, Rohlfing CL, Sacks DB; National Glycohemoglobin Standardization Program (NGSP) steering committee. Status of hemoglobin A1c measurement and goals for improvement: from chaos to order for improving diabetes care. Clin Chem. 2011;57(2):205-214. doi:10.1373/clinchem.2010.148841
Article PDF
Author and Disclosure Information

Kelsey Floerchinger, PharmDa; Kelley Oehlke, PharmD, BCACPa; Scott Bebensee, PharmD, BCPSa; Austin Hansen, PharmDa; Kelsey Oye, PharmD, BCACP, CDCESa

Correspondence: Kelsey Floerchinger ([email protected])

Author affiliations: aVeterans Affairs Sioux Falls Health Care System, South Dakota

Author disclosures: The authors report no actual or potential conflict of interest with regard to this article.

Disclaimer: The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the official position or policy of the Defense Health Agency, US Department of Defense, the US Government, or any of its agencies. This article may discuss unlabeled or investigational use of certain drugs. Please review the complete prescribing information for specific drugs or drug combinations—including indications, contraindications, warnings, and adverse effects—before administering pharmacologic therapy to patients.

Fed Pract. 2024;41(suppl 5). Published online November 15. doi:10.12788/fp.0525

Issue
Federal Practitioner - 41(11)s
Publications
Topics
Page Number
S1-S5
Sections
Author and Disclosure Information

Kelsey Floerchinger, PharmDa; Kelley Oehlke, PharmD, BCACPa; Scott Bebensee, PharmD, BCPSa; Austin Hansen, PharmDa; Kelsey Oye, PharmD, BCACP, CDCESa

Correspondence: Kelsey Floerchinger ([email protected])

Author affiliations: aVeterans Affairs Sioux Falls Health Care System, South Dakota

Author disclosures: The authors report no actual or potential conflict of interest with regard to this article.

Disclaimer: The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the official position or policy of the Defense Health Agency, US Department of Defense, the US Government, or any of its agencies. This article may discuss unlabeled or investigational use of certain drugs. Please review the complete prescribing information for specific drugs or drug combinations—including indications, contraindications, warnings, and adverse effects—before administering pharmacologic therapy to patients.

Fed Pract. 2024;41(suppl 5). Published online November 15. doi:10.12788/fp.0525

Author and Disclosure Information

Kelsey Floerchinger, PharmDa; Kelley Oehlke, PharmD, BCACPa; Scott Bebensee, PharmD, BCPSa; Austin Hansen, PharmDa; Kelsey Oye, PharmD, BCACP, CDCESa

Correspondence: Kelsey Floerchinger ([email protected])

Author affiliations: aVeterans Affairs Sioux Falls Health Care System, South Dakota

Author disclosures: The authors report no actual or potential conflict of interest with regard to this article.

Disclaimer: The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the official position or policy of the Defense Health Agency, US Department of Defense, the US Government, or any of its agencies. This article may discuss unlabeled or investigational use of certain drugs. Please review the complete prescribing information for specific drugs or drug combinations—including indications, contraindications, warnings, and adverse effects—before administering pharmacologic therapy to patients.

Fed Pract. 2024;41(suppl 5). Published online November 15. doi:10.12788/fp.0525

Article PDF
Article PDF

In the United States, 1 in 4 veterans lives with type 2 diabetes mellitus (T2DM), double the rate of the general population.1 Medications are important for the treatment of T2DM and preventing complications that may develop if not properly managed. Common classes of medications for diabetes include biguanides, sodiumglucose cotransporter-2 (SGLT-2) inhibitors, glucagon-like peptide-1 (GLP-1) receptor agonists, dipeptidyl peptidase-4 inhibitors, thiazolidinediones, sulfonylureas, and insulin. The selection of treatment depends on patient-specific factors including hemoglobin A1c (HbA1c) goal, potential effects on weight, risk of hypoglycemia, and comorbidities such as atherosclerotic cardiovascular disease, heart failure, or chronic kidney disease.2

HbA1c level reflects the mean blood glucose over the previous 3 months and serves as an indication of diabetes control. In patients with diabetes, it is recommended that HbA1c is checked ≥ 2 times annually for those meeting treatment goals, or more often if the patient needs to adjust medications to reach their HbA1c goal. The goal HbA1c level for most adults with diabetes is < 7%.3 This target can be adjusted based on age, comorbidities, or other patient factors. It is generally recommended that frequent glucose monitoring is not needed for patients with T2DM who are only taking oral agents and/or noninsulin injectables. However, for those on insulin regimens, it is advised to monitor glucose closely, with even more frequent testing for those with an intensive insulin regimen.3

Most patients with diabetes use fingerstick testing to self-monitor their blood glucose. However, continuous glucose monitors (CGMs) are becoming widely available and offer a solution to those who do not have the ability to check their glucose multiple times a day and throughout the night. The American Diabetes Association recommends that the frequency and timing of blood glucose monitoring, or the consideration of CGM use, should be based on the specific needs and goals of each patient.3 Guidelines also encourage those on intensive insulin regimens to check glucose levels when fasting, before and after meals, prior to exercise, and when hypoglycemia or hyperglycemia is suspected. Frequent testing can become a burden for patients, whereas once a CGM sensor is placed, it can be worn for 10 to 14 days. CGMs are also capable of transmitting glucose readings every 1 to 15 minutes to a receiver or mobile phone, allowing for further adaptability to a patient’s lifestyle.3

CGMs work by measuring the interstitial glucose with a small filament sensor and have demonstrated accuracy when compared to blood glucose readings. The ability of a CGM to accurately reflect HbA1c levels is a potential benefit, reducing the need for frequent testing to determine whether patients have achieved glycemic control.4 Another benefit of a CGM is the ease of sharing data; patient accounts can be linked with a health care site, allowing clinicians to access glucose data even if the patient is not able to be seen in clinic. This allows health care practitioners (HCPs) to more efficiently tailor medications and optimize regimens based on patient-specific data that was not available by fingerstick testing alone.

Vigersky and colleagues provided one of the few studies on the long-term effects of CGM in patients managing T2DM through diet and exercise alone, oral medications, or basal insulin and found significant improvement in HbA1c after only 3 months of CGM use.5

An important aspect of CGM use is the ability to alert the patient to low blood glucose readings, which can be dangerous for those unaware of hypoglycemia. Many studies have investigated the association between CGM use and acute metabolic events, demonstrating the potential for CGMs to prevent these emergencies. Karter and colleagues found a reduction in emergency department visits and hospitalizations for hypoglycemia associated with the use of CGMs in patients with type 1 DM (T1DM) and T2DM.6

There have been few studies on the use of CGM in veterans. Langford and colleagues found a reduction of HbA1c among veterans with T2DM using CGMs. However, > 50% of the patients in the study were not receiving insulin therapy, which currently is a US Department of Veterans Affairs (VA) CGM criteria for use.7 While current studies provide evidence that supports improvement in HbA1c levels with the use of CGMs, data are lacking for veterans with T2DM taking insulin. There is also minimal research that indicates which patients should be offered a CGM. The objective of this study was to evaluate glycemic control in veterans with T2DM on insulin using a CGM who were previously monitoring blood glucose with fingerstick testing. Secondary endpoints were explored to identify subgroups that may benefit from a CGM and other potential advantages of CGMs.

Methods

This was a retrospective study of veterans who transitioned from fingerstick testing to CGM for glucose monitoring. Each veteran served as their own control to limit confounding variables when comparing HbA1c levels. Veterans with an active or suspended CGM order were identified by reviewing outpatient prescription data. All data collection and analysis were done within the Veterans Affairs Sioux Falls Health Care System.

The primary objective of this study was to assess glycemic control from the use of a CGM by evaluating the change in HbA1c after transitioning to a CGM compared to the change in HbA1c with standard fingerstick monitoring. Three HbA1c values were collected for each veteran: before starting CGM, at initiation, and following CGM initiation (Figure 1). CGM start date was the date the CGM prescription order was placed. The pre-CGM HbA1c level was ≥ 1 year prior to the CGM start date or the HbA1c closest to 1 year. The start CGM HbA1c level was within 3 months before or 1 month after the CGM start date. The post-CGM HbA1c level was the most recent time of data collection and at least 6 months after CGM initiation. The change in HbA1c from fingerstick glucose monitoring was the difference between the pre-CGM and start CGM values. The change in HbA1c from use of a CGM was the difference between start CGM and post-CGM values, which were compared to determine HbA1c reduction from CGM use.

Abbreviations: CGM, continuous glucose monitor; HbA1c, hemoglobin A1c.

This study also explored secondary outcomes including changes in HbA1c by prescriber type, differences in HbA1c reduction based on age, and changes in diabetes medications, including total daily insulin doses. For secondary outcomes, diabetes medication information and the total daily dose of insulin were gathered at the start of CGM use and at the time of data collection. The most recent CGM order prescribed was also collected.

Veterans were included if they were aged ≥ 18 years, had an active order for a CGM, T2DM diagnosis, an insulin prescription, and previously used test strips for glucose monitoring. Patients with T1DM, those who accessed CGMs or care in the community, and patients without HbA1c values pre-CGM, were excluded.

Statistical Analysis

The primary endpoint of change in HbA1c level before and after CGM use was compared using a paired t test. A 0.5% change in HbA1c was considered clinically significant, as suggested in other studies.8,9P < .05 was considered statistically significant. Analysis for continuous baseline characteristics, including age and total daily insulin, were reported as mean values. Nominal characteristics including sex, race, diabetes medications, and prescriber type are reported as percentages.

Results

A total of 402 veterans were identified with an active CGM at the time of initial data collection in January 2024 and 175 met inclusion criteria. Sixty patients were excluded due to diabetes managed through a community HCP, 38 had T1DM, and 129 lacked HbA1c within all specified time periods. The 175 veterans were randomized, and 150 were selected to perform a chart review for data collection. The mean age was 70 years, most were male and identified as White (Table 1). The majority of patients were managed by endocrinology (53.3%), followed by primary care (24.0%), and pharmacy (22.7%) (Table 2). The mean baseline HbA1c was 8.6%.

The difference in HbA1c before and after use of CGM was -0.97% (P = .0001). Prior to use of a CGM the change in HbA1c was minimal, with an increase of 0.003% with the use of selfmonitoring glucose. After use of a CGM, HbA1c decreased by 0.971%. This reduction in HbA1c would also be considered clinically significant as the change was > 0.5%. The mean pre-, at start, and post-CGM HbA1c levels were 8.6%, 8.6%, and 7.6%, respectively (Figure 2). Pharmacy prescribers had a 0.7% reduction in HbA1c post-CGM, the least of all prescribers. While most age groups saw a reduction in HbA1c, those aged ≥ 80 years had an increase of 0.18% (Table 3). There was an overall mean reduction in insulin of 22 units, which was similar between all prescribers.

Abbreviation: CGM, continuous glucose monitor.

Discussion

The primary endpoint of difference in change of HbA1c before and after CGM use was found to be statistically and clinically significant, with a nearly 1% reduction in HbA1c, which was similar to the reduction found by Vigersky and colleagues. 5 Across all prescribers, post-CGM HbA1c levels were similar; however, patients with CGM prescribed by pharmacists had the smallest change in HbA1c. VA pharmacists primarily assess veterans taking insulin who have HbA1c levels that are below the goal with the aim of decreasing insulin to reduce the risk of hypoglycemia, which could result in increased HbA1c levels. This may also explain the observed increase in post-CGM HbA1c levels in patients aged ≥ 80 years. Patients under the care of pharmacists also had baseline mean HbA1c levels that were lower than primary care and endocrinology prescribers and were closer to their HbA1c goal at baseline, which likely was reflected in the smaller reduction in post-CGM HbA1c level.

While there was a decrease in HbA1c levels with CGM use, there were also changes to medications during this timeframe that also may have impacted HbA1c levels. The most common diabetes medications started during CGM use were GLP-1 agonists and SGLT2-inhibitors. Additionally, there was a reduction in the total daily dose of insulin in the study population. These results demonstrate the potential benefits of CGMs for prescribers who take advantage of the CGM glucose data available to assist with medication adjustments. Another consideration for differences in changes of HbA1c among prescriber types is the opportunity for more frequent follow- up visits with pharmacy or endocrinology compared with primary care. If veterans are followed more closely, it may be associated with improved HbA1c control. Further research investigating changes in HbA1c levels based on followup frequency may be useful.

Strengths and Limitations

The crossover design was a strength of this study. This design reduced confounding variables by having veterans serve as their own controls. In addition, the collection of multiple secondary outcomes adds to the knowledge base for future studies. This study focused on a unique population of veterans with T2DM who were taking insulin, an area that previously had very little data available to determine the benefits of CGM use.

Although the use of a CGM showed statistical significance in lowering HbA1c, many veterans were started on new diabetes medication during the period of CGM use, which also likely contributed to the reduction in HbA1c and may have confounded the results. The study was limited by its small population size due to time constraints of chart reviews and the limited generalizability of results outside of the VA system. The majority of patients were from a single site, male and identified as White, which may not be reflective of other VA and community health care systems. It was also noted that the time from the initiation of CGM use to the most recent HbA1c level varied from 6 months to several years. Additionally, veterans managed by community-based HCPs with complex diabetes cases were excluded.

Conclusions

This study demonstrated a clinically and statistically significant reduction in HbA1c with the use of a CGM compared to fingerstick monitoring in veterans with T2DM who were being treated with insulin. The change in post-CGM HbA1c levels across prescribers was similar. In the subgroup analysis of change in HbA1c among age groups, there was a lower HbA1c reduction in individuals aged ≥ 80 years. The results from this study support the idea that CGM use may be beneficial for patients who require a reduction in HbA1c by allowing more precise adjustments to medications and optimization of therapy, as well as the potential to reduce insulin requirements, which is especially valuable in the older adult veteran population.

In the United States, 1 in 4 veterans lives with type 2 diabetes mellitus (T2DM), double the rate of the general population.1 Medications are important for the treatment of T2DM and preventing complications that may develop if not properly managed. Common classes of medications for diabetes include biguanides, sodiumglucose cotransporter-2 (SGLT-2) inhibitors, glucagon-like peptide-1 (GLP-1) receptor agonists, dipeptidyl peptidase-4 inhibitors, thiazolidinediones, sulfonylureas, and insulin. The selection of treatment depends on patient-specific factors including hemoglobin A1c (HbA1c) goal, potential effects on weight, risk of hypoglycemia, and comorbidities such as atherosclerotic cardiovascular disease, heart failure, or chronic kidney disease.2

HbA1c level reflects the mean blood glucose over the previous 3 months and serves as an indication of diabetes control. In patients with diabetes, it is recommended that HbA1c is checked ≥ 2 times annually for those meeting treatment goals, or more often if the patient needs to adjust medications to reach their HbA1c goal. The goal HbA1c level for most adults with diabetes is < 7%.3 This target can be adjusted based on age, comorbidities, or other patient factors. It is generally recommended that frequent glucose monitoring is not needed for patients with T2DM who are only taking oral agents and/or noninsulin injectables. However, for those on insulin regimens, it is advised to monitor glucose closely, with even more frequent testing for those with an intensive insulin regimen.3

Most patients with diabetes use fingerstick testing to self-monitor their blood glucose. However, continuous glucose monitors (CGMs) are becoming widely available and offer a solution to those who do not have the ability to check their glucose multiple times a day and throughout the night. The American Diabetes Association recommends that the frequency and timing of blood glucose monitoring, or the consideration of CGM use, should be based on the specific needs and goals of each patient.3 Guidelines also encourage those on intensive insulin regimens to check glucose levels when fasting, before and after meals, prior to exercise, and when hypoglycemia or hyperglycemia is suspected. Frequent testing can become a burden for patients, whereas once a CGM sensor is placed, it can be worn for 10 to 14 days. CGMs are also capable of transmitting glucose readings every 1 to 15 minutes to a receiver or mobile phone, allowing for further adaptability to a patient’s lifestyle.3

CGMs work by measuring the interstitial glucose with a small filament sensor and have demonstrated accuracy when compared to blood glucose readings. The ability of a CGM to accurately reflect HbA1c levels is a potential benefit, reducing the need for frequent testing to determine whether patients have achieved glycemic control.4 Another benefit of a CGM is the ease of sharing data; patient accounts can be linked with a health care site, allowing clinicians to access glucose data even if the patient is not able to be seen in clinic. This allows health care practitioners (HCPs) to more efficiently tailor medications and optimize regimens based on patient-specific data that was not available by fingerstick testing alone.

Vigersky and colleagues provided one of the few studies on the long-term effects of CGM in patients managing T2DM through diet and exercise alone, oral medications, or basal insulin and found significant improvement in HbA1c after only 3 months of CGM use.5

An important aspect of CGM use is the ability to alert the patient to low blood glucose readings, which can be dangerous for those unaware of hypoglycemia. Many studies have investigated the association between CGM use and acute metabolic events, demonstrating the potential for CGMs to prevent these emergencies. Karter and colleagues found a reduction in emergency department visits and hospitalizations for hypoglycemia associated with the use of CGMs in patients with type 1 DM (T1DM) and T2DM.6

There have been few studies on the use of CGM in veterans. Langford and colleagues found a reduction of HbA1c among veterans with T2DM using CGMs. However, > 50% of the patients in the study were not receiving insulin therapy, which currently is a US Department of Veterans Affairs (VA) CGM criteria for use.7 While current studies provide evidence that supports improvement in HbA1c levels with the use of CGMs, data are lacking for veterans with T2DM taking insulin. There is also minimal research that indicates which patients should be offered a CGM. The objective of this study was to evaluate glycemic control in veterans with T2DM on insulin using a CGM who were previously monitoring blood glucose with fingerstick testing. Secondary endpoints were explored to identify subgroups that may benefit from a CGM and other potential advantages of CGMs.

Methods

This was a retrospective study of veterans who transitioned from fingerstick testing to CGM for glucose monitoring. Each veteran served as their own control to limit confounding variables when comparing HbA1c levels. Veterans with an active or suspended CGM order were identified by reviewing outpatient prescription data. All data collection and analysis were done within the Veterans Affairs Sioux Falls Health Care System.

The primary objective of this study was to assess glycemic control from the use of a CGM by evaluating the change in HbA1c after transitioning to a CGM compared to the change in HbA1c with standard fingerstick monitoring. Three HbA1c values were collected for each veteran: before starting CGM, at initiation, and following CGM initiation (Figure 1). CGM start date was the date the CGM prescription order was placed. The pre-CGM HbA1c level was ≥ 1 year prior to the CGM start date or the HbA1c closest to 1 year. The start CGM HbA1c level was within 3 months before or 1 month after the CGM start date. The post-CGM HbA1c level was the most recent time of data collection and at least 6 months after CGM initiation. The change in HbA1c from fingerstick glucose monitoring was the difference between the pre-CGM and start CGM values. The change in HbA1c from use of a CGM was the difference between start CGM and post-CGM values, which were compared to determine HbA1c reduction from CGM use.

Abbreviations: CGM, continuous glucose monitor; HbA1c, hemoglobin A1c.

This study also explored secondary outcomes including changes in HbA1c by prescriber type, differences in HbA1c reduction based on age, and changes in diabetes medications, including total daily insulin doses. For secondary outcomes, diabetes medication information and the total daily dose of insulin were gathered at the start of CGM use and at the time of data collection. The most recent CGM order prescribed was also collected.

Veterans were included if they were aged ≥ 18 years, had an active order for a CGM, T2DM diagnosis, an insulin prescription, and previously used test strips for glucose monitoring. Patients with T1DM, those who accessed CGMs or care in the community, and patients without HbA1c values pre-CGM, were excluded.

Statistical Analysis

The primary endpoint of change in HbA1c level before and after CGM use was compared using a paired t test. A 0.5% change in HbA1c was considered clinically significant, as suggested in other studies.8,9P < .05 was considered statistically significant. Analysis for continuous baseline characteristics, including age and total daily insulin, were reported as mean values. Nominal characteristics including sex, race, diabetes medications, and prescriber type are reported as percentages.

Results

A total of 402 veterans were identified with an active CGM at the time of initial data collection in January 2024 and 175 met inclusion criteria. Sixty patients were excluded due to diabetes managed through a community HCP, 38 had T1DM, and 129 lacked HbA1c within all specified time periods. The 175 veterans were randomized, and 150 were selected to perform a chart review for data collection. The mean age was 70 years, most were male and identified as White (Table 1). The majority of patients were managed by endocrinology (53.3%), followed by primary care (24.0%), and pharmacy (22.7%) (Table 2). The mean baseline HbA1c was 8.6%.

The difference in HbA1c before and after use of CGM was -0.97% (P = .0001). Prior to use of a CGM the change in HbA1c was minimal, with an increase of 0.003% with the use of selfmonitoring glucose. After use of a CGM, HbA1c decreased by 0.971%. This reduction in HbA1c would also be considered clinically significant as the change was > 0.5%. The mean pre-, at start, and post-CGM HbA1c levels were 8.6%, 8.6%, and 7.6%, respectively (Figure 2). Pharmacy prescribers had a 0.7% reduction in HbA1c post-CGM, the least of all prescribers. While most age groups saw a reduction in HbA1c, those aged ≥ 80 years had an increase of 0.18% (Table 3). There was an overall mean reduction in insulin of 22 units, which was similar between all prescribers.

Abbreviation: CGM, continuous glucose monitor.

Discussion

The primary endpoint of difference in change of HbA1c before and after CGM use was found to be statistically and clinically significant, with a nearly 1% reduction in HbA1c, which was similar to the reduction found by Vigersky and colleagues. 5 Across all prescribers, post-CGM HbA1c levels were similar; however, patients with CGM prescribed by pharmacists had the smallest change in HbA1c. VA pharmacists primarily assess veterans taking insulin who have HbA1c levels that are below the goal with the aim of decreasing insulin to reduce the risk of hypoglycemia, which could result in increased HbA1c levels. This may also explain the observed increase in post-CGM HbA1c levels in patients aged ≥ 80 years. Patients under the care of pharmacists also had baseline mean HbA1c levels that were lower than primary care and endocrinology prescribers and were closer to their HbA1c goal at baseline, which likely was reflected in the smaller reduction in post-CGM HbA1c level.

While there was a decrease in HbA1c levels with CGM use, there were also changes to medications during this timeframe that also may have impacted HbA1c levels. The most common diabetes medications started during CGM use were GLP-1 agonists and SGLT2-inhibitors. Additionally, there was a reduction in the total daily dose of insulin in the study population. These results demonstrate the potential benefits of CGMs for prescribers who take advantage of the CGM glucose data available to assist with medication adjustments. Another consideration for differences in changes of HbA1c among prescriber types is the opportunity for more frequent follow- up visits with pharmacy or endocrinology compared with primary care. If veterans are followed more closely, it may be associated with improved HbA1c control. Further research investigating changes in HbA1c levels based on followup frequency may be useful.

Strengths and Limitations

The crossover design was a strength of this study. This design reduced confounding variables by having veterans serve as their own controls. In addition, the collection of multiple secondary outcomes adds to the knowledge base for future studies. This study focused on a unique population of veterans with T2DM who were taking insulin, an area that previously had very little data available to determine the benefits of CGM use.

Although the use of a CGM showed statistical significance in lowering HbA1c, many veterans were started on new diabetes medication during the period of CGM use, which also likely contributed to the reduction in HbA1c and may have confounded the results. The study was limited by its small population size due to time constraints of chart reviews and the limited generalizability of results outside of the VA system. The majority of patients were from a single site, male and identified as White, which may not be reflective of other VA and community health care systems. It was also noted that the time from the initiation of CGM use to the most recent HbA1c level varied from 6 months to several years. Additionally, veterans managed by community-based HCPs with complex diabetes cases were excluded.

Conclusions

This study demonstrated a clinically and statistically significant reduction in HbA1c with the use of a CGM compared to fingerstick monitoring in veterans with T2DM who were being treated with insulin. The change in post-CGM HbA1c levels across prescribers was similar. In the subgroup analysis of change in HbA1c among age groups, there was a lower HbA1c reduction in individuals aged ≥ 80 years. The results from this study support the idea that CGM use may be beneficial for patients who require a reduction in HbA1c by allowing more precise adjustments to medications and optimization of therapy, as well as the potential to reduce insulin requirements, which is especially valuable in the older adult veteran population.

References
  1. US Department of Veterans Affairs. VA supports veterans who have type 2 diabetes. VA News. Accessed September 30, 2024. https://news.va.gov/107579/va-supports-veterans-who-have-type-2-diabetes/
  2. ElSayed NA, Aleppo G, Aroda VR, et al. 9. Pharmacologic approaches to glycemic treatment: standards of care in diabetes-2023. Diabetes Care. 2023;46(Suppl 1):S140- S157. doi:10.2337/dc23-S009
  3. ElSayed NA, Aleppo G, Aroda VR, et al. 6. Glycemic targets: standards of care in diabetes-2023. Diabetes Care. 2023;46(Suppl 1):S97-S110. doi:10.2337/dc23-S006
  4. Miller E, Gavin JR, Kruger DF, Brunton SA. Continuous glucose monitoring: optimizing diabetes care: executive summary. Clin Diabetes. 2022;40(4):394-398. doi:10.2337/cd22-0043
  5. Vigersky RA, Fonda SJ, Chellappa M, Walker MS, Ehrhardt NM. Short- and long-term effects of real-time continuous glucose monitoring in patients with type 2 diabetes. Diabetes Care. 2012;35(1):32-38. doi:10.2337/dc11-1438
  6. Karter AJ, Parker MM, Moffet HH, Gilliam LK, Dlott R. Association of real-time continuous glucose monitoring with glycemic control and acute metabolic events among patients with insulin-treated diabetes. JAMA. 2021;325(22):2273-2284. doi:10.1001/JAMA.2021.6530
  7. Langford SN, Lane M, Karounos D. Continuous blood glucose monitoring outcomes in veterans with type 2 diabetes. Fed Pract. 2021;38(Suppl 4):S14-S17. doi:10.12788/fp.0189
  8. Radin MS. Pitfalls in hemoglobin A1c measurement: when results may be misleading. J Gen Intern Med. 2014;29(2):388-394. doi:10.1007/s11606-013-2595-x.
  9. Little RR, Rohlfing CL, Sacks DB; National Glycohemoglobin Standardization Program (NGSP) steering committee. Status of hemoglobin A1c measurement and goals for improvement: from chaos to order for improving diabetes care. Clin Chem. 2011;57(2):205-214. doi:10.1373/clinchem.2010.148841
References
  1. US Department of Veterans Affairs. VA supports veterans who have type 2 diabetes. VA News. Accessed September 30, 2024. https://news.va.gov/107579/va-supports-veterans-who-have-type-2-diabetes/
  2. ElSayed NA, Aleppo G, Aroda VR, et al. 9. Pharmacologic approaches to glycemic treatment: standards of care in diabetes-2023. Diabetes Care. 2023;46(Suppl 1):S140- S157. doi:10.2337/dc23-S009
  3. ElSayed NA, Aleppo G, Aroda VR, et al. 6. Glycemic targets: standards of care in diabetes-2023. Diabetes Care. 2023;46(Suppl 1):S97-S110. doi:10.2337/dc23-S006
  4. Miller E, Gavin JR, Kruger DF, Brunton SA. Continuous glucose monitoring: optimizing diabetes care: executive summary. Clin Diabetes. 2022;40(4):394-398. doi:10.2337/cd22-0043
  5. Vigersky RA, Fonda SJ, Chellappa M, Walker MS, Ehrhardt NM. Short- and long-term effects of real-time continuous glucose monitoring in patients with type 2 diabetes. Diabetes Care. 2012;35(1):32-38. doi:10.2337/dc11-1438
  6. Karter AJ, Parker MM, Moffet HH, Gilliam LK, Dlott R. Association of real-time continuous glucose monitoring with glycemic control and acute metabolic events among patients with insulin-treated diabetes. JAMA. 2021;325(22):2273-2284. doi:10.1001/JAMA.2021.6530
  7. Langford SN, Lane M, Karounos D. Continuous blood glucose monitoring outcomes in veterans with type 2 diabetes. Fed Pract. 2021;38(Suppl 4):S14-S17. doi:10.12788/fp.0189
  8. Radin MS. Pitfalls in hemoglobin A1c measurement: when results may be misleading. J Gen Intern Med. 2014;29(2):388-394. doi:10.1007/s11606-013-2595-x.
  9. Little RR, Rohlfing CL, Sacks DB; National Glycohemoglobin Standardization Program (NGSP) steering committee. Status of hemoglobin A1c measurement and goals for improvement: from chaos to order for improving diabetes care. Clin Chem. 2011;57(2):205-214. doi:10.1373/clinchem.2010.148841
Issue
Federal Practitioner - 41(11)s
Issue
Federal Practitioner - 41(11)s
Page Number
S1-S5
Page Number
S1-S5
Publications
Publications
Topics
Article Type
Display Headline
Continuous Glucose Monitoring vs Fingerstick Monitoring for Hemoglobin A1c Control in Veterans
Display Headline
Continuous Glucose Monitoring vs Fingerstick Monitoring for Hemoglobin A1c Control in Veterans
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Tue, 10/29/2024 - 10:45
Un-Gate On Date
Tue, 10/29/2024 - 10:45
Use ProPublica
CFC Schedule Remove Status
Tue, 10/29/2024 - 10:45
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
survey writer start date
Wed, 11/06/2024 - 12:47
Article PDF Media

VA Cancer Clinical Trials as a Strategy for Increasing Accrual of Racial and Ethnic Underrepresented Groups

Article Type
Changed
Thu, 11/07/2024 - 09:29

Background

Cancer clinical trials (CCTs) are central to improving cancer care. However, generalizability of findings from CCTs is difficult due to the lack of diversity in most United States CCTs. Clinical trial accrual of underrepresented groups, is low throughout the United States and is approximately 4-5% in most CCTs. Reasons for low accrual in this population are multifactorial. Despite numerous factors related to accruing racial and ethnic underrepresented groups, many institutions have sought to address these barriers. We conducted a scoping review to identify evidence-based approaches to increase participation in cancer treatment clinical trials.

Methods

We reviewed the Salisbury VA Medical Center Oncology clinical trial database from October 2019 to June 2024. The participants in these clinical trials required consent. These clinical trials included treatment interventional as well as non-treatment interventional. Fifteen studies were included and over 260 Veterans participated.

Results

Key themes emerged that included a focus on patient education, cultural competency, and building capacity in the clinics to care for the Veteran population at three separate sites in the Salisbury VA system. The Black Veteran accrual rate of 29% was achieved. This accrual rate is representative of our VA catchment population of 33% for Black Veterans, and is five times the national average.

Conclusions

The research team’s success in enrolling Black Veterans in clinical trials is attributed to several factors. The demographic composition of Veterans served by the Salisbury, Charlotte, and Kernersville VA provided a diverse population that included a 33% Black group. The type of clinical trials focused on patients who were most impacted by the disease. The VA did afford less barriers to access to health care.

Issue
Federal Practitioner - 41(9)s
Publications
Topics
Page Number
S43
Sections

Background

Cancer clinical trials (CCTs) are central to improving cancer care. However, generalizability of findings from CCTs is difficult due to the lack of diversity in most United States CCTs. Clinical trial accrual of underrepresented groups, is low throughout the United States and is approximately 4-5% in most CCTs. Reasons for low accrual in this population are multifactorial. Despite numerous factors related to accruing racial and ethnic underrepresented groups, many institutions have sought to address these barriers. We conducted a scoping review to identify evidence-based approaches to increase participation in cancer treatment clinical trials.

Methods

We reviewed the Salisbury VA Medical Center Oncology clinical trial database from October 2019 to June 2024. The participants in these clinical trials required consent. These clinical trials included treatment interventional as well as non-treatment interventional. Fifteen studies were included and over 260 Veterans participated.

Results

Key themes emerged that included a focus on patient education, cultural competency, and building capacity in the clinics to care for the Veteran population at three separate sites in the Salisbury VA system. The Black Veteran accrual rate of 29% was achieved. This accrual rate is representative of our VA catchment population of 33% for Black Veterans, and is five times the national average.

Conclusions

The research team’s success in enrolling Black Veterans in clinical trials is attributed to several factors. The demographic composition of Veterans served by the Salisbury, Charlotte, and Kernersville VA provided a diverse population that included a 33% Black group. The type of clinical trials focused on patients who were most impacted by the disease. The VA did afford less barriers to access to health care.

Background

Cancer clinical trials (CCTs) are central to improving cancer care. However, generalizability of findings from CCTs is difficult due to the lack of diversity in most United States CCTs. Clinical trial accrual of underrepresented groups, is low throughout the United States and is approximately 4-5% in most CCTs. Reasons for low accrual in this population are multifactorial. Despite numerous factors related to accruing racial and ethnic underrepresented groups, many institutions have sought to address these barriers. We conducted a scoping review to identify evidence-based approaches to increase participation in cancer treatment clinical trials.

Methods

We reviewed the Salisbury VA Medical Center Oncology clinical trial database from October 2019 to June 2024. The participants in these clinical trials required consent. These clinical trials included treatment interventional as well as non-treatment interventional. Fifteen studies were included and over 260 Veterans participated.

Results

Key themes emerged that included a focus on patient education, cultural competency, and building capacity in the clinics to care for the Veteran population at three separate sites in the Salisbury VA system. The Black Veteran accrual rate of 29% was achieved. This accrual rate is representative of our VA catchment population of 33% for Black Veterans, and is five times the national average.

Conclusions

The research team’s success in enrolling Black Veterans in clinical trials is attributed to several factors. The demographic composition of Veterans served by the Salisbury, Charlotte, and Kernersville VA provided a diverse population that included a 33% Black group. The type of clinical trials focused on patients who were most impacted by the disease. The VA did afford less barriers to access to health care.

Issue
Federal Practitioner - 41(9)s
Issue
Federal Practitioner - 41(9)s
Page Number
S43
Page Number
S43
Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Eyebrow Default
Research
Gate On Date
Thu, 09/12/2024 - 14:00
Un-Gate On Date
Thu, 09/12/2024 - 14:00
Use ProPublica
CFC Schedule Remove Status
Thu, 09/12/2024 - 14:00
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
survey writer start date
Wed, 09/18/2024 - 20:58

Eating Disorder Risk Factors and the Impact of Obesity in Patients With Psoriasis

Article Type
Changed
Wed, 11/13/2024 - 10:45
Display Headline
Eating Disorder Risk Factors and the Impact of Obesity in Patients With Psoriasis

Current evidence indicates that obesity may initiate psoriasis or worsen existing disease. Various factors contribute to the development of obesity, including eating disorders (EDs). The aim of this study was to screen for and identify factors associated with EDs in patients with psoriasis and their impact on the development of obesity in this population. Demographic information including body mass index (BMI), Eating Attitude Test (EAT-26), Dermatology Life Quality Index (DLQI), Attitude Scale for Healthy Nutrition (ASHN), and Depression Anxiety Stress Scale 21 (DASS-21) scores were statistically analyzed for 82 participants with psoriasis at a tertiary dermatology clinic. It is important to manage obesity and other comorbidities of psoriasis in addition to treating its cutaneous manifestations, which may require a biopsychosocial approach.

Psoriasis is a chronic multisystemic inflammatory skin disease with a worldwide prevalence of 2% to 3%.1 Psoriasis can be accompanied by other conditions such as psoriatic arthritis, obesity, metabolic syndrome, diabetes mellitus, hypertension, dyslipidemia, atherosclerotic disease, inflammatory bowel disease, and anxiety/depression. It is important to manage comorbidities of psoriasis in addition to treating the cutaneous manifestations of the disease.1

Obesity is a major public health concern worldwide. Numerous observational and epidemiologic studies have reported a high prevalence of obesity among patients with psoriasis.2 Current evidence indicates that obesity may initiate or worsen psoriasis; furthermore, it is important to note that obesity may negatively impact the effectiveness of psoriasis-specific treatments or increase the incidence of adverse effects. Therefore, managing obesity is crucial in the treatment of psoriasis.3 Numerous studies have investigated the association between psoriasis and obesity, and they commonly conclude that both conditions share the same genetic metabolic pathways.2-4 However, it is important to consider environmental factors such as dietary habits, smoking, alcohol consumption, and a sedentary lifestyle—all of which are associated with psoriasis and also can contribute to the development of obesity.5 Because of the effects of obesity in psoriasis patients, factors that impact the development of obesity have become a popular research topic.

Eating disorders (EDs) are a crucial risk factor for both developing and maintaining obesity. In particular, two EDs that are associated with obesity include binge eating disorder and bulimia nervosa.6 According to the Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition,7 binge eating disorder can be diagnosed when a patient has at least 1 episode of binge eating per week over a 3-month period. Bulimia nervosa can be diagnosed when a patient is excessively concerned with their body weight and shape and engages in behaviors to prevent weight gain (eg, forced vomiting, excessive use of laxatives).7 Psychiatrists who specialize in EDs make diagnoses based on these criteria. In daily practice, there are several quick and simple questionnaires available to screen for EDs that can be used by nonpsychiatrist physicians, including the commonly used 26-item Eating Attitudes Test (EAT-26).8 The EAT-26 has been used to screen for EDs in patients with inflammatory disorders.9

The aim of this study was to screen for EDs in patients with psoriasis to identify potential risk factors for development of obesity.

Materials and Methods

This study included patients with psoriasis who were screened for EDs at a tertiary dermatology clinic in Turkey between January 2021 and December 2023. This study was approved by the local ethics committee and was in accordance with the Declaration of Helsinki (decision number E-93471371-514.99-225000079).

Study Design and Patient Inclusion Criteria—This quantitative cross-sectional study utilized EAT-26, Dermatology Life Quality Index (DLQI), Attitude Scale for Healthy Nutrition (ASHN), and Depression Anxiety Stress Scale-21 (DASS-21) scores. All the questionnaire scales used in the study were adapted and validated in Turkey.8,10-12 The inclusion criteria consisted of being older than 18 years of age, being literate, having psoriasis for at least 1 year that was not treated topically or systemically, and having no psychiatric diseases outside an ED. The questionnaires were presented in written format following the clinical examination. Literacy was an inclusion criterion in this study due to the absence of auxiliary health personnel.

Study Variables—The study variables included age, sex, marital status (single/divorced or married), education status (primary/secondary school or high school/university), employment status (employed or unemployed/retired), body mass index (BMI), smoking status, alcohol-consumption status, Psoriasis Area Severity Index score, presence of nail psoriasis and psoriatic arthritis, duration of psoriasis, family history of psoriasis, EAT-26 score, ASHN score, DLQI score, and DASS-21 score. Body mass index was calculated by taking a participant’s weight in kilograms and dividing it by their height in meters squared. The BMI values were classified into 3 categories: normal (18.5–24.9 kg/m2), overweight (25.0–29.9 kg/m2), and obese (≥30 kg/m2).13

Questionnaires—The EAT-26 questionnaire includes 26 questions that are used to detect EDs. Responses to each question include Likert-type answer options (ie, “always,” “usually,” “often,” “sometimes,” “rarely,” and “never.”) Patients with scores of 20 points or higher (range, 0–78) are classified as high risk for EDs.8 In our study, EAT-26 scores were grouped into 2 categories: patients scoring less than 20 points and those scoring 20 points or higher.

The DLQI questionnaire includes 10 questions to measure dermatologic symptoms and qualiy of life. Responses to each question include Likert-type answer options (ie, “not at all,” “a little,” “a lot,” or “very much.”) On the DLQI scale, the higher the score, the lower the quality of life (score range, 0–30).10

The ASHN questionnaire includes 21 questions that measure attitudes toward healthy nutrition with 5 possible answer options (“strongly disagree,” “disagree,” “undecided,” “agree,” and “strongly agree”). On this scale, higher scores indicate the participant is more knowledgeable about healthy nutrition (score range, 0–78).11

The DASS-21 questionnaire includes 21 questions that measure the severity of a range of symptoms common to depression, anxiety, and stress. Responses include Likert-type answer options (eg, “never,” “sometimes,” “often,” and “almost always.”) On this scale, a higher score (range of 0–21 for each) indicates higher levels of depression, anxiety, and stress.12

Statistical Analysis—Descriptive statistics were analyzed using SPSS software version 22.0 (IBM). The Shapiro-Wilk test was applied to determine whether the data were normally distributed. For categorical variables, frequency differences among groups were compared using the Pearson χ2 test. A t test was used to compare the means of 2 independent groups with a normal distribution. One-way analysis of variance and Tukey Honest Significant Difference post hoc analysis were used to test whether there was a statistically significant difference among the normally distributed means of independent groups. Pearson correlation analysis was used to determine whether there was a linear relationship between 2 numeric measurements and, if so, to determine the direction and severity of this relationship. P<.05 indicated statistical significance in this study.

Results

Study Participant Demographics—This study included 82 participants with a mean age of 44.3 years; 52.4% (43/82) were female, and 85.4% (70/82) were married. The questionnaire took an average of 4.2 minutes for participants to complete. A total of 57.3% (47/82) of patients had completed primary/secondary education and 59.8% (49/82) were employed. The mean BMI was 28.1 kg/m2. According to the BMI classification, 26.8% (22/82) participants had a normal weight, 36.6% (30/82) were overweight, and 43.9% (36/82) were obese. A total of 48.8% (40/82) of participants smoked, and 4.9% (4/82) consumed alcohol. The mean Psoriasis Area and Severity Index score was 5.4. A total of 54.9% (45/82) of participants had nail psoriasis, and 24.4% (20/82) had psoriatic arthritis. The mean duration of psoriasis was 153 months. A total of 29.3% (24/82) of participants had a positive family history of psoriasis. The mean EAT-26 score was 11.1. A total of 12.2% (10/82) of participants had an EAT-26 score of 20 points or higher and were considered at high risk for an ED. The mean ASHN score was 72.9; the mean DLQI score was 5.5; and on the DASS-21 scale, mean scores for depression, anxiety, and stress were 6.3, 8.7, and 10.0, respectively (Table).

Comparative Evaluation of the BMI Groups—The only statistically significant differences among the 3 BMI groups were related to marital status, EAT-26 score, and anxiety and stress scores (P=.02, <.01, <.01, and <.01, respectively)(eTable 1). The number of single/divorced participants in the overweight group was significantly (P=.02) greater than in the normal weight group. The mean EAT-26 score for the normal weight group was significantly (P<.01) lower than for the overweight and obese groups; there was no significant difference in mean EAT-26 scores between the overweight and obese groups. The mean anxiety score was significantly (P<.01) lower in the normal weight group compared with the overweight and obese groups. There was no significant difference between the overweight and obese groups according to the mean depression score. The mean stress and anxiety scores were significantly (P<.01) lower in the normal weight group than in the overweight and obese groups. There was no significant difference between the overweight and obese groups according to the mean anxiety score.

Comparative Evaluation of the EAT-26 Scores—There were statistically significant differences among the EAT-26 scores related to sex; BMI; and depression, anxiety, and stress scores (P=.04, .02, <.01, <.01, and <.01, respectively). The number of females in the group with a score of 20 points or higher was significantly (P=.04) less than that in the group scoring less than 20 points. The mean BMI in the group with a score of 20 points or higher was significantly (P=.02) greater than in group scoring less than 20 points. The mean depression, anxiety, and stress scores of the group scoring 20 points or higher were significantly (P<.01 for all) greater than in the group scoring less than 20 points (eTable 2).

Correlation Analysis of the Study Variables—The EAT-26 scores were positively correlated with BMI, anxiety, depression, and stress (P<.01 for all)(eTable 3).

Comment

Eating disorders are psychiatric conditions that require a multidisciplinary approach. Nonpsychiatric medical departments may be involved due to the severe consequences (eg, various skin changes14) of these disorders. Psoriasis is not known to be directly affected by the presence of an ED; however, it is possible that EDs could indirectly affect patients with psoriasis by influencing obesity. Therefore, this study aimed to examine the relationship between ED risk factors and obesity in this population.

The relationship between psoriasis and obesity has been a popular research topic in dermatology since the 1990s.15 Epidemiologic and observational studies have reported that patients with psoriasis are more likely to be overweight or have obesity, which is an independent risk factor for psoriasis.3,16 However, the causal relationship between psoriasis and obesity remains unclear. In a comprehensive review, Barros et al17 emphasized the causal relationship between obesity and psoriasis under several headings. Firstly, a higher BMI increases the risk for psoriasis by promoting cytokine release and immune system dysregulation. Secondly, a Western diet (eg, processed foods and fast food) triggers obesity and psoriasis by increasing adipose tissue. Thirdly, the alteration of the skin and gut microbiota triggers chronic inflammation as a result of bacterial translocation in patients with obesity. Fourthly, a high-fat diet and palmitic acid disrupt the intestinal integrity of the gut and increase the risk for psoriasis and obesity by triggering chronic inflammation of bacterial fragments that pass into the blood. Finally, the decrease in the amount of adiponectin and the increase in the amount of leptin in patients with obesity may cause psoriasis by increasing proinflammatory cytokines, which are similar to those involved in the pathogenesis of psoriasis.17 Additionally, psoriatic inflammation can cause insulin resistance and metabolic dysfunction, leading to obesity.18 The relationship between psoriasis and obesity cannot be solely explained by metabolic pathways. Smoking, alcohol consumption, and a sedentary lifestyle all are associated with psoriasis and also can contribute to obesity.5 Our study revealed no significant difference in smoking or alcohol consumption between the normal weight and overweight/obesity groups. Based on our data, we determined that smoking and alcohol consumption did not affect obesity in our patients with psoriasis.

Observational and epidemiologic studies have shown that patients with psoriasis experience increased rates of depression, anxiety, and stress.19 In studies of pathogenesis, a connection between depression and psoriatic inflammation has been established.20 It is known that inflammatory cytokines similar to those in psoriasis are involved in the development of obesity.18 In addition, depression and anxiety can lead to binge eating, unhealthy food choices, and a more sedentary lifestyle.5 All of these variables may contribute to the associations between depression and anxiety with psoriasis and obesity. Zafiriou et al21 conducted a study to investigate the relationship between psoriasis, obesity, and depression through inflammatory pathways with a focus on the importance of IL-17. Data showing that IL-17–producing Th17-cell subgroups play a considerable role in the development of obesity and depression prompted the authors to suggest that psoriasis, obesity, and ­anxiety/depression may be interconnected manifestations of immune dysregulation, potentially linked to IL-17 and its associated cells.21 Mrowietz et al22 also suggested that metabolic inflammation may contribute to obesity and depression in patients with psoriasis and highlighted the importance of several cytokines, including tumor necrosis factor α, IL-6, IL-8, IL-17, and IL-23. Our study revealed no significant differences in depression scores between BMI groups. Another meta-analysis reported conflicting findings on the incidence of depression in obese patients with psoriasis.23 Some of the studies had a small number of participants. Compared to depression, anxiety has received less attention in studies of patients with obesity with psoriasis. However, these studies have shown a positive correlation between anxiety scores and BMI in patients with psoriasis.24,25 In our study, similar to the findings of previous studies, overweight patients and those with obesitywho have psoriasis had significantly (P<.01) greater anxiety and stress scores than did normal weight patients with psoriasis.

Obesity should be assessed in patients with psoriasis via a biopsychosocial approach that takes into account genetic, behavioral, and environmental factors.26 Eating disorders are considered to be one of the factors contributing to obesity. Numerous studies in the literature have demonstrated a greater incidence of EDs in patients with obesity vs those without obesity.5,6,27 Obesity and EDs have a bidirectional relationship: individuals with obesity are at risk for EDs due to body dissatisfaction, dieting habits, and depressive states. Conversely, poor eating behaviors in individuals with a normal weight can lead to obesity.28

There are few studies in the literature exploring the relationship between psoriasis and EDs. Crosta et al29 demonstrated that patients with psoriasis had impaired results on ED screening tests and that these scores deteriorated further as BMI increased. Moreover, Altunay et al30 demonstrated that patients with psoriasis and metabolic syndrome had higher scores on the ED screening test. In this study, patients with higher scores also exhibited high levels of anxiety.30 In our study, similar to the findings of previous studies, patients with psoriasis who were overweight or had obesity had significantly (P<.01) greater EAT-26 scores than those in the normal weight group. Patients with high EAT-26 scores also exhibited elevated levels of depression, anxiety, and stress. Additionally, EAT-26 scores were positively correlated with BMI, anxiety, depression, and stress scores. Our study as well as other studies in the literature indicate that additional research is needed to determine the associations between EDs and obesity in psoriasis.

Conclusion

Managing obesity is crucial for patients with psoriasis. This study showed that EAT-26 scores were higher in patients with psoriasis who were overweight or had obesity than in those who were normal weight. Participants with high EAT-26 scores (≥20 points) were more likely to be female and have higher anxiety and stress scores. In addition, EAT-26 scores were positively correlated with BMI as well as depression, anxiety, and stress scores. Eating disorders may contribute to the development of obesity in patients with psoriasis. Although our study was limited by a small sample size, the results suggest that there is a need for large-scale multicenter studies to investigate the relationship between psoriasis and EDs.

References

1. Kalkan G. Comorbidities in psoriasis: the recognition of psoriasis as a systemic disease and current management. Turkderm-Turk Arch Dermatol Venereol. 2017;51:71-77.

2. Armstrong AW, Harskamp CT, Armstrong EJ. The association between psoriasis and obesity: a systematic review and meta-analysis of observational studies. Nutr Diabetes. 2012;2:E54.

3. Jensen P, Skov L. Psoriasis and obesity. Dermatology. 2016;232:633-639.

4. Mirghani H, Altemani AT, Altemani ST, et al. The cross talk between psoriasis, obesity, and dyslipidemia: a meta-analysis. Cureus. 2023;15:e49253.

5. Roehring M, Mashep MR, White MA, et al. The metabolic syndrome and behavioral correlates in obese patients with binge disorders. Obesity. 2009;17:481-486.

6. da Luz FQ, Hay P, Touyz S, et al. Obesity with comorbid eating disorders: associated health risks and treatment approaches. Nutrients. 2018;10:829.

7. American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition. American Psychiatric Association; 2013.

8. Ergüney Okumus¸ FE, Sertel Berk HÖ. The psychometric properties of the Eating Attitudes Test short form (EAT-26) in a college sample. Stud Psychol. 2020;40:57-78.

9. Stoleru G, Leopold A, Auerbach A, et al. Female gender, dissatisfaction with weight, and number of IBD related surgeries as independent risk factors for eating disorders among patients with inflammatory bowel diseases. BMC Gastroenterol. 2022;22:438.

10. Öztürkcan S, Ermertcan AT, Eser E, et al. Cross validation of the Turkish version of dermatology life quality index. Int J Dermatol. 2006;45:1300-1307.

11. Demir GT, Ciciog˘lu HI˙. Attitude scale for healthy nutrition (ASHN): validity and reliability study. Gaziantep Univ J Sport Sci. 2019;4:256-274.

12. Yılmaz O, Boz H, Arslan A. The validity and reliability of depression stress and anxiety scale (DASS 21) Turkish short form. Res Financial Econ Soc Stud. 2017;2:78-91.

13. Nuttall FQ. Body mass index: obesity, BMI, and health: a critical review. Nutr Today. 2015;50:117-128.

14. Strumia R, Manzata E, Gualandi M. Is there a role for dermatologists in eating disorders? Expert Rev Dermatol. 2017; 2:109-112.

15. Henseler T, Christophers E. Disease concomitance in psoriasis. J Am Acad Dermatol. 1995;32:982-986.

16. Naldi L, Addis A, Chimenti S, et al. Impact of body mass index and obesity on clinical response to systemic treatment for psoriasis. evidence from the Psocare project. Dermatology. 2008;217:365-373.

17. Barros G, Duran P, Vera I, et al. Exploring the links between obesity and psoriasis: a comprehensive review. Int J Mol Sci. 2022;23:7499.

18. Hao Y, Zhu YJ, Zou S, et al. Metabolic syndrome and psoriasis: mechanisms and future directions. Front Immunol. 2021;12:711060.

19. Jing D, Xiao H, Shen M, et al. Association of psoriasis with anxiety and depression: a case–control study in Chinese patients. Front Med (Lausanne). 2021;8:771645.

20. Sahi FM, Masood A, Danawar NA, et al. Association between psoriasis and depression: a traditional review. Cureus. 2020;12:E9708.

21. Zafiriou E, Daponte AI, Siokas V, et al. Depression and obesity in patients with psoriasis and psoriatic arthritis: is IL-17–mediated immune dysregulation the connecting link? Front Immunol. 2021;12:699848.

22. Mrowietz U, Sümbül M, Gerdes S. Depression, a major comorbidity of psoriatic disease, is caused by metabolic inflammation. J Eur Acad Dermatol Venereol. 2023;37:1731-1738.

23. Pavlova NT, Kioskli K, Smith C, et al. Psychosocial aspects of obesity in adults with psoriasis: a systematic review. Skin Health Dis. 2021;1:E33.

24. Innamorati M, Quinto RM, Imperatori C, et al. Health-related quality of life and its association with alexithymia and difficulties in emotion regulation in patients with psoriasis. Compr Psychiatry. 2016;70:200-208.

25. Tabolli S, Naldi L, Pagliarello C, et al. Evaluation of the impact of writing exercises interventions on quality of life in patients with psoriasis undergoing systemic treatments. Br J Dermatol. 2012;167:1254‐1264.

26. Albuquerque D, Nóbrega C, Manco L, et al. The contribution of genetics and environment to obesity. Br Med Bull. 2017;123:159‐173.

27. Balantekin KN, Grammer AC, Fitzsimmons-Craft EE, et al. Overweight and obesity are associated with increased eating disorder correlates and general psychopathology in university women with eating disorders. Eat Behav. 2021;41:101482.

28. Jebeile H, Lister NB, Baur LA, et al. Eating disorder risk in adolescents with obesity. Obes Rev. 2021;22:E13173.

29. Crosta ML, Caldarola G, Fraietta S, et al. Psychopathology and eating disorders in patients with psoriasis. G Ital Dermatol Venereol. 2014;149:355-361.

30. Altunay I, Demirci GT, Ates B, et al. Do eating disorders accompany metabolic syndrome in psoriasis patients? results of a preliminary study. Clin Cosmet Investig Dermatol. 2011;4:139-143.

Article PDF
Author and Disclosure Information

From the Department of Dermatology, Ministry of Health, Ankara Training and Research Hospital, Turkey.

The authors have no relevant financial disclosures to report.

The eTables are available in the Appendix online at www.mdedge.com/dermatology.

Correspondence: Berkay Temel, MD, Department of Dermatology, Ankara Training and Research Hospital, Ulucanlar St No: 89, Ankara, Turkey ([email protected]).

Cutis. 2024 November;114(5):164-168, E1-E5. doi:10.12788/cutis.1130

Publications
Topics
Page Number
164-168
Sections
Author and Disclosure Information

From the Department of Dermatology, Ministry of Health, Ankara Training and Research Hospital, Turkey.

The authors have no relevant financial disclosures to report.

The eTables are available in the Appendix online at www.mdedge.com/dermatology.

Correspondence: Berkay Temel, MD, Department of Dermatology, Ankara Training and Research Hospital, Ulucanlar St No: 89, Ankara, Turkey ([email protected]).

Cutis. 2024 November;114(5):164-168, E1-E5. doi:10.12788/cutis.1130

Author and Disclosure Information

From the Department of Dermatology, Ministry of Health, Ankara Training and Research Hospital, Turkey.

The authors have no relevant financial disclosures to report.

The eTables are available in the Appendix online at www.mdedge.com/dermatology.

Correspondence: Berkay Temel, MD, Department of Dermatology, Ankara Training and Research Hospital, Ulucanlar St No: 89, Ankara, Turkey ([email protected]).

Cutis. 2024 November;114(5):164-168, E1-E5. doi:10.12788/cutis.1130

Article PDF
Article PDF

Current evidence indicates that obesity may initiate psoriasis or worsen existing disease. Various factors contribute to the development of obesity, including eating disorders (EDs). The aim of this study was to screen for and identify factors associated with EDs in patients with psoriasis and their impact on the development of obesity in this population. Demographic information including body mass index (BMI), Eating Attitude Test (EAT-26), Dermatology Life Quality Index (DLQI), Attitude Scale for Healthy Nutrition (ASHN), and Depression Anxiety Stress Scale 21 (DASS-21) scores were statistically analyzed for 82 participants with psoriasis at a tertiary dermatology clinic. It is important to manage obesity and other comorbidities of psoriasis in addition to treating its cutaneous manifestations, which may require a biopsychosocial approach.

Psoriasis is a chronic multisystemic inflammatory skin disease with a worldwide prevalence of 2% to 3%.1 Psoriasis can be accompanied by other conditions such as psoriatic arthritis, obesity, metabolic syndrome, diabetes mellitus, hypertension, dyslipidemia, atherosclerotic disease, inflammatory bowel disease, and anxiety/depression. It is important to manage comorbidities of psoriasis in addition to treating the cutaneous manifestations of the disease.1

Obesity is a major public health concern worldwide. Numerous observational and epidemiologic studies have reported a high prevalence of obesity among patients with psoriasis.2 Current evidence indicates that obesity may initiate or worsen psoriasis; furthermore, it is important to note that obesity may negatively impact the effectiveness of psoriasis-specific treatments or increase the incidence of adverse effects. Therefore, managing obesity is crucial in the treatment of psoriasis.3 Numerous studies have investigated the association between psoriasis and obesity, and they commonly conclude that both conditions share the same genetic metabolic pathways.2-4 However, it is important to consider environmental factors such as dietary habits, smoking, alcohol consumption, and a sedentary lifestyle—all of which are associated with psoriasis and also can contribute to the development of obesity.5 Because of the effects of obesity in psoriasis patients, factors that impact the development of obesity have become a popular research topic.

Eating disorders (EDs) are a crucial risk factor for both developing and maintaining obesity. In particular, two EDs that are associated with obesity include binge eating disorder and bulimia nervosa.6 According to the Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition,7 binge eating disorder can be diagnosed when a patient has at least 1 episode of binge eating per week over a 3-month period. Bulimia nervosa can be diagnosed when a patient is excessively concerned with their body weight and shape and engages in behaviors to prevent weight gain (eg, forced vomiting, excessive use of laxatives).7 Psychiatrists who specialize in EDs make diagnoses based on these criteria. In daily practice, there are several quick and simple questionnaires available to screen for EDs that can be used by nonpsychiatrist physicians, including the commonly used 26-item Eating Attitudes Test (EAT-26).8 The EAT-26 has been used to screen for EDs in patients with inflammatory disorders.9

The aim of this study was to screen for EDs in patients with psoriasis to identify potential risk factors for development of obesity.

Materials and Methods

This study included patients with psoriasis who were screened for EDs at a tertiary dermatology clinic in Turkey between January 2021 and December 2023. This study was approved by the local ethics committee and was in accordance with the Declaration of Helsinki (decision number E-93471371-514.99-225000079).

Study Design and Patient Inclusion Criteria—This quantitative cross-sectional study utilized EAT-26, Dermatology Life Quality Index (DLQI), Attitude Scale for Healthy Nutrition (ASHN), and Depression Anxiety Stress Scale-21 (DASS-21) scores. All the questionnaire scales used in the study were adapted and validated in Turkey.8,10-12 The inclusion criteria consisted of being older than 18 years of age, being literate, having psoriasis for at least 1 year that was not treated topically or systemically, and having no psychiatric diseases outside an ED. The questionnaires were presented in written format following the clinical examination. Literacy was an inclusion criterion in this study due to the absence of auxiliary health personnel.

Study Variables—The study variables included age, sex, marital status (single/divorced or married), education status (primary/secondary school or high school/university), employment status (employed or unemployed/retired), body mass index (BMI), smoking status, alcohol-consumption status, Psoriasis Area Severity Index score, presence of nail psoriasis and psoriatic arthritis, duration of psoriasis, family history of psoriasis, EAT-26 score, ASHN score, DLQI score, and DASS-21 score. Body mass index was calculated by taking a participant’s weight in kilograms and dividing it by their height in meters squared. The BMI values were classified into 3 categories: normal (18.5–24.9 kg/m2), overweight (25.0–29.9 kg/m2), and obese (≥30 kg/m2).13

Questionnaires—The EAT-26 questionnaire includes 26 questions that are used to detect EDs. Responses to each question include Likert-type answer options (ie, “always,” “usually,” “often,” “sometimes,” “rarely,” and “never.”) Patients with scores of 20 points or higher (range, 0–78) are classified as high risk for EDs.8 In our study, EAT-26 scores were grouped into 2 categories: patients scoring less than 20 points and those scoring 20 points or higher.

The DLQI questionnaire includes 10 questions to measure dermatologic symptoms and qualiy of life. Responses to each question include Likert-type answer options (ie, “not at all,” “a little,” “a lot,” or “very much.”) On the DLQI scale, the higher the score, the lower the quality of life (score range, 0–30).10

The ASHN questionnaire includes 21 questions that measure attitudes toward healthy nutrition with 5 possible answer options (“strongly disagree,” “disagree,” “undecided,” “agree,” and “strongly agree”). On this scale, higher scores indicate the participant is more knowledgeable about healthy nutrition (score range, 0–78).11

The DASS-21 questionnaire includes 21 questions that measure the severity of a range of symptoms common to depression, anxiety, and stress. Responses include Likert-type answer options (eg, “never,” “sometimes,” “often,” and “almost always.”) On this scale, a higher score (range of 0–21 for each) indicates higher levels of depression, anxiety, and stress.12

Statistical Analysis—Descriptive statistics were analyzed using SPSS software version 22.0 (IBM). The Shapiro-Wilk test was applied to determine whether the data were normally distributed. For categorical variables, frequency differences among groups were compared using the Pearson χ2 test. A t test was used to compare the means of 2 independent groups with a normal distribution. One-way analysis of variance and Tukey Honest Significant Difference post hoc analysis were used to test whether there was a statistically significant difference among the normally distributed means of independent groups. Pearson correlation analysis was used to determine whether there was a linear relationship between 2 numeric measurements and, if so, to determine the direction and severity of this relationship. P<.05 indicated statistical significance in this study.

Results

Study Participant Demographics—This study included 82 participants with a mean age of 44.3 years; 52.4% (43/82) were female, and 85.4% (70/82) were married. The questionnaire took an average of 4.2 minutes for participants to complete. A total of 57.3% (47/82) of patients had completed primary/secondary education and 59.8% (49/82) were employed. The mean BMI was 28.1 kg/m2. According to the BMI classification, 26.8% (22/82) participants had a normal weight, 36.6% (30/82) were overweight, and 43.9% (36/82) were obese. A total of 48.8% (40/82) of participants smoked, and 4.9% (4/82) consumed alcohol. The mean Psoriasis Area and Severity Index score was 5.4. A total of 54.9% (45/82) of participants had nail psoriasis, and 24.4% (20/82) had psoriatic arthritis. The mean duration of psoriasis was 153 months. A total of 29.3% (24/82) of participants had a positive family history of psoriasis. The mean EAT-26 score was 11.1. A total of 12.2% (10/82) of participants had an EAT-26 score of 20 points or higher and were considered at high risk for an ED. The mean ASHN score was 72.9; the mean DLQI score was 5.5; and on the DASS-21 scale, mean scores for depression, anxiety, and stress were 6.3, 8.7, and 10.0, respectively (Table).

Comparative Evaluation of the BMI Groups—The only statistically significant differences among the 3 BMI groups were related to marital status, EAT-26 score, and anxiety and stress scores (P=.02, <.01, <.01, and <.01, respectively)(eTable 1). The number of single/divorced participants in the overweight group was significantly (P=.02) greater than in the normal weight group. The mean EAT-26 score for the normal weight group was significantly (P<.01) lower than for the overweight and obese groups; there was no significant difference in mean EAT-26 scores between the overweight and obese groups. The mean anxiety score was significantly (P<.01) lower in the normal weight group compared with the overweight and obese groups. There was no significant difference between the overweight and obese groups according to the mean depression score. The mean stress and anxiety scores were significantly (P<.01) lower in the normal weight group than in the overweight and obese groups. There was no significant difference between the overweight and obese groups according to the mean anxiety score.

Comparative Evaluation of the EAT-26 Scores—There were statistically significant differences among the EAT-26 scores related to sex; BMI; and depression, anxiety, and stress scores (P=.04, .02, <.01, <.01, and <.01, respectively). The number of females in the group with a score of 20 points or higher was significantly (P=.04) less than that in the group scoring less than 20 points. The mean BMI in the group with a score of 20 points or higher was significantly (P=.02) greater than in group scoring less than 20 points. The mean depression, anxiety, and stress scores of the group scoring 20 points or higher were significantly (P<.01 for all) greater than in the group scoring less than 20 points (eTable 2).

Correlation Analysis of the Study Variables—The EAT-26 scores were positively correlated with BMI, anxiety, depression, and stress (P<.01 for all)(eTable 3).

Comment

Eating disorders are psychiatric conditions that require a multidisciplinary approach. Nonpsychiatric medical departments may be involved due to the severe consequences (eg, various skin changes14) of these disorders. Psoriasis is not known to be directly affected by the presence of an ED; however, it is possible that EDs could indirectly affect patients with psoriasis by influencing obesity. Therefore, this study aimed to examine the relationship between ED risk factors and obesity in this population.

The relationship between psoriasis and obesity has been a popular research topic in dermatology since the 1990s.15 Epidemiologic and observational studies have reported that patients with psoriasis are more likely to be overweight or have obesity, which is an independent risk factor for psoriasis.3,16 However, the causal relationship between psoriasis and obesity remains unclear. In a comprehensive review, Barros et al17 emphasized the causal relationship between obesity and psoriasis under several headings. Firstly, a higher BMI increases the risk for psoriasis by promoting cytokine release and immune system dysregulation. Secondly, a Western diet (eg, processed foods and fast food) triggers obesity and psoriasis by increasing adipose tissue. Thirdly, the alteration of the skin and gut microbiota triggers chronic inflammation as a result of bacterial translocation in patients with obesity. Fourthly, a high-fat diet and palmitic acid disrupt the intestinal integrity of the gut and increase the risk for psoriasis and obesity by triggering chronic inflammation of bacterial fragments that pass into the blood. Finally, the decrease in the amount of adiponectin and the increase in the amount of leptin in patients with obesity may cause psoriasis by increasing proinflammatory cytokines, which are similar to those involved in the pathogenesis of psoriasis.17 Additionally, psoriatic inflammation can cause insulin resistance and metabolic dysfunction, leading to obesity.18 The relationship between psoriasis and obesity cannot be solely explained by metabolic pathways. Smoking, alcohol consumption, and a sedentary lifestyle all are associated with psoriasis and also can contribute to obesity.5 Our study revealed no significant difference in smoking or alcohol consumption between the normal weight and overweight/obesity groups. Based on our data, we determined that smoking and alcohol consumption did not affect obesity in our patients with psoriasis.

Observational and epidemiologic studies have shown that patients with psoriasis experience increased rates of depression, anxiety, and stress.19 In studies of pathogenesis, a connection between depression and psoriatic inflammation has been established.20 It is known that inflammatory cytokines similar to those in psoriasis are involved in the development of obesity.18 In addition, depression and anxiety can lead to binge eating, unhealthy food choices, and a more sedentary lifestyle.5 All of these variables may contribute to the associations between depression and anxiety with psoriasis and obesity. Zafiriou et al21 conducted a study to investigate the relationship between psoriasis, obesity, and depression through inflammatory pathways with a focus on the importance of IL-17. Data showing that IL-17–producing Th17-cell subgroups play a considerable role in the development of obesity and depression prompted the authors to suggest that psoriasis, obesity, and ­anxiety/depression may be interconnected manifestations of immune dysregulation, potentially linked to IL-17 and its associated cells.21 Mrowietz et al22 also suggested that metabolic inflammation may contribute to obesity and depression in patients with psoriasis and highlighted the importance of several cytokines, including tumor necrosis factor α, IL-6, IL-8, IL-17, and IL-23. Our study revealed no significant differences in depression scores between BMI groups. Another meta-analysis reported conflicting findings on the incidence of depression in obese patients with psoriasis.23 Some of the studies had a small number of participants. Compared to depression, anxiety has received less attention in studies of patients with obesity with psoriasis. However, these studies have shown a positive correlation between anxiety scores and BMI in patients with psoriasis.24,25 In our study, similar to the findings of previous studies, overweight patients and those with obesitywho have psoriasis had significantly (P<.01) greater anxiety and stress scores than did normal weight patients with psoriasis.

Obesity should be assessed in patients with psoriasis via a biopsychosocial approach that takes into account genetic, behavioral, and environmental factors.26 Eating disorders are considered to be one of the factors contributing to obesity. Numerous studies in the literature have demonstrated a greater incidence of EDs in patients with obesity vs those without obesity.5,6,27 Obesity and EDs have a bidirectional relationship: individuals with obesity are at risk for EDs due to body dissatisfaction, dieting habits, and depressive states. Conversely, poor eating behaviors in individuals with a normal weight can lead to obesity.28

There are few studies in the literature exploring the relationship between psoriasis and EDs. Crosta et al29 demonstrated that patients with psoriasis had impaired results on ED screening tests and that these scores deteriorated further as BMI increased. Moreover, Altunay et al30 demonstrated that patients with psoriasis and metabolic syndrome had higher scores on the ED screening test. In this study, patients with higher scores also exhibited high levels of anxiety.30 In our study, similar to the findings of previous studies, patients with psoriasis who were overweight or had obesity had significantly (P<.01) greater EAT-26 scores than those in the normal weight group. Patients with high EAT-26 scores also exhibited elevated levels of depression, anxiety, and stress. Additionally, EAT-26 scores were positively correlated with BMI, anxiety, depression, and stress scores. Our study as well as other studies in the literature indicate that additional research is needed to determine the associations between EDs and obesity in psoriasis.

Conclusion

Managing obesity is crucial for patients with psoriasis. This study showed that EAT-26 scores were higher in patients with psoriasis who were overweight or had obesity than in those who were normal weight. Participants with high EAT-26 scores (≥20 points) were more likely to be female and have higher anxiety and stress scores. In addition, EAT-26 scores were positively correlated with BMI as well as depression, anxiety, and stress scores. Eating disorders may contribute to the development of obesity in patients with psoriasis. Although our study was limited by a small sample size, the results suggest that there is a need for large-scale multicenter studies to investigate the relationship between psoriasis and EDs.

References

1. Kalkan G. Comorbidities in psoriasis: the recognition of psoriasis as a systemic disease and current management. Turkderm-Turk Arch Dermatol Venereol. 2017;51:71-77.

2. Armstrong AW, Harskamp CT, Armstrong EJ. The association between psoriasis and obesity: a systematic review and meta-analysis of observational studies. Nutr Diabetes. 2012;2:E54.

3. Jensen P, Skov L. Psoriasis and obesity. Dermatology. 2016;232:633-639.

4. Mirghani H, Altemani AT, Altemani ST, et al. The cross talk between psoriasis, obesity, and dyslipidemia: a meta-analysis. Cureus. 2023;15:e49253.

5. Roehring M, Mashep MR, White MA, et al. The metabolic syndrome and behavioral correlates in obese patients with binge disorders. Obesity. 2009;17:481-486.

6. da Luz FQ, Hay P, Touyz S, et al. Obesity with comorbid eating disorders: associated health risks and treatment approaches. Nutrients. 2018;10:829.

7. American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition. American Psychiatric Association; 2013.

8. Ergüney Okumus¸ FE, Sertel Berk HÖ. The psychometric properties of the Eating Attitudes Test short form (EAT-26) in a college sample. Stud Psychol. 2020;40:57-78.

9. Stoleru G, Leopold A, Auerbach A, et al. Female gender, dissatisfaction with weight, and number of IBD related surgeries as independent risk factors for eating disorders among patients with inflammatory bowel diseases. BMC Gastroenterol. 2022;22:438.

10. Öztürkcan S, Ermertcan AT, Eser E, et al. Cross validation of the Turkish version of dermatology life quality index. Int J Dermatol. 2006;45:1300-1307.

11. Demir GT, Ciciog˘lu HI˙. Attitude scale for healthy nutrition (ASHN): validity and reliability study. Gaziantep Univ J Sport Sci. 2019;4:256-274.

12. Yılmaz O, Boz H, Arslan A. The validity and reliability of depression stress and anxiety scale (DASS 21) Turkish short form. Res Financial Econ Soc Stud. 2017;2:78-91.

13. Nuttall FQ. Body mass index: obesity, BMI, and health: a critical review. Nutr Today. 2015;50:117-128.

14. Strumia R, Manzata E, Gualandi M. Is there a role for dermatologists in eating disorders? Expert Rev Dermatol. 2017; 2:109-112.

15. Henseler T, Christophers E. Disease concomitance in psoriasis. J Am Acad Dermatol. 1995;32:982-986.

16. Naldi L, Addis A, Chimenti S, et al. Impact of body mass index and obesity on clinical response to systemic treatment for psoriasis. evidence from the Psocare project. Dermatology. 2008;217:365-373.

17. Barros G, Duran P, Vera I, et al. Exploring the links between obesity and psoriasis: a comprehensive review. Int J Mol Sci. 2022;23:7499.

18. Hao Y, Zhu YJ, Zou S, et al. Metabolic syndrome and psoriasis: mechanisms and future directions. Front Immunol. 2021;12:711060.

19. Jing D, Xiao H, Shen M, et al. Association of psoriasis with anxiety and depression: a case–control study in Chinese patients. Front Med (Lausanne). 2021;8:771645.

20. Sahi FM, Masood A, Danawar NA, et al. Association between psoriasis and depression: a traditional review. Cureus. 2020;12:E9708.

21. Zafiriou E, Daponte AI, Siokas V, et al. Depression and obesity in patients with psoriasis and psoriatic arthritis: is IL-17–mediated immune dysregulation the connecting link? Front Immunol. 2021;12:699848.

22. Mrowietz U, Sümbül M, Gerdes S. Depression, a major comorbidity of psoriatic disease, is caused by metabolic inflammation. J Eur Acad Dermatol Venereol. 2023;37:1731-1738.

23. Pavlova NT, Kioskli K, Smith C, et al. Psychosocial aspects of obesity in adults with psoriasis: a systematic review. Skin Health Dis. 2021;1:E33.

24. Innamorati M, Quinto RM, Imperatori C, et al. Health-related quality of life and its association with alexithymia and difficulties in emotion regulation in patients with psoriasis. Compr Psychiatry. 2016;70:200-208.

25. Tabolli S, Naldi L, Pagliarello C, et al. Evaluation of the impact of writing exercises interventions on quality of life in patients with psoriasis undergoing systemic treatments. Br J Dermatol. 2012;167:1254‐1264.

26. Albuquerque D, Nóbrega C, Manco L, et al. The contribution of genetics and environment to obesity. Br Med Bull. 2017;123:159‐173.

27. Balantekin KN, Grammer AC, Fitzsimmons-Craft EE, et al. Overweight and obesity are associated with increased eating disorder correlates and general psychopathology in university women with eating disorders. Eat Behav. 2021;41:101482.

28. Jebeile H, Lister NB, Baur LA, et al. Eating disorder risk in adolescents with obesity. Obes Rev. 2021;22:E13173.

29. Crosta ML, Caldarola G, Fraietta S, et al. Psychopathology and eating disorders in patients with psoriasis. G Ital Dermatol Venereol. 2014;149:355-361.

30. Altunay I, Demirci GT, Ates B, et al. Do eating disorders accompany metabolic syndrome in psoriasis patients? results of a preliminary study. Clin Cosmet Investig Dermatol. 2011;4:139-143.

Current evidence indicates that obesity may initiate psoriasis or worsen existing disease. Various factors contribute to the development of obesity, including eating disorders (EDs). The aim of this study was to screen for and identify factors associated with EDs in patients with psoriasis and their impact on the development of obesity in this population. Demographic information including body mass index (BMI), Eating Attitude Test (EAT-26), Dermatology Life Quality Index (DLQI), Attitude Scale for Healthy Nutrition (ASHN), and Depression Anxiety Stress Scale 21 (DASS-21) scores were statistically analyzed for 82 participants with psoriasis at a tertiary dermatology clinic. It is important to manage obesity and other comorbidities of psoriasis in addition to treating its cutaneous manifestations, which may require a biopsychosocial approach.

Psoriasis is a chronic multisystemic inflammatory skin disease with a worldwide prevalence of 2% to 3%.1 Psoriasis can be accompanied by other conditions such as psoriatic arthritis, obesity, metabolic syndrome, diabetes mellitus, hypertension, dyslipidemia, atherosclerotic disease, inflammatory bowel disease, and anxiety/depression. It is important to manage comorbidities of psoriasis in addition to treating the cutaneous manifestations of the disease.1

Obesity is a major public health concern worldwide. Numerous observational and epidemiologic studies have reported a high prevalence of obesity among patients with psoriasis.2 Current evidence indicates that obesity may initiate or worsen psoriasis; furthermore, it is important to note that obesity may negatively impact the effectiveness of psoriasis-specific treatments or increase the incidence of adverse effects. Therefore, managing obesity is crucial in the treatment of psoriasis.3 Numerous studies have investigated the association between psoriasis and obesity, and they commonly conclude that both conditions share the same genetic metabolic pathways.2-4 However, it is important to consider environmental factors such as dietary habits, smoking, alcohol consumption, and a sedentary lifestyle—all of which are associated with psoriasis and also can contribute to the development of obesity.5 Because of the effects of obesity in psoriasis patients, factors that impact the development of obesity have become a popular research topic.

Eating disorders (EDs) are a crucial risk factor for both developing and maintaining obesity. In particular, two EDs that are associated with obesity include binge eating disorder and bulimia nervosa.6 According to the Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition,7 binge eating disorder can be diagnosed when a patient has at least 1 episode of binge eating per week over a 3-month period. Bulimia nervosa can be diagnosed when a patient is excessively concerned with their body weight and shape and engages in behaviors to prevent weight gain (eg, forced vomiting, excessive use of laxatives).7 Psychiatrists who specialize in EDs make diagnoses based on these criteria. In daily practice, there are several quick and simple questionnaires available to screen for EDs that can be used by nonpsychiatrist physicians, including the commonly used 26-item Eating Attitudes Test (EAT-26).8 The EAT-26 has been used to screen for EDs in patients with inflammatory disorders.9

The aim of this study was to screen for EDs in patients with psoriasis to identify potential risk factors for development of obesity.

Materials and Methods

This study included patients with psoriasis who were screened for EDs at a tertiary dermatology clinic in Turkey between January 2021 and December 2023. This study was approved by the local ethics committee and was in accordance with the Declaration of Helsinki (decision number E-93471371-514.99-225000079).

Study Design and Patient Inclusion Criteria—This quantitative cross-sectional study utilized EAT-26, Dermatology Life Quality Index (DLQI), Attitude Scale for Healthy Nutrition (ASHN), and Depression Anxiety Stress Scale-21 (DASS-21) scores. All the questionnaire scales used in the study were adapted and validated in Turkey.8,10-12 The inclusion criteria consisted of being older than 18 years of age, being literate, having psoriasis for at least 1 year that was not treated topically or systemically, and having no psychiatric diseases outside an ED. The questionnaires were presented in written format following the clinical examination. Literacy was an inclusion criterion in this study due to the absence of auxiliary health personnel.

Study Variables—The study variables included age, sex, marital status (single/divorced or married), education status (primary/secondary school or high school/university), employment status (employed or unemployed/retired), body mass index (BMI), smoking status, alcohol-consumption status, Psoriasis Area Severity Index score, presence of nail psoriasis and psoriatic arthritis, duration of psoriasis, family history of psoriasis, EAT-26 score, ASHN score, DLQI score, and DASS-21 score. Body mass index was calculated by taking a participant’s weight in kilograms and dividing it by their height in meters squared. The BMI values were classified into 3 categories: normal (18.5–24.9 kg/m2), overweight (25.0–29.9 kg/m2), and obese (≥30 kg/m2).13

Questionnaires—The EAT-26 questionnaire includes 26 questions that are used to detect EDs. Responses to each question include Likert-type answer options (ie, “always,” “usually,” “often,” “sometimes,” “rarely,” and “never.”) Patients with scores of 20 points or higher (range, 0–78) are classified as high risk for EDs.8 In our study, EAT-26 scores were grouped into 2 categories: patients scoring less than 20 points and those scoring 20 points or higher.

The DLQI questionnaire includes 10 questions to measure dermatologic symptoms and qualiy of life. Responses to each question include Likert-type answer options (ie, “not at all,” “a little,” “a lot,” or “very much.”) On the DLQI scale, the higher the score, the lower the quality of life (score range, 0–30).10

The ASHN questionnaire includes 21 questions that measure attitudes toward healthy nutrition with 5 possible answer options (“strongly disagree,” “disagree,” “undecided,” “agree,” and “strongly agree”). On this scale, higher scores indicate the participant is more knowledgeable about healthy nutrition (score range, 0–78).11

The DASS-21 questionnaire includes 21 questions that measure the severity of a range of symptoms common to depression, anxiety, and stress. Responses include Likert-type answer options (eg, “never,” “sometimes,” “often,” and “almost always.”) On this scale, a higher score (range of 0–21 for each) indicates higher levels of depression, anxiety, and stress.12

Statistical Analysis—Descriptive statistics were analyzed using SPSS software version 22.0 (IBM). The Shapiro-Wilk test was applied to determine whether the data were normally distributed. For categorical variables, frequency differences among groups were compared using the Pearson χ2 test. A t test was used to compare the means of 2 independent groups with a normal distribution. One-way analysis of variance and Tukey Honest Significant Difference post hoc analysis were used to test whether there was a statistically significant difference among the normally distributed means of independent groups. Pearson correlation analysis was used to determine whether there was a linear relationship between 2 numeric measurements and, if so, to determine the direction and severity of this relationship. P<.05 indicated statistical significance in this study.

Results

Study Participant Demographics—This study included 82 participants with a mean age of 44.3 years; 52.4% (43/82) were female, and 85.4% (70/82) were married. The questionnaire took an average of 4.2 minutes for participants to complete. A total of 57.3% (47/82) of patients had completed primary/secondary education and 59.8% (49/82) were employed. The mean BMI was 28.1 kg/m2. According to the BMI classification, 26.8% (22/82) participants had a normal weight, 36.6% (30/82) were overweight, and 43.9% (36/82) were obese. A total of 48.8% (40/82) of participants smoked, and 4.9% (4/82) consumed alcohol. The mean Psoriasis Area and Severity Index score was 5.4. A total of 54.9% (45/82) of participants had nail psoriasis, and 24.4% (20/82) had psoriatic arthritis. The mean duration of psoriasis was 153 months. A total of 29.3% (24/82) of participants had a positive family history of psoriasis. The mean EAT-26 score was 11.1. A total of 12.2% (10/82) of participants had an EAT-26 score of 20 points or higher and were considered at high risk for an ED. The mean ASHN score was 72.9; the mean DLQI score was 5.5; and on the DASS-21 scale, mean scores for depression, anxiety, and stress were 6.3, 8.7, and 10.0, respectively (Table).

Comparative Evaluation of the BMI Groups—The only statistically significant differences among the 3 BMI groups were related to marital status, EAT-26 score, and anxiety and stress scores (P=.02, <.01, <.01, and <.01, respectively)(eTable 1). The number of single/divorced participants in the overweight group was significantly (P=.02) greater than in the normal weight group. The mean EAT-26 score for the normal weight group was significantly (P<.01) lower than for the overweight and obese groups; there was no significant difference in mean EAT-26 scores between the overweight and obese groups. The mean anxiety score was significantly (P<.01) lower in the normal weight group compared with the overweight and obese groups. There was no significant difference between the overweight and obese groups according to the mean depression score. The mean stress and anxiety scores were significantly (P<.01) lower in the normal weight group than in the overweight and obese groups. There was no significant difference between the overweight and obese groups according to the mean anxiety score.

Comparative Evaluation of the EAT-26 Scores—There were statistically significant differences among the EAT-26 scores related to sex; BMI; and depression, anxiety, and stress scores (P=.04, .02, <.01, <.01, and <.01, respectively). The number of females in the group with a score of 20 points or higher was significantly (P=.04) less than that in the group scoring less than 20 points. The mean BMI in the group with a score of 20 points or higher was significantly (P=.02) greater than in group scoring less than 20 points. The mean depression, anxiety, and stress scores of the group scoring 20 points or higher were significantly (P<.01 for all) greater than in the group scoring less than 20 points (eTable 2).

Correlation Analysis of the Study Variables—The EAT-26 scores were positively correlated with BMI, anxiety, depression, and stress (P<.01 for all)(eTable 3).

Comment

Eating disorders are psychiatric conditions that require a multidisciplinary approach. Nonpsychiatric medical departments may be involved due to the severe consequences (eg, various skin changes14) of these disorders. Psoriasis is not known to be directly affected by the presence of an ED; however, it is possible that EDs could indirectly affect patients with psoriasis by influencing obesity. Therefore, this study aimed to examine the relationship between ED risk factors and obesity in this population.

The relationship between psoriasis and obesity has been a popular research topic in dermatology since the 1990s.15 Epidemiologic and observational studies have reported that patients with psoriasis are more likely to be overweight or have obesity, which is an independent risk factor for psoriasis.3,16 However, the causal relationship between psoriasis and obesity remains unclear. In a comprehensive review, Barros et al17 emphasized the causal relationship between obesity and psoriasis under several headings. Firstly, a higher BMI increases the risk for psoriasis by promoting cytokine release and immune system dysregulation. Secondly, a Western diet (eg, processed foods and fast food) triggers obesity and psoriasis by increasing adipose tissue. Thirdly, the alteration of the skin and gut microbiota triggers chronic inflammation as a result of bacterial translocation in patients with obesity. Fourthly, a high-fat diet and palmitic acid disrupt the intestinal integrity of the gut and increase the risk for psoriasis and obesity by triggering chronic inflammation of bacterial fragments that pass into the blood. Finally, the decrease in the amount of adiponectin and the increase in the amount of leptin in patients with obesity may cause psoriasis by increasing proinflammatory cytokines, which are similar to those involved in the pathogenesis of psoriasis.17 Additionally, psoriatic inflammation can cause insulin resistance and metabolic dysfunction, leading to obesity.18 The relationship between psoriasis and obesity cannot be solely explained by metabolic pathways. Smoking, alcohol consumption, and a sedentary lifestyle all are associated with psoriasis and also can contribute to obesity.5 Our study revealed no significant difference in smoking or alcohol consumption between the normal weight and overweight/obesity groups. Based on our data, we determined that smoking and alcohol consumption did not affect obesity in our patients with psoriasis.

Observational and epidemiologic studies have shown that patients with psoriasis experience increased rates of depression, anxiety, and stress.19 In studies of pathogenesis, a connection between depression and psoriatic inflammation has been established.20 It is known that inflammatory cytokines similar to those in psoriasis are involved in the development of obesity.18 In addition, depression and anxiety can lead to binge eating, unhealthy food choices, and a more sedentary lifestyle.5 All of these variables may contribute to the associations between depression and anxiety with psoriasis and obesity. Zafiriou et al21 conducted a study to investigate the relationship between psoriasis, obesity, and depression through inflammatory pathways with a focus on the importance of IL-17. Data showing that IL-17–producing Th17-cell subgroups play a considerable role in the development of obesity and depression prompted the authors to suggest that psoriasis, obesity, and ­anxiety/depression may be interconnected manifestations of immune dysregulation, potentially linked to IL-17 and its associated cells.21 Mrowietz et al22 also suggested that metabolic inflammation may contribute to obesity and depression in patients with psoriasis and highlighted the importance of several cytokines, including tumor necrosis factor α, IL-6, IL-8, IL-17, and IL-23. Our study revealed no significant differences in depression scores between BMI groups. Another meta-analysis reported conflicting findings on the incidence of depression in obese patients with psoriasis.23 Some of the studies had a small number of participants. Compared to depression, anxiety has received less attention in studies of patients with obesity with psoriasis. However, these studies have shown a positive correlation between anxiety scores and BMI in patients with psoriasis.24,25 In our study, similar to the findings of previous studies, overweight patients and those with obesitywho have psoriasis had significantly (P<.01) greater anxiety and stress scores than did normal weight patients with psoriasis.

Obesity should be assessed in patients with psoriasis via a biopsychosocial approach that takes into account genetic, behavioral, and environmental factors.26 Eating disorders are considered to be one of the factors contributing to obesity. Numerous studies in the literature have demonstrated a greater incidence of EDs in patients with obesity vs those without obesity.5,6,27 Obesity and EDs have a bidirectional relationship: individuals with obesity are at risk for EDs due to body dissatisfaction, dieting habits, and depressive states. Conversely, poor eating behaviors in individuals with a normal weight can lead to obesity.28

There are few studies in the literature exploring the relationship between psoriasis and EDs. Crosta et al29 demonstrated that patients with psoriasis had impaired results on ED screening tests and that these scores deteriorated further as BMI increased. Moreover, Altunay et al30 demonstrated that patients with psoriasis and metabolic syndrome had higher scores on the ED screening test. In this study, patients with higher scores also exhibited high levels of anxiety.30 In our study, similar to the findings of previous studies, patients with psoriasis who were overweight or had obesity had significantly (P<.01) greater EAT-26 scores than those in the normal weight group. Patients with high EAT-26 scores also exhibited elevated levels of depression, anxiety, and stress. Additionally, EAT-26 scores were positively correlated with BMI, anxiety, depression, and stress scores. Our study as well as other studies in the literature indicate that additional research is needed to determine the associations between EDs and obesity in psoriasis.

Conclusion

Managing obesity is crucial for patients with psoriasis. This study showed that EAT-26 scores were higher in patients with psoriasis who were overweight or had obesity than in those who were normal weight. Participants with high EAT-26 scores (≥20 points) were more likely to be female and have higher anxiety and stress scores. In addition, EAT-26 scores were positively correlated with BMI as well as depression, anxiety, and stress scores. Eating disorders may contribute to the development of obesity in patients with psoriasis. Although our study was limited by a small sample size, the results suggest that there is a need for large-scale multicenter studies to investigate the relationship between psoriasis and EDs.

References

1. Kalkan G. Comorbidities in psoriasis: the recognition of psoriasis as a systemic disease and current management. Turkderm-Turk Arch Dermatol Venereol. 2017;51:71-77.

2. Armstrong AW, Harskamp CT, Armstrong EJ. The association between psoriasis and obesity: a systematic review and meta-analysis of observational studies. Nutr Diabetes. 2012;2:E54.

3. Jensen P, Skov L. Psoriasis and obesity. Dermatology. 2016;232:633-639.

4. Mirghani H, Altemani AT, Altemani ST, et al. The cross talk between psoriasis, obesity, and dyslipidemia: a meta-analysis. Cureus. 2023;15:e49253.

5. Roehring M, Mashep MR, White MA, et al. The metabolic syndrome and behavioral correlates in obese patients with binge disorders. Obesity. 2009;17:481-486.

6. da Luz FQ, Hay P, Touyz S, et al. Obesity with comorbid eating disorders: associated health risks and treatment approaches. Nutrients. 2018;10:829.

7. American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition. American Psychiatric Association; 2013.

8. Ergüney Okumus¸ FE, Sertel Berk HÖ. The psychometric properties of the Eating Attitudes Test short form (EAT-26) in a college sample. Stud Psychol. 2020;40:57-78.

9. Stoleru G, Leopold A, Auerbach A, et al. Female gender, dissatisfaction with weight, and number of IBD related surgeries as independent risk factors for eating disorders among patients with inflammatory bowel diseases. BMC Gastroenterol. 2022;22:438.

10. Öztürkcan S, Ermertcan AT, Eser E, et al. Cross validation of the Turkish version of dermatology life quality index. Int J Dermatol. 2006;45:1300-1307.

11. Demir GT, Ciciog˘lu HI˙. Attitude scale for healthy nutrition (ASHN): validity and reliability study. Gaziantep Univ J Sport Sci. 2019;4:256-274.

12. Yılmaz O, Boz H, Arslan A. The validity and reliability of depression stress and anxiety scale (DASS 21) Turkish short form. Res Financial Econ Soc Stud. 2017;2:78-91.

13. Nuttall FQ. Body mass index: obesity, BMI, and health: a critical review. Nutr Today. 2015;50:117-128.

14. Strumia R, Manzata E, Gualandi M. Is there a role for dermatologists in eating disorders? Expert Rev Dermatol. 2017; 2:109-112.

15. Henseler T, Christophers E. Disease concomitance in psoriasis. J Am Acad Dermatol. 1995;32:982-986.

16. Naldi L, Addis A, Chimenti S, et al. Impact of body mass index and obesity on clinical response to systemic treatment for psoriasis. evidence from the Psocare project. Dermatology. 2008;217:365-373.

17. Barros G, Duran P, Vera I, et al. Exploring the links between obesity and psoriasis: a comprehensive review. Int J Mol Sci. 2022;23:7499.

18. Hao Y, Zhu YJ, Zou S, et al. Metabolic syndrome and psoriasis: mechanisms and future directions. Front Immunol. 2021;12:711060.

19. Jing D, Xiao H, Shen M, et al. Association of psoriasis with anxiety and depression: a case–control study in Chinese patients. Front Med (Lausanne). 2021;8:771645.

20. Sahi FM, Masood A, Danawar NA, et al. Association between psoriasis and depression: a traditional review. Cureus. 2020;12:E9708.

21. Zafiriou E, Daponte AI, Siokas V, et al. Depression and obesity in patients with psoriasis and psoriatic arthritis: is IL-17–mediated immune dysregulation the connecting link? Front Immunol. 2021;12:699848.

22. Mrowietz U, Sümbül M, Gerdes S. Depression, a major comorbidity of psoriatic disease, is caused by metabolic inflammation. J Eur Acad Dermatol Venereol. 2023;37:1731-1738.

23. Pavlova NT, Kioskli K, Smith C, et al. Psychosocial aspects of obesity in adults with psoriasis: a systematic review. Skin Health Dis. 2021;1:E33.

24. Innamorati M, Quinto RM, Imperatori C, et al. Health-related quality of life and its association with alexithymia and difficulties in emotion regulation in patients with psoriasis. Compr Psychiatry. 2016;70:200-208.

25. Tabolli S, Naldi L, Pagliarello C, et al. Evaluation of the impact of writing exercises interventions on quality of life in patients with psoriasis undergoing systemic treatments. Br J Dermatol. 2012;167:1254‐1264.

26. Albuquerque D, Nóbrega C, Manco L, et al. The contribution of genetics and environment to obesity. Br Med Bull. 2017;123:159‐173.

27. Balantekin KN, Grammer AC, Fitzsimmons-Craft EE, et al. Overweight and obesity are associated with increased eating disorder correlates and general psychopathology in university women with eating disorders. Eat Behav. 2021;41:101482.

28. Jebeile H, Lister NB, Baur LA, et al. Eating disorder risk in adolescents with obesity. Obes Rev. 2021;22:E13173.

29. Crosta ML, Caldarola G, Fraietta S, et al. Psychopathology and eating disorders in patients with psoriasis. G Ital Dermatol Venereol. 2014;149:355-361.

30. Altunay I, Demirci GT, Ates B, et al. Do eating disorders accompany metabolic syndrome in psoriasis patients? results of a preliminary study. Clin Cosmet Investig Dermatol. 2011;4:139-143.

Page Number
164-168
Page Number
164-168
Publications
Publications
Topics
Article Type
Display Headline
Eating Disorder Risk Factors and the Impact of Obesity in Patients With Psoriasis
Display Headline
Eating Disorder Risk Factors and the Impact of Obesity in Patients With Psoriasis
Sections
Inside the Article

<p class='insidehead'>Practice <strong>Points</strong></p> <ul class='insidebody'> <li>Eating disorders are considered a contributing factor in obesity.</li> <li>Obesity is prevalent in patients with psoriasis, and current evidence indicates that obesity may initiate psoriasis or worsen existing disease.</li> <li>Obesity should be considered as contributory to the development of psoriasis via a biopsychosocial approach that accounts for genetic, behavioral, and environmental factors.</li> </ul>

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

Evaluating Use of Empagliflozin for Diabetes Management in Veterans With Chronic Kidney Disease

Article Type
Changed
Thu, 11/14/2024 - 22:39
Display Headline
Evaluating Use of Empagliflozin for Diabetes Management in Veterans With Chronic Kidney Disease

More than 37 million Americans have diabetes mellitus (DM), and approximately 90% have type 2 DM (T2DM), including about 25% of veterans.1,2 The current guidelines suggest that therapy depends on a patient's comorbidities, management needs, and patient-centered treatment factors.3 About 1 in 3 adults with DM have chronic kidney disease (CKD), defined as the presence of kidney damage or an estimated glomerular filtration rate (eGFR) < 60 mL/min per 1.73 m2, persisting for ≥ 3 months.4

Sodium-glucose cotransporter-2 (SGLT-2) inhibitors are a class of antihyperglycemic agents acting on the SGLT-2 proteins expressed in the renal proximal convoluted tubules. They exert their effects by preventing the reabsorption of filtered glucose from the tubular lumen. There are 4 SGLT-2 inhibitors approved by the US Food and Drug Administration: canagliflozin, dapagliflozin, empagliflozin, and ertugliflozin. Empagliflozin is currently the preferred SGLT-2 inhibitor on the US Department of Veterans Affairs (VA) formulary.

According to the American Diabetes Association guidelines, empagliflozin is considered when an individual has or is at risk for atherosclerotic cardiovascular disease, heart failure, and CKD.3 SGLT-2 inhibitors are a favorable option due to their low risk for hypoglycemia while also promoting weight loss. The EMPEROR-Reduced trial demonstrated that, in addition to benefits for patients with heart failure, empagliflozin also slowed the progressive decline in kidney function in those with and without DM.5 The purpose of this study was to evaluate the effectiveness of empagliflozin on hemoglobin A1c (HbA1c) levels in patients with CKD at the Hershel “Woody” Williams VA Medical Center (HWWVAMC) in Huntington, West Virginia, along with other laboratory test markers.

Methods

The Marshall University Institutional Review Board #1 (Medical) and the HWWVAMC institutional review board and research and development committee each reviewed and approved this study. A retrospective chart review was conducted on patients diagnosed with T2DM and stage 3 CKD who were prescribed empagliflozin for DM management between January 1, 2015, and October 1, 2022, yielding 1771 patients. Data were obtained through the VHA Corporate Data Warehouse (CDW) and stored on the VA Informatics and Computing Infrastructure (VINCI) research server.

Patients were included if they were aged 18 to 89 years, prescribed empagliflozin by a VA clinician for the treatment of T2DM, had an eGFR between 30 and 59 mL/min/1.73 m2, and had an initial HbA1c between 7% and 10%. Using further random sampling, patients were either excluded or divided into, those with stage 3a CKD and those with stage 3b CKD. The primary endpoint of this study was the change in HbA1c levels in patients with stage 3b CKD (eGFR 30-44 mL/min/1.73 m2) compared with stage 3a (eGFR 45-59 mL/min/1.73 m2) after 12 months. The secondary endpoints included effects on renal function, weight, blood pressure, incidence of adverse drug events, and cardiovascular events. Of the excluded, 38 had HbA1c < 7%, 30 had HbA1c ≥ 10%, 21 did not have data at 1-year mark, 15 had the medication discontinued due to decline in renal function, 14 discontinued their medication without documented reason, 10 discontinued their medication due to adverse drug reactions (ADRs), 12 had eGFR > 60 mL/ min/1.73 m2, 9 died within 1 year of initiation, 4 had eGFR < 30 mL/min/1.73 m2, 1 had no baseline eGFR, and 1 was the spouse of a veteran.

Statistical Analysis

All statistical analyses were performed using STATA v.15. We used t tests to examine changes within each group, along with paired t tests to compare the 2 groups. Two-sample t tests were used to analyze the continuous data at both the primary and secondary endpoints.

Results

Of the 1771 patients included in the initial data set, a randomized sample of 255 charts were reviewed, 155 were excluded, and 100 were included. Fifty patients, had stage 3a CKD and 50 had stage 3b CKD. Baseline demographics were similar between the stage 3a and 3b groups (Table 1). Both groups were predominantly White and male, with mean age > 70 years.

The primary endpoint was the differences in HbA1c levels over time and between groups for patients with stage 3a and stage 3b CKD 1 year after initiation of empagliflozin. The starting doses of empagliflozin were either 12.5 mg or 25.0 mg. For both groups, the changes in HbA1c levels were statistically significant (Table 2). HbA1c levels dropped 0.65% for the stage 3a group and 0.48% for the 3b group. When compared to one another, the results were not statistically significant (P = .51).

Secondary Endpoint

There was no statistically significant difference in serum creatinine levels within each group between baselines and 1 year later for the stage 3a (P = .21) and stage 3b (P = .22) groups, or when compared to each other (P = .67). There were statistically significant changes in weight for patients in the stage 3a group (P < .05), but not for stage 3b group (P = .06) or when compared to each other (P = .41). A statistically significant change in systolic blood pressure was observed for the stage 3a group (P = .003), but not the stage 3b group (P = .16) or when compared to each other (P = .27). There were statistically significant changes in diastolic blood pressure within the stage 3a group (P = .04), but not within the stage 3b group (P = .61) or when compared to each other (P = .31).

Ten patients discontinued empagliflozin before the 1-year mark due to ADRs, including dizziness, increased incidence of urinary tract infections, rash, and tachycardia (Table 3). Additionally, 3 ADRs resulted in the empagliflozin discontinuation after 1 year (Table 3).

Discussion

This study showed a statistically significant change in HbA1c levels for patients with stage 3a and stage 3b CKD. With eGFR levels in these 2 groups > 30 mL/min/1.73 m2, patients were able to achieve glycemic benefits. There were no significant changes to the serum creatinine levels. Both groups saw statistically significant changes in weight loss within their own group; however, there were no statistically significant changes when compared to each other. With both systolic and diastolic blood pressure, the stage 3a group had statistically significant changes.

The EMPA-REG BP study demonstrated that empagliflozin was associated with significant and clinically meaningful reductions in blood pressure and HbA1c levels compared with placebo and was well tolerated in patients with T2DM and hypertension.6,7,8

Limitations

This study had a retrospective study design, which resulted in missing information for many patients and higher rates of exclusion. The population was predominantly older, White, and male and may not reflect other populations. The starting doses of empagliflozin varied between the groups. The VA employs tablet splitting for some patients, and the available doses were either 10.0 mg, 12.5 mg, or 25.0 mg. Some prescribers start veterans at lower doses and gradually increase to the higher dose of 25.0 mg, adding to the variability in starting doses.

Patients with eGFR < 30 mL/min/1.73 m2 make it difficult to determine any potential benefit in this population. The EMPA-KIDNEY trial demonstrated that the benefits of empagliflozin treatment were consistent among patients with or without DM and regardless of eGFR at randomization.9 Furthermore, many veterans had an initial HbA1c levels outside the inclusion criteria range, which was a factor in the smaller sample size.

Conclusions

While the reduction in HbA1c levels was less in patients with stage 3b CKD compared to patients stage 3a CKD, all patients experienced a benefit. The overall incidence of ADRs was low in the study population, showing empagliflozin as a favorable choice for those with T2DM and CKD. Based on the findings of this study, empagliflozin is a potentially beneficial option for reducing HbA1c levels in patients with CKD.

References
  1. Centers for Disease Control and Prevention. Type 2 diabetes. Updated May 25, 2024. Accessed September 27, 2024. https://www.cdc.gov/diabetes/about/about-type-2-diabetes.html?CDC_AAref_Val
  2. US Department of Veterans Affairs, VA research on diabetes. Updated September 2019. Accessed September 27, 2024. https://www.research.va.gov/pubs/docs/va_factsheets/Diabetes.pdf
  3. American Diabetes Association. Standards of Medical Care in Diabetes-2022 Abridged for Primary Care Providers. Clin Diabetes. 2022;40(1):10-38. doi:10.2337/cd22-as01
  4. Centers for Disease Control and Prevention. Diabetes, chronic kidney disease. Updated May 15, 2024. Accessed September 27, 2024. https://www.cdc.gov/diabetes/diabetes-complications/diabetes-and-chronic-kidney-disease.html
  5. Packer M, Anker SD, Butler J, et al. Cardiovascular and Renal Outcomes with Empagliflozin in Heart Failure. N Engl J Med. 2020;383(15):1413-1424. doi:10.1056/NEJMoa2022190
  6. Tikkanen I, Narko K, Zeller C, et al. Empagliflozin reduces blood pressure in patients with type 2 diabetes and hypertension. Diabetes Care. 2015;38(3):420-428. doi:10.2337/dc14-1096
  7. Zinman B, Wanner C, Lachin JM, et al. Empagliflozin, cardiovascular outcomes, and mortality in type 2 diabetes. N Engl J Med. 2015;373(22):2117-2128. doi:10.1056/NEJMoa1504720
  8. Chilton R, Tikkanen I, Cannon CP, et al. Effects of empagliflozin on blood pressure and markers of arterial stiffness and vascular resistance in patients with type 2 diabetes. Diabetes Obes Metab. 2015;17(12):1180-1193. doi:10.1111/dom.12572
  9. The EMPA-KIDNEY Collaborative Group, Herrington WG, Staplin N, et al. Empagliflozin in Patients with Chronic Kidney Disease. N Engl J Med. 2023;388(2):117-127. doi:10.1056/NEJMoa2204233
Article PDF
Author and Disclosure Information

Chelsey Williams, PharmD, BCACPa; Bobbie Bailey, PharmDa

Author affiliations: aHershel “Woody” Williams Veterans Affairs Medical Center, Huntington, West Virginia

Author disclosures: The authors report no actual or potential conflict of interest with regards to this article.

Funding: The authors report no outside source of funding.

Correspondence: Bobbie Bailey ([email protected])

Fed Pract. 2024;41(suppl 6). Published online November 17. doi:10.12788/fp.0524

Issue
Federal Practitioner - 41(11)s
Publications
Topics
Page Number
e1-e4
Sections
Author and Disclosure Information

Chelsey Williams, PharmD, BCACPa; Bobbie Bailey, PharmDa

Author affiliations: aHershel “Woody” Williams Veterans Affairs Medical Center, Huntington, West Virginia

Author disclosures: The authors report no actual or potential conflict of interest with regards to this article.

Funding: The authors report no outside source of funding.

Correspondence: Bobbie Bailey ([email protected])

Fed Pract. 2024;41(suppl 6). Published online November 17. doi:10.12788/fp.0524

Author and Disclosure Information

Chelsey Williams, PharmD, BCACPa; Bobbie Bailey, PharmDa

Author affiliations: aHershel “Woody” Williams Veterans Affairs Medical Center, Huntington, West Virginia

Author disclosures: The authors report no actual or potential conflict of interest with regards to this article.

Funding: The authors report no outside source of funding.

Correspondence: Bobbie Bailey ([email protected])

Fed Pract. 2024;41(suppl 6). Published online November 17. doi:10.12788/fp.0524

Article PDF
Article PDF

More than 37 million Americans have diabetes mellitus (DM), and approximately 90% have type 2 DM (T2DM), including about 25% of veterans.1,2 The current guidelines suggest that therapy depends on a patient's comorbidities, management needs, and patient-centered treatment factors.3 About 1 in 3 adults with DM have chronic kidney disease (CKD), defined as the presence of kidney damage or an estimated glomerular filtration rate (eGFR) < 60 mL/min per 1.73 m2, persisting for ≥ 3 months.4

Sodium-glucose cotransporter-2 (SGLT-2) inhibitors are a class of antihyperglycemic agents acting on the SGLT-2 proteins expressed in the renal proximal convoluted tubules. They exert their effects by preventing the reabsorption of filtered glucose from the tubular lumen. There are 4 SGLT-2 inhibitors approved by the US Food and Drug Administration: canagliflozin, dapagliflozin, empagliflozin, and ertugliflozin. Empagliflozin is currently the preferred SGLT-2 inhibitor on the US Department of Veterans Affairs (VA) formulary.

According to the American Diabetes Association guidelines, empagliflozin is considered when an individual has or is at risk for atherosclerotic cardiovascular disease, heart failure, and CKD.3 SGLT-2 inhibitors are a favorable option due to their low risk for hypoglycemia while also promoting weight loss. The EMPEROR-Reduced trial demonstrated that, in addition to benefits for patients with heart failure, empagliflozin also slowed the progressive decline in kidney function in those with and without DM.5 The purpose of this study was to evaluate the effectiveness of empagliflozin on hemoglobin A1c (HbA1c) levels in patients with CKD at the Hershel “Woody” Williams VA Medical Center (HWWVAMC) in Huntington, West Virginia, along with other laboratory test markers.

Methods

The Marshall University Institutional Review Board #1 (Medical) and the HWWVAMC institutional review board and research and development committee each reviewed and approved this study. A retrospective chart review was conducted on patients diagnosed with T2DM and stage 3 CKD who were prescribed empagliflozin for DM management between January 1, 2015, and October 1, 2022, yielding 1771 patients. Data were obtained through the VHA Corporate Data Warehouse (CDW) and stored on the VA Informatics and Computing Infrastructure (VINCI) research server.

Patients were included if they were aged 18 to 89 years, prescribed empagliflozin by a VA clinician for the treatment of T2DM, had an eGFR between 30 and 59 mL/min/1.73 m2, and had an initial HbA1c between 7% and 10%. Using further random sampling, patients were either excluded or divided into, those with stage 3a CKD and those with stage 3b CKD. The primary endpoint of this study was the change in HbA1c levels in patients with stage 3b CKD (eGFR 30-44 mL/min/1.73 m2) compared with stage 3a (eGFR 45-59 mL/min/1.73 m2) after 12 months. The secondary endpoints included effects on renal function, weight, blood pressure, incidence of adverse drug events, and cardiovascular events. Of the excluded, 38 had HbA1c < 7%, 30 had HbA1c ≥ 10%, 21 did not have data at 1-year mark, 15 had the medication discontinued due to decline in renal function, 14 discontinued their medication without documented reason, 10 discontinued their medication due to adverse drug reactions (ADRs), 12 had eGFR > 60 mL/ min/1.73 m2, 9 died within 1 year of initiation, 4 had eGFR < 30 mL/min/1.73 m2, 1 had no baseline eGFR, and 1 was the spouse of a veteran.

Statistical Analysis

All statistical analyses were performed using STATA v.15. We used t tests to examine changes within each group, along with paired t tests to compare the 2 groups. Two-sample t tests were used to analyze the continuous data at both the primary and secondary endpoints.

Results

Of the 1771 patients included in the initial data set, a randomized sample of 255 charts were reviewed, 155 were excluded, and 100 were included. Fifty patients, had stage 3a CKD and 50 had stage 3b CKD. Baseline demographics were similar between the stage 3a and 3b groups (Table 1). Both groups were predominantly White and male, with mean age > 70 years.

The primary endpoint was the differences in HbA1c levels over time and between groups for patients with stage 3a and stage 3b CKD 1 year after initiation of empagliflozin. The starting doses of empagliflozin were either 12.5 mg or 25.0 mg. For both groups, the changes in HbA1c levels were statistically significant (Table 2). HbA1c levels dropped 0.65% for the stage 3a group and 0.48% for the 3b group. When compared to one another, the results were not statistically significant (P = .51).

Secondary Endpoint

There was no statistically significant difference in serum creatinine levels within each group between baselines and 1 year later for the stage 3a (P = .21) and stage 3b (P = .22) groups, or when compared to each other (P = .67). There were statistically significant changes in weight for patients in the stage 3a group (P < .05), but not for stage 3b group (P = .06) or when compared to each other (P = .41). A statistically significant change in systolic blood pressure was observed for the stage 3a group (P = .003), but not the stage 3b group (P = .16) or when compared to each other (P = .27). There were statistically significant changes in diastolic blood pressure within the stage 3a group (P = .04), but not within the stage 3b group (P = .61) or when compared to each other (P = .31).

Ten patients discontinued empagliflozin before the 1-year mark due to ADRs, including dizziness, increased incidence of urinary tract infections, rash, and tachycardia (Table 3). Additionally, 3 ADRs resulted in the empagliflozin discontinuation after 1 year (Table 3).

Discussion

This study showed a statistically significant change in HbA1c levels for patients with stage 3a and stage 3b CKD. With eGFR levels in these 2 groups > 30 mL/min/1.73 m2, patients were able to achieve glycemic benefits. There were no significant changes to the serum creatinine levels. Both groups saw statistically significant changes in weight loss within their own group; however, there were no statistically significant changes when compared to each other. With both systolic and diastolic blood pressure, the stage 3a group had statistically significant changes.

The EMPA-REG BP study demonstrated that empagliflozin was associated with significant and clinically meaningful reductions in blood pressure and HbA1c levels compared with placebo and was well tolerated in patients with T2DM and hypertension.6,7,8

Limitations

This study had a retrospective study design, which resulted in missing information for many patients and higher rates of exclusion. The population was predominantly older, White, and male and may not reflect other populations. The starting doses of empagliflozin varied between the groups. The VA employs tablet splitting for some patients, and the available doses were either 10.0 mg, 12.5 mg, or 25.0 mg. Some prescribers start veterans at lower doses and gradually increase to the higher dose of 25.0 mg, adding to the variability in starting doses.

Patients with eGFR < 30 mL/min/1.73 m2 make it difficult to determine any potential benefit in this population. The EMPA-KIDNEY trial demonstrated that the benefits of empagliflozin treatment were consistent among patients with or without DM and regardless of eGFR at randomization.9 Furthermore, many veterans had an initial HbA1c levels outside the inclusion criteria range, which was a factor in the smaller sample size.

Conclusions

While the reduction in HbA1c levels was less in patients with stage 3b CKD compared to patients stage 3a CKD, all patients experienced a benefit. The overall incidence of ADRs was low in the study population, showing empagliflozin as a favorable choice for those with T2DM and CKD. Based on the findings of this study, empagliflozin is a potentially beneficial option for reducing HbA1c levels in patients with CKD.

More than 37 million Americans have diabetes mellitus (DM), and approximately 90% have type 2 DM (T2DM), including about 25% of veterans.1,2 The current guidelines suggest that therapy depends on a patient's comorbidities, management needs, and patient-centered treatment factors.3 About 1 in 3 adults with DM have chronic kidney disease (CKD), defined as the presence of kidney damage or an estimated glomerular filtration rate (eGFR) < 60 mL/min per 1.73 m2, persisting for ≥ 3 months.4

Sodium-glucose cotransporter-2 (SGLT-2) inhibitors are a class of antihyperglycemic agents acting on the SGLT-2 proteins expressed in the renal proximal convoluted tubules. They exert their effects by preventing the reabsorption of filtered glucose from the tubular lumen. There are 4 SGLT-2 inhibitors approved by the US Food and Drug Administration: canagliflozin, dapagliflozin, empagliflozin, and ertugliflozin. Empagliflozin is currently the preferred SGLT-2 inhibitor on the US Department of Veterans Affairs (VA) formulary.

According to the American Diabetes Association guidelines, empagliflozin is considered when an individual has or is at risk for atherosclerotic cardiovascular disease, heart failure, and CKD.3 SGLT-2 inhibitors are a favorable option due to their low risk for hypoglycemia while also promoting weight loss. The EMPEROR-Reduced trial demonstrated that, in addition to benefits for patients with heart failure, empagliflozin also slowed the progressive decline in kidney function in those with and without DM.5 The purpose of this study was to evaluate the effectiveness of empagliflozin on hemoglobin A1c (HbA1c) levels in patients with CKD at the Hershel “Woody” Williams VA Medical Center (HWWVAMC) in Huntington, West Virginia, along with other laboratory test markers.

Methods

The Marshall University Institutional Review Board #1 (Medical) and the HWWVAMC institutional review board and research and development committee each reviewed and approved this study. A retrospective chart review was conducted on patients diagnosed with T2DM and stage 3 CKD who were prescribed empagliflozin for DM management between January 1, 2015, and October 1, 2022, yielding 1771 patients. Data were obtained through the VHA Corporate Data Warehouse (CDW) and stored on the VA Informatics and Computing Infrastructure (VINCI) research server.

Patients were included if they were aged 18 to 89 years, prescribed empagliflozin by a VA clinician for the treatment of T2DM, had an eGFR between 30 and 59 mL/min/1.73 m2, and had an initial HbA1c between 7% and 10%. Using further random sampling, patients were either excluded or divided into, those with stage 3a CKD and those with stage 3b CKD. The primary endpoint of this study was the change in HbA1c levels in patients with stage 3b CKD (eGFR 30-44 mL/min/1.73 m2) compared with stage 3a (eGFR 45-59 mL/min/1.73 m2) after 12 months. The secondary endpoints included effects on renal function, weight, blood pressure, incidence of adverse drug events, and cardiovascular events. Of the excluded, 38 had HbA1c < 7%, 30 had HbA1c ≥ 10%, 21 did not have data at 1-year mark, 15 had the medication discontinued due to decline in renal function, 14 discontinued their medication without documented reason, 10 discontinued their medication due to adverse drug reactions (ADRs), 12 had eGFR > 60 mL/ min/1.73 m2, 9 died within 1 year of initiation, 4 had eGFR < 30 mL/min/1.73 m2, 1 had no baseline eGFR, and 1 was the spouse of a veteran.

Statistical Analysis

All statistical analyses were performed using STATA v.15. We used t tests to examine changes within each group, along with paired t tests to compare the 2 groups. Two-sample t tests were used to analyze the continuous data at both the primary and secondary endpoints.

Results

Of the 1771 patients included in the initial data set, a randomized sample of 255 charts were reviewed, 155 were excluded, and 100 were included. Fifty patients, had stage 3a CKD and 50 had stage 3b CKD. Baseline demographics were similar between the stage 3a and 3b groups (Table 1). Both groups were predominantly White and male, with mean age > 70 years.

The primary endpoint was the differences in HbA1c levels over time and between groups for patients with stage 3a and stage 3b CKD 1 year after initiation of empagliflozin. The starting doses of empagliflozin were either 12.5 mg or 25.0 mg. For both groups, the changes in HbA1c levels were statistically significant (Table 2). HbA1c levels dropped 0.65% for the stage 3a group and 0.48% for the 3b group. When compared to one another, the results were not statistically significant (P = .51).

Secondary Endpoint

There was no statistically significant difference in serum creatinine levels within each group between baselines and 1 year later for the stage 3a (P = .21) and stage 3b (P = .22) groups, or when compared to each other (P = .67). There were statistically significant changes in weight for patients in the stage 3a group (P < .05), but not for stage 3b group (P = .06) or when compared to each other (P = .41). A statistically significant change in systolic blood pressure was observed for the stage 3a group (P = .003), but not the stage 3b group (P = .16) or when compared to each other (P = .27). There were statistically significant changes in diastolic blood pressure within the stage 3a group (P = .04), but not within the stage 3b group (P = .61) or when compared to each other (P = .31).

Ten patients discontinued empagliflozin before the 1-year mark due to ADRs, including dizziness, increased incidence of urinary tract infections, rash, and tachycardia (Table 3). Additionally, 3 ADRs resulted in the empagliflozin discontinuation after 1 year (Table 3).

Discussion

This study showed a statistically significant change in HbA1c levels for patients with stage 3a and stage 3b CKD. With eGFR levels in these 2 groups > 30 mL/min/1.73 m2, patients were able to achieve glycemic benefits. There were no significant changes to the serum creatinine levels. Both groups saw statistically significant changes in weight loss within their own group; however, there were no statistically significant changes when compared to each other. With both systolic and diastolic blood pressure, the stage 3a group had statistically significant changes.

The EMPA-REG BP study demonstrated that empagliflozin was associated with significant and clinically meaningful reductions in blood pressure and HbA1c levels compared with placebo and was well tolerated in patients with T2DM and hypertension.6,7,8

Limitations

This study had a retrospective study design, which resulted in missing information for many patients and higher rates of exclusion. The population was predominantly older, White, and male and may not reflect other populations. The starting doses of empagliflozin varied between the groups. The VA employs tablet splitting for some patients, and the available doses were either 10.0 mg, 12.5 mg, or 25.0 mg. Some prescribers start veterans at lower doses and gradually increase to the higher dose of 25.0 mg, adding to the variability in starting doses.

Patients with eGFR < 30 mL/min/1.73 m2 make it difficult to determine any potential benefit in this population. The EMPA-KIDNEY trial demonstrated that the benefits of empagliflozin treatment were consistent among patients with or without DM and regardless of eGFR at randomization.9 Furthermore, many veterans had an initial HbA1c levels outside the inclusion criteria range, which was a factor in the smaller sample size.

Conclusions

While the reduction in HbA1c levels was less in patients with stage 3b CKD compared to patients stage 3a CKD, all patients experienced a benefit. The overall incidence of ADRs was low in the study population, showing empagliflozin as a favorable choice for those with T2DM and CKD. Based on the findings of this study, empagliflozin is a potentially beneficial option for reducing HbA1c levels in patients with CKD.

References
  1. Centers for Disease Control and Prevention. Type 2 diabetes. Updated May 25, 2024. Accessed September 27, 2024. https://www.cdc.gov/diabetes/about/about-type-2-diabetes.html?CDC_AAref_Val
  2. US Department of Veterans Affairs, VA research on diabetes. Updated September 2019. Accessed September 27, 2024. https://www.research.va.gov/pubs/docs/va_factsheets/Diabetes.pdf
  3. American Diabetes Association. Standards of Medical Care in Diabetes-2022 Abridged for Primary Care Providers. Clin Diabetes. 2022;40(1):10-38. doi:10.2337/cd22-as01
  4. Centers for Disease Control and Prevention. Diabetes, chronic kidney disease. Updated May 15, 2024. Accessed September 27, 2024. https://www.cdc.gov/diabetes/diabetes-complications/diabetes-and-chronic-kidney-disease.html
  5. Packer M, Anker SD, Butler J, et al. Cardiovascular and Renal Outcomes with Empagliflozin in Heart Failure. N Engl J Med. 2020;383(15):1413-1424. doi:10.1056/NEJMoa2022190
  6. Tikkanen I, Narko K, Zeller C, et al. Empagliflozin reduces blood pressure in patients with type 2 diabetes and hypertension. Diabetes Care. 2015;38(3):420-428. doi:10.2337/dc14-1096
  7. Zinman B, Wanner C, Lachin JM, et al. Empagliflozin, cardiovascular outcomes, and mortality in type 2 diabetes. N Engl J Med. 2015;373(22):2117-2128. doi:10.1056/NEJMoa1504720
  8. Chilton R, Tikkanen I, Cannon CP, et al. Effects of empagliflozin on blood pressure and markers of arterial stiffness and vascular resistance in patients with type 2 diabetes. Diabetes Obes Metab. 2015;17(12):1180-1193. doi:10.1111/dom.12572
  9. The EMPA-KIDNEY Collaborative Group, Herrington WG, Staplin N, et al. Empagliflozin in Patients with Chronic Kidney Disease. N Engl J Med. 2023;388(2):117-127. doi:10.1056/NEJMoa2204233
References
  1. Centers for Disease Control and Prevention. Type 2 diabetes. Updated May 25, 2024. Accessed September 27, 2024. https://www.cdc.gov/diabetes/about/about-type-2-diabetes.html?CDC_AAref_Val
  2. US Department of Veterans Affairs, VA research on diabetes. Updated September 2019. Accessed September 27, 2024. https://www.research.va.gov/pubs/docs/va_factsheets/Diabetes.pdf
  3. American Diabetes Association. Standards of Medical Care in Diabetes-2022 Abridged for Primary Care Providers. Clin Diabetes. 2022;40(1):10-38. doi:10.2337/cd22-as01
  4. Centers for Disease Control and Prevention. Diabetes, chronic kidney disease. Updated May 15, 2024. Accessed September 27, 2024. https://www.cdc.gov/diabetes/diabetes-complications/diabetes-and-chronic-kidney-disease.html
  5. Packer M, Anker SD, Butler J, et al. Cardiovascular and Renal Outcomes with Empagliflozin in Heart Failure. N Engl J Med. 2020;383(15):1413-1424. doi:10.1056/NEJMoa2022190
  6. Tikkanen I, Narko K, Zeller C, et al. Empagliflozin reduces blood pressure in patients with type 2 diabetes and hypertension. Diabetes Care. 2015;38(3):420-428. doi:10.2337/dc14-1096
  7. Zinman B, Wanner C, Lachin JM, et al. Empagliflozin, cardiovascular outcomes, and mortality in type 2 diabetes. N Engl J Med. 2015;373(22):2117-2128. doi:10.1056/NEJMoa1504720
  8. Chilton R, Tikkanen I, Cannon CP, et al. Effects of empagliflozin on blood pressure and markers of arterial stiffness and vascular resistance in patients with type 2 diabetes. Diabetes Obes Metab. 2015;17(12):1180-1193. doi:10.1111/dom.12572
  9. The EMPA-KIDNEY Collaborative Group, Herrington WG, Staplin N, et al. Empagliflozin in Patients with Chronic Kidney Disease. N Engl J Med. 2023;388(2):117-127. doi:10.1056/NEJMoa2204233
Issue
Federal Practitioner - 41(11)s
Issue
Federal Practitioner - 41(11)s
Page Number
e1-e4
Page Number
e1-e4
Publications
Publications
Topics
Article Type
Display Headline
Evaluating Use of Empagliflozin for Diabetes Management in Veterans With Chronic Kidney Disease
Display Headline
Evaluating Use of Empagliflozin for Diabetes Management in Veterans With Chronic Kidney Disease
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Wed, 11/06/2024 - 10:30
Un-Gate On Date
Wed, 11/06/2024 - 10:30
Use ProPublica
CFC Schedule Remove Status
Wed, 11/06/2024 - 10:30
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
survey writer start date
Thu, 11/14/2024 - 15:21
Article PDF Media

VHA Support for Home Health Agency Staff and Patients During Natural Disasters

Article Type
Changed
Wed, 11/06/2024 - 10:03

As large-scale natural disasters become more common, health care coalitions and the engagement of health systems with local, state, and federal public health departments have effectively bolstered communities’ resilience via collective sharing and distribution of resources.1 These resources may include supplies and the dissemination of emergency information, education, and training.2 The COVID-19 pandemic demonstrated that larger health care systems including hospital networks and nursing homes are better connected to health care coalition resources than smaller, independent systems, such as community home health agencies.3 This leaves some organizations on their own to meet requirements that maintain continuity of care and support their patients and staff throughout a natural disaster.

Home health care workers play important roles in the care of older adults.4 Older adults experience high levels of disability and comorbidities that put them at risk during emergencies; they often require support from paid, family, and neighborhood caregivers to live independently.5 More than 9.3 million US adults receive paid care from 2.6 million home health care workers (eg, home health aides and personal care assistants).6 Many of these individuals are hired through small independent home health agencies (HHAs), while others may work directly for an individual. When neighborhood resources and family caregiving are disrupted during emergencies, the critical services these workers administer become even more essential to ensuring continued access to medical care and social services.

The importance of these services was underscored by the Centers for Medicare and Medicaid Services 2017 inclusion of HHAs in federal emergency preparedness guidelines.7,8 The fractured and decentralized nature of the home health care industry means many HHAs struggle to maintain continuous care during emergencies and protect their staff. HHAs, and health care workers in the home, are often isolated, under-resourced, and disconnected from broader emergency planning efforts. Additionally, home care jobs are largely part-time, unstable, and low paying, making the workers themselves vulnerable during emergencies.3,9-13

This is a significant issue for the Veterans Health Administration (VHA), which annually purchases 10.5 million home health care worker visits for 150,000 veterans from community-based HHAs to enable those individuals to live independently. Figure 1 illustrates the existing structure of directly provided and contracted VHA services for community-dwelling veterans, highlighting the circle of care around the veteran.8,9 Home health care workers anchored health care teams during the COVID-19 pandemic, observing and reporting on patients’ well-being to family caregivers, primary care practitioners, and HHAs. They also provided critical emotional support and companionship to patients isolated from family and friends.9 These workers also exposed themselves and their families to considerable risk and often lacked the protection afforded by personal protective equipment (PPE) in accordance with infection prevention guidance.3,12

FIGURE 1. Circle of Care for Community-Dwelling Veterans
Abbreviations: HBPC, home based primary care; HHA, home health agency; VHA, Veterans Health Administration.
aAdapted with permission from Wyte-Lake and Franzosa.8,9

Through a combination of its national and local health care networks, the VHA has a robust and well-positioned emergency infrastructure to supportcommunity-dwelling older adults during disasters.14 This network is supported by the VHA Office of Emergency Management, which shares resources and guidance with local emergency managers at each facility as well as individual programs such as the VHA Home Based Primary Care (HBPC) program, which provides 38,000 seriously ill veterans with home medical visits.15 Working closely with their local and national hospital networks and emergency managers, individual VHA HBPC programs were able to maintain the safety of staff and continuity of care for patients enrolled in HBPC by rapidly administering COVID-19 vaccines to patients, caregivers, and staff, and providing emergency assistance during the 2017 hurricane season.16,17 These efforts were successful because HBPC practitioners and their patients, had access to a level of emergency-related information, resources, and technology that are often out of reach for individual community-based health care practitioners (HCPs). The US Department of Veterans Affairs (VA) also supports local communities through its Fourth Mission, which provides emergency resources to non-VHA health care facilities (ie, hospitals and nursing homes) during national emergencies and natural disasters.17 Although there has been an expansion in the definition of shared resources, such as extending behavioral health support to local communities, the VHA has not historically provided these resources to HHAs.14



This study examines opportunities to leverage VHA emergency management resources to support contracted HHAs and inform other large health system emergency planning efforts. The findings from the exploratory phase are described in this article. We interviewed VHA emergency managers, HBPC and VA staff who coordinate home health care worker services, as well as administrators at contracted HHAs within a Veterans Integrated Services Network (VISN). These findings will inform the second (single-site pilot study) and third (feasibility study) phases. Our intent was to (1) better understand the relationships between VA medical centers (VAMCs) and their contracted HHAs; (2) identify existing VHA emergency protocols to support community-dwelling older adults; and (3) determine opportunities to build on existing infrastructure and relationships to better support contracted HHAs and their staff in emergencies.

 

Methods

The 18 VISNs act as regional systems of care that are loosely connected to better meet local health needs and maximize access to care. This study was conducted at 6 of 9 VAMCs within VISN 2, the New York/New Jersey VHA Health Care Network.18 VAMCs that serve urban, rural, and mixed urban/rural catchment areas were included.

Each VAMC has an emergency management program led by an emergency manager, an HBPC program led by a program director and medical director, and a community care or purchased care office that has a liaison who manages contracted home health care worker services. The studyfocused on HBPC programs because they are most likely to interact with veterans’ home health care workers in the home and care for community-dwelling veterans during emergencies. Each VHA also contracts with a series of local HHAs that generally have a dedicated staff member who interfaces with the VHA liaison. Our goal was to interview ≥ 1 emergency manager, ≥ 1 HBPC team member, ≥ 1 community care staff person, and ≥ 1 contracted home health agency administrator at each site to gain multiple perspectives from the range of HCPs serving veterans in the community.

 

Recruitment and Data Collection

The 6 sites were selected in consultation with VISN 2 leadership for their strong HBPC and emergency management programs. To recruit respondents, we contacted VISN and VAMC leads and used our professional networks to identify a sample of multidisciplinary individuals who represent both community care and HBPC programs who were contacted via email.

Since each VAMC is organized differently, we utilized a snowball sampling approach to identify the appropriate contacts.19 At the completion of each interview, we asked the participant to suggest additional contacts and introduce us to any remaining stakeholders (eg, the emergency manager) at that site or colleagues at other VISN facilities. Because roles vary among VAMCs, we contacted the person who most closely resembled the identified role and asked them to direct us to a more appropriate contact, if necessary. We asked community care managers to identify 1 to 2 agencies serving the highest volume of patients who are veterans at their site and requested interviews with those liaisons. This resulted in the recruitment of key stakeholders from 4 teams across the 6 sites (Table).

A semistructured interview guide was jointly developed based on constructs of interest, including relationships within VAMCs and between VAMCs and HHAs; existing emergency protocols and experience during disasters; and suggestions and opportunities for supporting agencies during emergencies and potential barriers. Two researchers (TWL and EF) who were trained in qualitative methods jointly conducted interviews using the interview guide, with 1 researcher leading and another taking notes and asking clarifying questions.

Interviews were conducted virtually via Microsoft Teams with respondents at their work locations between September 2022 and January 2023. Interviews were audio recorded and transcribed and 2 authors (TWL and ESO) reviewed transcripts for accuracy. Interviews averaged 47 minutes in length (range, 20-59).

The study was reviewed and determined to be exempt by institutional review boards at the James J. Peters VAMC and Greater Los Angeles VAMC. We asked participants for verbal consent to participate and preserved their confidentiality.

Analysis

Data were analyzed via an inductive approach, which involves drawing salient themes rather than imposing preconceived theories.20 Three researchers (TWL, EF, and ES) listened to and discussed 2 staff interviews and tagged text with specific codes (eg, communication between the VHA and HHA, internal communication, and barriers to case fulfillment) so the team could selectively return to the interview text for deeper analysis, allowing for the development of a final codebook. The project team synthesized the findings to identify higher-level themes, drawing comparisons across and within the respondent groups, including within and between health care systems. Throughout the analysis, we maintained analytic memos, documented discussions, and engaged in analyst triangulation to ensure trustworthiness.21,22 To ensure the analysis accurately reflected the participants’ understanding, we held 2 virtual member-checking sessions with participants to share preliminary findings and conclusions and solicit feedback. Analysis was conducted using ATLAS.ti version 20.

Results

VHA-based participants described internal emergency management systems that are deployed during a disaster to support patients and staff. Agency participants described their own internal emergency management protocols. Respondents discussed how and when the 2 intersected, as well as opportunities for future mutual support. The analysis identified several themes: (1) relationships between VAMC teams; (2) relationships between VHA and HHAs; (3) VHA and agencies responses during emergencies; (4) receptivity and opportunities for extending VHA resources into the community; and (5) barriers and facilitators to deeper engagement.

Relationships Within VHA (n = 17)

Staff at all VHA sites described close relationships between the internal emergency management and HBPC teams. HBPC teams identified patients who were most at risk during emergencies to triage those with the highest medical needs (eg, patients dependent on home infusion, oxygen, or electronic medical devices) and worked alongside emergency managers to develop plans to continue care during an emergency. HBPC representatives were part of their facilities’ local emergency response committees. Due to this close collaboration, VHA emergency managers were familiar with the needs of homebound veterans and caregivers. “I invite our [HBPC] program manager to attend [committee] meetings and … they’re part of the EOC [emergency operations center]," an emergency manager said. “We work together and I’m constantly in contact with that individual, especially during natural disasters and so forth, to ensure that everybody’s prepared in the community.”

On the other hand, community caremanagers—who described frequent interactions with HBPC teams, largely around coordinating and managing non-VHA home care services—were less likely to have direct relationships with their facility emergency managers. For example, when asked if they had a relationship with their emergency manager, a community care manager admitted, “I [only] know who he is.” They also did not report having structured protocols for veteran outreach during emergencies, “because all those veterans who are receiving [home health care worker] services also belong to a primary care team,” and considered the outreach to be the responsibility of the primary care team and HHA.

Relationships Between the VHA and HHAs (n = 17)

Communication between VAMCs and contracted agencies primarily went through community care managers, who described established long-term relationships with agency administrators. Communication was commonly restricted to operational activities, such as processing referrals and occasional troubleshooting. According to a community care manager most communication is “why haven’t you signed my orders?” There was a general sense from participants that communication was promptly answered, problems were addressed, and professional collegiality existed between the agencies as patients were referred and placed for services. One community care manager reported meeting with agencies regularly, noting, “I talk to them pretty much daily.”

If problems arose, community care managers described themselves as “the liaison” between agencies and VHA HCPs who ordered the referrals. This is particularly the case if the agency needed help finding a VHA clinician or addressing differences in care delivery protocols.

Responding During Emergencies (n = 19)

During emergencies, VHA and agency staff described following their own organization’s protocols and communicating with each other only on a case-by-case basis rather than through formal or systematic channels and had little knowledge of their counterpart’s emergency protocols. Beyond patient care, there was no evidence of information sharing between VHA and agency staff. Regarding sharing information with their local community, an HBPC Program Director said, “it’s almost like the VHA had become siloed” and operated on its own without engaging with community health systems or emergency managers.

 

Beyond the guidance provided by state departments of public health, HHAs described collaborating with other agencies in their network and relying on their informal professional network to manage the volume of information and updates they followed during emergencies like the COVID-19 pandemic. One agency administrator did not frequently communicate with VHA partners during the pandemic but explained that the local public health department helped work through challenges. However, “we realized pretty quickly they were overloaded and there was only so much they could do.” The agency administrator turned to a “sister agency” and local hospitals, noting, “Wherever you have connections in the field or in the industry, you know you’re going to reach out to people for guidance on policies and… protocol.”

Opportunities for Extending VHA Resources to the Community (n = 16)

All VHA emergency managers were receptive to extending support to community-based HCPS and, in some cases, felt strongly that they were an essential part of veterans’ care networks. Emergency managers offered examples for how they supportedcommunity-based HCPs, such as helping those in the VAMC medical foster home program develop and evaluate emergency plans. Many said they had not explicitly considered HHAs before (Appendix).

Emergency managers also described how supporting community-based HCPs could be considered within the scope of the VHA role and mission, specifically the Fourth Mission. “I think that we should be making our best effort to make sure that we’re also providing that same level [of protection] to the people taking care of the veteran [as our VHA staff],” an emergency manager said. “It’s our responsibility to provide the best for the staff that are going into those homes to take care of that patient.”

In many cases, emergency managers had already developed practical tools that could be easily shared outside the VHA, including weather alerts, trainings, emergency plan templates, and lists of community resources and shelters (Figure 2). A number of these examples built on existing communication channels. One emergency manager said that the extension of resources could be an opportunity to decrease the perceived isolation of home health care workers through regular training for agencies that are providing health care aides, so that they know that “some bigger folks are keeping an eye on it.”

FIGURE 2. Suggestions Received for Extended Resources to Contracted VA Organizations
Abbreviations: PPE, personal protective equipment; VA, US Department of Veterans Affairs.

On the agency side, participants noted that some HHAs could benefit more from support than others. While some agencies are well staffed and have good protocols and keep up to date, “There are smaller agencies, agencies that are starting up that may not have the resources to just disseminate all the information. Those are the agencies [that] could well benefit from the VHA,” an HBPC medical director explained. Agency administrators suggested several areas where they would welcome support, including a deeper understanding of available community resources and access to PPE for staff. Regarding informational resources, an administrator said, “Anytime we can get information, it’s good to have it come to you and not always have to go out searching for it.”

Barriers and Facilitators to Partnering With Community Agencies (n = 16)

A primary barrier regarding resource sharing was potential misalignment between each organization’s policies. HHAs followed state and federal public health guidelines, which sometimes differed from VHA policies. Given that agencies care for both VHA and non-VHA clients, questions also arose around how agencies would prioritize information from the VHA, if they were already receiving information from other sources. When asked about information sharing, both VHA staff and agencies agreed staff time to support any additional activities should be weighed against the value of the information gained.

 

Six participants also shared that education around emergency preparedness could be an opportunity to bridge gaps between VAMCs and their surrounding communities. One local Chief of Community Care noted, “Any opportunity to just give information is going to make it a lot better for the veteran patient … to have something that’s a little more robust.”

Two emergency managers noted the need to be sensitive in the way they engaged with partners, respecting and building on the work that agencies were already doing in this area to ensure VHA was seen as a trusted partner and resource rather than trying to impose new policies or rules on community-based HCPs. “I know that like all leadership in various organizations, there’s a little bit of bristling going on when other people try and tell them what to do,” an HBPC medical director said. “However, if it is established that as a sort of greater level like a state level or a federal level, that VHA can be a resource. I think that as long as that’s recognized by their own professional organizations within each state, then I think that that would be a tremendous advantage to many agencies.”

In terms of sharing physical resources, emergency managers raised concerns around potential liability, although they also acknowledged this issue was important enough to think about potential workarounds. As one emergency manager said, “I want to know that my PPE is not compromised in any way shape or form and that I am in charge of that PPE, so to rely upon going to a home and hoping that [the PPE] wasn’t compromised … would kind of make me a little uneasy.” This emergency manager suggested possible solutions, such as creating a sealed PPE package to give directly to an aide.

Discussion

As the prevalence of climate-related disasters increases, the need to ensure the safety and independence of older adults during emergencies grows more urgent. Health systems must think beyond the direct services they provide and consider the community resources upon which their patients rely. While relationships did not formally exist between VHA emergency managers and community home health HCPs in the sample analyzed in this article, there is precedent and interest in supporting contracted home health agencies caring for veterans in the community. Although not historically part of the VA Fourth Mission, creating a pipeline of support for contracted HHAs by leveraging existing relationships and resources can potentially strengthen its mission to protect older veterans in emergencies, help them age safely in place, and provide a model for health systems to collaborate with community-based HCPs around emergency planning and response (Figure 3).23

FIGURE 3. Support Pipeline for Contracted US Department of Veterans Affairs Organizations

Existing research on the value of health care coalitions highlights the need for established and growing partnerships with a focus on ensuring they are value-added, which echoes concerns we heard in interviews.24 Investment in community partnerships not only includes sharing supplies but also relying on bidirectional support that can be a trusted form of timely information.1,25 The findings in this study exhibit strong communication practices within the VHA during periods of nonemergency and underscore the untapped value of the pre-existing relationship between VAMCs and their contracted HHAs as an area of potential growth for health care coalitions.

Sharing resources in a way that does not put new demands on partners contributes to the sustainability and value-added nature of coalitions. Examples include establishing new low-investment practices (ie, information sharing) that support capacity and compliance with existing requirements rather than create new responsibilities for either member of the coalition. The relationship between the VHA emergency managers and the VHA HBPC program can act as a guide. The emergency managers interviewed for this study are currently engaged with HBPC programs and therefore understand the needs of homebound older adults and their caregivers. Extending the information already available to the HBPC teams via existing channels strengthens workforce practices and increased security for the shared patient, even without direct relationships between emergency managers and agencies. It is important to understand the limitations of these practices, including concerns around conflicting federal and state mandates, legal concerns around the liability of sharing physical resources (such as PPE), and awareness that the objective is not for the VHA to increase burdens (eg, increasing compliance requirements) but rather to serve as a resource for a mutual population in a shared community.

Offering training and practical resources to HHA home health care workers can help them meet disaster preparedness requirements. This is particularly important considering the growing home care workforce shortages, a topic mentioned by all HBPC and community care participants interviewed for this study.26,27 Home health care workers report feeling underprepared and isolated while on the job in normal conditions, a sentiment exacerbated by the COVID-19 pandemic.3,10 Supporting these individuals may help them feel more prepared and connected to their work, improving stability and quality of care.

While these issues are priorities within the VHA, there is growing recognition at the state and federal level of the importance of including older adults and their HCPs in disaster preparedness and response.5,28 The US Department of Health and Human Services, for example, includes older adults and organizations that serve them on its National Advisory Committee on Seniors and Disasters. The Senate version of the 2023 reauthorization of the Pandemic and All-Hazards Preparedness and Response Act included specific provisions to support community-dwelling older adults and people with disabilities, incorporating funding for community organizations to support continuity of services and avoid institutionalization in an emergency.29 Other proposed legislation includes the Real Emergency Access for Aging and Disability Inclusion for Disasters Act, which would ensure the needs of older adults and people with disabilities are explicitly included in all phases of emergency planning and response.30

The VHA expansion of the its VEText program to include disaster response is an effort to more efficiently extend outreach to older and vulnerable patients who are veterans.31 Given these growing efforts, the VHA and other health systems have an opportunity to expand internal emergency preparedness efforts to ensure the health and safety of individuals living in the community.

Limitations

VISN 2 has been a target of terrorism and other disasters. In addition to the sites being initially recruited for their strong emergency management protocols, this context may have biased respondents who are favorable to extending their resources into the community. At the time of recruitment, contracted HHAs were still experiencing staff shortages due to the COVID-19 pandemic, which limited the ability of agency staff to participate in interviews. Additionally, while the comprehensive exploration of VISN 2 facilities allows for confidence of the organizational structures described, the qualitative research design and small study sample, the study findings cannot be immediately generalized to all VISNs.

Conclusions

Many older veterans increasingly rely on home health care workers to age safely. The VHA, as a large national health care system and leader in emergency preparedness, could play an important role in supporting home health care workers and ameliorating their sense of isolation during emergencies and natural disasters. Leveraging existing resources and relationships may be a low-cost, low-effort opportunity to build higher-level interventions that support the needs of patients. Future research and work in this field, including the authors’ ongoing work, will expand agency participation and engage agency staff in conceptualizing pilot projects to ensure they are viable and feasible for the field.

References
  1. Barnett DJ, Knieser L, Errett NA, Rosenblum AJ, Seshamani M, Kirsch TD. Reexamining health-care coalitions in light of COVID-19. Disaster Med public Health Prep. 2022;16(3):859-863. doi:10.1017/dmp.2020.431
  2. Wulff K, Donato D, Lurie N. What is health resilience and how can we build it? Annu Rev Public Health. 2015;36:361-374. doi:10.1146/annurev-publhealth-031914-122829
  3. Franzosa E, Wyte-Lake T, Tsui EK, Reckrey JM, Sterling MR. Essential but excluded: building disaster preparedness capacity for home health care workers and home care agencies. J Am Med Dir Assoc. 2022;23(12):1990-1996. doi:10.1016/j.jamda.2022.09.012
  4. Miner S, Masci L, Chimenti C, Rin N, Mann A, Noonan B. An outreach phone call project: using home health to reach isolated community dwelling adults during the COVID 19 lockdown. J Community Health. 2022;47(2):266-272. doi:10.1007/s10900-021-01044-6
  5. National Institute on Aging. Protecting older adults from the effects of natural disasters and extreme weather. October 18, 2022. Accessed August 19, 2024. https://www.nia.nih.gov/news/protecting-older-adults-effects-natural-disasters-and-extreme-weather
  6. PHI. Direct Care Workers in the United States: Key Facts. September 7, 2021. Accessed August 19, 2024. https://www.phinational.org/resource/direct-care-workers-in-the-united-states-key-facts-2/
  7. Centers for Medicare & Medicaid Services. Emergency Preparedness Rule. September 8, 2016. Updated September 6, 2023. Accessed August 19, 2024. https://www.cms.gov/medicare/health-safety-standards/quality-safety-oversight-emergency-preparedness/emergency-preparedness-rule
  8. Wyte-Lake T, Claver M, Tubbesing S, Davis D, Dobalian A. Development of a home health patient assessment tool for disaster planning. Gerontology. 2019;65(4):353-361. doi:10.1159/000494971
  9. Franzosa E, Judon KM, Gottesman EM, et al. Home health aides’ increased role in supporting older veterans and primary healthcare teams during COVID-19: a qualitative analysis. J Gen Intern Med. 2022;37(8):1830-1837. doi:10.1007/s11606-021-07271-w
  10. Franzosa E, Tsui EK, Baron S. “Who’s caring for us?”: understanding and addressing the effects of emotional labor on home health aides’ well-being. Gerontologist. 2019;59(6):1055-1064. doi:10.1093/geront/gny099
  11. Osakwe ZT, Osborne JC, Samuel T, et al. All alone: a qualitative study of home health aides’ experiences during the COVID-19 pandemic in New York. Am J Infect Control. 2021;49(11):1362-1368. doi:10.1016/j.ajic.2021.08.004
  12. Feldman PH, Russell D, Onorato N, et al. Ensuring the safety of the home health aide workforce and the continuation of essential patient care through sustainable pandemic preparedness. July 2022. Accessed August 19, 2024. https://www.vnshealth.org/wp-content/uploads/2022/08/Pandemic_Preparedness_IB_07_21_22.pdf
  13. Sterling MR, Tseng E, Poon A, et al. Experiences of home health care workers in New York City during the coronavirus disease 2019 pandemic: a qualitative analysis. JAMA Internal Med. 2020;180(11):1453-1459. doi:10.1001/jamainternmed.2020.3930
  14. Wyte-Lake T, Schmitz S, Kornegay RJ, Acevedo F, Dobalian A. Three case studies of community behavioral health support from the US Department of Veterans Affairs after disasters. BMC Public Health. 2021;21(1):639. doi:10.1186/s12889-021-10650-x
  15. Beales JL, Edes T. Veteran’s affairs home based primary care. Clin Geriatr Med. 2009;25(1):149-ix. doi:10.1016/j.cger.2008.11.002
  16. Wyte-Lake T, Manheim C, Gillespie SM, Dobalian A, Haverhals LM. COVID-19 vaccination in VA home based primary care: experience of interdisciplinary team members. J Am Med Dir Assoc. 2022;23(6):917-922. doi:10.1016/j.jamda.2022.03.014
  17. Wyte-Lake T, Schmitz S, Cosme Torres-Sabater R, Dobalian A. Case study of VA Caribbean Healthcare System’s community response to Hurricane Maria. J Emerg Manag. 2022;19(8):189-199. doi:10.5055/jem.0536
  18. US Department of Veterans Affairs. New York/New Jersey VA Health Care Network, VISN 2 Locations. Updated January 3, 2024. Accessed August 19, 2024. https://www.visn2.va.gov/visn2/facilities.asp
  19. Noy C. Sampling knowledge: the hermeneutics of snowball sampling in qualitative research. Int J Soc Res Methodol. 2008;11(4):327-344. doi:10.1080/13645570701401305
  20. Ritchie J, Lewis J, Nicholls CM, Ormston R, eds. Qualitative Research Practice: A Guide for Social Science Students and Researchers. 2nd ed. Sage; 2013.
  21. Morrow SL. Quality and trustworthiness in qualitative research in counseling psychology. J Couns Psychol. 2005;52(2):250-260. doi:10.1037/0022-0167.52.2.250
  22. Rolfe G. Validity, trustworthiness and rigour: quality and the idea of qualitative research. J Adv Nurs. 2006;53(3):304-310. doi:10.1111/j.1365-2648.2006.03727.x
  23. Schmitz S, Wyte-Lake T, Dobalian A. Facilitators and barriers to preparedness partnerships: a veterans affairs medical center perspective. Disaster Med Public Health Prep. 2018;12(4):431-436. doi:10.1017/dmp.2017.92
  24. Koch AE, Bohn J, Corvin JA, Seaberg J. Maturing into high-functioning health-care coalitions: a qualitative Nationwide study of emergency preparedness and response leadership. Disaster Med Public Health Prep. 2022;17:e111. doi:10.1017/dmp.2022.13
  25. Lin JS, Webber EM, Bean SI, Martin AM, Davies MC. Rapid evidence review: policy actions for the integration of public health and health care in the United States. Front Public Health. 2023;11:1098431. doi:10.3389/fpubh.2023.1098431
  26. Watts MOM, Burns A, Ammula M. Ongoing impacts of the pandemic on medicaid home & community-based services (HCBS) programs: findings from a 50-state survey. November 28, 2022. Accessed August 19, 2024. https://www.kff.org/medicaid/issue-brief/ongoing-impacts-of-the-pandemic-on-medicaid-home-community-based-services-hcbs-programs-findings-from-a-50-state-survey/
  27. Kreider AR, Werner RM. The home care workforce has not kept pace with growth in home and community-based services. Health Aff (Millwood). 2023;42(5):650-657. doi:10.1377/hlthaff.2022.01351
  28. FEMA introduces disaster preparedness guide for older adults. News release. FEMA. September 20, 2023. Accessed August 19, 2024. https://www.fema.gov/press-release/20230920/fema-introduces-disaster-preparedness-guide-older-adults
  29. Pandemic and All-Hazards Preparedness and Response Act, S 2333, 118th Cong, 1st Sess (2023). https://www.congress.gov/bill/118th-congress/senate-bill/2333/text
  30. REAADI for Disasters Act, HR 2371, 118th Cong, 1st Sess (2023). https://www.congress.gov/bill/118th-congress/house-bill/2371
  31. Wyte-Lake T, Brewster P, Hubert T, Gin J, Davis D, Dobalian A. VA’s experience building capability to conduct outreach to vulnerable patients during emergencies. Innov Aging. 2023;7(suppl 1):209. doi:10.1093/geroni/igad104.0690
Article PDF
Author and Disclosure Information

Author affiliations

aVeterans Emergency Management Evaluation Center, US Department of Veterans Affairs, North Hills, California

bThe Ohio State University, Columbus

cJames J. Peters Department of Veterans Affairs Medical Center, Bronx, New York

dIcahn School of Medicine at Mount Sinai, New York

Author disclosures

The author(s) disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: This material is based on work supported by the US Department of Veterans Affairs, Veterans Health Administration, Office of Emergency Management and the Office of Population Health. The views expressed in this article are those of the authors and do not necessarily reflect the position or policy of the Department of Veterans Affairs or the United States Government.

Disclaimer

The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the US Government, or any of its agencies.

Ethics and consent

The study was reviewed and determined to be exempt by the James J. Peters Department of Veterans Affairs Medical Center Institutional Review Board and Greater Los Angeles Veterans Affairs Medical Center Institutional Review Board.

Author contributions

Concept and design: Wyte-Lake, Dobalian, and Franzosa. Material preparation, data collection, and analysis: Wyte-Lake, Franzosa, and Solorzano. Drafting of the manuscript: Wyte-Lake and Franzosa. Critical revision of the manuscript: Solorzano, Hall, and Dobalian. All authors read and approved the final manuscript.

Issue
Federal Practitioner - 41(10)a
Publications
Topics
Page Number
1-9
Sections
Author and Disclosure Information

Author affiliations

aVeterans Emergency Management Evaluation Center, US Department of Veterans Affairs, North Hills, California

bThe Ohio State University, Columbus

cJames J. Peters Department of Veterans Affairs Medical Center, Bronx, New York

dIcahn School of Medicine at Mount Sinai, New York

Author disclosures

The author(s) disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: This material is based on work supported by the US Department of Veterans Affairs, Veterans Health Administration, Office of Emergency Management and the Office of Population Health. The views expressed in this article are those of the authors and do not necessarily reflect the position or policy of the Department of Veterans Affairs or the United States Government.

Disclaimer

The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the US Government, or any of its agencies.

Ethics and consent

The study was reviewed and determined to be exempt by the James J. Peters Department of Veterans Affairs Medical Center Institutional Review Board and Greater Los Angeles Veterans Affairs Medical Center Institutional Review Board.

Author contributions

Concept and design: Wyte-Lake, Dobalian, and Franzosa. Material preparation, data collection, and analysis: Wyte-Lake, Franzosa, and Solorzano. Drafting of the manuscript: Wyte-Lake and Franzosa. Critical revision of the manuscript: Solorzano, Hall, and Dobalian. All authors read and approved the final manuscript.

Author and Disclosure Information

Author affiliations

aVeterans Emergency Management Evaluation Center, US Department of Veterans Affairs, North Hills, California

bThe Ohio State University, Columbus

cJames J. Peters Department of Veterans Affairs Medical Center, Bronx, New York

dIcahn School of Medicine at Mount Sinai, New York

Author disclosures

The author(s) disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: This material is based on work supported by the US Department of Veterans Affairs, Veterans Health Administration, Office of Emergency Management and the Office of Population Health. The views expressed in this article are those of the authors and do not necessarily reflect the position or policy of the Department of Veterans Affairs or the United States Government.

Disclaimer

The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the US Government, or any of its agencies.

Ethics and consent

The study was reviewed and determined to be exempt by the James J. Peters Department of Veterans Affairs Medical Center Institutional Review Board and Greater Los Angeles Veterans Affairs Medical Center Institutional Review Board.

Author contributions

Concept and design: Wyte-Lake, Dobalian, and Franzosa. Material preparation, data collection, and analysis: Wyte-Lake, Franzosa, and Solorzano. Drafting of the manuscript: Wyte-Lake and Franzosa. Critical revision of the manuscript: Solorzano, Hall, and Dobalian. All authors read and approved the final manuscript.

Article PDF
Article PDF

As large-scale natural disasters become more common, health care coalitions and the engagement of health systems with local, state, and federal public health departments have effectively bolstered communities’ resilience via collective sharing and distribution of resources.1 These resources may include supplies and the dissemination of emergency information, education, and training.2 The COVID-19 pandemic demonstrated that larger health care systems including hospital networks and nursing homes are better connected to health care coalition resources than smaller, independent systems, such as community home health agencies.3 This leaves some organizations on their own to meet requirements that maintain continuity of care and support their patients and staff throughout a natural disaster.

Home health care workers play important roles in the care of older adults.4 Older adults experience high levels of disability and comorbidities that put them at risk during emergencies; they often require support from paid, family, and neighborhood caregivers to live independently.5 More than 9.3 million US adults receive paid care from 2.6 million home health care workers (eg, home health aides and personal care assistants).6 Many of these individuals are hired through small independent home health agencies (HHAs), while others may work directly for an individual. When neighborhood resources and family caregiving are disrupted during emergencies, the critical services these workers administer become even more essential to ensuring continued access to medical care and social services.

The importance of these services was underscored by the Centers for Medicare and Medicaid Services 2017 inclusion of HHAs in federal emergency preparedness guidelines.7,8 The fractured and decentralized nature of the home health care industry means many HHAs struggle to maintain continuous care during emergencies and protect their staff. HHAs, and health care workers in the home, are often isolated, under-resourced, and disconnected from broader emergency planning efforts. Additionally, home care jobs are largely part-time, unstable, and low paying, making the workers themselves vulnerable during emergencies.3,9-13

This is a significant issue for the Veterans Health Administration (VHA), which annually purchases 10.5 million home health care worker visits for 150,000 veterans from community-based HHAs to enable those individuals to live independently. Figure 1 illustrates the existing structure of directly provided and contracted VHA services for community-dwelling veterans, highlighting the circle of care around the veteran.8,9 Home health care workers anchored health care teams during the COVID-19 pandemic, observing and reporting on patients’ well-being to family caregivers, primary care practitioners, and HHAs. They also provided critical emotional support and companionship to patients isolated from family and friends.9 These workers also exposed themselves and their families to considerable risk and often lacked the protection afforded by personal protective equipment (PPE) in accordance with infection prevention guidance.3,12

FIGURE 1. Circle of Care for Community-Dwelling Veterans
Abbreviations: HBPC, home based primary care; HHA, home health agency; VHA, Veterans Health Administration.
aAdapted with permission from Wyte-Lake and Franzosa.8,9

Through a combination of its national and local health care networks, the VHA has a robust and well-positioned emergency infrastructure to supportcommunity-dwelling older adults during disasters.14 This network is supported by the VHA Office of Emergency Management, which shares resources and guidance with local emergency managers at each facility as well as individual programs such as the VHA Home Based Primary Care (HBPC) program, which provides 38,000 seriously ill veterans with home medical visits.15 Working closely with their local and national hospital networks and emergency managers, individual VHA HBPC programs were able to maintain the safety of staff and continuity of care for patients enrolled in HBPC by rapidly administering COVID-19 vaccines to patients, caregivers, and staff, and providing emergency assistance during the 2017 hurricane season.16,17 These efforts were successful because HBPC practitioners and their patients, had access to a level of emergency-related information, resources, and technology that are often out of reach for individual community-based health care practitioners (HCPs). The US Department of Veterans Affairs (VA) also supports local communities through its Fourth Mission, which provides emergency resources to non-VHA health care facilities (ie, hospitals and nursing homes) during national emergencies and natural disasters.17 Although there has been an expansion in the definition of shared resources, such as extending behavioral health support to local communities, the VHA has not historically provided these resources to HHAs.14



This study examines opportunities to leverage VHA emergency management resources to support contracted HHAs and inform other large health system emergency planning efforts. The findings from the exploratory phase are described in this article. We interviewed VHA emergency managers, HBPC and VA staff who coordinate home health care worker services, as well as administrators at contracted HHAs within a Veterans Integrated Services Network (VISN). These findings will inform the second (single-site pilot study) and third (feasibility study) phases. Our intent was to (1) better understand the relationships between VA medical centers (VAMCs) and their contracted HHAs; (2) identify existing VHA emergency protocols to support community-dwelling older adults; and (3) determine opportunities to build on existing infrastructure and relationships to better support contracted HHAs and their staff in emergencies.

 

Methods

The 18 VISNs act as regional systems of care that are loosely connected to better meet local health needs and maximize access to care. This study was conducted at 6 of 9 VAMCs within VISN 2, the New York/New Jersey VHA Health Care Network.18 VAMCs that serve urban, rural, and mixed urban/rural catchment areas were included.

Each VAMC has an emergency management program led by an emergency manager, an HBPC program led by a program director and medical director, and a community care or purchased care office that has a liaison who manages contracted home health care worker services. The studyfocused on HBPC programs because they are most likely to interact with veterans’ home health care workers in the home and care for community-dwelling veterans during emergencies. Each VHA also contracts with a series of local HHAs that generally have a dedicated staff member who interfaces with the VHA liaison. Our goal was to interview ≥ 1 emergency manager, ≥ 1 HBPC team member, ≥ 1 community care staff person, and ≥ 1 contracted home health agency administrator at each site to gain multiple perspectives from the range of HCPs serving veterans in the community.

 

Recruitment and Data Collection

The 6 sites were selected in consultation with VISN 2 leadership for their strong HBPC and emergency management programs. To recruit respondents, we contacted VISN and VAMC leads and used our professional networks to identify a sample of multidisciplinary individuals who represent both community care and HBPC programs who were contacted via email.

Since each VAMC is organized differently, we utilized a snowball sampling approach to identify the appropriate contacts.19 At the completion of each interview, we asked the participant to suggest additional contacts and introduce us to any remaining stakeholders (eg, the emergency manager) at that site or colleagues at other VISN facilities. Because roles vary among VAMCs, we contacted the person who most closely resembled the identified role and asked them to direct us to a more appropriate contact, if necessary. We asked community care managers to identify 1 to 2 agencies serving the highest volume of patients who are veterans at their site and requested interviews with those liaisons. This resulted in the recruitment of key stakeholders from 4 teams across the 6 sites (Table).

A semistructured interview guide was jointly developed based on constructs of interest, including relationships within VAMCs and between VAMCs and HHAs; existing emergency protocols and experience during disasters; and suggestions and opportunities for supporting agencies during emergencies and potential barriers. Two researchers (TWL and EF) who were trained in qualitative methods jointly conducted interviews using the interview guide, with 1 researcher leading and another taking notes and asking clarifying questions.

Interviews were conducted virtually via Microsoft Teams with respondents at their work locations between September 2022 and January 2023. Interviews were audio recorded and transcribed and 2 authors (TWL and ESO) reviewed transcripts for accuracy. Interviews averaged 47 minutes in length (range, 20-59).

The study was reviewed and determined to be exempt by institutional review boards at the James J. Peters VAMC and Greater Los Angeles VAMC. We asked participants for verbal consent to participate and preserved their confidentiality.

Analysis

Data were analyzed via an inductive approach, which involves drawing salient themes rather than imposing preconceived theories.20 Three researchers (TWL, EF, and ES) listened to and discussed 2 staff interviews and tagged text with specific codes (eg, communication between the VHA and HHA, internal communication, and barriers to case fulfillment) so the team could selectively return to the interview text for deeper analysis, allowing for the development of a final codebook. The project team synthesized the findings to identify higher-level themes, drawing comparisons across and within the respondent groups, including within and between health care systems. Throughout the analysis, we maintained analytic memos, documented discussions, and engaged in analyst triangulation to ensure trustworthiness.21,22 To ensure the analysis accurately reflected the participants’ understanding, we held 2 virtual member-checking sessions with participants to share preliminary findings and conclusions and solicit feedback. Analysis was conducted using ATLAS.ti version 20.

Results

VHA-based participants described internal emergency management systems that are deployed during a disaster to support patients and staff. Agency participants described their own internal emergency management protocols. Respondents discussed how and when the 2 intersected, as well as opportunities for future mutual support. The analysis identified several themes: (1) relationships between VAMC teams; (2) relationships between VHA and HHAs; (3) VHA and agencies responses during emergencies; (4) receptivity and opportunities for extending VHA resources into the community; and (5) barriers and facilitators to deeper engagement.

Relationships Within VHA (n = 17)

Staff at all VHA sites described close relationships between the internal emergency management and HBPC teams. HBPC teams identified patients who were most at risk during emergencies to triage those with the highest medical needs (eg, patients dependent on home infusion, oxygen, or electronic medical devices) and worked alongside emergency managers to develop plans to continue care during an emergency. HBPC representatives were part of their facilities’ local emergency response committees. Due to this close collaboration, VHA emergency managers were familiar with the needs of homebound veterans and caregivers. “I invite our [HBPC] program manager to attend [committee] meetings and … they’re part of the EOC [emergency operations center]," an emergency manager said. “We work together and I’m constantly in contact with that individual, especially during natural disasters and so forth, to ensure that everybody’s prepared in the community.”

On the other hand, community caremanagers—who described frequent interactions with HBPC teams, largely around coordinating and managing non-VHA home care services—were less likely to have direct relationships with their facility emergency managers. For example, when asked if they had a relationship with their emergency manager, a community care manager admitted, “I [only] know who he is.” They also did not report having structured protocols for veteran outreach during emergencies, “because all those veterans who are receiving [home health care worker] services also belong to a primary care team,” and considered the outreach to be the responsibility of the primary care team and HHA.

Relationships Between the VHA and HHAs (n = 17)

Communication between VAMCs and contracted agencies primarily went through community care managers, who described established long-term relationships with agency administrators. Communication was commonly restricted to operational activities, such as processing referrals and occasional troubleshooting. According to a community care manager most communication is “why haven’t you signed my orders?” There was a general sense from participants that communication was promptly answered, problems were addressed, and professional collegiality existed between the agencies as patients were referred and placed for services. One community care manager reported meeting with agencies regularly, noting, “I talk to them pretty much daily.”

If problems arose, community care managers described themselves as “the liaison” between agencies and VHA HCPs who ordered the referrals. This is particularly the case if the agency needed help finding a VHA clinician or addressing differences in care delivery protocols.

Responding During Emergencies (n = 19)

During emergencies, VHA and agency staff described following their own organization’s protocols and communicating with each other only on a case-by-case basis rather than through formal or systematic channels and had little knowledge of their counterpart’s emergency protocols. Beyond patient care, there was no evidence of information sharing between VHA and agency staff. Regarding sharing information with their local community, an HBPC Program Director said, “it’s almost like the VHA had become siloed” and operated on its own without engaging with community health systems or emergency managers.

 

Beyond the guidance provided by state departments of public health, HHAs described collaborating with other agencies in their network and relying on their informal professional network to manage the volume of information and updates they followed during emergencies like the COVID-19 pandemic. One agency administrator did not frequently communicate with VHA partners during the pandemic but explained that the local public health department helped work through challenges. However, “we realized pretty quickly they were overloaded and there was only so much they could do.” The agency administrator turned to a “sister agency” and local hospitals, noting, “Wherever you have connections in the field or in the industry, you know you’re going to reach out to people for guidance on policies and… protocol.”

Opportunities for Extending VHA Resources to the Community (n = 16)

All VHA emergency managers were receptive to extending support to community-based HCPS and, in some cases, felt strongly that they were an essential part of veterans’ care networks. Emergency managers offered examples for how they supportedcommunity-based HCPs, such as helping those in the VAMC medical foster home program develop and evaluate emergency plans. Many said they had not explicitly considered HHAs before (Appendix).

Emergency managers also described how supporting community-based HCPs could be considered within the scope of the VHA role and mission, specifically the Fourth Mission. “I think that we should be making our best effort to make sure that we’re also providing that same level [of protection] to the people taking care of the veteran [as our VHA staff],” an emergency manager said. “It’s our responsibility to provide the best for the staff that are going into those homes to take care of that patient.”

In many cases, emergency managers had already developed practical tools that could be easily shared outside the VHA, including weather alerts, trainings, emergency plan templates, and lists of community resources and shelters (Figure 2). A number of these examples built on existing communication channels. One emergency manager said that the extension of resources could be an opportunity to decrease the perceived isolation of home health care workers through regular training for agencies that are providing health care aides, so that they know that “some bigger folks are keeping an eye on it.”

FIGURE 2. Suggestions Received for Extended Resources to Contracted VA Organizations
Abbreviations: PPE, personal protective equipment; VA, US Department of Veterans Affairs.

On the agency side, participants noted that some HHAs could benefit more from support than others. While some agencies are well staffed and have good protocols and keep up to date, “There are smaller agencies, agencies that are starting up that may not have the resources to just disseminate all the information. Those are the agencies [that] could well benefit from the VHA,” an HBPC medical director explained. Agency administrators suggested several areas where they would welcome support, including a deeper understanding of available community resources and access to PPE for staff. Regarding informational resources, an administrator said, “Anytime we can get information, it’s good to have it come to you and not always have to go out searching for it.”

Barriers and Facilitators to Partnering With Community Agencies (n = 16)

A primary barrier regarding resource sharing was potential misalignment between each organization’s policies. HHAs followed state and federal public health guidelines, which sometimes differed from VHA policies. Given that agencies care for both VHA and non-VHA clients, questions also arose around how agencies would prioritize information from the VHA, if they were already receiving information from other sources. When asked about information sharing, both VHA staff and agencies agreed staff time to support any additional activities should be weighed against the value of the information gained.

 

Six participants also shared that education around emergency preparedness could be an opportunity to bridge gaps between VAMCs and their surrounding communities. One local Chief of Community Care noted, “Any opportunity to just give information is going to make it a lot better for the veteran patient … to have something that’s a little more robust.”

Two emergency managers noted the need to be sensitive in the way they engaged with partners, respecting and building on the work that agencies were already doing in this area to ensure VHA was seen as a trusted partner and resource rather than trying to impose new policies or rules on community-based HCPs. “I know that like all leadership in various organizations, there’s a little bit of bristling going on when other people try and tell them what to do,” an HBPC medical director said. “However, if it is established that as a sort of greater level like a state level or a federal level, that VHA can be a resource. I think that as long as that’s recognized by their own professional organizations within each state, then I think that that would be a tremendous advantage to many agencies.”

In terms of sharing physical resources, emergency managers raised concerns around potential liability, although they also acknowledged this issue was important enough to think about potential workarounds. As one emergency manager said, “I want to know that my PPE is not compromised in any way shape or form and that I am in charge of that PPE, so to rely upon going to a home and hoping that [the PPE] wasn’t compromised … would kind of make me a little uneasy.” This emergency manager suggested possible solutions, such as creating a sealed PPE package to give directly to an aide.

Discussion

As the prevalence of climate-related disasters increases, the need to ensure the safety and independence of older adults during emergencies grows more urgent. Health systems must think beyond the direct services they provide and consider the community resources upon which their patients rely. While relationships did not formally exist between VHA emergency managers and community home health HCPs in the sample analyzed in this article, there is precedent and interest in supporting contracted home health agencies caring for veterans in the community. Although not historically part of the VA Fourth Mission, creating a pipeline of support for contracted HHAs by leveraging existing relationships and resources can potentially strengthen its mission to protect older veterans in emergencies, help them age safely in place, and provide a model for health systems to collaborate with community-based HCPs around emergency planning and response (Figure 3).23

FIGURE 3. Support Pipeline for Contracted US Department of Veterans Affairs Organizations

Existing research on the value of health care coalitions highlights the need for established and growing partnerships with a focus on ensuring they are value-added, which echoes concerns we heard in interviews.24 Investment in community partnerships not only includes sharing supplies but also relying on bidirectional support that can be a trusted form of timely information.1,25 The findings in this study exhibit strong communication practices within the VHA during periods of nonemergency and underscore the untapped value of the pre-existing relationship between VAMCs and their contracted HHAs as an area of potential growth for health care coalitions.

Sharing resources in a way that does not put new demands on partners contributes to the sustainability and value-added nature of coalitions. Examples include establishing new low-investment practices (ie, information sharing) that support capacity and compliance with existing requirements rather than create new responsibilities for either member of the coalition. The relationship between the VHA emergency managers and the VHA HBPC program can act as a guide. The emergency managers interviewed for this study are currently engaged with HBPC programs and therefore understand the needs of homebound older adults and their caregivers. Extending the information already available to the HBPC teams via existing channels strengthens workforce practices and increased security for the shared patient, even without direct relationships between emergency managers and agencies. It is important to understand the limitations of these practices, including concerns around conflicting federal and state mandates, legal concerns around the liability of sharing physical resources (such as PPE), and awareness that the objective is not for the VHA to increase burdens (eg, increasing compliance requirements) but rather to serve as a resource for a mutual population in a shared community.

Offering training and practical resources to HHA home health care workers can help them meet disaster preparedness requirements. This is particularly important considering the growing home care workforce shortages, a topic mentioned by all HBPC and community care participants interviewed for this study.26,27 Home health care workers report feeling underprepared and isolated while on the job in normal conditions, a sentiment exacerbated by the COVID-19 pandemic.3,10 Supporting these individuals may help them feel more prepared and connected to their work, improving stability and quality of care.

While these issues are priorities within the VHA, there is growing recognition at the state and federal level of the importance of including older adults and their HCPs in disaster preparedness and response.5,28 The US Department of Health and Human Services, for example, includes older adults and organizations that serve them on its National Advisory Committee on Seniors and Disasters. The Senate version of the 2023 reauthorization of the Pandemic and All-Hazards Preparedness and Response Act included specific provisions to support community-dwelling older adults and people with disabilities, incorporating funding for community organizations to support continuity of services and avoid institutionalization in an emergency.29 Other proposed legislation includes the Real Emergency Access for Aging and Disability Inclusion for Disasters Act, which would ensure the needs of older adults and people with disabilities are explicitly included in all phases of emergency planning and response.30

The VHA expansion of the its VEText program to include disaster response is an effort to more efficiently extend outreach to older and vulnerable patients who are veterans.31 Given these growing efforts, the VHA and other health systems have an opportunity to expand internal emergency preparedness efforts to ensure the health and safety of individuals living in the community.

Limitations

VISN 2 has been a target of terrorism and other disasters. In addition to the sites being initially recruited for their strong emergency management protocols, this context may have biased respondents who are favorable to extending their resources into the community. At the time of recruitment, contracted HHAs were still experiencing staff shortages due to the COVID-19 pandemic, which limited the ability of agency staff to participate in interviews. Additionally, while the comprehensive exploration of VISN 2 facilities allows for confidence of the organizational structures described, the qualitative research design and small study sample, the study findings cannot be immediately generalized to all VISNs.

Conclusions

Many older veterans increasingly rely on home health care workers to age safely. The VHA, as a large national health care system and leader in emergency preparedness, could play an important role in supporting home health care workers and ameliorating their sense of isolation during emergencies and natural disasters. Leveraging existing resources and relationships may be a low-cost, low-effort opportunity to build higher-level interventions that support the needs of patients. Future research and work in this field, including the authors’ ongoing work, will expand agency participation and engage agency staff in conceptualizing pilot projects to ensure they are viable and feasible for the field.

As large-scale natural disasters become more common, health care coalitions and the engagement of health systems with local, state, and federal public health departments have effectively bolstered communities’ resilience via collective sharing and distribution of resources.1 These resources may include supplies and the dissemination of emergency information, education, and training.2 The COVID-19 pandemic demonstrated that larger health care systems including hospital networks and nursing homes are better connected to health care coalition resources than smaller, independent systems, such as community home health agencies.3 This leaves some organizations on their own to meet requirements that maintain continuity of care and support their patients and staff throughout a natural disaster.

Home health care workers play important roles in the care of older adults.4 Older adults experience high levels of disability and comorbidities that put them at risk during emergencies; they often require support from paid, family, and neighborhood caregivers to live independently.5 More than 9.3 million US adults receive paid care from 2.6 million home health care workers (eg, home health aides and personal care assistants).6 Many of these individuals are hired through small independent home health agencies (HHAs), while others may work directly for an individual. When neighborhood resources and family caregiving are disrupted during emergencies, the critical services these workers administer become even more essential to ensuring continued access to medical care and social services.

The importance of these services was underscored by the Centers for Medicare and Medicaid Services 2017 inclusion of HHAs in federal emergency preparedness guidelines.7,8 The fractured and decentralized nature of the home health care industry means many HHAs struggle to maintain continuous care during emergencies and protect their staff. HHAs, and health care workers in the home, are often isolated, under-resourced, and disconnected from broader emergency planning efforts. Additionally, home care jobs are largely part-time, unstable, and low paying, making the workers themselves vulnerable during emergencies.3,9-13

This is a significant issue for the Veterans Health Administration (VHA), which annually purchases 10.5 million home health care worker visits for 150,000 veterans from community-based HHAs to enable those individuals to live independently. Figure 1 illustrates the existing structure of directly provided and contracted VHA services for community-dwelling veterans, highlighting the circle of care around the veteran.8,9 Home health care workers anchored health care teams during the COVID-19 pandemic, observing and reporting on patients’ well-being to family caregivers, primary care practitioners, and HHAs. They also provided critical emotional support and companionship to patients isolated from family and friends.9 These workers also exposed themselves and their families to considerable risk and often lacked the protection afforded by personal protective equipment (PPE) in accordance with infection prevention guidance.3,12

FIGURE 1. Circle of Care for Community-Dwelling Veterans
Abbreviations: HBPC, home based primary care; HHA, home health agency; VHA, Veterans Health Administration.
aAdapted with permission from Wyte-Lake and Franzosa.8,9

Through a combination of its national and local health care networks, the VHA has a robust and well-positioned emergency infrastructure to supportcommunity-dwelling older adults during disasters.14 This network is supported by the VHA Office of Emergency Management, which shares resources and guidance with local emergency managers at each facility as well as individual programs such as the VHA Home Based Primary Care (HBPC) program, which provides 38,000 seriously ill veterans with home medical visits.15 Working closely with their local and national hospital networks and emergency managers, individual VHA HBPC programs were able to maintain the safety of staff and continuity of care for patients enrolled in HBPC by rapidly administering COVID-19 vaccines to patients, caregivers, and staff, and providing emergency assistance during the 2017 hurricane season.16,17 These efforts were successful because HBPC practitioners and their patients, had access to a level of emergency-related information, resources, and technology that are often out of reach for individual community-based health care practitioners (HCPs). The US Department of Veterans Affairs (VA) also supports local communities through its Fourth Mission, which provides emergency resources to non-VHA health care facilities (ie, hospitals and nursing homes) during national emergencies and natural disasters.17 Although there has been an expansion in the definition of shared resources, such as extending behavioral health support to local communities, the VHA has not historically provided these resources to HHAs.14



This study examines opportunities to leverage VHA emergency management resources to support contracted HHAs and inform other large health system emergency planning efforts. The findings from the exploratory phase are described in this article. We interviewed VHA emergency managers, HBPC and VA staff who coordinate home health care worker services, as well as administrators at contracted HHAs within a Veterans Integrated Services Network (VISN). These findings will inform the second (single-site pilot study) and third (feasibility study) phases. Our intent was to (1) better understand the relationships between VA medical centers (VAMCs) and their contracted HHAs; (2) identify existing VHA emergency protocols to support community-dwelling older adults; and (3) determine opportunities to build on existing infrastructure and relationships to better support contracted HHAs and their staff in emergencies.

 

Methods

The 18 VISNs act as regional systems of care that are loosely connected to better meet local health needs and maximize access to care. This study was conducted at 6 of 9 VAMCs within VISN 2, the New York/New Jersey VHA Health Care Network.18 VAMCs that serve urban, rural, and mixed urban/rural catchment areas were included.

Each VAMC has an emergency management program led by an emergency manager, an HBPC program led by a program director and medical director, and a community care or purchased care office that has a liaison who manages contracted home health care worker services. The studyfocused on HBPC programs because they are most likely to interact with veterans’ home health care workers in the home and care for community-dwelling veterans during emergencies. Each VHA also contracts with a series of local HHAs that generally have a dedicated staff member who interfaces with the VHA liaison. Our goal was to interview ≥ 1 emergency manager, ≥ 1 HBPC team member, ≥ 1 community care staff person, and ≥ 1 contracted home health agency administrator at each site to gain multiple perspectives from the range of HCPs serving veterans in the community.

 

Recruitment and Data Collection

The 6 sites were selected in consultation with VISN 2 leadership for their strong HBPC and emergency management programs. To recruit respondents, we contacted VISN and VAMC leads and used our professional networks to identify a sample of multidisciplinary individuals who represent both community care and HBPC programs who were contacted via email.

Since each VAMC is organized differently, we utilized a snowball sampling approach to identify the appropriate contacts.19 At the completion of each interview, we asked the participant to suggest additional contacts and introduce us to any remaining stakeholders (eg, the emergency manager) at that site or colleagues at other VISN facilities. Because roles vary among VAMCs, we contacted the person who most closely resembled the identified role and asked them to direct us to a more appropriate contact, if necessary. We asked community care managers to identify 1 to 2 agencies serving the highest volume of patients who are veterans at their site and requested interviews with those liaisons. This resulted in the recruitment of key stakeholders from 4 teams across the 6 sites (Table).

A semistructured interview guide was jointly developed based on constructs of interest, including relationships within VAMCs and between VAMCs and HHAs; existing emergency protocols and experience during disasters; and suggestions and opportunities for supporting agencies during emergencies and potential barriers. Two researchers (TWL and EF) who were trained in qualitative methods jointly conducted interviews using the interview guide, with 1 researcher leading and another taking notes and asking clarifying questions.

Interviews were conducted virtually via Microsoft Teams with respondents at their work locations between September 2022 and January 2023. Interviews were audio recorded and transcribed and 2 authors (TWL and ESO) reviewed transcripts for accuracy. Interviews averaged 47 minutes in length (range, 20-59).

The study was reviewed and determined to be exempt by institutional review boards at the James J. Peters VAMC and Greater Los Angeles VAMC. We asked participants for verbal consent to participate and preserved their confidentiality.

Analysis

Data were analyzed via an inductive approach, which involves drawing salient themes rather than imposing preconceived theories.20 Three researchers (TWL, EF, and ES) listened to and discussed 2 staff interviews and tagged text with specific codes (eg, communication between the VHA and HHA, internal communication, and barriers to case fulfillment) so the team could selectively return to the interview text for deeper analysis, allowing for the development of a final codebook. The project team synthesized the findings to identify higher-level themes, drawing comparisons across and within the respondent groups, including within and between health care systems. Throughout the analysis, we maintained analytic memos, documented discussions, and engaged in analyst triangulation to ensure trustworthiness.21,22 To ensure the analysis accurately reflected the participants’ understanding, we held 2 virtual member-checking sessions with participants to share preliminary findings and conclusions and solicit feedback. Analysis was conducted using ATLAS.ti version 20.

Results

VHA-based participants described internal emergency management systems that are deployed during a disaster to support patients and staff. Agency participants described their own internal emergency management protocols. Respondents discussed how and when the 2 intersected, as well as opportunities for future mutual support. The analysis identified several themes: (1) relationships between VAMC teams; (2) relationships between VHA and HHAs; (3) VHA and agencies responses during emergencies; (4) receptivity and opportunities for extending VHA resources into the community; and (5) barriers and facilitators to deeper engagement.

Relationships Within VHA (n = 17)

Staff at all VHA sites described close relationships between the internal emergency management and HBPC teams. HBPC teams identified patients who were most at risk during emergencies to triage those with the highest medical needs (eg, patients dependent on home infusion, oxygen, or electronic medical devices) and worked alongside emergency managers to develop plans to continue care during an emergency. HBPC representatives were part of their facilities’ local emergency response committees. Due to this close collaboration, VHA emergency managers were familiar with the needs of homebound veterans and caregivers. “I invite our [HBPC] program manager to attend [committee] meetings and … they’re part of the EOC [emergency operations center]," an emergency manager said. “We work together and I’m constantly in contact with that individual, especially during natural disasters and so forth, to ensure that everybody’s prepared in the community.”

On the other hand, community caremanagers—who described frequent interactions with HBPC teams, largely around coordinating and managing non-VHA home care services—were less likely to have direct relationships with their facility emergency managers. For example, when asked if they had a relationship with their emergency manager, a community care manager admitted, “I [only] know who he is.” They also did not report having structured protocols for veteran outreach during emergencies, “because all those veterans who are receiving [home health care worker] services also belong to a primary care team,” and considered the outreach to be the responsibility of the primary care team and HHA.

Relationships Between the VHA and HHAs (n = 17)

Communication between VAMCs and contracted agencies primarily went through community care managers, who described established long-term relationships with agency administrators. Communication was commonly restricted to operational activities, such as processing referrals and occasional troubleshooting. According to a community care manager most communication is “why haven’t you signed my orders?” There was a general sense from participants that communication was promptly answered, problems were addressed, and professional collegiality existed between the agencies as patients were referred and placed for services. One community care manager reported meeting with agencies regularly, noting, “I talk to them pretty much daily.”

If problems arose, community care managers described themselves as “the liaison” between agencies and VHA HCPs who ordered the referrals. This is particularly the case if the agency needed help finding a VHA clinician or addressing differences in care delivery protocols.

Responding During Emergencies (n = 19)

During emergencies, VHA and agency staff described following their own organization’s protocols and communicating with each other only on a case-by-case basis rather than through formal or systematic channels and had little knowledge of their counterpart’s emergency protocols. Beyond patient care, there was no evidence of information sharing between VHA and agency staff. Regarding sharing information with their local community, an HBPC Program Director said, “it’s almost like the VHA had become siloed” and operated on its own without engaging with community health systems or emergency managers.

 

Beyond the guidance provided by state departments of public health, HHAs described collaborating with other agencies in their network and relying on their informal professional network to manage the volume of information and updates they followed during emergencies like the COVID-19 pandemic. One agency administrator did not frequently communicate with VHA partners during the pandemic but explained that the local public health department helped work through challenges. However, “we realized pretty quickly they were overloaded and there was only so much they could do.” The agency administrator turned to a “sister agency” and local hospitals, noting, “Wherever you have connections in the field or in the industry, you know you’re going to reach out to people for guidance on policies and… protocol.”

Opportunities for Extending VHA Resources to the Community (n = 16)

All VHA emergency managers were receptive to extending support to community-based HCPS and, in some cases, felt strongly that they were an essential part of veterans’ care networks. Emergency managers offered examples for how they supportedcommunity-based HCPs, such as helping those in the VAMC medical foster home program develop and evaluate emergency plans. Many said they had not explicitly considered HHAs before (Appendix).

Emergency managers also described how supporting community-based HCPs could be considered within the scope of the VHA role and mission, specifically the Fourth Mission. “I think that we should be making our best effort to make sure that we’re also providing that same level [of protection] to the people taking care of the veteran [as our VHA staff],” an emergency manager said. “It’s our responsibility to provide the best for the staff that are going into those homes to take care of that patient.”

In many cases, emergency managers had already developed practical tools that could be easily shared outside the VHA, including weather alerts, trainings, emergency plan templates, and lists of community resources and shelters (Figure 2). A number of these examples built on existing communication channels. One emergency manager said that the extension of resources could be an opportunity to decrease the perceived isolation of home health care workers through regular training for agencies that are providing health care aides, so that they know that “some bigger folks are keeping an eye on it.”

FIGURE 2. Suggestions Received for Extended Resources to Contracted VA Organizations
Abbreviations: PPE, personal protective equipment; VA, US Department of Veterans Affairs.

On the agency side, participants noted that some HHAs could benefit more from support than others. While some agencies are well staffed and have good protocols and keep up to date, “There are smaller agencies, agencies that are starting up that may not have the resources to just disseminate all the information. Those are the agencies [that] could well benefit from the VHA,” an HBPC medical director explained. Agency administrators suggested several areas where they would welcome support, including a deeper understanding of available community resources and access to PPE for staff. Regarding informational resources, an administrator said, “Anytime we can get information, it’s good to have it come to you and not always have to go out searching for it.”

Barriers and Facilitators to Partnering With Community Agencies (n = 16)

A primary barrier regarding resource sharing was potential misalignment between each organization’s policies. HHAs followed state and federal public health guidelines, which sometimes differed from VHA policies. Given that agencies care for both VHA and non-VHA clients, questions also arose around how agencies would prioritize information from the VHA, if they were already receiving information from other sources. When asked about information sharing, both VHA staff and agencies agreed staff time to support any additional activities should be weighed against the value of the information gained.

 

Six participants also shared that education around emergency preparedness could be an opportunity to bridge gaps between VAMCs and their surrounding communities. One local Chief of Community Care noted, “Any opportunity to just give information is going to make it a lot better for the veteran patient … to have something that’s a little more robust.”

Two emergency managers noted the need to be sensitive in the way they engaged with partners, respecting and building on the work that agencies were already doing in this area to ensure VHA was seen as a trusted partner and resource rather than trying to impose new policies or rules on community-based HCPs. “I know that like all leadership in various organizations, there’s a little bit of bristling going on when other people try and tell them what to do,” an HBPC medical director said. “However, if it is established that as a sort of greater level like a state level or a federal level, that VHA can be a resource. I think that as long as that’s recognized by their own professional organizations within each state, then I think that that would be a tremendous advantage to many agencies.”

In terms of sharing physical resources, emergency managers raised concerns around potential liability, although they also acknowledged this issue was important enough to think about potential workarounds. As one emergency manager said, “I want to know that my PPE is not compromised in any way shape or form and that I am in charge of that PPE, so to rely upon going to a home and hoping that [the PPE] wasn’t compromised … would kind of make me a little uneasy.” This emergency manager suggested possible solutions, such as creating a sealed PPE package to give directly to an aide.

Discussion

As the prevalence of climate-related disasters increases, the need to ensure the safety and independence of older adults during emergencies grows more urgent. Health systems must think beyond the direct services they provide and consider the community resources upon which their patients rely. While relationships did not formally exist between VHA emergency managers and community home health HCPs in the sample analyzed in this article, there is precedent and interest in supporting contracted home health agencies caring for veterans in the community. Although not historically part of the VA Fourth Mission, creating a pipeline of support for contracted HHAs by leveraging existing relationships and resources can potentially strengthen its mission to protect older veterans in emergencies, help them age safely in place, and provide a model for health systems to collaborate with community-based HCPs around emergency planning and response (Figure 3).23

FIGURE 3. Support Pipeline for Contracted US Department of Veterans Affairs Organizations

Existing research on the value of health care coalitions highlights the need for established and growing partnerships with a focus on ensuring they are value-added, which echoes concerns we heard in interviews.24 Investment in community partnerships not only includes sharing supplies but also relying on bidirectional support that can be a trusted form of timely information.1,25 The findings in this study exhibit strong communication practices within the VHA during periods of nonemergency and underscore the untapped value of the pre-existing relationship between VAMCs and their contracted HHAs as an area of potential growth for health care coalitions.

Sharing resources in a way that does not put new demands on partners contributes to the sustainability and value-added nature of coalitions. Examples include establishing new low-investment practices (ie, information sharing) that support capacity and compliance with existing requirements rather than create new responsibilities for either member of the coalition. The relationship between the VHA emergency managers and the VHA HBPC program can act as a guide. The emergency managers interviewed for this study are currently engaged with HBPC programs and therefore understand the needs of homebound older adults and their caregivers. Extending the information already available to the HBPC teams via existing channels strengthens workforce practices and increased security for the shared patient, even without direct relationships between emergency managers and agencies. It is important to understand the limitations of these practices, including concerns around conflicting federal and state mandates, legal concerns around the liability of sharing physical resources (such as PPE), and awareness that the objective is not for the VHA to increase burdens (eg, increasing compliance requirements) but rather to serve as a resource for a mutual population in a shared community.

Offering training and practical resources to HHA home health care workers can help them meet disaster preparedness requirements. This is particularly important considering the growing home care workforce shortages, a topic mentioned by all HBPC and community care participants interviewed for this study.26,27 Home health care workers report feeling underprepared and isolated while on the job in normal conditions, a sentiment exacerbated by the COVID-19 pandemic.3,10 Supporting these individuals may help them feel more prepared and connected to their work, improving stability and quality of care.

While these issues are priorities within the VHA, there is growing recognition at the state and federal level of the importance of including older adults and their HCPs in disaster preparedness and response.5,28 The US Department of Health and Human Services, for example, includes older adults and organizations that serve them on its National Advisory Committee on Seniors and Disasters. The Senate version of the 2023 reauthorization of the Pandemic and All-Hazards Preparedness and Response Act included specific provisions to support community-dwelling older adults and people with disabilities, incorporating funding for community organizations to support continuity of services and avoid institutionalization in an emergency.29 Other proposed legislation includes the Real Emergency Access for Aging and Disability Inclusion for Disasters Act, which would ensure the needs of older adults and people with disabilities are explicitly included in all phases of emergency planning and response.30

The VHA expansion of the its VEText program to include disaster response is an effort to more efficiently extend outreach to older and vulnerable patients who are veterans.31 Given these growing efforts, the VHA and other health systems have an opportunity to expand internal emergency preparedness efforts to ensure the health and safety of individuals living in the community.

Limitations

VISN 2 has been a target of terrorism and other disasters. In addition to the sites being initially recruited for their strong emergency management protocols, this context may have biased respondents who are favorable to extending their resources into the community. At the time of recruitment, contracted HHAs were still experiencing staff shortages due to the COVID-19 pandemic, which limited the ability of agency staff to participate in interviews. Additionally, while the comprehensive exploration of VISN 2 facilities allows for confidence of the organizational structures described, the qualitative research design and small study sample, the study findings cannot be immediately generalized to all VISNs.

Conclusions

Many older veterans increasingly rely on home health care workers to age safely. The VHA, as a large national health care system and leader in emergency preparedness, could play an important role in supporting home health care workers and ameliorating their sense of isolation during emergencies and natural disasters. Leveraging existing resources and relationships may be a low-cost, low-effort opportunity to build higher-level interventions that support the needs of patients. Future research and work in this field, including the authors’ ongoing work, will expand agency participation and engage agency staff in conceptualizing pilot projects to ensure they are viable and feasible for the field.

References
  1. Barnett DJ, Knieser L, Errett NA, Rosenblum AJ, Seshamani M, Kirsch TD. Reexamining health-care coalitions in light of COVID-19. Disaster Med public Health Prep. 2022;16(3):859-863. doi:10.1017/dmp.2020.431
  2. Wulff K, Donato D, Lurie N. What is health resilience and how can we build it? Annu Rev Public Health. 2015;36:361-374. doi:10.1146/annurev-publhealth-031914-122829
  3. Franzosa E, Wyte-Lake T, Tsui EK, Reckrey JM, Sterling MR. Essential but excluded: building disaster preparedness capacity for home health care workers and home care agencies. J Am Med Dir Assoc. 2022;23(12):1990-1996. doi:10.1016/j.jamda.2022.09.012
  4. Miner S, Masci L, Chimenti C, Rin N, Mann A, Noonan B. An outreach phone call project: using home health to reach isolated community dwelling adults during the COVID 19 lockdown. J Community Health. 2022;47(2):266-272. doi:10.1007/s10900-021-01044-6
  5. National Institute on Aging. Protecting older adults from the effects of natural disasters and extreme weather. October 18, 2022. Accessed August 19, 2024. https://www.nia.nih.gov/news/protecting-older-adults-effects-natural-disasters-and-extreme-weather
  6. PHI. Direct Care Workers in the United States: Key Facts. September 7, 2021. Accessed August 19, 2024. https://www.phinational.org/resource/direct-care-workers-in-the-united-states-key-facts-2/
  7. Centers for Medicare & Medicaid Services. Emergency Preparedness Rule. September 8, 2016. Updated September 6, 2023. Accessed August 19, 2024. https://www.cms.gov/medicare/health-safety-standards/quality-safety-oversight-emergency-preparedness/emergency-preparedness-rule
  8. Wyte-Lake T, Claver M, Tubbesing S, Davis D, Dobalian A. Development of a home health patient assessment tool for disaster planning. Gerontology. 2019;65(4):353-361. doi:10.1159/000494971
  9. Franzosa E, Judon KM, Gottesman EM, et al. Home health aides’ increased role in supporting older veterans and primary healthcare teams during COVID-19: a qualitative analysis. J Gen Intern Med. 2022;37(8):1830-1837. doi:10.1007/s11606-021-07271-w
  10. Franzosa E, Tsui EK, Baron S. “Who’s caring for us?”: understanding and addressing the effects of emotional labor on home health aides’ well-being. Gerontologist. 2019;59(6):1055-1064. doi:10.1093/geront/gny099
  11. Osakwe ZT, Osborne JC, Samuel T, et al. All alone: a qualitative study of home health aides’ experiences during the COVID-19 pandemic in New York. Am J Infect Control. 2021;49(11):1362-1368. doi:10.1016/j.ajic.2021.08.004
  12. Feldman PH, Russell D, Onorato N, et al. Ensuring the safety of the home health aide workforce and the continuation of essential patient care through sustainable pandemic preparedness. July 2022. Accessed August 19, 2024. https://www.vnshealth.org/wp-content/uploads/2022/08/Pandemic_Preparedness_IB_07_21_22.pdf
  13. Sterling MR, Tseng E, Poon A, et al. Experiences of home health care workers in New York City during the coronavirus disease 2019 pandemic: a qualitative analysis. JAMA Internal Med. 2020;180(11):1453-1459. doi:10.1001/jamainternmed.2020.3930
  14. Wyte-Lake T, Schmitz S, Kornegay RJ, Acevedo F, Dobalian A. Three case studies of community behavioral health support from the US Department of Veterans Affairs after disasters. BMC Public Health. 2021;21(1):639. doi:10.1186/s12889-021-10650-x
  15. Beales JL, Edes T. Veteran’s affairs home based primary care. Clin Geriatr Med. 2009;25(1):149-ix. doi:10.1016/j.cger.2008.11.002
  16. Wyte-Lake T, Manheim C, Gillespie SM, Dobalian A, Haverhals LM. COVID-19 vaccination in VA home based primary care: experience of interdisciplinary team members. J Am Med Dir Assoc. 2022;23(6):917-922. doi:10.1016/j.jamda.2022.03.014
  17. Wyte-Lake T, Schmitz S, Cosme Torres-Sabater R, Dobalian A. Case study of VA Caribbean Healthcare System’s community response to Hurricane Maria. J Emerg Manag. 2022;19(8):189-199. doi:10.5055/jem.0536
  18. US Department of Veterans Affairs. New York/New Jersey VA Health Care Network, VISN 2 Locations. Updated January 3, 2024. Accessed August 19, 2024. https://www.visn2.va.gov/visn2/facilities.asp
  19. Noy C. Sampling knowledge: the hermeneutics of snowball sampling in qualitative research. Int J Soc Res Methodol. 2008;11(4):327-344. doi:10.1080/13645570701401305
  20. Ritchie J, Lewis J, Nicholls CM, Ormston R, eds. Qualitative Research Practice: A Guide for Social Science Students and Researchers. 2nd ed. Sage; 2013.
  21. Morrow SL. Quality and trustworthiness in qualitative research in counseling psychology. J Couns Psychol. 2005;52(2):250-260. doi:10.1037/0022-0167.52.2.250
  22. Rolfe G. Validity, trustworthiness and rigour: quality and the idea of qualitative research. J Adv Nurs. 2006;53(3):304-310. doi:10.1111/j.1365-2648.2006.03727.x
  23. Schmitz S, Wyte-Lake T, Dobalian A. Facilitators and barriers to preparedness partnerships: a veterans affairs medical center perspective. Disaster Med Public Health Prep. 2018;12(4):431-436. doi:10.1017/dmp.2017.92
  24. Koch AE, Bohn J, Corvin JA, Seaberg J. Maturing into high-functioning health-care coalitions: a qualitative Nationwide study of emergency preparedness and response leadership. Disaster Med Public Health Prep. 2022;17:e111. doi:10.1017/dmp.2022.13
  25. Lin JS, Webber EM, Bean SI, Martin AM, Davies MC. Rapid evidence review: policy actions for the integration of public health and health care in the United States. Front Public Health. 2023;11:1098431. doi:10.3389/fpubh.2023.1098431
  26. Watts MOM, Burns A, Ammula M. Ongoing impacts of the pandemic on medicaid home & community-based services (HCBS) programs: findings from a 50-state survey. November 28, 2022. Accessed August 19, 2024. https://www.kff.org/medicaid/issue-brief/ongoing-impacts-of-the-pandemic-on-medicaid-home-community-based-services-hcbs-programs-findings-from-a-50-state-survey/
  27. Kreider AR, Werner RM. The home care workforce has not kept pace with growth in home and community-based services. Health Aff (Millwood). 2023;42(5):650-657. doi:10.1377/hlthaff.2022.01351
  28. FEMA introduces disaster preparedness guide for older adults. News release. FEMA. September 20, 2023. Accessed August 19, 2024. https://www.fema.gov/press-release/20230920/fema-introduces-disaster-preparedness-guide-older-adults
  29. Pandemic and All-Hazards Preparedness and Response Act, S 2333, 118th Cong, 1st Sess (2023). https://www.congress.gov/bill/118th-congress/senate-bill/2333/text
  30. REAADI for Disasters Act, HR 2371, 118th Cong, 1st Sess (2023). https://www.congress.gov/bill/118th-congress/house-bill/2371
  31. Wyte-Lake T, Brewster P, Hubert T, Gin J, Davis D, Dobalian A. VA’s experience building capability to conduct outreach to vulnerable patients during emergencies. Innov Aging. 2023;7(suppl 1):209. doi:10.1093/geroni/igad104.0690
References
  1. Barnett DJ, Knieser L, Errett NA, Rosenblum AJ, Seshamani M, Kirsch TD. Reexamining health-care coalitions in light of COVID-19. Disaster Med public Health Prep. 2022;16(3):859-863. doi:10.1017/dmp.2020.431
  2. Wulff K, Donato D, Lurie N. What is health resilience and how can we build it? Annu Rev Public Health. 2015;36:361-374. doi:10.1146/annurev-publhealth-031914-122829
  3. Franzosa E, Wyte-Lake T, Tsui EK, Reckrey JM, Sterling MR. Essential but excluded: building disaster preparedness capacity for home health care workers and home care agencies. J Am Med Dir Assoc. 2022;23(12):1990-1996. doi:10.1016/j.jamda.2022.09.012
  4. Miner S, Masci L, Chimenti C, Rin N, Mann A, Noonan B. An outreach phone call project: using home health to reach isolated community dwelling adults during the COVID 19 lockdown. J Community Health. 2022;47(2):266-272. doi:10.1007/s10900-021-01044-6
  5. National Institute on Aging. Protecting older adults from the effects of natural disasters and extreme weather. October 18, 2022. Accessed August 19, 2024. https://www.nia.nih.gov/news/protecting-older-adults-effects-natural-disasters-and-extreme-weather
  6. PHI. Direct Care Workers in the United States: Key Facts. September 7, 2021. Accessed August 19, 2024. https://www.phinational.org/resource/direct-care-workers-in-the-united-states-key-facts-2/
  7. Centers for Medicare & Medicaid Services. Emergency Preparedness Rule. September 8, 2016. Updated September 6, 2023. Accessed August 19, 2024. https://www.cms.gov/medicare/health-safety-standards/quality-safety-oversight-emergency-preparedness/emergency-preparedness-rule
  8. Wyte-Lake T, Claver M, Tubbesing S, Davis D, Dobalian A. Development of a home health patient assessment tool for disaster planning. Gerontology. 2019;65(4):353-361. doi:10.1159/000494971
  9. Franzosa E, Judon KM, Gottesman EM, et al. Home health aides’ increased role in supporting older veterans and primary healthcare teams during COVID-19: a qualitative analysis. J Gen Intern Med. 2022;37(8):1830-1837. doi:10.1007/s11606-021-07271-w
  10. Franzosa E, Tsui EK, Baron S. “Who’s caring for us?”: understanding and addressing the effects of emotional labor on home health aides’ well-being. Gerontologist. 2019;59(6):1055-1064. doi:10.1093/geront/gny099
  11. Osakwe ZT, Osborne JC, Samuel T, et al. All alone: a qualitative study of home health aides’ experiences during the COVID-19 pandemic in New York. Am J Infect Control. 2021;49(11):1362-1368. doi:10.1016/j.ajic.2021.08.004
  12. Feldman PH, Russell D, Onorato N, et al. Ensuring the safety of the home health aide workforce and the continuation of essential patient care through sustainable pandemic preparedness. July 2022. Accessed August 19, 2024. https://www.vnshealth.org/wp-content/uploads/2022/08/Pandemic_Preparedness_IB_07_21_22.pdf
  13. Sterling MR, Tseng E, Poon A, et al. Experiences of home health care workers in New York City during the coronavirus disease 2019 pandemic: a qualitative analysis. JAMA Internal Med. 2020;180(11):1453-1459. doi:10.1001/jamainternmed.2020.3930
  14. Wyte-Lake T, Schmitz S, Kornegay RJ, Acevedo F, Dobalian A. Three case studies of community behavioral health support from the US Department of Veterans Affairs after disasters. BMC Public Health. 2021;21(1):639. doi:10.1186/s12889-021-10650-x
  15. Beales JL, Edes T. Veteran’s affairs home based primary care. Clin Geriatr Med. 2009;25(1):149-ix. doi:10.1016/j.cger.2008.11.002
  16. Wyte-Lake T, Manheim C, Gillespie SM, Dobalian A, Haverhals LM. COVID-19 vaccination in VA home based primary care: experience of interdisciplinary team members. J Am Med Dir Assoc. 2022;23(6):917-922. doi:10.1016/j.jamda.2022.03.014
  17. Wyte-Lake T, Schmitz S, Cosme Torres-Sabater R, Dobalian A. Case study of VA Caribbean Healthcare System’s community response to Hurricane Maria. J Emerg Manag. 2022;19(8):189-199. doi:10.5055/jem.0536
  18. US Department of Veterans Affairs. New York/New Jersey VA Health Care Network, VISN 2 Locations. Updated January 3, 2024. Accessed August 19, 2024. https://www.visn2.va.gov/visn2/facilities.asp
  19. Noy C. Sampling knowledge: the hermeneutics of snowball sampling in qualitative research. Int J Soc Res Methodol. 2008;11(4):327-344. doi:10.1080/13645570701401305
  20. Ritchie J, Lewis J, Nicholls CM, Ormston R, eds. Qualitative Research Practice: A Guide for Social Science Students and Researchers. 2nd ed. Sage; 2013.
  21. Morrow SL. Quality and trustworthiness in qualitative research in counseling psychology. J Couns Psychol. 2005;52(2):250-260. doi:10.1037/0022-0167.52.2.250
  22. Rolfe G. Validity, trustworthiness and rigour: quality and the idea of qualitative research. J Adv Nurs. 2006;53(3):304-310. doi:10.1111/j.1365-2648.2006.03727.x
  23. Schmitz S, Wyte-Lake T, Dobalian A. Facilitators and barriers to preparedness partnerships: a veterans affairs medical center perspective. Disaster Med Public Health Prep. 2018;12(4):431-436. doi:10.1017/dmp.2017.92
  24. Koch AE, Bohn J, Corvin JA, Seaberg J. Maturing into high-functioning health-care coalitions: a qualitative Nationwide study of emergency preparedness and response leadership. Disaster Med Public Health Prep. 2022;17:e111. doi:10.1017/dmp.2022.13
  25. Lin JS, Webber EM, Bean SI, Martin AM, Davies MC. Rapid evidence review: policy actions for the integration of public health and health care in the United States. Front Public Health. 2023;11:1098431. doi:10.3389/fpubh.2023.1098431
  26. Watts MOM, Burns A, Ammula M. Ongoing impacts of the pandemic on medicaid home & community-based services (HCBS) programs: findings from a 50-state survey. November 28, 2022. Accessed August 19, 2024. https://www.kff.org/medicaid/issue-brief/ongoing-impacts-of-the-pandemic-on-medicaid-home-community-based-services-hcbs-programs-findings-from-a-50-state-survey/
  27. Kreider AR, Werner RM. The home care workforce has not kept pace with growth in home and community-based services. Health Aff (Millwood). 2023;42(5):650-657. doi:10.1377/hlthaff.2022.01351
  28. FEMA introduces disaster preparedness guide for older adults. News release. FEMA. September 20, 2023. Accessed August 19, 2024. https://www.fema.gov/press-release/20230920/fema-introduces-disaster-preparedness-guide-older-adults
  29. Pandemic and All-Hazards Preparedness and Response Act, S 2333, 118th Cong, 1st Sess (2023). https://www.congress.gov/bill/118th-congress/senate-bill/2333/text
  30. REAADI for Disasters Act, HR 2371, 118th Cong, 1st Sess (2023). https://www.congress.gov/bill/118th-congress/house-bill/2371
  31. Wyte-Lake T, Brewster P, Hubert T, Gin J, Davis D, Dobalian A. VA’s experience building capability to conduct outreach to vulnerable patients during emergencies. Innov Aging. 2023;7(suppl 1):209. doi:10.1093/geroni/igad104.0690
Issue
Federal Practitioner - 41(10)a
Issue
Federal Practitioner - 41(10)a
Page Number
1-9
Page Number
1-9
Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

The Impact of a Metformin Recall on Patient Hemoglobin A1c Levels at a VA Network

Article Type
Changed
Mon, 11/04/2024 - 14:08
Display Headline
The Impact of a Metformin Recall on Patient Hemoglobin A1c Levels at a VA Network

About 1 in 10 Americans have diabetes mellitus (DM), of which about 90% to 95% are diagnosed with type 2 DM (T2DM) and veterans are disproportionately affected.1,2 About 25% enrolled in the Veterans Health Administration (VHA) have T2DM, which has been attributed to exposure to herbicides (eg, Agent Orange), decreased physical activity resulting from past physical strain, chronic pain, and other physical limitations resulting from military service.3-5

Pharmacologic management of DM is guided by the effectiveness of lifestyle interventions and comorbid diagnoses. Current DM management guidelines recommend patients with comorbid atherosclerotic cardiovascular disease, chronic kidney disease, or congestive heart failure receive first-line diabetes therapy with a sodium-glucose cotransporter-2 (SGLT-2) inhibitor or glucagon-like peptide-1 receptor (GLP-1) agonist.

Metformin remains a first-line pharmacologic option for the treatment of T2DM with the goal of achieving glycemic management when lifestyle interventions are insufficient.6,7 Newer antihyperglycemic therapies have been studied as adjunct therapy to metformin. However, there is limited literature comparing metformin directly to other medication classes for the treatment of T2DM.8-13 A systematic review of treatment-naive patients found HbA1c reductions were similar whether patients received metformin vs an SGLT-2 inhibitor, GLP-1 agonist, sulfonylurea, or thiazolidinedione monotherapy.10 The analysis found dipeptidyl-peptidase-4 (DPP-4) inhibitors had inferior HbA1c reduction compared to metformin.10 A Japanese systematic review compared metformin to thiazolidinediones, sulfonylureas, glinides, DPP-4 inhibitors, α-glucosidase inhibitors, or SGLT-2 inhibitors for ≥ 12 weeks but found no statistically significant differences in HbA1c reduction.11 The AWARD-3 trial compared once-weekly dulaglutide to metformin in treatment-experienced patients and found greater improvement in HbA1c and achievement of HbA1c goal with dulaglutide.13 While these studies show some comparisons of metformin to alternative pharmacologic therapy, researchers have not looked at what happens to patients’ HbA1c levels when an event, such as a recall, prompts a rapid change to a different antihyperglycemic agent.

On May 28, 2020, the US Food and Drug Administration (FDA) asked 5 pharmaceutical companies to voluntarily recall certain formulations of metformin. This action was taken when FDA testing revealed unacceptably high levels of N-Nitrosodimethylamine, a probable carcinogen.14 This FDA recall of metformin extended-release, referred to as metformin sustained-action (SA) within the VHA electronic medication file but the same type of formulation, prompted clinicians to revisit and revise the pharmacologic regimens of patients taking the drug. Because of the paucity of head-to-head trials comparing metformin with newer alternative antihyperglycemic therapies, the effect of treatment change was unknown. In response, we aimed to establish a data registry within Veterans Integrated Service Network (VISN) 6.

Registry Development

The VISN 6 registry was established to gather long-term, observational, head-to-head data that would allow review of HbA1c levels before and after the recall, as well as HbA1c levels broken down by the agent that patients were switched to after the recall. Another goal was to explore prescribing trends following the recall.

Data Access Request Tracker approval was obtained and a US Department of Veterans Affairs (VA) Information and Computing Infrastructure workspace was developed to host the registry data. The research cohort was established from this data, and the registry framework was finalized using Structured Query Language (SQL). The SQL coding allows for recurring data updates for all individuals within the cohort including date of birth, race, sex, ethnicity, VHA facility visited, weight, body mass index, HbA1c level, creatinine clearance, serum creatinine, antihyperglycemic medication prescriptions, adverse drug reactions, medication adherence (as defined by ≥ 80% refill history), and hospitalizations related to diabetes. For the purposes of this initial analysis, registry data included demographics, diabetes medications, and HbA1c results.

METHODS

This study was a concurrent, observational, multicenter, registry-based study conducted at the Western North Carolina VA Health Care System (WNCVAHCS). The study was approved by the WNCVAHCS institutional review board and research and development committees.

All patients aged ≥ 18 years with T2DM and receiving health care from VISN 6 facilities who had an active metformin SA prescription on, and 1 year prior to, June 1, 2020 (the initial date VHA began implementing the FDA metformin recall) were entered into the registry. Data from 1 year prior were collected to provide a baseline. Veterans were excluded if they received metformin SA for any indication other than T2DM, there was no pre- or postrecall HbA1c measurement, or death. We included 15,594 VISN 6 veterans.

Registry data were analyzed to determine whether a significant change in HbA1c level occurred after the metformin recall and in response to alternative agents being prescribed. Data from veterans who met all inclusion criteria were assessed during the year before and after June 1, 2020. Demographic data were analyzed using frequency and descriptive statistics. The Shapiro Wilkes test was performed, and data were found to be nonparametric; therefore the Wilcoxon signed-rank test was used to evaluate the hypothesis that HbA1c levels were not impacted by the recall.

Our sample size allowed us to create exact matched pairs of 9130 individuals and utilize rank-biserial correlation to establish effect size. Following this initial population-level test, we constructed 2 models. The first, a linear mixed-effects model, focused solely on the interaction effects between the pre- and postrecall periods and various medication classes on HbA1c levels. Second, we constructed a random-effects within-between model (REWB) to evaluate the impact ofmedication classes and demographic variables. Statistical significance was measured at P < .05 with conservative power at .90. The effect size was set to 1.0, reflecting a minimum clinically important difference. Literature establishes 0.5 as a modest level of HbA1c improvement and 1.0 as a clinically significant improvement.

RESULTS

Preliminary results included 15,594 veterans who received a metformin SA prescription as of June 1, 2020 from VISN 6 facilities; 15,392 veterans had a drug exposure end on June 1, 2020, indicating their standard therapy of metformin SA was discontinued following the FDA recall. Two hundred and two veterans were excluded from the registry because they continued to receive metformin SA from existing stock at a VISN6 facility. After identifying veterans with data for 1 year prior (June 1, 2019) to the index date and 1 year after (June 1, 2021) the study population was adjusted to 9130. The population was predominantly males aged> 60 years. Roughly 55% of the registry identified as White and nearly 40% as Black, and 2% indentified as Hispanic (Table 1).

Wilcoxon Signed-Rank Test

We created exact pairs by iterating the data and finding the closest measurements for each patient before and after the recall. This has the advantage over averaging a patient’s pre- and post-HbA1c levels, as it allows for a rank-biserial correlation. Using the nonparametric Wilcoxon signed-rank test, V was 20,100,707 (P < .001), indicating a significant effect. The –0.29 rank-biserial correlation, which was computed to assess the effect size of the recall, suggests that the median HbA1c level was lower postrecall vs prerecall. The magnitude of the correlation suggests a moderate effect size, and while the recall had a noticeable impact at a population level, it was not extreme (Table 2).

Linear Mixed-Effects Model

The binary variable for medication class exposure suggests the use of a logit link function for binary outcomes within the multilevel modeling framework.15 We employed a linear mixed-effects model to investigate the impact that switching from metformin SA to other T2DM medications had on HbA1c levels. The model was adjusted for patient-specific random effects and included interaction terms between the recall period (before and after) and the usage of different T2DM medications.

Model Fit and Random Effects

The model demonstrated a residual maximum likelihood criterion of 100,219.7, indicating its fit to the data. Notably, the random effects analysis revealed a substantial variability in baseline HbA1c levels across patients (SD, 0.94), highlighting the importance of individual differences in DM management. Medication classes with zero or near-zero exposure rate were removed. Due to demographic homogeneity, the model did not converge on demographic variables. Veterans were taking a mean of 1.8 T2DM medications and metformin SA was most common (Table 3).

During the postrecall period, metformin SA remained the most frequently prescribed medication class. This may be attributed to the existence of multiple manufacturers of metformin SA, some of which may not have been impacted by the recall. VISN 6 medical centers could have sought metformin SA outside of the usual procurement path following the recall.

Complex Random Effects Model

We employed a complex REWB model that evaluated the impact of medication classes on HbA1c levels, accounting for both within and between subject effects of these medications, along with demographic variables (sex, race, and ethnicity) (eAppendix). This model accounts for individual-level changes over time (within-patient effects) and between groups of patients (between-patient effects). This is a more comprehensive model aimed at understanding the broader impact of medications on HbA1c levels across diverse patient groups.

Most demographic categories did not demonstrate significant effects in this model. Black individuals experienced a slight increase in HbA1c levels compared with other racial categories that was not statistically significant. However, this model confirms the findings from the linear mixed-effects model that GLP-1 agonists showed a substantial decrease in HbA1c levels within patients (coefficient –0.5; 95% CI, –0.56 to –0.44; P < .001) and a moderate increase between patients (coefficient, 0.21; 95% CI, 0.12-0.31; P < .001). Additionally, SGLT-2 inhibitors had a notable decrease within patients (coefficient, –0.27; 95% CI, –0.32 to –0.22; P < .001).Another notable finding with our REWB model is insulin usage was associated with high HbA1c levels, but only between subjects. Long-acting insulin (coefficient, 0.96; 95% CI, 0.90-1.01; P <. 001) and mixed insulin (coefficient, 1.09; 95% CI, 0.94-1.24; P < .001) both displayed marked increases between patients, suggesting future analysis may benefit from stratifying across insulin users and nonusers.

Fixed Effect Analysis

The fixed effects analysis yielded several notable findings. The intercept, representing the mean baseline HbA1c level, was estimated at 7.8% (58 mmol/mol). The coefficient for the period (postrecall) was not statistically significant, indicating no overall change in HbA1c levels from before to after the recall when specific medication classes were not considered (Table 4). Among medication classes examined, several showed significant associations with HbA1c levels. DPP-4 inhibitors and GLP-1 agonists were associated with a decrease in HbA1c levels, with coefficients of −0.08 and −0.24, respectively. Long-acting insulin and metformin immediate-release (IR) were associated with an increase in HbA1c levels, as indicated by their positive coefficients of 0.38 and 0.16, respectively. Mixed insulin formulations and sulfonylureas showed an association with decreased HbA1c levels.

Interaction Effects

The interaction terms between the recall period and the medication classes provided insights into the differential impact of the medication switch postrecall. Notably, the interaction term for long-acting insulin (coefficient, −0.10) was significant, suggesting a differential effect on HbA1c levels postrecall. Other medications, like metformin IR, also exhibited significant interaction effects, indicating changes in the impact on HbA1c levels in the postrecall period. The binary variable for medication class exposure suggests the use of a logit link function for binary outcomes within the multilevel modeling framework.15 We did not address the potential for cross cluster heterogeneity due to different medication classes.

DISCUSSION

This study is an ongoing, concurrent, observational, multicenter, registry-based study consisting of VISN 6 veterans who have T2DM and were prescribed metformin SA on June 1, 2020. This initial aim was to evaluate change in HbA1c levels following the FDA metformin recall. While there was substantial variability in baseline HbA1c levels across the patients, the mean baseline HbA1c level at 7.5% (58 mmol/mol). Patients taking GLP-1 agonists showed substantial decrease in HbA1c levels (coefficient; –0.5; 95% CI, –0.56 to –0.44; P <. 001). Patients taking SGLT-2 inhibitors had a notable decrease in HbA1c (coefficient, –0.27; 95% CI, –0.32 to –0.22; P < .001). Despite this, the coefficient for the postrecall period was not statistically significant, indicating no overall change in HbA1c levels from pre- to postrecall when specific medication classes were not considered.

Further analysis included assessment of prescribing trends postrecall. There was an increase in SGLT-2 inhibitor, GLP-1 agonist, and DPP-4 inhibitor prescribing. Considering the growing evidence of the cardiovascular and renal benefits of these medication classes, specifically the GLP-1 agonists and SGLT-2 inhibitors, this trend would be expected.

Limitations

This study cohort did not capture veterans with T2DM who transferred their health care to VISN 6 after June 1, 2020, and continued to receive metformin SA from the prior facility. Inclusion of these veterans would have increased the registry population. Additionally, the cohort did not identify veterans who continued to receive metformin SA through a source other than the VA. Without that information, the registry cohort may include veterans thought to have either transitioned to a different therapy or to no other T2DM therapy after the recall.

Given that DM can progress over time, it is possible the transition to a new medication after the recall was the result of suboptimal management, or in response to an adverse effect from a previous medication, and not solely due to the metformin SA recall. In addition, there are several factors that could impact HbA1c level over time that were not accounted for in this study, such as medication adherence and lifestyle modifications.

The notable level of metformin SA prescriptions, despite the recall, may be attributed to several factors. First, not all patients stopped metformin completely. Review of the prescription data indicated that some veterans were provided with limited refills at select VA medical centers that had supplies (medication lots not recalled). Access to a safe supply of metformin SA after the recall may have varied among VISN 6 facilities. It is also possible that as new supplies of metformin SA became available, veterans restarted metformin SA. This may have been resumed while continuing a new medication prescribed at the beginning of the recall. As the year progressed after the recall, an increase in metformin SA prescriptions likely occurred as supplies became available and clinicians/veterans chose to resume this medication therapy.

Conclusions

Results of this initial registry study found no difference in HbA1c levels across the study population after the metformin SA recall. However, there was clinical difference in the HbA1c within veterans prescribed SGLT-2 inhibitors and GLP-1 agonists. As expected, prescribing trends showed an increase in these agents after the recall. With the known benefits of these medications beyond glucose lowering, it is anticipated the cohort of veterans prescribed these medications will continue to grow.

The VISN 6 research registry allowed this study to gain an important snapshot in time following the metformin SA recall, and will serve as an important resource for future DM research endeavors. It will allow for ongoing evaluation of the impact of the transition to alternative T2DM medications after the metformin SA recall. Future exploration will include evaluation of adverse drug reactions, DM-related hospitalizations, emergency department visits related to T2DM, changes in renal function, and cardiovascular events among all diabetes medication classes.

Acknowledgments

The study team thanks the Veterans Affairs Informatics and Computing Infrastructure for their help and expertise throughout this project. The authors acknowledge the contributions of Philip Nelson, PharmD, and Brian Peek, PharmD.

References
  1. Centers for Disease Control and Prevention. Type 2 diabetes. Updated April 18, 2023. Accessed September 18, 2023. https://www.cdc.gov/diabetes/basics/type2.html 
  2. ElSayed NA, Aleppo G, Aroda VR, et al. 2. Classification and diagnosis of diabetes: standards of care in diabetes—2023. Diabetes Care. 2023;46(Supplement_1):S19-S40. doi:10.2337/dc23-S002
  3. Liu Y, Sayam S, Shao X, et al. Prevalence of and trends in diabetes among veterans, United States, 2005–2014. Prev Chronic Dis. 2017;14:E135. doi:10.5888/pcd14.170230
  4. Yi SW, Hong JS, Ohrr H, Yi JJ. Agent Orange exposure and disease prevalence in Korean Vietnam veterans: the Korean veterans health study. Environ Res. 2014;133:56-65. doi:10.1016/j.envres.2014.04.027
  5. Price LE, Gephart S, Shea K. The VA’s Corporate Data Warehouse: Uses and Implications for Nursing Research and Practice. Nurs Adm Q. 2015;39(4):311-318. doi:10.1097/NAQ.0000000000000118
  6. ElSayed NA, Aleppo G, Aroda VR, et al. 9. Pharmacologic approaches to glycemic treatment: standards of care in diabetes-2023. Diabetes Care. 2023;46(suppl 1):S140-S157. doi:10.2337/dc23-S009
  7. Samson SL, Vellanki P, Blonde L, et al. American Association of Clinical Endocrinology Consensus Statement: Comprehensive Type 2 Diabetes Management Algorithm - 2023 Update. Endocr Pract. 2023;29(5):305-340. doi:10.1016/j.eprac.2023.02.001
  8. Bennett WL, Maruthur NM, Singh S, et al. Comparative effectiveness and safety of medications for type 2 diabetes: an update including new drugs and 2-drug combinations. Ann Intern Med. 2011;154(9):602-613. doi:10.7326/0003-4819-154-9-201105030-00336
  9. Bolen S, Feldman L, Vassy J, et al. Systematic review: comparative effectiveness and safety of oral medications for type 2 diabetes mellitus. Ann Intern Med. 2007;147(6):386-399. doi:10.7326/0003-4819-147-6-200709180-00178
  10. Tsapas A, Avgerinos I, Karagiannis T, et al. Comparative effectiveness of glucose-lowering drugs for type 2 diabetes: a systematic review and network meta-analysis. Ann Intern Med. 2020;173(4):278-286. doi:10.7326/M20-0864
  11. Nishimura R, Taniguchi M, Takeshima T, Iwasaki K. Efficacy and safety of metformin versus the other oral antidiabetic drugs in Japanese type 2 diabetes patients: a network meta-analysis. Adv Ther. 2022;39(1):632-654. doi:10.1007/s12325-021-01979-1
  12. Russell-Jones D, Cuddihy RM, Hanefeld M, et al. Efficacy and safety of exenatide once weekly versus metformin, pioglitazone, and sitagliptin used as monotherapy in drug-naive patients with type 2 diabetes (DURATION-4): a 26-week double-blind study. Diabetes Care. 2012;35(2):252-258. doi:10.2337/dc11-1107
  13. Umpierrez G, Tofé Povedano S, Pérez Manghi F, Shurzinske L, Pechtner V. Efficacy and safety of dulaglutide monotherapy versus metformin in type 2 diabetes in a randomized controlled trial (AWARD-3). Diabetes Care. 2014;37(8):2168-2176. doi:10.2337/dc13-2759
  14. US Food and Drug Administration. FDA alerts patients and health care professionals to nitrosamine impurity findings in certain metformin extended-release products [press release]. May 28, 2020. Accessed October 16, 2024. https://www.fda.gov/news-events/press-announcements/fda-alerts-patients-and-health-care-professionals-nitrosamine-impurity-findings-certain-metformin
  15. Bell A, Jones K. Explaining fixed effects: random effects modeling of time-series cross-sectional and panel data. PSRM. 2015;3(1):133-153. doi:10.1017/psrm.2014.7
Article PDF
Author and Disclosure Information

Beth D. Greck, PharmD, BCACP, CDCESa; Aimee Pehrson, MHA, MPHb; Hayden Spence, MSb

Author affiliations

aWestern North Carolina Veterans Affairs Health Care System, Asheville

bAptive Resources, Alexandria, Virginia

Author disclosures

The authors report no actual or potential conflicts of interest or outside sources of funding with regard to this article.

Disclaimer

The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the US Government, or any of its agencies. This article may discuss unlabeled or investigational use of certain drugs. Please review the complete prescribing information for specific drugs or drug combinations—including indications, contraindications, warnings, and adverse effects— before administering pharmacologic therapy to patients.

Ethics and consent

All authors adhered to ethical principles for medical research involving human subjects as outlined in the World Medical Association’s Declaration of Helsinki. All relevant guidelines and federal regulations were followed for conducting research at the Western North Carolina Veterans Affairs Health Care System (WNCVAHCS)/Charles George VA Medical Center. This research study was submitted and approved by the WNCVAHCS Institutional Review Board and Research and Development committees.

Issue
Federal Practitioner - 41(11)s
Publications
Topics
Page Number
S6-S11
Sections
Author and Disclosure Information

Beth D. Greck, PharmD, BCACP, CDCESa; Aimee Pehrson, MHA, MPHb; Hayden Spence, MSb

Author affiliations

aWestern North Carolina Veterans Affairs Health Care System, Asheville

bAptive Resources, Alexandria, Virginia

Author disclosures

The authors report no actual or potential conflicts of interest or outside sources of funding with regard to this article.

Disclaimer

The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the US Government, or any of its agencies. This article may discuss unlabeled or investigational use of certain drugs. Please review the complete prescribing information for specific drugs or drug combinations—including indications, contraindications, warnings, and adverse effects— before administering pharmacologic therapy to patients.

Ethics and consent

All authors adhered to ethical principles for medical research involving human subjects as outlined in the World Medical Association’s Declaration of Helsinki. All relevant guidelines and federal regulations were followed for conducting research at the Western North Carolina Veterans Affairs Health Care System (WNCVAHCS)/Charles George VA Medical Center. This research study was submitted and approved by the WNCVAHCS Institutional Review Board and Research and Development committees.

Author and Disclosure Information

Beth D. Greck, PharmD, BCACP, CDCESa; Aimee Pehrson, MHA, MPHb; Hayden Spence, MSb

Author affiliations

aWestern North Carolina Veterans Affairs Health Care System, Asheville

bAptive Resources, Alexandria, Virginia

Author disclosures

The authors report no actual or potential conflicts of interest or outside sources of funding with regard to this article.

Disclaimer

The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the US Government, or any of its agencies. This article may discuss unlabeled or investigational use of certain drugs. Please review the complete prescribing information for specific drugs or drug combinations—including indications, contraindications, warnings, and adverse effects— before administering pharmacologic therapy to patients.

Ethics and consent

All authors adhered to ethical principles for medical research involving human subjects as outlined in the World Medical Association’s Declaration of Helsinki. All relevant guidelines and federal regulations were followed for conducting research at the Western North Carolina Veterans Affairs Health Care System (WNCVAHCS)/Charles George VA Medical Center. This research study was submitted and approved by the WNCVAHCS Institutional Review Board and Research and Development committees.

Article PDF
Article PDF

About 1 in 10 Americans have diabetes mellitus (DM), of which about 90% to 95% are diagnosed with type 2 DM (T2DM) and veterans are disproportionately affected.1,2 About 25% enrolled in the Veterans Health Administration (VHA) have T2DM, which has been attributed to exposure to herbicides (eg, Agent Orange), decreased physical activity resulting from past physical strain, chronic pain, and other physical limitations resulting from military service.3-5

Pharmacologic management of DM is guided by the effectiveness of lifestyle interventions and comorbid diagnoses. Current DM management guidelines recommend patients with comorbid atherosclerotic cardiovascular disease, chronic kidney disease, or congestive heart failure receive first-line diabetes therapy with a sodium-glucose cotransporter-2 (SGLT-2) inhibitor or glucagon-like peptide-1 receptor (GLP-1) agonist.

Metformin remains a first-line pharmacologic option for the treatment of T2DM with the goal of achieving glycemic management when lifestyle interventions are insufficient.6,7 Newer antihyperglycemic therapies have been studied as adjunct therapy to metformin. However, there is limited literature comparing metformin directly to other medication classes for the treatment of T2DM.8-13 A systematic review of treatment-naive patients found HbA1c reductions were similar whether patients received metformin vs an SGLT-2 inhibitor, GLP-1 agonist, sulfonylurea, or thiazolidinedione monotherapy.10 The analysis found dipeptidyl-peptidase-4 (DPP-4) inhibitors had inferior HbA1c reduction compared to metformin.10 A Japanese systematic review compared metformin to thiazolidinediones, sulfonylureas, glinides, DPP-4 inhibitors, α-glucosidase inhibitors, or SGLT-2 inhibitors for ≥ 12 weeks but found no statistically significant differences in HbA1c reduction.11 The AWARD-3 trial compared once-weekly dulaglutide to metformin in treatment-experienced patients and found greater improvement in HbA1c and achievement of HbA1c goal with dulaglutide.13 While these studies show some comparisons of metformin to alternative pharmacologic therapy, researchers have not looked at what happens to patients’ HbA1c levels when an event, such as a recall, prompts a rapid change to a different antihyperglycemic agent.

On May 28, 2020, the US Food and Drug Administration (FDA) asked 5 pharmaceutical companies to voluntarily recall certain formulations of metformin. This action was taken when FDA testing revealed unacceptably high levels of N-Nitrosodimethylamine, a probable carcinogen.14 This FDA recall of metformin extended-release, referred to as metformin sustained-action (SA) within the VHA electronic medication file but the same type of formulation, prompted clinicians to revisit and revise the pharmacologic regimens of patients taking the drug. Because of the paucity of head-to-head trials comparing metformin with newer alternative antihyperglycemic therapies, the effect of treatment change was unknown. In response, we aimed to establish a data registry within Veterans Integrated Service Network (VISN) 6.

Registry Development

The VISN 6 registry was established to gather long-term, observational, head-to-head data that would allow review of HbA1c levels before and after the recall, as well as HbA1c levels broken down by the agent that patients were switched to after the recall. Another goal was to explore prescribing trends following the recall.

Data Access Request Tracker approval was obtained and a US Department of Veterans Affairs (VA) Information and Computing Infrastructure workspace was developed to host the registry data. The research cohort was established from this data, and the registry framework was finalized using Structured Query Language (SQL). The SQL coding allows for recurring data updates for all individuals within the cohort including date of birth, race, sex, ethnicity, VHA facility visited, weight, body mass index, HbA1c level, creatinine clearance, serum creatinine, antihyperglycemic medication prescriptions, adverse drug reactions, medication adherence (as defined by ≥ 80% refill history), and hospitalizations related to diabetes. For the purposes of this initial analysis, registry data included demographics, diabetes medications, and HbA1c results.

METHODS

This study was a concurrent, observational, multicenter, registry-based study conducted at the Western North Carolina VA Health Care System (WNCVAHCS). The study was approved by the WNCVAHCS institutional review board and research and development committees.

All patients aged ≥ 18 years with T2DM and receiving health care from VISN 6 facilities who had an active metformin SA prescription on, and 1 year prior to, June 1, 2020 (the initial date VHA began implementing the FDA metformin recall) were entered into the registry. Data from 1 year prior were collected to provide a baseline. Veterans were excluded if they received metformin SA for any indication other than T2DM, there was no pre- or postrecall HbA1c measurement, or death. We included 15,594 VISN 6 veterans.

Registry data were analyzed to determine whether a significant change in HbA1c level occurred after the metformin recall and in response to alternative agents being prescribed. Data from veterans who met all inclusion criteria were assessed during the year before and after June 1, 2020. Demographic data were analyzed using frequency and descriptive statistics. The Shapiro Wilkes test was performed, and data were found to be nonparametric; therefore the Wilcoxon signed-rank test was used to evaluate the hypothesis that HbA1c levels were not impacted by the recall.

Our sample size allowed us to create exact matched pairs of 9130 individuals and utilize rank-biserial correlation to establish effect size. Following this initial population-level test, we constructed 2 models. The first, a linear mixed-effects model, focused solely on the interaction effects between the pre- and postrecall periods and various medication classes on HbA1c levels. Second, we constructed a random-effects within-between model (REWB) to evaluate the impact ofmedication classes and demographic variables. Statistical significance was measured at P < .05 with conservative power at .90. The effect size was set to 1.0, reflecting a minimum clinically important difference. Literature establishes 0.5 as a modest level of HbA1c improvement and 1.0 as a clinically significant improvement.

RESULTS

Preliminary results included 15,594 veterans who received a metformin SA prescription as of June 1, 2020 from VISN 6 facilities; 15,392 veterans had a drug exposure end on June 1, 2020, indicating their standard therapy of metformin SA was discontinued following the FDA recall. Two hundred and two veterans were excluded from the registry because they continued to receive metformin SA from existing stock at a VISN6 facility. After identifying veterans with data for 1 year prior (June 1, 2019) to the index date and 1 year after (June 1, 2021) the study population was adjusted to 9130. The population was predominantly males aged> 60 years. Roughly 55% of the registry identified as White and nearly 40% as Black, and 2% indentified as Hispanic (Table 1).

Wilcoxon Signed-Rank Test

We created exact pairs by iterating the data and finding the closest measurements for each patient before and after the recall. This has the advantage over averaging a patient’s pre- and post-HbA1c levels, as it allows for a rank-biserial correlation. Using the nonparametric Wilcoxon signed-rank test, V was 20,100,707 (P < .001), indicating a significant effect. The –0.29 rank-biserial correlation, which was computed to assess the effect size of the recall, suggests that the median HbA1c level was lower postrecall vs prerecall. The magnitude of the correlation suggests a moderate effect size, and while the recall had a noticeable impact at a population level, it was not extreme (Table 2).

Linear Mixed-Effects Model

The binary variable for medication class exposure suggests the use of a logit link function for binary outcomes within the multilevel modeling framework.15 We employed a linear mixed-effects model to investigate the impact that switching from metformin SA to other T2DM medications had on HbA1c levels. The model was adjusted for patient-specific random effects and included interaction terms between the recall period (before and after) and the usage of different T2DM medications.

Model Fit and Random Effects

The model demonstrated a residual maximum likelihood criterion of 100,219.7, indicating its fit to the data. Notably, the random effects analysis revealed a substantial variability in baseline HbA1c levels across patients (SD, 0.94), highlighting the importance of individual differences in DM management. Medication classes with zero or near-zero exposure rate were removed. Due to demographic homogeneity, the model did not converge on demographic variables. Veterans were taking a mean of 1.8 T2DM medications and metformin SA was most common (Table 3).

During the postrecall period, metformin SA remained the most frequently prescribed medication class. This may be attributed to the existence of multiple manufacturers of metformin SA, some of which may not have been impacted by the recall. VISN 6 medical centers could have sought metformin SA outside of the usual procurement path following the recall.

Complex Random Effects Model

We employed a complex REWB model that evaluated the impact of medication classes on HbA1c levels, accounting for both within and between subject effects of these medications, along with demographic variables (sex, race, and ethnicity) (eAppendix). This model accounts for individual-level changes over time (within-patient effects) and between groups of patients (between-patient effects). This is a more comprehensive model aimed at understanding the broader impact of medications on HbA1c levels across diverse patient groups.

Most demographic categories did not demonstrate significant effects in this model. Black individuals experienced a slight increase in HbA1c levels compared with other racial categories that was not statistically significant. However, this model confirms the findings from the linear mixed-effects model that GLP-1 agonists showed a substantial decrease in HbA1c levels within patients (coefficient –0.5; 95% CI, –0.56 to –0.44; P < .001) and a moderate increase between patients (coefficient, 0.21; 95% CI, 0.12-0.31; P < .001). Additionally, SGLT-2 inhibitors had a notable decrease within patients (coefficient, –0.27; 95% CI, –0.32 to –0.22; P < .001).Another notable finding with our REWB model is insulin usage was associated with high HbA1c levels, but only between subjects. Long-acting insulin (coefficient, 0.96; 95% CI, 0.90-1.01; P <. 001) and mixed insulin (coefficient, 1.09; 95% CI, 0.94-1.24; P < .001) both displayed marked increases between patients, suggesting future analysis may benefit from stratifying across insulin users and nonusers.

Fixed Effect Analysis

The fixed effects analysis yielded several notable findings. The intercept, representing the mean baseline HbA1c level, was estimated at 7.8% (58 mmol/mol). The coefficient for the period (postrecall) was not statistically significant, indicating no overall change in HbA1c levels from before to after the recall when specific medication classes were not considered (Table 4). Among medication classes examined, several showed significant associations with HbA1c levels. DPP-4 inhibitors and GLP-1 agonists were associated with a decrease in HbA1c levels, with coefficients of −0.08 and −0.24, respectively. Long-acting insulin and metformin immediate-release (IR) were associated with an increase in HbA1c levels, as indicated by their positive coefficients of 0.38 and 0.16, respectively. Mixed insulin formulations and sulfonylureas showed an association with decreased HbA1c levels.

Interaction Effects

The interaction terms between the recall period and the medication classes provided insights into the differential impact of the medication switch postrecall. Notably, the interaction term for long-acting insulin (coefficient, −0.10) was significant, suggesting a differential effect on HbA1c levels postrecall. Other medications, like metformin IR, also exhibited significant interaction effects, indicating changes in the impact on HbA1c levels in the postrecall period. The binary variable for medication class exposure suggests the use of a logit link function for binary outcomes within the multilevel modeling framework.15 We did not address the potential for cross cluster heterogeneity due to different medication classes.

DISCUSSION

This study is an ongoing, concurrent, observational, multicenter, registry-based study consisting of VISN 6 veterans who have T2DM and were prescribed metformin SA on June 1, 2020. This initial aim was to evaluate change in HbA1c levels following the FDA metformin recall. While there was substantial variability in baseline HbA1c levels across the patients, the mean baseline HbA1c level at 7.5% (58 mmol/mol). Patients taking GLP-1 agonists showed substantial decrease in HbA1c levels (coefficient; –0.5; 95% CI, –0.56 to –0.44; P <. 001). Patients taking SGLT-2 inhibitors had a notable decrease in HbA1c (coefficient, –0.27; 95% CI, –0.32 to –0.22; P < .001). Despite this, the coefficient for the postrecall period was not statistically significant, indicating no overall change in HbA1c levels from pre- to postrecall when specific medication classes were not considered.

Further analysis included assessment of prescribing trends postrecall. There was an increase in SGLT-2 inhibitor, GLP-1 agonist, and DPP-4 inhibitor prescribing. Considering the growing evidence of the cardiovascular and renal benefits of these medication classes, specifically the GLP-1 agonists and SGLT-2 inhibitors, this trend would be expected.

Limitations

This study cohort did not capture veterans with T2DM who transferred their health care to VISN 6 after June 1, 2020, and continued to receive metformin SA from the prior facility. Inclusion of these veterans would have increased the registry population. Additionally, the cohort did not identify veterans who continued to receive metformin SA through a source other than the VA. Without that information, the registry cohort may include veterans thought to have either transitioned to a different therapy or to no other T2DM therapy after the recall.

Given that DM can progress over time, it is possible the transition to a new medication after the recall was the result of suboptimal management, or in response to an adverse effect from a previous medication, and not solely due to the metformin SA recall. In addition, there are several factors that could impact HbA1c level over time that were not accounted for in this study, such as medication adherence and lifestyle modifications.

The notable level of metformin SA prescriptions, despite the recall, may be attributed to several factors. First, not all patients stopped metformin completely. Review of the prescription data indicated that some veterans were provided with limited refills at select VA medical centers that had supplies (medication lots not recalled). Access to a safe supply of metformin SA after the recall may have varied among VISN 6 facilities. It is also possible that as new supplies of metformin SA became available, veterans restarted metformin SA. This may have been resumed while continuing a new medication prescribed at the beginning of the recall. As the year progressed after the recall, an increase in metformin SA prescriptions likely occurred as supplies became available and clinicians/veterans chose to resume this medication therapy.

Conclusions

Results of this initial registry study found no difference in HbA1c levels across the study population after the metformin SA recall. However, there was clinical difference in the HbA1c within veterans prescribed SGLT-2 inhibitors and GLP-1 agonists. As expected, prescribing trends showed an increase in these agents after the recall. With the known benefits of these medications beyond glucose lowering, it is anticipated the cohort of veterans prescribed these medications will continue to grow.

The VISN 6 research registry allowed this study to gain an important snapshot in time following the metformin SA recall, and will serve as an important resource for future DM research endeavors. It will allow for ongoing evaluation of the impact of the transition to alternative T2DM medications after the metformin SA recall. Future exploration will include evaluation of adverse drug reactions, DM-related hospitalizations, emergency department visits related to T2DM, changes in renal function, and cardiovascular events among all diabetes medication classes.

Acknowledgments

The study team thanks the Veterans Affairs Informatics and Computing Infrastructure for their help and expertise throughout this project. The authors acknowledge the contributions of Philip Nelson, PharmD, and Brian Peek, PharmD.

About 1 in 10 Americans have diabetes mellitus (DM), of which about 90% to 95% are diagnosed with type 2 DM (T2DM) and veterans are disproportionately affected.1,2 About 25% enrolled in the Veterans Health Administration (VHA) have T2DM, which has been attributed to exposure to herbicides (eg, Agent Orange), decreased physical activity resulting from past physical strain, chronic pain, and other physical limitations resulting from military service.3-5

Pharmacologic management of DM is guided by the effectiveness of lifestyle interventions and comorbid diagnoses. Current DM management guidelines recommend patients with comorbid atherosclerotic cardiovascular disease, chronic kidney disease, or congestive heart failure receive first-line diabetes therapy with a sodium-glucose cotransporter-2 (SGLT-2) inhibitor or glucagon-like peptide-1 receptor (GLP-1) agonist.

Metformin remains a first-line pharmacologic option for the treatment of T2DM with the goal of achieving glycemic management when lifestyle interventions are insufficient.6,7 Newer antihyperglycemic therapies have been studied as adjunct therapy to metformin. However, there is limited literature comparing metformin directly to other medication classes for the treatment of T2DM.8-13 A systematic review of treatment-naive patients found HbA1c reductions were similar whether patients received metformin vs an SGLT-2 inhibitor, GLP-1 agonist, sulfonylurea, or thiazolidinedione monotherapy.10 The analysis found dipeptidyl-peptidase-4 (DPP-4) inhibitors had inferior HbA1c reduction compared to metformin.10 A Japanese systematic review compared metformin to thiazolidinediones, sulfonylureas, glinides, DPP-4 inhibitors, α-glucosidase inhibitors, or SGLT-2 inhibitors for ≥ 12 weeks but found no statistically significant differences in HbA1c reduction.11 The AWARD-3 trial compared once-weekly dulaglutide to metformin in treatment-experienced patients and found greater improvement in HbA1c and achievement of HbA1c goal with dulaglutide.13 While these studies show some comparisons of metformin to alternative pharmacologic therapy, researchers have not looked at what happens to patients’ HbA1c levels when an event, such as a recall, prompts a rapid change to a different antihyperglycemic agent.

On May 28, 2020, the US Food and Drug Administration (FDA) asked 5 pharmaceutical companies to voluntarily recall certain formulations of metformin. This action was taken when FDA testing revealed unacceptably high levels of N-Nitrosodimethylamine, a probable carcinogen.14 This FDA recall of metformin extended-release, referred to as metformin sustained-action (SA) within the VHA electronic medication file but the same type of formulation, prompted clinicians to revisit and revise the pharmacologic regimens of patients taking the drug. Because of the paucity of head-to-head trials comparing metformin with newer alternative antihyperglycemic therapies, the effect of treatment change was unknown. In response, we aimed to establish a data registry within Veterans Integrated Service Network (VISN) 6.

Registry Development

The VISN 6 registry was established to gather long-term, observational, head-to-head data that would allow review of HbA1c levels before and after the recall, as well as HbA1c levels broken down by the agent that patients were switched to after the recall. Another goal was to explore prescribing trends following the recall.

Data Access Request Tracker approval was obtained and a US Department of Veterans Affairs (VA) Information and Computing Infrastructure workspace was developed to host the registry data. The research cohort was established from this data, and the registry framework was finalized using Structured Query Language (SQL). The SQL coding allows for recurring data updates for all individuals within the cohort including date of birth, race, sex, ethnicity, VHA facility visited, weight, body mass index, HbA1c level, creatinine clearance, serum creatinine, antihyperglycemic medication prescriptions, adverse drug reactions, medication adherence (as defined by ≥ 80% refill history), and hospitalizations related to diabetes. For the purposes of this initial analysis, registry data included demographics, diabetes medications, and HbA1c results.

METHODS

This study was a concurrent, observational, multicenter, registry-based study conducted at the Western North Carolina VA Health Care System (WNCVAHCS). The study was approved by the WNCVAHCS institutional review board and research and development committees.

All patients aged ≥ 18 years with T2DM and receiving health care from VISN 6 facilities who had an active metformin SA prescription on, and 1 year prior to, June 1, 2020 (the initial date VHA began implementing the FDA metformin recall) were entered into the registry. Data from 1 year prior were collected to provide a baseline. Veterans were excluded if they received metformin SA for any indication other than T2DM, there was no pre- or postrecall HbA1c measurement, or death. We included 15,594 VISN 6 veterans.

Registry data were analyzed to determine whether a significant change in HbA1c level occurred after the metformin recall and in response to alternative agents being prescribed. Data from veterans who met all inclusion criteria were assessed during the year before and after June 1, 2020. Demographic data were analyzed using frequency and descriptive statistics. The Shapiro Wilkes test was performed, and data were found to be nonparametric; therefore the Wilcoxon signed-rank test was used to evaluate the hypothesis that HbA1c levels were not impacted by the recall.

Our sample size allowed us to create exact matched pairs of 9130 individuals and utilize rank-biserial correlation to establish effect size. Following this initial population-level test, we constructed 2 models. The first, a linear mixed-effects model, focused solely on the interaction effects between the pre- and postrecall periods and various medication classes on HbA1c levels. Second, we constructed a random-effects within-between model (REWB) to evaluate the impact ofmedication classes and demographic variables. Statistical significance was measured at P < .05 with conservative power at .90. The effect size was set to 1.0, reflecting a minimum clinically important difference. Literature establishes 0.5 as a modest level of HbA1c improvement and 1.0 as a clinically significant improvement.

RESULTS

Preliminary results included 15,594 veterans who received a metformin SA prescription as of June 1, 2020 from VISN 6 facilities; 15,392 veterans had a drug exposure end on June 1, 2020, indicating their standard therapy of metformin SA was discontinued following the FDA recall. Two hundred and two veterans were excluded from the registry because they continued to receive metformin SA from existing stock at a VISN6 facility. After identifying veterans with data for 1 year prior (June 1, 2019) to the index date and 1 year after (June 1, 2021) the study population was adjusted to 9130. The population was predominantly males aged> 60 years. Roughly 55% of the registry identified as White and nearly 40% as Black, and 2% indentified as Hispanic (Table 1).

Wilcoxon Signed-Rank Test

We created exact pairs by iterating the data and finding the closest measurements for each patient before and after the recall. This has the advantage over averaging a patient’s pre- and post-HbA1c levels, as it allows for a rank-biserial correlation. Using the nonparametric Wilcoxon signed-rank test, V was 20,100,707 (P < .001), indicating a significant effect. The –0.29 rank-biserial correlation, which was computed to assess the effect size of the recall, suggests that the median HbA1c level was lower postrecall vs prerecall. The magnitude of the correlation suggests a moderate effect size, and while the recall had a noticeable impact at a population level, it was not extreme (Table 2).

Linear Mixed-Effects Model

The binary variable for medication class exposure suggests the use of a logit link function for binary outcomes within the multilevel modeling framework.15 We employed a linear mixed-effects model to investigate the impact that switching from metformin SA to other T2DM medications had on HbA1c levels. The model was adjusted for patient-specific random effects and included interaction terms between the recall period (before and after) and the usage of different T2DM medications.

Model Fit and Random Effects

The model demonstrated a residual maximum likelihood criterion of 100,219.7, indicating its fit to the data. Notably, the random effects analysis revealed a substantial variability in baseline HbA1c levels across patients (SD, 0.94), highlighting the importance of individual differences in DM management. Medication classes with zero or near-zero exposure rate were removed. Due to demographic homogeneity, the model did not converge on demographic variables. Veterans were taking a mean of 1.8 T2DM medications and metformin SA was most common (Table 3).

During the postrecall period, metformin SA remained the most frequently prescribed medication class. This may be attributed to the existence of multiple manufacturers of metformin SA, some of which may not have been impacted by the recall. VISN 6 medical centers could have sought metformin SA outside of the usual procurement path following the recall.

Complex Random Effects Model

We employed a complex REWB model that evaluated the impact of medication classes on HbA1c levels, accounting for both within and between subject effects of these medications, along with demographic variables (sex, race, and ethnicity) (eAppendix). This model accounts for individual-level changes over time (within-patient effects) and between groups of patients (between-patient effects). This is a more comprehensive model aimed at understanding the broader impact of medications on HbA1c levels across diverse patient groups.

Most demographic categories did not demonstrate significant effects in this model. Black individuals experienced a slight increase in HbA1c levels compared with other racial categories that was not statistically significant. However, this model confirms the findings from the linear mixed-effects model that GLP-1 agonists showed a substantial decrease in HbA1c levels within patients (coefficient –0.5; 95% CI, –0.56 to –0.44; P < .001) and a moderate increase between patients (coefficient, 0.21; 95% CI, 0.12-0.31; P < .001). Additionally, SGLT-2 inhibitors had a notable decrease within patients (coefficient, –0.27; 95% CI, –0.32 to –0.22; P < .001).Another notable finding with our REWB model is insulin usage was associated with high HbA1c levels, but only between subjects. Long-acting insulin (coefficient, 0.96; 95% CI, 0.90-1.01; P <. 001) and mixed insulin (coefficient, 1.09; 95% CI, 0.94-1.24; P < .001) both displayed marked increases between patients, suggesting future analysis may benefit from stratifying across insulin users and nonusers.

Fixed Effect Analysis

The fixed effects analysis yielded several notable findings. The intercept, representing the mean baseline HbA1c level, was estimated at 7.8% (58 mmol/mol). The coefficient for the period (postrecall) was not statistically significant, indicating no overall change in HbA1c levels from before to after the recall when specific medication classes were not considered (Table 4). Among medication classes examined, several showed significant associations with HbA1c levels. DPP-4 inhibitors and GLP-1 agonists were associated with a decrease in HbA1c levels, with coefficients of −0.08 and −0.24, respectively. Long-acting insulin and metformin immediate-release (IR) were associated with an increase in HbA1c levels, as indicated by their positive coefficients of 0.38 and 0.16, respectively. Mixed insulin formulations and sulfonylureas showed an association with decreased HbA1c levels.

Interaction Effects

The interaction terms between the recall period and the medication classes provided insights into the differential impact of the medication switch postrecall. Notably, the interaction term for long-acting insulin (coefficient, −0.10) was significant, suggesting a differential effect on HbA1c levels postrecall. Other medications, like metformin IR, also exhibited significant interaction effects, indicating changes in the impact on HbA1c levels in the postrecall period. The binary variable for medication class exposure suggests the use of a logit link function for binary outcomes within the multilevel modeling framework.15 We did not address the potential for cross cluster heterogeneity due to different medication classes.

DISCUSSION

This study is an ongoing, concurrent, observational, multicenter, registry-based study consisting of VISN 6 veterans who have T2DM and were prescribed metformin SA on June 1, 2020. This initial aim was to evaluate change in HbA1c levels following the FDA metformin recall. While there was substantial variability in baseline HbA1c levels across the patients, the mean baseline HbA1c level at 7.5% (58 mmol/mol). Patients taking GLP-1 agonists showed substantial decrease in HbA1c levels (coefficient; –0.5; 95% CI, –0.56 to –0.44; P <. 001). Patients taking SGLT-2 inhibitors had a notable decrease in HbA1c (coefficient, –0.27; 95% CI, –0.32 to –0.22; P < .001). Despite this, the coefficient for the postrecall period was not statistically significant, indicating no overall change in HbA1c levels from pre- to postrecall when specific medication classes were not considered.

Further analysis included assessment of prescribing trends postrecall. There was an increase in SGLT-2 inhibitor, GLP-1 agonist, and DPP-4 inhibitor prescribing. Considering the growing evidence of the cardiovascular and renal benefits of these medication classes, specifically the GLP-1 agonists and SGLT-2 inhibitors, this trend would be expected.

Limitations

This study cohort did not capture veterans with T2DM who transferred their health care to VISN 6 after June 1, 2020, and continued to receive metformin SA from the prior facility. Inclusion of these veterans would have increased the registry population. Additionally, the cohort did not identify veterans who continued to receive metformin SA through a source other than the VA. Without that information, the registry cohort may include veterans thought to have either transitioned to a different therapy or to no other T2DM therapy after the recall.

Given that DM can progress over time, it is possible the transition to a new medication after the recall was the result of suboptimal management, or in response to an adverse effect from a previous medication, and not solely due to the metformin SA recall. In addition, there are several factors that could impact HbA1c level over time that were not accounted for in this study, such as medication adherence and lifestyle modifications.

The notable level of metformin SA prescriptions, despite the recall, may be attributed to several factors. First, not all patients stopped metformin completely. Review of the prescription data indicated that some veterans were provided with limited refills at select VA medical centers that had supplies (medication lots not recalled). Access to a safe supply of metformin SA after the recall may have varied among VISN 6 facilities. It is also possible that as new supplies of metformin SA became available, veterans restarted metformin SA. This may have been resumed while continuing a new medication prescribed at the beginning of the recall. As the year progressed after the recall, an increase in metformin SA prescriptions likely occurred as supplies became available and clinicians/veterans chose to resume this medication therapy.

Conclusions

Results of this initial registry study found no difference in HbA1c levels across the study population after the metformin SA recall. However, there was clinical difference in the HbA1c within veterans prescribed SGLT-2 inhibitors and GLP-1 agonists. As expected, prescribing trends showed an increase in these agents after the recall. With the known benefits of these medications beyond glucose lowering, it is anticipated the cohort of veterans prescribed these medications will continue to grow.

The VISN 6 research registry allowed this study to gain an important snapshot in time following the metformin SA recall, and will serve as an important resource for future DM research endeavors. It will allow for ongoing evaluation of the impact of the transition to alternative T2DM medications after the metformin SA recall. Future exploration will include evaluation of adverse drug reactions, DM-related hospitalizations, emergency department visits related to T2DM, changes in renal function, and cardiovascular events among all diabetes medication classes.

Acknowledgments

The study team thanks the Veterans Affairs Informatics and Computing Infrastructure for their help and expertise throughout this project. The authors acknowledge the contributions of Philip Nelson, PharmD, and Brian Peek, PharmD.

References
  1. Centers for Disease Control and Prevention. Type 2 diabetes. Updated April 18, 2023. Accessed September 18, 2023. https://www.cdc.gov/diabetes/basics/type2.html 
  2. ElSayed NA, Aleppo G, Aroda VR, et al. 2. Classification and diagnosis of diabetes: standards of care in diabetes—2023. Diabetes Care. 2023;46(Supplement_1):S19-S40. doi:10.2337/dc23-S002
  3. Liu Y, Sayam S, Shao X, et al. Prevalence of and trends in diabetes among veterans, United States, 2005–2014. Prev Chronic Dis. 2017;14:E135. doi:10.5888/pcd14.170230
  4. Yi SW, Hong JS, Ohrr H, Yi JJ. Agent Orange exposure and disease prevalence in Korean Vietnam veterans: the Korean veterans health study. Environ Res. 2014;133:56-65. doi:10.1016/j.envres.2014.04.027
  5. Price LE, Gephart S, Shea K. The VA’s Corporate Data Warehouse: Uses and Implications for Nursing Research and Practice. Nurs Adm Q. 2015;39(4):311-318. doi:10.1097/NAQ.0000000000000118
  6. ElSayed NA, Aleppo G, Aroda VR, et al. 9. Pharmacologic approaches to glycemic treatment: standards of care in diabetes-2023. Diabetes Care. 2023;46(suppl 1):S140-S157. doi:10.2337/dc23-S009
  7. Samson SL, Vellanki P, Blonde L, et al. American Association of Clinical Endocrinology Consensus Statement: Comprehensive Type 2 Diabetes Management Algorithm - 2023 Update. Endocr Pract. 2023;29(5):305-340. doi:10.1016/j.eprac.2023.02.001
  8. Bennett WL, Maruthur NM, Singh S, et al. Comparative effectiveness and safety of medications for type 2 diabetes: an update including new drugs and 2-drug combinations. Ann Intern Med. 2011;154(9):602-613. doi:10.7326/0003-4819-154-9-201105030-00336
  9. Bolen S, Feldman L, Vassy J, et al. Systematic review: comparative effectiveness and safety of oral medications for type 2 diabetes mellitus. Ann Intern Med. 2007;147(6):386-399. doi:10.7326/0003-4819-147-6-200709180-00178
  10. Tsapas A, Avgerinos I, Karagiannis T, et al. Comparative effectiveness of glucose-lowering drugs for type 2 diabetes: a systematic review and network meta-analysis. Ann Intern Med. 2020;173(4):278-286. doi:10.7326/M20-0864
  11. Nishimura R, Taniguchi M, Takeshima T, Iwasaki K. Efficacy and safety of metformin versus the other oral antidiabetic drugs in Japanese type 2 diabetes patients: a network meta-analysis. Adv Ther. 2022;39(1):632-654. doi:10.1007/s12325-021-01979-1
  12. Russell-Jones D, Cuddihy RM, Hanefeld M, et al. Efficacy and safety of exenatide once weekly versus metformin, pioglitazone, and sitagliptin used as monotherapy in drug-naive patients with type 2 diabetes (DURATION-4): a 26-week double-blind study. Diabetes Care. 2012;35(2):252-258. doi:10.2337/dc11-1107
  13. Umpierrez G, Tofé Povedano S, Pérez Manghi F, Shurzinske L, Pechtner V. Efficacy and safety of dulaglutide monotherapy versus metformin in type 2 diabetes in a randomized controlled trial (AWARD-3). Diabetes Care. 2014;37(8):2168-2176. doi:10.2337/dc13-2759
  14. US Food and Drug Administration. FDA alerts patients and health care professionals to nitrosamine impurity findings in certain metformin extended-release products [press release]. May 28, 2020. Accessed October 16, 2024. https://www.fda.gov/news-events/press-announcements/fda-alerts-patients-and-health-care-professionals-nitrosamine-impurity-findings-certain-metformin
  15. Bell A, Jones K. Explaining fixed effects: random effects modeling of time-series cross-sectional and panel data. PSRM. 2015;3(1):133-153. doi:10.1017/psrm.2014.7
References
  1. Centers for Disease Control and Prevention. Type 2 diabetes. Updated April 18, 2023. Accessed September 18, 2023. https://www.cdc.gov/diabetes/basics/type2.html 
  2. ElSayed NA, Aleppo G, Aroda VR, et al. 2. Classification and diagnosis of diabetes: standards of care in diabetes—2023. Diabetes Care. 2023;46(Supplement_1):S19-S40. doi:10.2337/dc23-S002
  3. Liu Y, Sayam S, Shao X, et al. Prevalence of and trends in diabetes among veterans, United States, 2005–2014. Prev Chronic Dis. 2017;14:E135. doi:10.5888/pcd14.170230
  4. Yi SW, Hong JS, Ohrr H, Yi JJ. Agent Orange exposure and disease prevalence in Korean Vietnam veterans: the Korean veterans health study. Environ Res. 2014;133:56-65. doi:10.1016/j.envres.2014.04.027
  5. Price LE, Gephart S, Shea K. The VA’s Corporate Data Warehouse: Uses and Implications for Nursing Research and Practice. Nurs Adm Q. 2015;39(4):311-318. doi:10.1097/NAQ.0000000000000118
  6. ElSayed NA, Aleppo G, Aroda VR, et al. 9. Pharmacologic approaches to glycemic treatment: standards of care in diabetes-2023. Diabetes Care. 2023;46(suppl 1):S140-S157. doi:10.2337/dc23-S009
  7. Samson SL, Vellanki P, Blonde L, et al. American Association of Clinical Endocrinology Consensus Statement: Comprehensive Type 2 Diabetes Management Algorithm - 2023 Update. Endocr Pract. 2023;29(5):305-340. doi:10.1016/j.eprac.2023.02.001
  8. Bennett WL, Maruthur NM, Singh S, et al. Comparative effectiveness and safety of medications for type 2 diabetes: an update including new drugs and 2-drug combinations. Ann Intern Med. 2011;154(9):602-613. doi:10.7326/0003-4819-154-9-201105030-00336
  9. Bolen S, Feldman L, Vassy J, et al. Systematic review: comparative effectiveness and safety of oral medications for type 2 diabetes mellitus. Ann Intern Med. 2007;147(6):386-399. doi:10.7326/0003-4819-147-6-200709180-00178
  10. Tsapas A, Avgerinos I, Karagiannis T, et al. Comparative effectiveness of glucose-lowering drugs for type 2 diabetes: a systematic review and network meta-analysis. Ann Intern Med. 2020;173(4):278-286. doi:10.7326/M20-0864
  11. Nishimura R, Taniguchi M, Takeshima T, Iwasaki K. Efficacy and safety of metformin versus the other oral antidiabetic drugs in Japanese type 2 diabetes patients: a network meta-analysis. Adv Ther. 2022;39(1):632-654. doi:10.1007/s12325-021-01979-1
  12. Russell-Jones D, Cuddihy RM, Hanefeld M, et al. Efficacy and safety of exenatide once weekly versus metformin, pioglitazone, and sitagliptin used as monotherapy in drug-naive patients with type 2 diabetes (DURATION-4): a 26-week double-blind study. Diabetes Care. 2012;35(2):252-258. doi:10.2337/dc11-1107
  13. Umpierrez G, Tofé Povedano S, Pérez Manghi F, Shurzinske L, Pechtner V. Efficacy and safety of dulaglutide monotherapy versus metformin in type 2 diabetes in a randomized controlled trial (AWARD-3). Diabetes Care. 2014;37(8):2168-2176. doi:10.2337/dc13-2759
  14. US Food and Drug Administration. FDA alerts patients and health care professionals to nitrosamine impurity findings in certain metformin extended-release products [press release]. May 28, 2020. Accessed October 16, 2024. https://www.fda.gov/news-events/press-announcements/fda-alerts-patients-and-health-care-professionals-nitrosamine-impurity-findings-certain-metformin
  15. Bell A, Jones K. Explaining fixed effects: random effects modeling of time-series cross-sectional and panel data. PSRM. 2015;3(1):133-153. doi:10.1017/psrm.2014.7
Issue
Federal Practitioner - 41(11)s
Issue
Federal Practitioner - 41(11)s
Page Number
S6-S11
Page Number
S6-S11
Publications
Publications
Topics
Article Type
Display Headline
The Impact of a Metformin Recall on Patient Hemoglobin A1c Levels at a VA Network
Display Headline
The Impact of a Metformin Recall on Patient Hemoglobin A1c Levels at a VA Network
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

Projected 2023 Cost Reduction From Tumor Necrosis Factor α Inhibitor Biosimilars in Dermatology: A National Medicare Analysis

Article Type
Changed
Tue, 10/22/2024 - 09:30
Display Headline
Projected 2023 Cost Reduction From Tumor Necrosis Factor α Inhibitor Biosimilars in Dermatology: A National Medicare Analysis

To the Editor:

Although biologics provide major therapeutic benefits for dermatologic conditions, they also come with a substantial cost, making them among the most expensive medications available. Medicare and Medicaid spending on biologics for dermatologic conditions increased by 320% from 2012 to 2018, reaching a staggering $10.6 billion in 2018 alone.1 Biosimilars show promise in reducing health care spending for dermatologic conditions; however, their utilization has been limited due to multiple factors, including delayed market entry from patent thickets, exclusionary formulary contracts, and prescriber skepticism regarding their safety and efficacy.2 For instance, a national survey of 1201 US physicians in specialties that are high prescribers of biologics reported that 55% doubted the safety and appropriateness of biosimilars.3

US Food and Drug Administration approval of biosimilars for adalimumab and etanercept offers the potential to reduce health care spending for dermatologic conditions. However, this cost reduction is dependent on utilization rates among dermatologists. In this national cross-sectional review of Medicare data, we predicted the impact of these biosimilars on dermatologic Medicare costs and demonstrated how differing utilization rates among dermatologists can influence potential savings.

To model 2023 utilization and cost reduction from biosimilars, we analyzed Medicare Part D data from 2020 on existing biosimilars, including granulocyte colony–stimulating factors, erythropoiesis-stimulating agents, and tumor necrosis factor α inhibitors.4 Methods in line with a 2021 report from the US Department of Health and Human Services5 as well as those of Yazdany et al6 were used. For each class, we calculated the 2020 distribution of biosimilar and originator drug claims as well as biosimilar cost reduction per 30-day claim. We utilized 2018-2021 annual growth rates for branded adalimumab and etanercept to estimate 30-day claims for 2023 and the cost of these branded agents in the absence of biosimilars. The hypothetical 2023 cost reduction from adalimumab and etanercept biosimilars was estimated by assuming 2020 biosimilar utilization rates and mean cost reduction per claim. This study utilized publicly available or aggregate summary data (not attributable to specific patients) and did not qualify as human subject research; therefore, institutional review board approval was not required.

In 2020, biosimilar utilization proportions ranged from 6.4% (tumor necrosis factor α inhibitors) to 82.7% (granulocyte colony–stimulating factors), with a mean across all classes of 35.7%. On average, the cost per 30-day claim of biosimilars was 66.8% of originator agents (Table 1). In 2021, we identified 57,868 30-day claims for branded adalimumab and etanercept submitted by dermatologists. From 2018 to 2021, 30-day branded adalimumab claims increased by 1.27% annually (cost + 10.62% annually), while claims for branded etanercept decreased by 13.0% annually (cost + 5.68% annually). Assuming these trends, the cost of branded adalimumab and etanercept was estimated to be $539 million in 2023. Applying the aforementioned 35.7% utilization, the introduction of biosimilars in dermatology would yield a cost reduction of approximately $118 million (21.9%). A high utilization rate (82.7%) of biosimilars among dermatologists would increase cost savings to $199 million (36.9%)(Table 2).



Our study demonstrates that the introduction of 2 biosimilars into dermatology may result in a notable reduction in Medicare expenditures. The savings observed are likely to translate to substantial cost savings for patients. A cross-sectional analysis of 2020 Medicare data indicated that coverage for psoriasis medications was 10.0% to 99.8% across different products and Medicare Part D plans. Consequently, patients faced considerable out-of-pocket expenses, amounting to $5653 and $5714 per year for adalimumab and etanercept, respectively.7 


We found that the extent of savings from biosimilars was dependent on the utilization rates among dermatologists, with the highest utilization rate almost doubling the total savings of average utilization rates. Given the impact of high utilization and the wide variation observed, understanding the factors that have influenced uptake of biosimilars is important to increasing utilization as these medications become integrated into dermatology. For instance, limited uptake of infliximab initially may have been influenced by concerns about efficacy and increased adverse events.8,9 In contrast, the high utilization of filgrastim biosimilars (82.7%) may be attributed to its longevity in the market and familiarity to prescribers, as filgrastim was the first biosimilar to be approved in the United States.10

Promoting reasonable utilization of biosimilars may require prescriber education on their safety and approval processes, which could foster increased utilization and reduce skepticism.4 Under the Biologics Price Competition and Innovation Act, the US Food and Drug Administration approves biosimilars only when they exhibit “high similarity” and show no “clinically meaningful differences” compared to the reference biologic, with no added safety risks or reduced efficacy.11 Moreover, a 2023 systematic review of 17 studies found no major difference in efficacy and safety between biosimilars and originators of etanercept, infliximab, and other biologics.12 Understanding these findings may reassure dermatologists and patients about the reliability and safety of biosimilars.

A limitation of our study is that it solely assesses Medicare data and estimates derived from existing (separate) biologic classes. It also does not account for potential expenditure shifts to newer biologic agents (eg, IL-12/17/23 inhibitors) or changes in manufacturer behavior or promotions. Nevertheless, it indicates notable financial savings from new biosimilar agents in dermatology; along with their compelling efficacy and safety profiles, this could represent a substantial benefit to patients and the health care system.

References
  1. Price KN, Atluri S, Hsiao JL, et al. Medicare and medicaid spending trends for immunomodulators prescribed for dermatologic conditions. J Dermatolog Treat. 2020;33:575-579.
  2. Zhai MZ, Sarpatwari A, Kesselheim AS. Why are biosimilars not living up to their promise in the US? AMA J Ethics. 2019;21:E668-E678. doi:10.1001/amajethics.2019.668
  3. Cohen H, Beydoun D, Chien D, et al. Awareness, knowledge, and perceptions of biosimilars among specialty physicians. Adv Ther. 2017;33:2160-2172.
  4. Centers for Medicare & Medicaid Services. Medicare Part D prescribers— by provider and drug. Accessed September 11, 2024. https://data.cms.gov/provider-summary-by-type-of-service/medicare-part-d-prescribers/medicare-part-d-prescribers-by-provider-and-drug/data
  5. US Department of Health and Human Services. Office of Inspector General. Medicare Part D and beneficiaries could realize significant spending reductions with increased biosimilar use. Accessed September 11, 2024. https://oig.hhs.gov/oei/reports/OEI-05-20-00480.pdf
  6. Yazdany J, Dudley RA, Lin GA, et al. Out-of-pocket costs for infliximab and its biosimilar for rheumatoid arthritis under Medicare Part D. JAMA. 2018;320:931-933. doi:10.1001/jama.2018.7316
  7. Pourali SP, Nshuti L, Dusetzina SB. Out-of-pocket costs of specialty medications for psoriasis and psoriatic arthritis treatment in the medicare population. JAMA Dermatol. 2021;157:1239-1241. doi:10.1001/ jamadermatol.2021.3616
  8. Lebwohl M. Biosimilars in dermatology. JAMA Dermatol. 2021; 157:641-642. doi:10.1001/jamadermatol.2021.0219
  9. Westerkam LL, Tackett KJ, Sayed CJ. Comparing the effectiveness and safety associated with infliximab vs infliximab-abda therapy for patients with hidradenitis suppurativa. JAMA Dermatol. 2021;157:708-711. doi:10.1001/jamadermatol.2021.0220
  10. Awad M, Singh P, Hilas O. Zarxio (Filgrastim-sndz): the first biosimilar approved by the FDA. P T. 2017;42:19-23.
  11. Development of therapeutic protein biosimilars: comparative analytical assessment and other quality-related considerations guidance for industry. US Department of Health and Human Services website. Updated June 15, 2022. Accessed October 21, 2024. https://www.fda.gov/regulatory-information/search-fda-guidance-documents/development-therapeutic-protein-biosimilars-comparative-analyticalassessment-and-other-quality
  12. Phan DB, Elyoussfi S, Stevenson M, et al. Biosimilars for the treatment of psoriasis: a systematic review of clinical trials and observational studies. JAMA Dermatol. 2023;159:763-771. doi:10.1001/jamadermatol.2023.1338
Article PDF
Author and Disclosure Information

Dr. Roster is from the Department of Dermatology, Georgetown University School of Medicine, Medstar Washington Hospital Center, Washington, DC. Drs. Gronbeck and Feng are from the Department of Dermatology, University of Connecticut Health Center, Farmington.

Drs. Roster and Gronbeck have no relevant financial disclosures to report. Dr. Feng is a consultant for Cytrellis Biosystems, Inc, and Soliton Inc.

Correspondence: Hao Feng, MD, MHS, Department of Dermatology, University of Connecticut Health Center, 21 South Rd, 2nd Floor, Farmington, CT 06032 ([email protected]).

Cutis. 2024 October;114(4):E8-E11. doi:10.12788/cutis.1107

Publications
Topics
Page Number
E8-E11
Sections
Author and Disclosure Information

Dr. Roster is from the Department of Dermatology, Georgetown University School of Medicine, Medstar Washington Hospital Center, Washington, DC. Drs. Gronbeck and Feng are from the Department of Dermatology, University of Connecticut Health Center, Farmington.

Drs. Roster and Gronbeck have no relevant financial disclosures to report. Dr. Feng is a consultant for Cytrellis Biosystems, Inc, and Soliton Inc.

Correspondence: Hao Feng, MD, MHS, Department of Dermatology, University of Connecticut Health Center, 21 South Rd, 2nd Floor, Farmington, CT 06032 ([email protected]).

Cutis. 2024 October;114(4):E8-E11. doi:10.12788/cutis.1107

Author and Disclosure Information

Dr. Roster is from the Department of Dermatology, Georgetown University School of Medicine, Medstar Washington Hospital Center, Washington, DC. Drs. Gronbeck and Feng are from the Department of Dermatology, University of Connecticut Health Center, Farmington.

Drs. Roster and Gronbeck have no relevant financial disclosures to report. Dr. Feng is a consultant for Cytrellis Biosystems, Inc, and Soliton Inc.

Correspondence: Hao Feng, MD, MHS, Department of Dermatology, University of Connecticut Health Center, 21 South Rd, 2nd Floor, Farmington, CT 06032 ([email protected]).

Cutis. 2024 October;114(4):E8-E11. doi:10.12788/cutis.1107

Article PDF
Article PDF

To the Editor:

Although biologics provide major therapeutic benefits for dermatologic conditions, they also come with a substantial cost, making them among the most expensive medications available. Medicare and Medicaid spending on biologics for dermatologic conditions increased by 320% from 2012 to 2018, reaching a staggering $10.6 billion in 2018 alone.1 Biosimilars show promise in reducing health care spending for dermatologic conditions; however, their utilization has been limited due to multiple factors, including delayed market entry from patent thickets, exclusionary formulary contracts, and prescriber skepticism regarding their safety and efficacy.2 For instance, a national survey of 1201 US physicians in specialties that are high prescribers of biologics reported that 55% doubted the safety and appropriateness of biosimilars.3

US Food and Drug Administration approval of biosimilars for adalimumab and etanercept offers the potential to reduce health care spending for dermatologic conditions. However, this cost reduction is dependent on utilization rates among dermatologists. In this national cross-sectional review of Medicare data, we predicted the impact of these biosimilars on dermatologic Medicare costs and demonstrated how differing utilization rates among dermatologists can influence potential savings.

To model 2023 utilization and cost reduction from biosimilars, we analyzed Medicare Part D data from 2020 on existing biosimilars, including granulocyte colony–stimulating factors, erythropoiesis-stimulating agents, and tumor necrosis factor α inhibitors.4 Methods in line with a 2021 report from the US Department of Health and Human Services5 as well as those of Yazdany et al6 were used. For each class, we calculated the 2020 distribution of biosimilar and originator drug claims as well as biosimilar cost reduction per 30-day claim. We utilized 2018-2021 annual growth rates for branded adalimumab and etanercept to estimate 30-day claims for 2023 and the cost of these branded agents in the absence of biosimilars. The hypothetical 2023 cost reduction from adalimumab and etanercept biosimilars was estimated by assuming 2020 biosimilar utilization rates and mean cost reduction per claim. This study utilized publicly available or aggregate summary data (not attributable to specific patients) and did not qualify as human subject research; therefore, institutional review board approval was not required.

In 2020, biosimilar utilization proportions ranged from 6.4% (tumor necrosis factor α inhibitors) to 82.7% (granulocyte colony–stimulating factors), with a mean across all classes of 35.7%. On average, the cost per 30-day claim of biosimilars was 66.8% of originator agents (Table 1). In 2021, we identified 57,868 30-day claims for branded adalimumab and etanercept submitted by dermatologists. From 2018 to 2021, 30-day branded adalimumab claims increased by 1.27% annually (cost + 10.62% annually), while claims for branded etanercept decreased by 13.0% annually (cost + 5.68% annually). Assuming these trends, the cost of branded adalimumab and etanercept was estimated to be $539 million in 2023. Applying the aforementioned 35.7% utilization, the introduction of biosimilars in dermatology would yield a cost reduction of approximately $118 million (21.9%). A high utilization rate (82.7%) of biosimilars among dermatologists would increase cost savings to $199 million (36.9%)(Table 2).



Our study demonstrates that the introduction of 2 biosimilars into dermatology may result in a notable reduction in Medicare expenditures. The savings observed are likely to translate to substantial cost savings for patients. A cross-sectional analysis of 2020 Medicare data indicated that coverage for psoriasis medications was 10.0% to 99.8% across different products and Medicare Part D plans. Consequently, patients faced considerable out-of-pocket expenses, amounting to $5653 and $5714 per year for adalimumab and etanercept, respectively.7 


We found that the extent of savings from biosimilars was dependent on the utilization rates among dermatologists, with the highest utilization rate almost doubling the total savings of average utilization rates. Given the impact of high utilization and the wide variation observed, understanding the factors that have influenced uptake of biosimilars is important to increasing utilization as these medications become integrated into dermatology. For instance, limited uptake of infliximab initially may have been influenced by concerns about efficacy and increased adverse events.8,9 In contrast, the high utilization of filgrastim biosimilars (82.7%) may be attributed to its longevity in the market and familiarity to prescribers, as filgrastim was the first biosimilar to be approved in the United States.10

Promoting reasonable utilization of biosimilars may require prescriber education on their safety and approval processes, which could foster increased utilization and reduce skepticism.4 Under the Biologics Price Competition and Innovation Act, the US Food and Drug Administration approves biosimilars only when they exhibit “high similarity” and show no “clinically meaningful differences” compared to the reference biologic, with no added safety risks or reduced efficacy.11 Moreover, a 2023 systematic review of 17 studies found no major difference in efficacy and safety between biosimilars and originators of etanercept, infliximab, and other biologics.12 Understanding these findings may reassure dermatologists and patients about the reliability and safety of biosimilars.

A limitation of our study is that it solely assesses Medicare data and estimates derived from existing (separate) biologic classes. It also does not account for potential expenditure shifts to newer biologic agents (eg, IL-12/17/23 inhibitors) or changes in manufacturer behavior or promotions. Nevertheless, it indicates notable financial savings from new biosimilar agents in dermatology; along with their compelling efficacy and safety profiles, this could represent a substantial benefit to patients and the health care system.

To the Editor:

Although biologics provide major therapeutic benefits for dermatologic conditions, they also come with a substantial cost, making them among the most expensive medications available. Medicare and Medicaid spending on biologics for dermatologic conditions increased by 320% from 2012 to 2018, reaching a staggering $10.6 billion in 2018 alone.1 Biosimilars show promise in reducing health care spending for dermatologic conditions; however, their utilization has been limited due to multiple factors, including delayed market entry from patent thickets, exclusionary formulary contracts, and prescriber skepticism regarding their safety and efficacy.2 For instance, a national survey of 1201 US physicians in specialties that are high prescribers of biologics reported that 55% doubted the safety and appropriateness of biosimilars.3

US Food and Drug Administration approval of biosimilars for adalimumab and etanercept offers the potential to reduce health care spending for dermatologic conditions. However, this cost reduction is dependent on utilization rates among dermatologists. In this national cross-sectional review of Medicare data, we predicted the impact of these biosimilars on dermatologic Medicare costs and demonstrated how differing utilization rates among dermatologists can influence potential savings.

To model 2023 utilization and cost reduction from biosimilars, we analyzed Medicare Part D data from 2020 on existing biosimilars, including granulocyte colony–stimulating factors, erythropoiesis-stimulating agents, and tumor necrosis factor α inhibitors.4 Methods in line with a 2021 report from the US Department of Health and Human Services5 as well as those of Yazdany et al6 were used. For each class, we calculated the 2020 distribution of biosimilar and originator drug claims as well as biosimilar cost reduction per 30-day claim. We utilized 2018-2021 annual growth rates for branded adalimumab and etanercept to estimate 30-day claims for 2023 and the cost of these branded agents in the absence of biosimilars. The hypothetical 2023 cost reduction from adalimumab and etanercept biosimilars was estimated by assuming 2020 biosimilar utilization rates and mean cost reduction per claim. This study utilized publicly available or aggregate summary data (not attributable to specific patients) and did not qualify as human subject research; therefore, institutional review board approval was not required.

In 2020, biosimilar utilization proportions ranged from 6.4% (tumor necrosis factor α inhibitors) to 82.7% (granulocyte colony–stimulating factors), with a mean across all classes of 35.7%. On average, the cost per 30-day claim of biosimilars was 66.8% of originator agents (Table 1). In 2021, we identified 57,868 30-day claims for branded adalimumab and etanercept submitted by dermatologists. From 2018 to 2021, 30-day branded adalimumab claims increased by 1.27% annually (cost + 10.62% annually), while claims for branded etanercept decreased by 13.0% annually (cost + 5.68% annually). Assuming these trends, the cost of branded adalimumab and etanercept was estimated to be $539 million in 2023. Applying the aforementioned 35.7% utilization, the introduction of biosimilars in dermatology would yield a cost reduction of approximately $118 million (21.9%). A high utilization rate (82.7%) of biosimilars among dermatologists would increase cost savings to $199 million (36.9%)(Table 2).



Our study demonstrates that the introduction of 2 biosimilars into dermatology may result in a notable reduction in Medicare expenditures. The savings observed are likely to translate to substantial cost savings for patients. A cross-sectional analysis of 2020 Medicare data indicated that coverage for psoriasis medications was 10.0% to 99.8% across different products and Medicare Part D plans. Consequently, patients faced considerable out-of-pocket expenses, amounting to $5653 and $5714 per year for adalimumab and etanercept, respectively.7 


We found that the extent of savings from biosimilars was dependent on the utilization rates among dermatologists, with the highest utilization rate almost doubling the total savings of average utilization rates. Given the impact of high utilization and the wide variation observed, understanding the factors that have influenced uptake of biosimilars is important to increasing utilization as these medications become integrated into dermatology. For instance, limited uptake of infliximab initially may have been influenced by concerns about efficacy and increased adverse events.8,9 In contrast, the high utilization of filgrastim biosimilars (82.7%) may be attributed to its longevity in the market and familiarity to prescribers, as filgrastim was the first biosimilar to be approved in the United States.10

Promoting reasonable utilization of biosimilars may require prescriber education on their safety and approval processes, which could foster increased utilization and reduce skepticism.4 Under the Biologics Price Competition and Innovation Act, the US Food and Drug Administration approves biosimilars only when they exhibit “high similarity” and show no “clinically meaningful differences” compared to the reference biologic, with no added safety risks or reduced efficacy.11 Moreover, a 2023 systematic review of 17 studies found no major difference in efficacy and safety between biosimilars and originators of etanercept, infliximab, and other biologics.12 Understanding these findings may reassure dermatologists and patients about the reliability and safety of biosimilars.

A limitation of our study is that it solely assesses Medicare data and estimates derived from existing (separate) biologic classes. It also does not account for potential expenditure shifts to newer biologic agents (eg, IL-12/17/23 inhibitors) or changes in manufacturer behavior or promotions. Nevertheless, it indicates notable financial savings from new biosimilar agents in dermatology; along with their compelling efficacy and safety profiles, this could represent a substantial benefit to patients and the health care system.

References
  1. Price KN, Atluri S, Hsiao JL, et al. Medicare and medicaid spending trends for immunomodulators prescribed for dermatologic conditions. J Dermatolog Treat. 2020;33:575-579.
  2. Zhai MZ, Sarpatwari A, Kesselheim AS. Why are biosimilars not living up to their promise in the US? AMA J Ethics. 2019;21:E668-E678. doi:10.1001/amajethics.2019.668
  3. Cohen H, Beydoun D, Chien D, et al. Awareness, knowledge, and perceptions of biosimilars among specialty physicians. Adv Ther. 2017;33:2160-2172.
  4. Centers for Medicare & Medicaid Services. Medicare Part D prescribers— by provider and drug. Accessed September 11, 2024. https://data.cms.gov/provider-summary-by-type-of-service/medicare-part-d-prescribers/medicare-part-d-prescribers-by-provider-and-drug/data
  5. US Department of Health and Human Services. Office of Inspector General. Medicare Part D and beneficiaries could realize significant spending reductions with increased biosimilar use. Accessed September 11, 2024. https://oig.hhs.gov/oei/reports/OEI-05-20-00480.pdf
  6. Yazdany J, Dudley RA, Lin GA, et al. Out-of-pocket costs for infliximab and its biosimilar for rheumatoid arthritis under Medicare Part D. JAMA. 2018;320:931-933. doi:10.1001/jama.2018.7316
  7. Pourali SP, Nshuti L, Dusetzina SB. Out-of-pocket costs of specialty medications for psoriasis and psoriatic arthritis treatment in the medicare population. JAMA Dermatol. 2021;157:1239-1241. doi:10.1001/ jamadermatol.2021.3616
  8. Lebwohl M. Biosimilars in dermatology. JAMA Dermatol. 2021; 157:641-642. doi:10.1001/jamadermatol.2021.0219
  9. Westerkam LL, Tackett KJ, Sayed CJ. Comparing the effectiveness and safety associated with infliximab vs infliximab-abda therapy for patients with hidradenitis suppurativa. JAMA Dermatol. 2021;157:708-711. doi:10.1001/jamadermatol.2021.0220
  10. Awad M, Singh P, Hilas O. Zarxio (Filgrastim-sndz): the first biosimilar approved by the FDA. P T. 2017;42:19-23.
  11. Development of therapeutic protein biosimilars: comparative analytical assessment and other quality-related considerations guidance for industry. US Department of Health and Human Services website. Updated June 15, 2022. Accessed October 21, 2024. https://www.fda.gov/regulatory-information/search-fda-guidance-documents/development-therapeutic-protein-biosimilars-comparative-analyticalassessment-and-other-quality
  12. Phan DB, Elyoussfi S, Stevenson M, et al. Biosimilars for the treatment of psoriasis: a systematic review of clinical trials and observational studies. JAMA Dermatol. 2023;159:763-771. doi:10.1001/jamadermatol.2023.1338
References
  1. Price KN, Atluri S, Hsiao JL, et al. Medicare and medicaid spending trends for immunomodulators prescribed for dermatologic conditions. J Dermatolog Treat. 2020;33:575-579.
  2. Zhai MZ, Sarpatwari A, Kesselheim AS. Why are biosimilars not living up to their promise in the US? AMA J Ethics. 2019;21:E668-E678. doi:10.1001/amajethics.2019.668
  3. Cohen H, Beydoun D, Chien D, et al. Awareness, knowledge, and perceptions of biosimilars among specialty physicians. Adv Ther. 2017;33:2160-2172.
  4. Centers for Medicare & Medicaid Services. Medicare Part D prescribers— by provider and drug. Accessed September 11, 2024. https://data.cms.gov/provider-summary-by-type-of-service/medicare-part-d-prescribers/medicare-part-d-prescribers-by-provider-and-drug/data
  5. US Department of Health and Human Services. Office of Inspector General. Medicare Part D and beneficiaries could realize significant spending reductions with increased biosimilar use. Accessed September 11, 2024. https://oig.hhs.gov/oei/reports/OEI-05-20-00480.pdf
  6. Yazdany J, Dudley RA, Lin GA, et al. Out-of-pocket costs for infliximab and its biosimilar for rheumatoid arthritis under Medicare Part D. JAMA. 2018;320:931-933. doi:10.1001/jama.2018.7316
  7. Pourali SP, Nshuti L, Dusetzina SB. Out-of-pocket costs of specialty medications for psoriasis and psoriatic arthritis treatment in the medicare population. JAMA Dermatol. 2021;157:1239-1241. doi:10.1001/ jamadermatol.2021.3616
  8. Lebwohl M. Biosimilars in dermatology. JAMA Dermatol. 2021; 157:641-642. doi:10.1001/jamadermatol.2021.0219
  9. Westerkam LL, Tackett KJ, Sayed CJ. Comparing the effectiveness and safety associated with infliximab vs infliximab-abda therapy for patients with hidradenitis suppurativa. JAMA Dermatol. 2021;157:708-711. doi:10.1001/jamadermatol.2021.0220
  10. Awad M, Singh P, Hilas O. Zarxio (Filgrastim-sndz): the first biosimilar approved by the FDA. P T. 2017;42:19-23.
  11. Development of therapeutic protein biosimilars: comparative analytical assessment and other quality-related considerations guidance for industry. US Department of Health and Human Services website. Updated June 15, 2022. Accessed October 21, 2024. https://www.fda.gov/regulatory-information/search-fda-guidance-documents/development-therapeutic-protein-biosimilars-comparative-analyticalassessment-and-other-quality
  12. Phan DB, Elyoussfi S, Stevenson M, et al. Biosimilars for the treatment of psoriasis: a systematic review of clinical trials and observational studies. JAMA Dermatol. 2023;159:763-771. doi:10.1001/jamadermatol.2023.1338
Page Number
E8-E11
Page Number
E8-E11
Publications
Publications
Topics
Article Type
Display Headline
Projected 2023 Cost Reduction From Tumor Necrosis Factor α Inhibitor Biosimilars in Dermatology: A National Medicare Analysis
Display Headline
Projected 2023 Cost Reduction From Tumor Necrosis Factor α Inhibitor Biosimilars in Dermatology: A National Medicare Analysis
Sections
Inside the Article

Practice Points

  • Biosimilars for adalimumab and etanercept are safe and effective alternatives with the potential to reduce health care costs in dermatology by approximately $118 million.
  • A high utilization rate of biosimilars by dermatologists would increase cost savings even further.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

Utilization, Cost, and Prescription Trends of Antipsychotics Prescribed by Dermatologists for Medicare Patients

Article Type
Changed
Fri, 10/11/2024 - 16:15
Display Headline
Utilization, Cost, and Prescription Trends of Antipsychotics Prescribed by Dermatologists for Medicare Patients

To the Editor:

Patients with primary psychiatric disorders with dermatologic manifestations often seek treatment from dermatologists instead of psychiatrists.1 For example, patients with delusions of parasitosis may lack insight into the underlying etiology of their disease and instead fixate on establishing an organic cause for their symptoms. As a result, it is an increasingly common practice for dermatologists to diagnose and treat psychiatric conditions.1 The goal of this study was to evaluate trends for the top 5 antipsychotics most frequently prescribed by dermatologists in the Medicare Part D database.

In this retrospective analysis, we consulted the Medicare Provider Utilization and Payment Data for January 2013 through December 2020, which is provided to the public by the Centers for Medicare & Medicaid Services.2 Only prescribing data from dermatologists were included in this study by using the built-in filter on the website to select “dermatology” as the prescriber type. All other provider types were excluded. We chose the top 5 most prescribed antipsychotics based on the number of supply days reported. Supply days—defined by Medicare as the number of days’ worth of medication that is prescribed—were used as a metric for ­utilization; therefore, each drug’s total supply days prescribed by dermatologists were calculated using this combined filter of drug name and total supply days using the database.

To analyze utilization over time, the annual average growth rate (AAGR) was calculated by determining the growth rate in total supply days annually from 2013 to 2020 and then averaging those rates to determine the overall AAGR. For greater clinical relevance, we calculated the average growth in supply days for the entire study period by determining the difference in the number of supply days for each year and then averaging these values. This was done to consider overall trends across dermatology rather than individual dermatologist prescribing patterns.

Based on our analysis, the antipsychotics most frequently prescribed by dermatologists for Medicare patients from January 2013 to December 2020 were pimozide, quetiapine, risperidone, olanzapine, and aripiprazole. The AAGR for each drug was 2.35%, 4.89%, 5.59%, 9.48%, and 20.72%, respectively, which is consistent with increased utilization over the study period for all 5 drugs (Table 1). The change in cost per supply day for the same period was 1.3%, 66.1%, 60.2%, 81.7%, and84.3%, respectively. The net difference in cost per supply day over this entire period was $0.02, $2.79, $1.06, $5.37, and $21.22, respectively (Table 2).



There were several limitations to our study. Our analysis was limited to the Medicare population. Uninsured patients and those with Medicare Advantage or private health insurance plans were not included. In the Medicare database, only prescribers who prescribed a medication 10 times or more were recorded; therefore, some prescribers were not captured.

Although there was an increase in the dermatologic use of all 5 drugs in this study, perhaps the most marked growth was exhibited by aripiprazole, which had an AAGR of 20.72% (Table 1). Affordability may have been a factor, as the most marked reduction in price per supply day was noted for aripiprazole during the study period. Pimozide, which traditionally has been the first-line therapy for delusions of parasitosis, is the only first-generation antipsychotic drug among the 5 most frequently prescribed antipsychotics.3 Interestingly, pimozide had the lowest AAGR compared with the 4 second-generation antipsychotics. This finding also is corroborated by the average growth in supply days. While pimozide is a first-generation antipsychotic and had the lowest AAGR, pimozide still was the most prescribed antipsychotic in this study. Considering the average growth in Medicare beneficiaries during the study period was 2.70% per year,2 the AAGR of the 4 other drugs excluding pimozide shows that this growth was larger than what can be attributed to an increase in population size.

The most common conditions for which dermatologists prescribe antipsychotics are primary delusional infestation disorders as well as a range of self-inflicted dermatologic manifestations of dermatitis artefacta.4 Particularly, dermatologist-prescribed antipsychotics are first-line for these conditions in which perception of a persistent disease state is present.4 Importantly, dermatologists must differentiate between other dermatology-related psychiatric conditions such as trichotillomania and body dysmorphic disorder, which tend to respond better to selective serotonin reuptake inhibitors.4 Our data suggest that dermatologists are increasing their utilization of second-generation antipsychotics at a higher rate than first-generation antipsychotics, likely due to the lower risk of extrapyramidal symptoms. Patients are more willing to initiate a trial of psychiatric medication when it is prescribed by a dermatologist vs a psychiatrist due to lack of perceived stigma, which can lead to greater treatment compliance rates.5 As mentioned previously, as part of the differential, dermatologists also can effectively prescribe medications such as selective serotonin reuptake inhibitors for symptoms including anxiety, trichotillomania, body dysmorphic disorder, or secondary psychiatric disorders as a result of the burden of skin disease.5

In many cases, a dermatologist may be the first and only specialist to evaluate patients with conditions that overlap within the jurisdiction of dermatology and psychiatry. It is imperative that dermatologists feel comfortable treating this vulnerable patient population. As demonstrated by Medicare prescription data, the increasing utilization of antipsychotics in our specialty demands that dermatologists possess an adequate working knowledge of psychopharmacology, which may be accomplished during residency training through several directives, including focused didactic sessions, elective rotations in psychiatry, increased exposure to psychocutaneous lectures at national conferences, and finally through the establishment of joint dermatology-psychiatry clinics with interdepartmental collaboration.

References
  1. Weber MB, Recuero JK, Almeida CS. Use of psychiatric drugs in dermatology. An Bras Dermatol. 2020;95:133-143. doi:10.1016/j.abd.2019.12.002
  2. Centers for Medicare & Medicaid Services. Medicare provider utilization and payment data: part D prescriber. Updated September 10, 2024. Accessed October 7, 2024. https://www.cms.gov/data -research/statistics-trends-and-reports/medicare-provider-utilization-payment-data/part-d-prescriber
  3. Bolognia J, Schaffe JV, Lorenzo C. Dermatology. In: Duncan KO, Koo JYM, eds. Psychocutaneous Diseases. Elsevier; 2017:128-136.
  4. Gupta MA, Vujcic B, Pur DR, et al. Use of antipsychotic drugs in dermatology. Clin Dermatol. 2018;36:765-773. doi:10.1016/j.clindermatol.2018.08.006
  5. Jafferany M, Stamu-O’Brien C, Mkhoyan R, et al. Psychotropic drugs in dermatology: a dermatologist’s approach and choice of medications. Dermatol Ther. 2020;33:E13385. doi:10.1111/dth.13385
Article PDF
Author and Disclosure Information

Dr. Maheshwari is from the University of Texas Medical Branch at Galveston. Drs. Wang, Edminister, Haidari, and Feldman are from the Department of Dermatology, Wake Forest University School of Medicine, Winston-Salem, North Carolina. Dr. Wang also is from the Departments of Pathology; Social Sciences and Health Policy; and Dermatology and Allergy Centre, University of Southern Denmark, Odense. Dr. Pang is from the Department of Psychiatry, University of Texas Health, Houston.

Drs. Maheshwari, Wang, Edminister, Haidari, and Pang have no relevant financial disclosures to report. Dr. Feldman is a researcher, speaker, and/or consultant for AbbVie; Advance Medical; Almirall; Boehringer Ingelheim; Celgene; CVS Caremark; Eli Lilly and Company; Galderma; GlaxoSmithKline/Stiefel; Informa; Janssen Pharmaceuticals; LEO Pharma; Merck & Co, Inc; Mylan N.V.; NatBio; National Psoriasis Foundation; Novan Inc; Novartis; Pfizer; Qurient Co; Regeneron Pharmaceuticals; Samsung; Sanofi; Sun Pharmaceutical Industries Ltd; Suncare Research Laboratories, LLC; UpToDate, Inc; and Valeant Pharmaceuticals. Dr. Feldman also is the founder and majority owner of www.DrScore.com as well as the founder and part owner of Causa Research.

Correspondence: Kush Maheshwari, MD, 301 University Blvd, Galveston, TX 77555 ([email protected]).

Cutis. 2024 October;114(4):E2-E4. doi: 10.12788/cutis.1116

Publications
Topics
Page Number
E2-E4
Sections
Author and Disclosure Information

Dr. Maheshwari is from the University of Texas Medical Branch at Galveston. Drs. Wang, Edminister, Haidari, and Feldman are from the Department of Dermatology, Wake Forest University School of Medicine, Winston-Salem, North Carolina. Dr. Wang also is from the Departments of Pathology; Social Sciences and Health Policy; and Dermatology and Allergy Centre, University of Southern Denmark, Odense. Dr. Pang is from the Department of Psychiatry, University of Texas Health, Houston.

Drs. Maheshwari, Wang, Edminister, Haidari, and Pang have no relevant financial disclosures to report. Dr. Feldman is a researcher, speaker, and/or consultant for AbbVie; Advance Medical; Almirall; Boehringer Ingelheim; Celgene; CVS Caremark; Eli Lilly and Company; Galderma; GlaxoSmithKline/Stiefel; Informa; Janssen Pharmaceuticals; LEO Pharma; Merck & Co, Inc; Mylan N.V.; NatBio; National Psoriasis Foundation; Novan Inc; Novartis; Pfizer; Qurient Co; Regeneron Pharmaceuticals; Samsung; Sanofi; Sun Pharmaceutical Industries Ltd; Suncare Research Laboratories, LLC; UpToDate, Inc; and Valeant Pharmaceuticals. Dr. Feldman also is the founder and majority owner of www.DrScore.com as well as the founder and part owner of Causa Research.

Correspondence: Kush Maheshwari, MD, 301 University Blvd, Galveston, TX 77555 ([email protected]).

Cutis. 2024 October;114(4):E2-E4. doi: 10.12788/cutis.1116

Author and Disclosure Information

Dr. Maheshwari is from the University of Texas Medical Branch at Galveston. Drs. Wang, Edminister, Haidari, and Feldman are from the Department of Dermatology, Wake Forest University School of Medicine, Winston-Salem, North Carolina. Dr. Wang also is from the Departments of Pathology; Social Sciences and Health Policy; and Dermatology and Allergy Centre, University of Southern Denmark, Odense. Dr. Pang is from the Department of Psychiatry, University of Texas Health, Houston.

Drs. Maheshwari, Wang, Edminister, Haidari, and Pang have no relevant financial disclosures to report. Dr. Feldman is a researcher, speaker, and/or consultant for AbbVie; Advance Medical; Almirall; Boehringer Ingelheim; Celgene; CVS Caremark; Eli Lilly and Company; Galderma; GlaxoSmithKline/Stiefel; Informa; Janssen Pharmaceuticals; LEO Pharma; Merck & Co, Inc; Mylan N.V.; NatBio; National Psoriasis Foundation; Novan Inc; Novartis; Pfizer; Qurient Co; Regeneron Pharmaceuticals; Samsung; Sanofi; Sun Pharmaceutical Industries Ltd; Suncare Research Laboratories, LLC; UpToDate, Inc; and Valeant Pharmaceuticals. Dr. Feldman also is the founder and majority owner of www.DrScore.com as well as the founder and part owner of Causa Research.

Correspondence: Kush Maheshwari, MD, 301 University Blvd, Galveston, TX 77555 ([email protected]).

Cutis. 2024 October;114(4):E2-E4. doi: 10.12788/cutis.1116

Article PDF
Article PDF

To the Editor:

Patients with primary psychiatric disorders with dermatologic manifestations often seek treatment from dermatologists instead of psychiatrists.1 For example, patients with delusions of parasitosis may lack insight into the underlying etiology of their disease and instead fixate on establishing an organic cause for their symptoms. As a result, it is an increasingly common practice for dermatologists to diagnose and treat psychiatric conditions.1 The goal of this study was to evaluate trends for the top 5 antipsychotics most frequently prescribed by dermatologists in the Medicare Part D database.

In this retrospective analysis, we consulted the Medicare Provider Utilization and Payment Data for January 2013 through December 2020, which is provided to the public by the Centers for Medicare & Medicaid Services.2 Only prescribing data from dermatologists were included in this study by using the built-in filter on the website to select “dermatology” as the prescriber type. All other provider types were excluded. We chose the top 5 most prescribed antipsychotics based on the number of supply days reported. Supply days—defined by Medicare as the number of days’ worth of medication that is prescribed—were used as a metric for ­utilization; therefore, each drug’s total supply days prescribed by dermatologists were calculated using this combined filter of drug name and total supply days using the database.

To analyze utilization over time, the annual average growth rate (AAGR) was calculated by determining the growth rate in total supply days annually from 2013 to 2020 and then averaging those rates to determine the overall AAGR. For greater clinical relevance, we calculated the average growth in supply days for the entire study period by determining the difference in the number of supply days for each year and then averaging these values. This was done to consider overall trends across dermatology rather than individual dermatologist prescribing patterns.

Based on our analysis, the antipsychotics most frequently prescribed by dermatologists for Medicare patients from January 2013 to December 2020 were pimozide, quetiapine, risperidone, olanzapine, and aripiprazole. The AAGR for each drug was 2.35%, 4.89%, 5.59%, 9.48%, and 20.72%, respectively, which is consistent with increased utilization over the study period for all 5 drugs (Table 1). The change in cost per supply day for the same period was 1.3%, 66.1%, 60.2%, 81.7%, and84.3%, respectively. The net difference in cost per supply day over this entire period was $0.02, $2.79, $1.06, $5.37, and $21.22, respectively (Table 2).



There were several limitations to our study. Our analysis was limited to the Medicare population. Uninsured patients and those with Medicare Advantage or private health insurance plans were not included. In the Medicare database, only prescribers who prescribed a medication 10 times or more were recorded; therefore, some prescribers were not captured.

Although there was an increase in the dermatologic use of all 5 drugs in this study, perhaps the most marked growth was exhibited by aripiprazole, which had an AAGR of 20.72% (Table 1). Affordability may have been a factor, as the most marked reduction in price per supply day was noted for aripiprazole during the study period. Pimozide, which traditionally has been the first-line therapy for delusions of parasitosis, is the only first-generation antipsychotic drug among the 5 most frequently prescribed antipsychotics.3 Interestingly, pimozide had the lowest AAGR compared with the 4 second-generation antipsychotics. This finding also is corroborated by the average growth in supply days. While pimozide is a first-generation antipsychotic and had the lowest AAGR, pimozide still was the most prescribed antipsychotic in this study. Considering the average growth in Medicare beneficiaries during the study period was 2.70% per year,2 the AAGR of the 4 other drugs excluding pimozide shows that this growth was larger than what can be attributed to an increase in population size.

The most common conditions for which dermatologists prescribe antipsychotics are primary delusional infestation disorders as well as a range of self-inflicted dermatologic manifestations of dermatitis artefacta.4 Particularly, dermatologist-prescribed antipsychotics are first-line for these conditions in which perception of a persistent disease state is present.4 Importantly, dermatologists must differentiate between other dermatology-related psychiatric conditions such as trichotillomania and body dysmorphic disorder, which tend to respond better to selective serotonin reuptake inhibitors.4 Our data suggest that dermatologists are increasing their utilization of second-generation antipsychotics at a higher rate than first-generation antipsychotics, likely due to the lower risk of extrapyramidal symptoms. Patients are more willing to initiate a trial of psychiatric medication when it is prescribed by a dermatologist vs a psychiatrist due to lack of perceived stigma, which can lead to greater treatment compliance rates.5 As mentioned previously, as part of the differential, dermatologists also can effectively prescribe medications such as selective serotonin reuptake inhibitors for symptoms including anxiety, trichotillomania, body dysmorphic disorder, or secondary psychiatric disorders as a result of the burden of skin disease.5

In many cases, a dermatologist may be the first and only specialist to evaluate patients with conditions that overlap within the jurisdiction of dermatology and psychiatry. It is imperative that dermatologists feel comfortable treating this vulnerable patient population. As demonstrated by Medicare prescription data, the increasing utilization of antipsychotics in our specialty demands that dermatologists possess an adequate working knowledge of psychopharmacology, which may be accomplished during residency training through several directives, including focused didactic sessions, elective rotations in psychiatry, increased exposure to psychocutaneous lectures at national conferences, and finally through the establishment of joint dermatology-psychiatry clinics with interdepartmental collaboration.

To the Editor:

Patients with primary psychiatric disorders with dermatologic manifestations often seek treatment from dermatologists instead of psychiatrists.1 For example, patients with delusions of parasitosis may lack insight into the underlying etiology of their disease and instead fixate on establishing an organic cause for their symptoms. As a result, it is an increasingly common practice for dermatologists to diagnose and treat psychiatric conditions.1 The goal of this study was to evaluate trends for the top 5 antipsychotics most frequently prescribed by dermatologists in the Medicare Part D database.

In this retrospective analysis, we consulted the Medicare Provider Utilization and Payment Data for January 2013 through December 2020, which is provided to the public by the Centers for Medicare & Medicaid Services.2 Only prescribing data from dermatologists were included in this study by using the built-in filter on the website to select “dermatology” as the prescriber type. All other provider types were excluded. We chose the top 5 most prescribed antipsychotics based on the number of supply days reported. Supply days—defined by Medicare as the number of days’ worth of medication that is prescribed—were used as a metric for ­utilization; therefore, each drug’s total supply days prescribed by dermatologists were calculated using this combined filter of drug name and total supply days using the database.

To analyze utilization over time, the annual average growth rate (AAGR) was calculated by determining the growth rate in total supply days annually from 2013 to 2020 and then averaging those rates to determine the overall AAGR. For greater clinical relevance, we calculated the average growth in supply days for the entire study period by determining the difference in the number of supply days for each year and then averaging these values. This was done to consider overall trends across dermatology rather than individual dermatologist prescribing patterns.

Based on our analysis, the antipsychotics most frequently prescribed by dermatologists for Medicare patients from January 2013 to December 2020 were pimozide, quetiapine, risperidone, olanzapine, and aripiprazole. The AAGR for each drug was 2.35%, 4.89%, 5.59%, 9.48%, and 20.72%, respectively, which is consistent with increased utilization over the study period for all 5 drugs (Table 1). The change in cost per supply day for the same period was 1.3%, 66.1%, 60.2%, 81.7%, and84.3%, respectively. The net difference in cost per supply day over this entire period was $0.02, $2.79, $1.06, $5.37, and $21.22, respectively (Table 2).



There were several limitations to our study. Our analysis was limited to the Medicare population. Uninsured patients and those with Medicare Advantage or private health insurance plans were not included. In the Medicare database, only prescribers who prescribed a medication 10 times or more were recorded; therefore, some prescribers were not captured.

Although there was an increase in the dermatologic use of all 5 drugs in this study, perhaps the most marked growth was exhibited by aripiprazole, which had an AAGR of 20.72% (Table 1). Affordability may have been a factor, as the most marked reduction in price per supply day was noted for aripiprazole during the study period. Pimozide, which traditionally has been the first-line therapy for delusions of parasitosis, is the only first-generation antipsychotic drug among the 5 most frequently prescribed antipsychotics.3 Interestingly, pimozide had the lowest AAGR compared with the 4 second-generation antipsychotics. This finding also is corroborated by the average growth in supply days. While pimozide is a first-generation antipsychotic and had the lowest AAGR, pimozide still was the most prescribed antipsychotic in this study. Considering the average growth in Medicare beneficiaries during the study period was 2.70% per year,2 the AAGR of the 4 other drugs excluding pimozide shows that this growth was larger than what can be attributed to an increase in population size.

The most common conditions for which dermatologists prescribe antipsychotics are primary delusional infestation disorders as well as a range of self-inflicted dermatologic manifestations of dermatitis artefacta.4 Particularly, dermatologist-prescribed antipsychotics are first-line for these conditions in which perception of a persistent disease state is present.4 Importantly, dermatologists must differentiate between other dermatology-related psychiatric conditions such as trichotillomania and body dysmorphic disorder, which tend to respond better to selective serotonin reuptake inhibitors.4 Our data suggest that dermatologists are increasing their utilization of second-generation antipsychotics at a higher rate than first-generation antipsychotics, likely due to the lower risk of extrapyramidal symptoms. Patients are more willing to initiate a trial of psychiatric medication when it is prescribed by a dermatologist vs a psychiatrist due to lack of perceived stigma, which can lead to greater treatment compliance rates.5 As mentioned previously, as part of the differential, dermatologists also can effectively prescribe medications such as selective serotonin reuptake inhibitors for symptoms including anxiety, trichotillomania, body dysmorphic disorder, or secondary psychiatric disorders as a result of the burden of skin disease.5

In many cases, a dermatologist may be the first and only specialist to evaluate patients with conditions that overlap within the jurisdiction of dermatology and psychiatry. It is imperative that dermatologists feel comfortable treating this vulnerable patient population. As demonstrated by Medicare prescription data, the increasing utilization of antipsychotics in our specialty demands that dermatologists possess an adequate working knowledge of psychopharmacology, which may be accomplished during residency training through several directives, including focused didactic sessions, elective rotations in psychiatry, increased exposure to psychocutaneous lectures at national conferences, and finally through the establishment of joint dermatology-psychiatry clinics with interdepartmental collaboration.

References
  1. Weber MB, Recuero JK, Almeida CS. Use of psychiatric drugs in dermatology. An Bras Dermatol. 2020;95:133-143. doi:10.1016/j.abd.2019.12.002
  2. Centers for Medicare & Medicaid Services. Medicare provider utilization and payment data: part D prescriber. Updated September 10, 2024. Accessed October 7, 2024. https://www.cms.gov/data -research/statistics-trends-and-reports/medicare-provider-utilization-payment-data/part-d-prescriber
  3. Bolognia J, Schaffe JV, Lorenzo C. Dermatology. In: Duncan KO, Koo JYM, eds. Psychocutaneous Diseases. Elsevier; 2017:128-136.
  4. Gupta MA, Vujcic B, Pur DR, et al. Use of antipsychotic drugs in dermatology. Clin Dermatol. 2018;36:765-773. doi:10.1016/j.clindermatol.2018.08.006
  5. Jafferany M, Stamu-O’Brien C, Mkhoyan R, et al. Psychotropic drugs in dermatology: a dermatologist’s approach and choice of medications. Dermatol Ther. 2020;33:E13385. doi:10.1111/dth.13385
References
  1. Weber MB, Recuero JK, Almeida CS. Use of psychiatric drugs in dermatology. An Bras Dermatol. 2020;95:133-143. doi:10.1016/j.abd.2019.12.002
  2. Centers for Medicare & Medicaid Services. Medicare provider utilization and payment data: part D prescriber. Updated September 10, 2024. Accessed October 7, 2024. https://www.cms.gov/data -research/statistics-trends-and-reports/medicare-provider-utilization-payment-data/part-d-prescriber
  3. Bolognia J, Schaffe JV, Lorenzo C. Dermatology. In: Duncan KO, Koo JYM, eds. Psychocutaneous Diseases. Elsevier; 2017:128-136.
  4. Gupta MA, Vujcic B, Pur DR, et al. Use of antipsychotic drugs in dermatology. Clin Dermatol. 2018;36:765-773. doi:10.1016/j.clindermatol.2018.08.006
  5. Jafferany M, Stamu-O’Brien C, Mkhoyan R, et al. Psychotropic drugs in dermatology: a dermatologist’s approach and choice of medications. Dermatol Ther. 2020;33:E13385. doi:10.1111/dth.13385
Page Number
E2-E4
Page Number
E2-E4
Publications
Publications
Topics
Article Type
Display Headline
Utilization, Cost, and Prescription Trends of Antipsychotics Prescribed by Dermatologists for Medicare Patients
Display Headline
Utilization, Cost, and Prescription Trends of Antipsychotics Prescribed by Dermatologists for Medicare Patients
Sections
Citation Override
Cutis. 2024 October;114(4):E2-E4
Inside the Article

Practice Points

  • Dermatologists are frontline medical providers who can be useful in screening for primary psychiatric disorders in patients with dermatologic manifestations.
  • Second-generation antipsychotics are effective for treating many psychiatric disorders.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

Treat-to-Target Outcomes With Tapinarof Cream 1% in Phase 3 Trials for Plaque Psoriasis

Article Type
Changed
Mon, 10/07/2024 - 09:51

Psoriasis is a chronic inflammatory disease affecting approximately 8 million adults in the United States and 2% of the global population.1,2 Psoriasis causes pain, itching, and disfigurement and is associated with a physical, psychological, and economic burden that substantially affects health-related quality of life.3-5

Setting treatment goals and treating to target are evidence-based approaches that have been successfully applied to several chronic diseases to improve patient outcomes, including diabetes, hypertension, and rheumatoid arthritis.6-9 Treat-to-target strategies generally set low disease activity (or remission) as an overall goal and seek to achieve this using available therapeutic options as necessary. Introduced following the availability of biologics and targeted systemic therapies, treat-to-target strategies generally provide guidance on expectations of treatment but not specific treatments, as personalized treatment decisions depend on an assessment of individual patients and consider clinical and demographic features as well as preferences for available therapeutic options. If targets are not achieved in the assigned time span, adjustments can be made to the treatment approach in close consultation with the patient. If the target is reached, follow-up visits can be scheduled to ensure improvement is maintained or to establish if more aggressive goals could be selected.

Treat-to-target strategies for the management of psoriasis developed by the National Psoriasis Foundation (NPF) Medical Board include reducing the extent of psoriasis to 1% or lower total body surface area (BSA) after 3 months of treatment.10 Treatment targets endorsed by the European Academy of Dermatology and Venereology (EADV) in guidelines on the use of systemic therapies in psoriasis include achieving a 75% or greater reduction in Psoriasis Area and Severity Index (PASI) score within 3 to 4 months of treatment.11

In clinical practice, many patients do not achieve these treatment targets, and topical treatments alone generally are insufficient in achieving treatment goals for psoriasis.12,13 Moreover, conventional topical treatments (eg, topical corticosteroids) used by most patients with psoriasis regardless of disease severity are associated with adverse events that can limit their use. Most topical corticosteroids have US Food and Drug Administration label restrictions relating to sites of application, duration and extent of use, and frequency of administration.14,15

Tapinarof cream 1% (VTAMA [Dermavant Sciences, Inc]) is a first-in-class topical nonsteroidal aryl hydrocarbon receptor agonist that was approved by the US Food and Drug Administration for the treatment of plaque psoriasis in adults16 and is being studied for the treatment of plaque psoriasis in children 2 years and older as well as for atopic dermatitis in adults and children 2 years and older. In PSOARING 1 (ClinicalTrials .gov identifier NCT03956355) and PSOARING 2 (NCT03983980)—identical 12-week pivotal phase 3 trials—monotherapy with tapinarof cream 1% once daily (QD) demonstrated statistically significant efficacy vs vehicle cream and was well tolerated in adults with mild to severe plaque psoriasis (Supplementary Figure S1).17 Lebwohl et al17 reported that significantly higher PASI75 responses were observed at week 12 with tapinarof cream vs vehicle in PSOARING 1 and PSOARING 2 (36% and 48% vs 10% and 7%, respectively; both P<.0001). A significantly higher PASI90 response of 19% and 21% at week 12 also was observed with tapinarof cream vs 2% and 3% with vehicle in PSOARING 1 and PSOARING 2, respectively (P=.0005 and P<.0001).17

In PSOARING 3 (NCT04053387)—the long-term extension trial (Supplementary Figure S1)—efficacy continued to improve or was maintained beyond the two 12-week trials, with improvements in total BSA affected and PASI scores for up to 52 weeks.18 Tapinarof cream 1% QD demonstrated positive, rapid, and durable outcomes in PSOARING 3, including high rates of complete disease clearance (Physician Global Assessment [PGA] score=0 [clear])(40.9% [312/763]), durability of response on treatment with no evidence of tachyphylaxis, and a remittive effect of approximately 4 months when off therapy (defined as maintenance of a PGA score of 0 [clear] or 1 [almost clear] after first achieving a PGA score of 0).18

Herein, we report absolute treatment targets for patients with plaque psoriasis who received tapinarof cream 1% QD in the PSOARING trials that are at least as stringent as the corresponding NPF and EADV targets of achieving a total BSA affected of 1% or lower or a PASI75 response within 3 to 4 months, respectively.

 

 

METHODS

Study Design

The pooled efficacy analyses included all patients with a baseline PGA score of 2 or higher (mild or worse) before treatment with tapinarof cream 1% QD in the PSOARING trials. This included patients who received tapinarof cream 1% in PSOARING 1 and PSOARING 2 who may or may not have continued into PSOARING 3, as well as those who received the vehicle in PSOARING 1 and PSOARING 2 who enrolled in PSOARING 3 and had a PGA score of 2 or higher before receiving tapinarof cream 1%.

Trial Participants

Full methods, including inclusion and exclusion criteria, for the PSOARING trials have been previously reported.17,18 Patients were aged 18 to 75 years and had chronic plaque psoriasis that was stable for at least 6 months before randomization; 3% to 20% total BSA affected (excluding the scalp, palms, fingernails, toenails, and soles); and a PGA score of 2 (mild), 3 (moderate), or 4 (severe) at baseline.

The clinical trials were conducted in compliance with the guidelines for Good Clinical Practice and the Declaration of Helsinki. Approval was obtained from local ethics committees or institutional review boards at each center. All patients provided written informed consent.

Trial Treatment

In PSOARING 1 and PSOARING 2, patients were randomized (2:1) to receive tapinarof cream 1% or vehicle QD for 12 weeks. In PSOARING 3 (the long-term extension trial), patients received up to 40 weeks of open-label tapinarof, followed by 4 weeks of follow-up off treatment. Patients received intermittent or continuous treatment with tapinarof cream 1% in PSOARING 3 based on PGA score: those entering the trial with a PGA score of 1 or higher received tapinarof cream 1% until complete disease clearance was achieved (defined as a PGA score of 0 [clear]). Those entering PSOARING 3 with or achieving a PGA score of 0 (clear) discontinued treatment and were observed for the duration of maintenance of a PGA score of 0 (clear) or 1 (almost clear) while off therapy (the protocol-defined “duration of remittive effect”). If disease worsening (defined as a PGA score 2 or higher) occurred, tapinarof cream 1% was restarted and continued until a PGA score of 0 (clear) was achieved. This pattern of treatment, discontinuation on achieving a PGA score of 0 (clear), and retreatment on disease worsening continued until the end of the trial. As a result, patients in PSOARING 3 could receive tapinarof cream 1% continuously or intermittently for 40 weeks.

Outcome Measures and Statistical Analyses

The assessment of total BSA affected by plaque psoriasis is an estimate of the total extent of disease as a percentage of total skin area. In the PSOARING trials, the skin surface of one hand (palm and digits) was assumed to be approximately equivalent to 1% BSA. The total BSA affected by psoriasis was evaluated from 0% to 100%, with greater total BSA affected being an indication of more extensive disease. The BSA efficacy outcomes used in these analyses were based post hoc on the proportion of patients who achieved a 1% or lower or 0.5% or lower total BSA affected. The smallest BSA affected increment that investigators were trained to measure and could record was 0.1%.

 

 

Psoriasis Area and Severity Index scores assess both the severity and extent of psoriasis. A PASI score lower than 5 often is considered indicative of mild psoriasis, a score of 5 to 10 indicates moderate disease, and a score higher than 10 indicates severe disease.19 The maximum PASI score is 72. The PASI efficacy outcomes used in these analyses were based post hoc on the proportion of patients who achieved an absolute total PASI score of 3 or lower, 2 or lower, and 1 or lower.

Efficacy analyses were based on pooled data for all patients in the PSOARING trials who had a PGA score of 2 to 4 (mild to severe) before treatment with tapinarof cream 1% in the intention-to-treat population using observed cases. Time-to-target analyses were based on Kaplan-Meier (KM) estimates using observed cases.

Safety analyses included the incidence and frequency of adverse events and were based on all patients who received tapinarof cream 1% in the PSOARING trials.

RESULTS

Baseline Patient Demographics and Disease Characteristics

The pooled efficacy analyses included 915 eligible patients (Table). At baseline, the mean (SD) age was 50.2 (13.25) years, 58.7% were male, the mean (SD) weight was 92.2 (23.67) kg, and the mean (SD) body mass index was 31.6 (7.53) kg/m2. The percentage of patients with a PGA score of 2 (mild), 3 (moderate), or 4 (severe) was 13.9%, 78.1%, and 8.0%, respectively. The mean (SD) PASI score was 8.7 (4.23) and mean (SD) total BSA affected was 7.8% (4.98).

Efficacy

Achievement of BSA-Affected Targets—The NPF-recommended target of 1% or lower total BSA affected within 3 months was achieved by 40% of patients (KM estimate [95% CI, 37%-43%])(Figure 1). Across the total trial period of up to 52 weeks, a total BSA affected of 1% or lower was achieved by 61% of patients (561/915), with the median time to target of approximately 4 months (KM estimate: 120 days [95% CI, 113-141])(Supplementary Figure S2a). Approximately 50% of patients (455/915) achieved a total BSA affected of 0.5% or lower, with a median time to target of 199 days (KM estimate [95% CI, 172-228)(Figure 1; Supplementary Figure S2b).

FIGURE 1. Pooled analysis of total body surface area (BSA) affected targets achieved by patients with mild to severe plaque psoriasis treated with tapinarof cream 1% once daily (QD) across a trial period up to 52 weeks in PSOARING 1, PSOARING 2, and PSOARING 3 (target total BSA affected, ≤1% [National Psoriasis Foundation [NPF]−recommended target]; target total BSA affected, ≤.5%)(N=915). These analyses included patients receiving continuous or intermittent tapinarof monotherapy in the 12-week pivotal trials (PSOARING 1 and PSOARING 2) and in the forced-withdrawal design of PSOARING 3 (treatment was stopped when patients achieved a Physician Global Assessment score of 0).

FIGURE 2. Total Psoriasis Area and Severity Index (PASI) score targets achieved by patients with mild to severe plaque psoriasis treated with tapinarof cream 1% once daily across a trial period up to 52 weeks in PSOARING 1, PSOARING 2 (target PASI score), and PSOARING 3 (target PASI score ≤3, ≤2, and ≤1)(N=915). These analyses included patients receiving continuous or intermittent tapinarof monotherapy in the 12-week pivotal trials (PSOARING 1 and PSOARING 2) and in the forced-withdrawal design of PSOARING 3 (treatment was stopped when patients achieved a Physician Global Assessment score of 0).

Achievement of Absolute PASI Targets—Across the total trial period (up to 52 weeks), an absolute total PASI score of 3 or lower was achieved by 75% of patients (686/915), with a median time to achieve this of 2 months (KM estimate: 58 days [95% CI, 57-63]); approximately 67% of patients (612/915) achieved a total PASI score of 2 or lower, with a median time to achieve of 3 months (KM estimate: 87 days [95% CI, 85-110])(Figure 2; Supplementary Figures S3a and S3b). A PASI score of 1 or lower was achieved by approximately 50% of patients (460/915), with a median time to achieve of approximately 6 months (KM estimate: 185 days [95% CI, 169-218])(Figure 2, Supplementary Figure S3c).

Illustrative Case—Case photography showing the clinical response in a 63-year-old man with moderate plaque psoriasis in PSOARING 2 is shown in Figure 3. After 12 weeks of treatment with tapinarof cream 1% QD, the patient achieved all primary and secondary efficacy end points. In addition to achieving the regulatory end point of a PGA score of 0 (clear) or 1 (almost clear) and a decrease from baseline of at least 2 points, achievement of 0% total BSA affected and a total PASI score of 0 at week 12 exceeded the NPF and EADV consensus treatment targets.10,11 Targets were achieved as early as week 4, with a total BSA affected of 0.5% or lower and a total PASI score of 1 or lower, illustrated by marked skin clearing and only faint residual erythema that completely resolved at week 12, with the absence of postinflammatory hyperpigmentation.

 

 

Safety

Safety data for the PSOARING trials have been previously reported.17,18 The most common treatment-emergent adverse events were folliculitis, contact dermatitis, upper respiratory tract infection, and nasopharyngitis. Treatment-emergent adverse events generally were mild or moderate in severity and did not lead to trial discontinuation.17,18

FIGURE 3. Moderate plaque psoriasis on the abdomen in a patient treated with tapinarof cream 1% once daily in PSOARING 2 who achieved the primary end point at week 4. A, At baseline, wellcircumscribed erythematous patches, plaques, and scaling were visible. B, The patient achieved the primary end point and National Psoriasis Foundation (NPF) and European Academy of Dermatology and Venereology (EADV) treatment targets by week 4, at which point there was marked clearing with faint residual erythema C, By week 12, the patient had 0% total body surface area affected and a total Psoriasis Area and Severity Index score of 0, exceeding NPF/EADV consensus treatment targets. Faint residual erythema completely resolved with the absence of postinflammatory hyperpigmentation.

COMMENT

Treat-to-target management approaches aim to improve patient outcomes by striving to achieve optimal goals. The treat-to-target approach supports shared decision-making between clinicians and patients based on common expectations of what constitutes treatment success.

The findings of this analysis based on pooled data from a large cohort of patients demonstrate that a high proportion of patients can achieve or exceed recommended treatment targets with tapinarof cream 1% QD and maintain improvements long-term. The NPF-recommended treatment target of 1% or lower BSA affected within approximately 3 months (90 days) of treatment was achieved by 40% of tapinarof-treated patients. In addition, 1% or lower BSA affected at any time during the trials was achieved by 61% of patients (median, approximately 4 months). The analyses also indicated that PASI total scores of 3 or lower and 2 or lower were achieved by 75% and 67% of tapinarof-treated patients, respectively, within 2 to 3 months.

These findings support the previously reported efficacy of tapinarof cream, including high rates of complete disease clearance (40.9% [312/763]), durable response following treatment interruption, an off-therapy remittive effect of approximately 4 months, and good disease control on therapy with no evidence of tachyphylaxis.17,18

CONCLUSION

Taken together with previously reported tapinarof efficacy and safety results, our findings demonstrate that a high proportion of patients treated with tapinarof cream as monotherapy can achieve aggressive treatment targets set by both US and European guidelines developed for systemic and biologic therapies. Tapinarof cream 1% QD is an effective topical treatment option for patients with plaque psoriasis that has been approved without restrictions relating to severity or extent of disease treated, duration of use, or application sites, including application to sensitive and intertriginous skin.

Acknowledgments—Editorial and medical writing support under the guidance of the authors was provided by Melanie Govender, MSc (Med), ApotheCom (United Kingdom), and was funded by Dermavant Sciences, Inc, in accordance with Good Publication Practice (GPP) guidelines.

Files
References
  1. Armstrong AW, Mehta MD, Schupp CW, et al. Psoriasis prevalence in adults in the United States. JAMA Dermatol. 2021;157:940-946.
  2. Parisi R, Iskandar IYK, Kontopantelis E, et al. National, regional, and worldwide epidemiology of psoriasis: systematic analysis and modelling study. BMJ. 2020;369:m1590.
  3. Pilon D, Teeple A, Zhdanava M, et al. The economic burden of psoriasis with high comorbidity among privately insured patients in the United States. J Med Econ. 2019;22:196-203.
  4. Singh S, Taylor C, Kornmehl H, et al. Psoriasis and suicidality: a systematic review and meta-analysis. J Am Acad Dermatol. 2017;77:425-440.e2.
  5. Feldman SR, Goffe B, Rice G, et al. The challenge of managing psoriasis: unmet medical needs and stakeholder perspectives. Am Health Drug Benefits. 2016;9:504-513.
  6. Ford JA, Solomon DH. Challenges in implementing treat-to-target strategies in rheumatology. Rheum Dis Clin North Am. 2019;45:101-112.
  7. Sitbon O, Galiè N. Treat-to-target strategies in pulmonary arterial hypertension: the importance of using multiple goals. Eur Respir Rev. 2010;19:272-278.
  8. Smolen JS, Aletaha D, Bijlsma JW, et al. Treating rheumatoid arthritis to target: recommendations of an international task force. Ann Rheum Dis. 2010;69:631-637.
  9. Wangnoo SK, Sethi B, Sahay RK, et al. Treat-to-target trials in diabetes. Indian J Endocrinol Metab. 2014;18:166-174.
  10. Armstrong AW, Siegel MP, Bagel J, et al. From the Medical Board of the National Psoriasis Foundation: treatment targets for plaque psoriasis. J Am Acad Dermatol. 2017;76:290-298.
  11. Pathirana D, Ormerod AD, Saiag P, et al. European S3-guidelines on the systemic treatment of psoriasis vulgaris. J Eur Acad Dermatol Venereol. 2009;23(Suppl 2):1-70.
  12. Strober BE, van der Walt JM, Armstrong AW, et al. Clinical goals and barriers to effective psoriasis care. Dermatol Ther (Heidelb). 2019; 9:5-18.
  13. Bagel J, Gold LS. Combining topical psoriasis treatment to enhance systemic and phototherapy: a review of the literature. J Drugs Dermatol. 2017;16:1209-1222.
  14. Elmets CA, Korman NJ, Prater EF, et al. Joint AAD-NPF Guidelines of care for the management and treatment of psoriasis with topical therapy and alternative medicine modalities for psoriasis severity measures. J Am Acad Dermatol. 2021;84:432-470.
  15. Stein Gold LF. Topical therapies for psoriasis: improving management strategies and patient adherence. Semin Cutan Med Surg. 2016;35 (2 Suppl 2):S36-S44; quiz S45.
  16. VTAMA® (tapinarof) cream. Prescribing information. Dermavant Sciences; 2022. Accessed September 13, 2024. https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/215272s000lbl.pdf
  17. Lebwohl MG, Stein Gold L, Strober B, et al. Phase 3 trials of tapinarof cream for plaque psoriasis. N Engl J Med. 2021;385:2219-2229 and supplementary appendix.
  18. Strober B, Stein Gold L, Bissonnette R, et al. One-year safety and efficacy of tapinarof cream for the treatment of plaque psoriasis: results from the PSOARING 3 trial. J Am Acad Dermatol. 2022;87:800-806.
  19. Clinical Review Report: Guselkumab (Tremfya) [Internet]. Canadian Agency for Drugs and Technologies in Health; 2018. Accessed September 13, 2024. https://www.ncbi.nlm.nih.gov/books/NBK534047/pdf/Bookshelf_NBK534047.pdf
Article PDF
Author and Disclosure Information

 

Dr. Armstrong is from the Division of Dermatology, University of California Los Angeles. Dr. Bissonnette is from Innovaderm Research Inc, Montreal, Quebec, Canada. Dr. Chovatiya is from Chicago Medical School, Rosalind Franklin University of Medicine and Science, Illinois, and the Center for Medical Dermatology and Immunology Research, Chicago. Dr. Bhutani is from the Department of Dermatology, University of California, San Francisco. Drs. Brown and Tallman are from Dermavant Sciences, Inc, Morrisville, North Carolina. Dr. Papp is from Probity Medical Research Inc and Alliance Clinical Trials, Waterloo, Ontario, Canada, and the University of Toronto, Ontario.

Several of the authors have relevant financial disclosures to report. Due to their length, the disclosures are listed in their entirety in the Appendix online at www.mdedge.com/dermatology.

This study was funded by Dermavant Sciences, Inc.

Supplemental information—Supplementary Figures S1-S3—is available online at www.mdedge.com/dermatology. This material has been provided by the authors to give readers additional information about their work.

Trial registration with the following ClinicalTrials.gov identifiers: NCT03956355, NCT03983980, and NCT04053387.

ORCID: April W. Armstrong, MD, MPH: 0000-0003-0064-8707; Robert Bissonnette, MD: 0000-0001-5927-6587; Raj Chovatiya, MD, PhD: 0000-0001-6510-399X; Tina Bhutani, MD: 0000-0001-8187-1024; Anna M. Tallman, PharmD: 0000-0001-9535-0414; Kim A. Papp, MD, PhD: 0000-0001-9557-3642.

Correspondence: April W. Armstrong, MD, MPH, University of California Los Angeles, 405 Hilgard Ave, Los Angeles, CA 90095 ([email protected]).

Cutis. 2024 October;114(4):122-127, E1. doi:10.12788/cutis.1112

Issue
Cutis - 114(4)
Publications
Topics
Page Number
122-127
Sections
Files
Files
Author and Disclosure Information

 

Dr. Armstrong is from the Division of Dermatology, University of California Los Angeles. Dr. Bissonnette is from Innovaderm Research Inc, Montreal, Quebec, Canada. Dr. Chovatiya is from Chicago Medical School, Rosalind Franklin University of Medicine and Science, Illinois, and the Center for Medical Dermatology and Immunology Research, Chicago. Dr. Bhutani is from the Department of Dermatology, University of California, San Francisco. Drs. Brown and Tallman are from Dermavant Sciences, Inc, Morrisville, North Carolina. Dr. Papp is from Probity Medical Research Inc and Alliance Clinical Trials, Waterloo, Ontario, Canada, and the University of Toronto, Ontario.

Several of the authors have relevant financial disclosures to report. Due to their length, the disclosures are listed in their entirety in the Appendix online at www.mdedge.com/dermatology.

This study was funded by Dermavant Sciences, Inc.

Supplemental information—Supplementary Figures S1-S3—is available online at www.mdedge.com/dermatology. This material has been provided by the authors to give readers additional information about their work.

Trial registration with the following ClinicalTrials.gov identifiers: NCT03956355, NCT03983980, and NCT04053387.

ORCID: April W. Armstrong, MD, MPH: 0000-0003-0064-8707; Robert Bissonnette, MD: 0000-0001-5927-6587; Raj Chovatiya, MD, PhD: 0000-0001-6510-399X; Tina Bhutani, MD: 0000-0001-8187-1024; Anna M. Tallman, PharmD: 0000-0001-9535-0414; Kim A. Papp, MD, PhD: 0000-0001-9557-3642.

Correspondence: April W. Armstrong, MD, MPH, University of California Los Angeles, 405 Hilgard Ave, Los Angeles, CA 90095 ([email protected]).

Cutis. 2024 October;114(4):122-127, E1. doi:10.12788/cutis.1112

Author and Disclosure Information

 

Dr. Armstrong is from the Division of Dermatology, University of California Los Angeles. Dr. Bissonnette is from Innovaderm Research Inc, Montreal, Quebec, Canada. Dr. Chovatiya is from Chicago Medical School, Rosalind Franklin University of Medicine and Science, Illinois, and the Center for Medical Dermatology and Immunology Research, Chicago. Dr. Bhutani is from the Department of Dermatology, University of California, San Francisco. Drs. Brown and Tallman are from Dermavant Sciences, Inc, Morrisville, North Carolina. Dr. Papp is from Probity Medical Research Inc and Alliance Clinical Trials, Waterloo, Ontario, Canada, and the University of Toronto, Ontario.

Several of the authors have relevant financial disclosures to report. Due to their length, the disclosures are listed in their entirety in the Appendix online at www.mdedge.com/dermatology.

This study was funded by Dermavant Sciences, Inc.

Supplemental information—Supplementary Figures S1-S3—is available online at www.mdedge.com/dermatology. This material has been provided by the authors to give readers additional information about their work.

Trial registration with the following ClinicalTrials.gov identifiers: NCT03956355, NCT03983980, and NCT04053387.

ORCID: April W. Armstrong, MD, MPH: 0000-0003-0064-8707; Robert Bissonnette, MD: 0000-0001-5927-6587; Raj Chovatiya, MD, PhD: 0000-0001-6510-399X; Tina Bhutani, MD: 0000-0001-8187-1024; Anna M. Tallman, PharmD: 0000-0001-9535-0414; Kim A. Papp, MD, PhD: 0000-0001-9557-3642.

Correspondence: April W. Armstrong, MD, MPH, University of California Los Angeles, 405 Hilgard Ave, Los Angeles, CA 90095 ([email protected]).

Cutis. 2024 October;114(4):122-127, E1. doi:10.12788/cutis.1112

Article PDF
Article PDF

Psoriasis is a chronic inflammatory disease affecting approximately 8 million adults in the United States and 2% of the global population.1,2 Psoriasis causes pain, itching, and disfigurement and is associated with a physical, psychological, and economic burden that substantially affects health-related quality of life.3-5

Setting treatment goals and treating to target are evidence-based approaches that have been successfully applied to several chronic diseases to improve patient outcomes, including diabetes, hypertension, and rheumatoid arthritis.6-9 Treat-to-target strategies generally set low disease activity (or remission) as an overall goal and seek to achieve this using available therapeutic options as necessary. Introduced following the availability of biologics and targeted systemic therapies, treat-to-target strategies generally provide guidance on expectations of treatment but not specific treatments, as personalized treatment decisions depend on an assessment of individual patients and consider clinical and demographic features as well as preferences for available therapeutic options. If targets are not achieved in the assigned time span, adjustments can be made to the treatment approach in close consultation with the patient. If the target is reached, follow-up visits can be scheduled to ensure improvement is maintained or to establish if more aggressive goals could be selected.

Treat-to-target strategies for the management of psoriasis developed by the National Psoriasis Foundation (NPF) Medical Board include reducing the extent of psoriasis to 1% or lower total body surface area (BSA) after 3 months of treatment.10 Treatment targets endorsed by the European Academy of Dermatology and Venereology (EADV) in guidelines on the use of systemic therapies in psoriasis include achieving a 75% or greater reduction in Psoriasis Area and Severity Index (PASI) score within 3 to 4 months of treatment.11

In clinical practice, many patients do not achieve these treatment targets, and topical treatments alone generally are insufficient in achieving treatment goals for psoriasis.12,13 Moreover, conventional topical treatments (eg, topical corticosteroids) used by most patients with psoriasis regardless of disease severity are associated with adverse events that can limit their use. Most topical corticosteroids have US Food and Drug Administration label restrictions relating to sites of application, duration and extent of use, and frequency of administration.14,15

Tapinarof cream 1% (VTAMA [Dermavant Sciences, Inc]) is a first-in-class topical nonsteroidal aryl hydrocarbon receptor agonist that was approved by the US Food and Drug Administration for the treatment of plaque psoriasis in adults16 and is being studied for the treatment of plaque psoriasis in children 2 years and older as well as for atopic dermatitis in adults and children 2 years and older. In PSOARING 1 (ClinicalTrials .gov identifier NCT03956355) and PSOARING 2 (NCT03983980)—identical 12-week pivotal phase 3 trials—monotherapy with tapinarof cream 1% once daily (QD) demonstrated statistically significant efficacy vs vehicle cream and was well tolerated in adults with mild to severe plaque psoriasis (Supplementary Figure S1).17 Lebwohl et al17 reported that significantly higher PASI75 responses were observed at week 12 with tapinarof cream vs vehicle in PSOARING 1 and PSOARING 2 (36% and 48% vs 10% and 7%, respectively; both P<.0001). A significantly higher PASI90 response of 19% and 21% at week 12 also was observed with tapinarof cream vs 2% and 3% with vehicle in PSOARING 1 and PSOARING 2, respectively (P=.0005 and P<.0001).17

In PSOARING 3 (NCT04053387)—the long-term extension trial (Supplementary Figure S1)—efficacy continued to improve or was maintained beyond the two 12-week trials, with improvements in total BSA affected and PASI scores for up to 52 weeks.18 Tapinarof cream 1% QD demonstrated positive, rapid, and durable outcomes in PSOARING 3, including high rates of complete disease clearance (Physician Global Assessment [PGA] score=0 [clear])(40.9% [312/763]), durability of response on treatment with no evidence of tachyphylaxis, and a remittive effect of approximately 4 months when off therapy (defined as maintenance of a PGA score of 0 [clear] or 1 [almost clear] after first achieving a PGA score of 0).18

Herein, we report absolute treatment targets for patients with plaque psoriasis who received tapinarof cream 1% QD in the PSOARING trials that are at least as stringent as the corresponding NPF and EADV targets of achieving a total BSA affected of 1% or lower or a PASI75 response within 3 to 4 months, respectively.

 

 

METHODS

Study Design

The pooled efficacy analyses included all patients with a baseline PGA score of 2 or higher (mild or worse) before treatment with tapinarof cream 1% QD in the PSOARING trials. This included patients who received tapinarof cream 1% in PSOARING 1 and PSOARING 2 who may or may not have continued into PSOARING 3, as well as those who received the vehicle in PSOARING 1 and PSOARING 2 who enrolled in PSOARING 3 and had a PGA score of 2 or higher before receiving tapinarof cream 1%.

Trial Participants

Full methods, including inclusion and exclusion criteria, for the PSOARING trials have been previously reported.17,18 Patients were aged 18 to 75 years and had chronic plaque psoriasis that was stable for at least 6 months before randomization; 3% to 20% total BSA affected (excluding the scalp, palms, fingernails, toenails, and soles); and a PGA score of 2 (mild), 3 (moderate), or 4 (severe) at baseline.

The clinical trials were conducted in compliance with the guidelines for Good Clinical Practice and the Declaration of Helsinki. Approval was obtained from local ethics committees or institutional review boards at each center. All patients provided written informed consent.

Trial Treatment

In PSOARING 1 and PSOARING 2, patients were randomized (2:1) to receive tapinarof cream 1% or vehicle QD for 12 weeks. In PSOARING 3 (the long-term extension trial), patients received up to 40 weeks of open-label tapinarof, followed by 4 weeks of follow-up off treatment. Patients received intermittent or continuous treatment with tapinarof cream 1% in PSOARING 3 based on PGA score: those entering the trial with a PGA score of 1 or higher received tapinarof cream 1% until complete disease clearance was achieved (defined as a PGA score of 0 [clear]). Those entering PSOARING 3 with or achieving a PGA score of 0 (clear) discontinued treatment and were observed for the duration of maintenance of a PGA score of 0 (clear) or 1 (almost clear) while off therapy (the protocol-defined “duration of remittive effect”). If disease worsening (defined as a PGA score 2 or higher) occurred, tapinarof cream 1% was restarted and continued until a PGA score of 0 (clear) was achieved. This pattern of treatment, discontinuation on achieving a PGA score of 0 (clear), and retreatment on disease worsening continued until the end of the trial. As a result, patients in PSOARING 3 could receive tapinarof cream 1% continuously or intermittently for 40 weeks.

Outcome Measures and Statistical Analyses

The assessment of total BSA affected by plaque psoriasis is an estimate of the total extent of disease as a percentage of total skin area. In the PSOARING trials, the skin surface of one hand (palm and digits) was assumed to be approximately equivalent to 1% BSA. The total BSA affected by psoriasis was evaluated from 0% to 100%, with greater total BSA affected being an indication of more extensive disease. The BSA efficacy outcomes used in these analyses were based post hoc on the proportion of patients who achieved a 1% or lower or 0.5% or lower total BSA affected. The smallest BSA affected increment that investigators were trained to measure and could record was 0.1%.

 

 

Psoriasis Area and Severity Index scores assess both the severity and extent of psoriasis. A PASI score lower than 5 often is considered indicative of mild psoriasis, a score of 5 to 10 indicates moderate disease, and a score higher than 10 indicates severe disease.19 The maximum PASI score is 72. The PASI efficacy outcomes used in these analyses were based post hoc on the proportion of patients who achieved an absolute total PASI score of 3 or lower, 2 or lower, and 1 or lower.

Efficacy analyses were based on pooled data for all patients in the PSOARING trials who had a PGA score of 2 to 4 (mild to severe) before treatment with tapinarof cream 1% in the intention-to-treat population using observed cases. Time-to-target analyses were based on Kaplan-Meier (KM) estimates using observed cases.

Safety analyses included the incidence and frequency of adverse events and were based on all patients who received tapinarof cream 1% in the PSOARING trials.

RESULTS

Baseline Patient Demographics and Disease Characteristics

The pooled efficacy analyses included 915 eligible patients (Table). At baseline, the mean (SD) age was 50.2 (13.25) years, 58.7% were male, the mean (SD) weight was 92.2 (23.67) kg, and the mean (SD) body mass index was 31.6 (7.53) kg/m2. The percentage of patients with a PGA score of 2 (mild), 3 (moderate), or 4 (severe) was 13.9%, 78.1%, and 8.0%, respectively. The mean (SD) PASI score was 8.7 (4.23) and mean (SD) total BSA affected was 7.8% (4.98).

Efficacy

Achievement of BSA-Affected Targets—The NPF-recommended target of 1% or lower total BSA affected within 3 months was achieved by 40% of patients (KM estimate [95% CI, 37%-43%])(Figure 1). Across the total trial period of up to 52 weeks, a total BSA affected of 1% or lower was achieved by 61% of patients (561/915), with the median time to target of approximately 4 months (KM estimate: 120 days [95% CI, 113-141])(Supplementary Figure S2a). Approximately 50% of patients (455/915) achieved a total BSA affected of 0.5% or lower, with a median time to target of 199 days (KM estimate [95% CI, 172-228)(Figure 1; Supplementary Figure S2b).

FIGURE 1. Pooled analysis of total body surface area (BSA) affected targets achieved by patients with mild to severe plaque psoriasis treated with tapinarof cream 1% once daily (QD) across a trial period up to 52 weeks in PSOARING 1, PSOARING 2, and PSOARING 3 (target total BSA affected, ≤1% [National Psoriasis Foundation [NPF]−recommended target]; target total BSA affected, ≤.5%)(N=915). These analyses included patients receiving continuous or intermittent tapinarof monotherapy in the 12-week pivotal trials (PSOARING 1 and PSOARING 2) and in the forced-withdrawal design of PSOARING 3 (treatment was stopped when patients achieved a Physician Global Assessment score of 0).

FIGURE 2. Total Psoriasis Area and Severity Index (PASI) score targets achieved by patients with mild to severe plaque psoriasis treated with tapinarof cream 1% once daily across a trial period up to 52 weeks in PSOARING 1, PSOARING 2 (target PASI score), and PSOARING 3 (target PASI score ≤3, ≤2, and ≤1)(N=915). These analyses included patients receiving continuous or intermittent tapinarof monotherapy in the 12-week pivotal trials (PSOARING 1 and PSOARING 2) and in the forced-withdrawal design of PSOARING 3 (treatment was stopped when patients achieved a Physician Global Assessment score of 0).

Achievement of Absolute PASI Targets—Across the total trial period (up to 52 weeks), an absolute total PASI score of 3 or lower was achieved by 75% of patients (686/915), with a median time to achieve this of 2 months (KM estimate: 58 days [95% CI, 57-63]); approximately 67% of patients (612/915) achieved a total PASI score of 2 or lower, with a median time to achieve of 3 months (KM estimate: 87 days [95% CI, 85-110])(Figure 2; Supplementary Figures S3a and S3b). A PASI score of 1 or lower was achieved by approximately 50% of patients (460/915), with a median time to achieve of approximately 6 months (KM estimate: 185 days [95% CI, 169-218])(Figure 2, Supplementary Figure S3c).

Illustrative Case—Case photography showing the clinical response in a 63-year-old man with moderate plaque psoriasis in PSOARING 2 is shown in Figure 3. After 12 weeks of treatment with tapinarof cream 1% QD, the patient achieved all primary and secondary efficacy end points. In addition to achieving the regulatory end point of a PGA score of 0 (clear) or 1 (almost clear) and a decrease from baseline of at least 2 points, achievement of 0% total BSA affected and a total PASI score of 0 at week 12 exceeded the NPF and EADV consensus treatment targets.10,11 Targets were achieved as early as week 4, with a total BSA affected of 0.5% or lower and a total PASI score of 1 or lower, illustrated by marked skin clearing and only faint residual erythema that completely resolved at week 12, with the absence of postinflammatory hyperpigmentation.

 

 

Safety

Safety data for the PSOARING trials have been previously reported.17,18 The most common treatment-emergent adverse events were folliculitis, contact dermatitis, upper respiratory tract infection, and nasopharyngitis. Treatment-emergent adverse events generally were mild or moderate in severity and did not lead to trial discontinuation.17,18

FIGURE 3. Moderate plaque psoriasis on the abdomen in a patient treated with tapinarof cream 1% once daily in PSOARING 2 who achieved the primary end point at week 4. A, At baseline, wellcircumscribed erythematous patches, plaques, and scaling were visible. B, The patient achieved the primary end point and National Psoriasis Foundation (NPF) and European Academy of Dermatology and Venereology (EADV) treatment targets by week 4, at which point there was marked clearing with faint residual erythema C, By week 12, the patient had 0% total body surface area affected and a total Psoriasis Area and Severity Index score of 0, exceeding NPF/EADV consensus treatment targets. Faint residual erythema completely resolved with the absence of postinflammatory hyperpigmentation.

COMMENT

Treat-to-target management approaches aim to improve patient outcomes by striving to achieve optimal goals. The treat-to-target approach supports shared decision-making between clinicians and patients based on common expectations of what constitutes treatment success.

The findings of this analysis based on pooled data from a large cohort of patients demonstrate that a high proportion of patients can achieve or exceed recommended treatment targets with tapinarof cream 1% QD and maintain improvements long-term. The NPF-recommended treatment target of 1% or lower BSA affected within approximately 3 months (90 days) of treatment was achieved by 40% of tapinarof-treated patients. In addition, 1% or lower BSA affected at any time during the trials was achieved by 61% of patients (median, approximately 4 months). The analyses also indicated that PASI total scores of 3 or lower and 2 or lower were achieved by 75% and 67% of tapinarof-treated patients, respectively, within 2 to 3 months.

These findings support the previously reported efficacy of tapinarof cream, including high rates of complete disease clearance (40.9% [312/763]), durable response following treatment interruption, an off-therapy remittive effect of approximately 4 months, and good disease control on therapy with no evidence of tachyphylaxis.17,18

CONCLUSION

Taken together with previously reported tapinarof efficacy and safety results, our findings demonstrate that a high proportion of patients treated with tapinarof cream as monotherapy can achieve aggressive treatment targets set by both US and European guidelines developed for systemic and biologic therapies. Tapinarof cream 1% QD is an effective topical treatment option for patients with plaque psoriasis that has been approved without restrictions relating to severity or extent of disease treated, duration of use, or application sites, including application to sensitive and intertriginous skin.

Acknowledgments—Editorial and medical writing support under the guidance of the authors was provided by Melanie Govender, MSc (Med), ApotheCom (United Kingdom), and was funded by Dermavant Sciences, Inc, in accordance with Good Publication Practice (GPP) guidelines.

Psoriasis is a chronic inflammatory disease affecting approximately 8 million adults in the United States and 2% of the global population.1,2 Psoriasis causes pain, itching, and disfigurement and is associated with a physical, psychological, and economic burden that substantially affects health-related quality of life.3-5

Setting treatment goals and treating to target are evidence-based approaches that have been successfully applied to several chronic diseases to improve patient outcomes, including diabetes, hypertension, and rheumatoid arthritis.6-9 Treat-to-target strategies generally set low disease activity (or remission) as an overall goal and seek to achieve this using available therapeutic options as necessary. Introduced following the availability of biologics and targeted systemic therapies, treat-to-target strategies generally provide guidance on expectations of treatment but not specific treatments, as personalized treatment decisions depend on an assessment of individual patients and consider clinical and demographic features as well as preferences for available therapeutic options. If targets are not achieved in the assigned time span, adjustments can be made to the treatment approach in close consultation with the patient. If the target is reached, follow-up visits can be scheduled to ensure improvement is maintained or to establish if more aggressive goals could be selected.

Treat-to-target strategies for the management of psoriasis developed by the National Psoriasis Foundation (NPF) Medical Board include reducing the extent of psoriasis to 1% or lower total body surface area (BSA) after 3 months of treatment.10 Treatment targets endorsed by the European Academy of Dermatology and Venereology (EADV) in guidelines on the use of systemic therapies in psoriasis include achieving a 75% or greater reduction in Psoriasis Area and Severity Index (PASI) score within 3 to 4 months of treatment.11

In clinical practice, many patients do not achieve these treatment targets, and topical treatments alone generally are insufficient in achieving treatment goals for psoriasis.12,13 Moreover, conventional topical treatments (eg, topical corticosteroids) used by most patients with psoriasis regardless of disease severity are associated with adverse events that can limit their use. Most topical corticosteroids have US Food and Drug Administration label restrictions relating to sites of application, duration and extent of use, and frequency of administration.14,15

Tapinarof cream 1% (VTAMA [Dermavant Sciences, Inc]) is a first-in-class topical nonsteroidal aryl hydrocarbon receptor agonist that was approved by the US Food and Drug Administration for the treatment of plaque psoriasis in adults16 and is being studied for the treatment of plaque psoriasis in children 2 years and older as well as for atopic dermatitis in adults and children 2 years and older. In PSOARING 1 (ClinicalTrials .gov identifier NCT03956355) and PSOARING 2 (NCT03983980)—identical 12-week pivotal phase 3 trials—monotherapy with tapinarof cream 1% once daily (QD) demonstrated statistically significant efficacy vs vehicle cream and was well tolerated in adults with mild to severe plaque psoriasis (Supplementary Figure S1).17 Lebwohl et al17 reported that significantly higher PASI75 responses were observed at week 12 with tapinarof cream vs vehicle in PSOARING 1 and PSOARING 2 (36% and 48% vs 10% and 7%, respectively; both P<.0001). A significantly higher PASI90 response of 19% and 21% at week 12 also was observed with tapinarof cream vs 2% and 3% with vehicle in PSOARING 1 and PSOARING 2, respectively (P=.0005 and P<.0001).17

In PSOARING 3 (NCT04053387)—the long-term extension trial (Supplementary Figure S1)—efficacy continued to improve or was maintained beyond the two 12-week trials, with improvements in total BSA affected and PASI scores for up to 52 weeks.18 Tapinarof cream 1% QD demonstrated positive, rapid, and durable outcomes in PSOARING 3, including high rates of complete disease clearance (Physician Global Assessment [PGA] score=0 [clear])(40.9% [312/763]), durability of response on treatment with no evidence of tachyphylaxis, and a remittive effect of approximately 4 months when off therapy (defined as maintenance of a PGA score of 0 [clear] or 1 [almost clear] after first achieving a PGA score of 0).18

Herein, we report absolute treatment targets for patients with plaque psoriasis who received tapinarof cream 1% QD in the PSOARING trials that are at least as stringent as the corresponding NPF and EADV targets of achieving a total BSA affected of 1% or lower or a PASI75 response within 3 to 4 months, respectively.

 

 

METHODS

Study Design

The pooled efficacy analyses included all patients with a baseline PGA score of 2 or higher (mild or worse) before treatment with tapinarof cream 1% QD in the PSOARING trials. This included patients who received tapinarof cream 1% in PSOARING 1 and PSOARING 2 who may or may not have continued into PSOARING 3, as well as those who received the vehicle in PSOARING 1 and PSOARING 2 who enrolled in PSOARING 3 and had a PGA score of 2 or higher before receiving tapinarof cream 1%.

Trial Participants

Full methods, including inclusion and exclusion criteria, for the PSOARING trials have been previously reported.17,18 Patients were aged 18 to 75 years and had chronic plaque psoriasis that was stable for at least 6 months before randomization; 3% to 20% total BSA affected (excluding the scalp, palms, fingernails, toenails, and soles); and a PGA score of 2 (mild), 3 (moderate), or 4 (severe) at baseline.

The clinical trials were conducted in compliance with the guidelines for Good Clinical Practice and the Declaration of Helsinki. Approval was obtained from local ethics committees or institutional review boards at each center. All patients provided written informed consent.

Trial Treatment

In PSOARING 1 and PSOARING 2, patients were randomized (2:1) to receive tapinarof cream 1% or vehicle QD for 12 weeks. In PSOARING 3 (the long-term extension trial), patients received up to 40 weeks of open-label tapinarof, followed by 4 weeks of follow-up off treatment. Patients received intermittent or continuous treatment with tapinarof cream 1% in PSOARING 3 based on PGA score: those entering the trial with a PGA score of 1 or higher received tapinarof cream 1% until complete disease clearance was achieved (defined as a PGA score of 0 [clear]). Those entering PSOARING 3 with or achieving a PGA score of 0 (clear) discontinued treatment and were observed for the duration of maintenance of a PGA score of 0 (clear) or 1 (almost clear) while off therapy (the protocol-defined “duration of remittive effect”). If disease worsening (defined as a PGA score 2 or higher) occurred, tapinarof cream 1% was restarted and continued until a PGA score of 0 (clear) was achieved. This pattern of treatment, discontinuation on achieving a PGA score of 0 (clear), and retreatment on disease worsening continued until the end of the trial. As a result, patients in PSOARING 3 could receive tapinarof cream 1% continuously or intermittently for 40 weeks.

Outcome Measures and Statistical Analyses

The assessment of total BSA affected by plaque psoriasis is an estimate of the total extent of disease as a percentage of total skin area. In the PSOARING trials, the skin surface of one hand (palm and digits) was assumed to be approximately equivalent to 1% BSA. The total BSA affected by psoriasis was evaluated from 0% to 100%, with greater total BSA affected being an indication of more extensive disease. The BSA efficacy outcomes used in these analyses were based post hoc on the proportion of patients who achieved a 1% or lower or 0.5% or lower total BSA affected. The smallest BSA affected increment that investigators were trained to measure and could record was 0.1%.

 

 

Psoriasis Area and Severity Index scores assess both the severity and extent of psoriasis. A PASI score lower than 5 often is considered indicative of mild psoriasis, a score of 5 to 10 indicates moderate disease, and a score higher than 10 indicates severe disease.19 The maximum PASI score is 72. The PASI efficacy outcomes used in these analyses were based post hoc on the proportion of patients who achieved an absolute total PASI score of 3 or lower, 2 or lower, and 1 or lower.

Efficacy analyses were based on pooled data for all patients in the PSOARING trials who had a PGA score of 2 to 4 (mild to severe) before treatment with tapinarof cream 1% in the intention-to-treat population using observed cases. Time-to-target analyses were based on Kaplan-Meier (KM) estimates using observed cases.

Safety analyses included the incidence and frequency of adverse events and were based on all patients who received tapinarof cream 1% in the PSOARING trials.

RESULTS

Baseline Patient Demographics and Disease Characteristics

The pooled efficacy analyses included 915 eligible patients (Table). At baseline, the mean (SD) age was 50.2 (13.25) years, 58.7% were male, the mean (SD) weight was 92.2 (23.67) kg, and the mean (SD) body mass index was 31.6 (7.53) kg/m2. The percentage of patients with a PGA score of 2 (mild), 3 (moderate), or 4 (severe) was 13.9%, 78.1%, and 8.0%, respectively. The mean (SD) PASI score was 8.7 (4.23) and mean (SD) total BSA affected was 7.8% (4.98).

Efficacy

Achievement of BSA-Affected Targets—The NPF-recommended target of 1% or lower total BSA affected within 3 months was achieved by 40% of patients (KM estimate [95% CI, 37%-43%])(Figure 1). Across the total trial period of up to 52 weeks, a total BSA affected of 1% or lower was achieved by 61% of patients (561/915), with the median time to target of approximately 4 months (KM estimate: 120 days [95% CI, 113-141])(Supplementary Figure S2a). Approximately 50% of patients (455/915) achieved a total BSA affected of 0.5% or lower, with a median time to target of 199 days (KM estimate [95% CI, 172-228)(Figure 1; Supplementary Figure S2b).

FIGURE 1. Pooled analysis of total body surface area (BSA) affected targets achieved by patients with mild to severe plaque psoriasis treated with tapinarof cream 1% once daily (QD) across a trial period up to 52 weeks in PSOARING 1, PSOARING 2, and PSOARING 3 (target total BSA affected, ≤1% [National Psoriasis Foundation [NPF]−recommended target]; target total BSA affected, ≤.5%)(N=915). These analyses included patients receiving continuous or intermittent tapinarof monotherapy in the 12-week pivotal trials (PSOARING 1 and PSOARING 2) and in the forced-withdrawal design of PSOARING 3 (treatment was stopped when patients achieved a Physician Global Assessment score of 0).

FIGURE 2. Total Psoriasis Area and Severity Index (PASI) score targets achieved by patients with mild to severe plaque psoriasis treated with tapinarof cream 1% once daily across a trial period up to 52 weeks in PSOARING 1, PSOARING 2 (target PASI score), and PSOARING 3 (target PASI score ≤3, ≤2, and ≤1)(N=915). These analyses included patients receiving continuous or intermittent tapinarof monotherapy in the 12-week pivotal trials (PSOARING 1 and PSOARING 2) and in the forced-withdrawal design of PSOARING 3 (treatment was stopped when patients achieved a Physician Global Assessment score of 0).

Achievement of Absolute PASI Targets—Across the total trial period (up to 52 weeks), an absolute total PASI score of 3 or lower was achieved by 75% of patients (686/915), with a median time to achieve this of 2 months (KM estimate: 58 days [95% CI, 57-63]); approximately 67% of patients (612/915) achieved a total PASI score of 2 or lower, with a median time to achieve of 3 months (KM estimate: 87 days [95% CI, 85-110])(Figure 2; Supplementary Figures S3a and S3b). A PASI score of 1 or lower was achieved by approximately 50% of patients (460/915), with a median time to achieve of approximately 6 months (KM estimate: 185 days [95% CI, 169-218])(Figure 2, Supplementary Figure S3c).

Illustrative Case—Case photography showing the clinical response in a 63-year-old man with moderate plaque psoriasis in PSOARING 2 is shown in Figure 3. After 12 weeks of treatment with tapinarof cream 1% QD, the patient achieved all primary and secondary efficacy end points. In addition to achieving the regulatory end point of a PGA score of 0 (clear) or 1 (almost clear) and a decrease from baseline of at least 2 points, achievement of 0% total BSA affected and a total PASI score of 0 at week 12 exceeded the NPF and EADV consensus treatment targets.10,11 Targets were achieved as early as week 4, with a total BSA affected of 0.5% or lower and a total PASI score of 1 or lower, illustrated by marked skin clearing and only faint residual erythema that completely resolved at week 12, with the absence of postinflammatory hyperpigmentation.

 

 

Safety

Safety data for the PSOARING trials have been previously reported.17,18 The most common treatment-emergent adverse events were folliculitis, contact dermatitis, upper respiratory tract infection, and nasopharyngitis. Treatment-emergent adverse events generally were mild or moderate in severity and did not lead to trial discontinuation.17,18

FIGURE 3. Moderate plaque psoriasis on the abdomen in a patient treated with tapinarof cream 1% once daily in PSOARING 2 who achieved the primary end point at week 4. A, At baseline, wellcircumscribed erythematous patches, plaques, and scaling were visible. B, The patient achieved the primary end point and National Psoriasis Foundation (NPF) and European Academy of Dermatology and Venereology (EADV) treatment targets by week 4, at which point there was marked clearing with faint residual erythema C, By week 12, the patient had 0% total body surface area affected and a total Psoriasis Area and Severity Index score of 0, exceeding NPF/EADV consensus treatment targets. Faint residual erythema completely resolved with the absence of postinflammatory hyperpigmentation.

COMMENT

Treat-to-target management approaches aim to improve patient outcomes by striving to achieve optimal goals. The treat-to-target approach supports shared decision-making between clinicians and patients based on common expectations of what constitutes treatment success.

The findings of this analysis based on pooled data from a large cohort of patients demonstrate that a high proportion of patients can achieve or exceed recommended treatment targets with tapinarof cream 1% QD and maintain improvements long-term. The NPF-recommended treatment target of 1% or lower BSA affected within approximately 3 months (90 days) of treatment was achieved by 40% of tapinarof-treated patients. In addition, 1% or lower BSA affected at any time during the trials was achieved by 61% of patients (median, approximately 4 months). The analyses also indicated that PASI total scores of 3 or lower and 2 or lower were achieved by 75% and 67% of tapinarof-treated patients, respectively, within 2 to 3 months.

These findings support the previously reported efficacy of tapinarof cream, including high rates of complete disease clearance (40.9% [312/763]), durable response following treatment interruption, an off-therapy remittive effect of approximately 4 months, and good disease control on therapy with no evidence of tachyphylaxis.17,18

CONCLUSION

Taken together with previously reported tapinarof efficacy and safety results, our findings demonstrate that a high proportion of patients treated with tapinarof cream as monotherapy can achieve aggressive treatment targets set by both US and European guidelines developed for systemic and biologic therapies. Tapinarof cream 1% QD is an effective topical treatment option for patients with plaque psoriasis that has been approved without restrictions relating to severity or extent of disease treated, duration of use, or application sites, including application to sensitive and intertriginous skin.

Acknowledgments—Editorial and medical writing support under the guidance of the authors was provided by Melanie Govender, MSc (Med), ApotheCom (United Kingdom), and was funded by Dermavant Sciences, Inc, in accordance with Good Publication Practice (GPP) guidelines.

References
  1. Armstrong AW, Mehta MD, Schupp CW, et al. Psoriasis prevalence in adults in the United States. JAMA Dermatol. 2021;157:940-946.
  2. Parisi R, Iskandar IYK, Kontopantelis E, et al. National, regional, and worldwide epidemiology of psoriasis: systematic analysis and modelling study. BMJ. 2020;369:m1590.
  3. Pilon D, Teeple A, Zhdanava M, et al. The economic burden of psoriasis with high comorbidity among privately insured patients in the United States. J Med Econ. 2019;22:196-203.
  4. Singh S, Taylor C, Kornmehl H, et al. Psoriasis and suicidality: a systematic review and meta-analysis. J Am Acad Dermatol. 2017;77:425-440.e2.
  5. Feldman SR, Goffe B, Rice G, et al. The challenge of managing psoriasis: unmet medical needs and stakeholder perspectives. Am Health Drug Benefits. 2016;9:504-513.
  6. Ford JA, Solomon DH. Challenges in implementing treat-to-target strategies in rheumatology. Rheum Dis Clin North Am. 2019;45:101-112.
  7. Sitbon O, Galiè N. Treat-to-target strategies in pulmonary arterial hypertension: the importance of using multiple goals. Eur Respir Rev. 2010;19:272-278.
  8. Smolen JS, Aletaha D, Bijlsma JW, et al. Treating rheumatoid arthritis to target: recommendations of an international task force. Ann Rheum Dis. 2010;69:631-637.
  9. Wangnoo SK, Sethi B, Sahay RK, et al. Treat-to-target trials in diabetes. Indian J Endocrinol Metab. 2014;18:166-174.
  10. Armstrong AW, Siegel MP, Bagel J, et al. From the Medical Board of the National Psoriasis Foundation: treatment targets for plaque psoriasis. J Am Acad Dermatol. 2017;76:290-298.
  11. Pathirana D, Ormerod AD, Saiag P, et al. European S3-guidelines on the systemic treatment of psoriasis vulgaris. J Eur Acad Dermatol Venereol. 2009;23(Suppl 2):1-70.
  12. Strober BE, van der Walt JM, Armstrong AW, et al. Clinical goals and barriers to effective psoriasis care. Dermatol Ther (Heidelb). 2019; 9:5-18.
  13. Bagel J, Gold LS. Combining topical psoriasis treatment to enhance systemic and phototherapy: a review of the literature. J Drugs Dermatol. 2017;16:1209-1222.
  14. Elmets CA, Korman NJ, Prater EF, et al. Joint AAD-NPF Guidelines of care for the management and treatment of psoriasis with topical therapy and alternative medicine modalities for psoriasis severity measures. J Am Acad Dermatol. 2021;84:432-470.
  15. Stein Gold LF. Topical therapies for psoriasis: improving management strategies and patient adherence. Semin Cutan Med Surg. 2016;35 (2 Suppl 2):S36-S44; quiz S45.
  16. VTAMA® (tapinarof) cream. Prescribing information. Dermavant Sciences; 2022. Accessed September 13, 2024. https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/215272s000lbl.pdf
  17. Lebwohl MG, Stein Gold L, Strober B, et al. Phase 3 trials of tapinarof cream for plaque psoriasis. N Engl J Med. 2021;385:2219-2229 and supplementary appendix.
  18. Strober B, Stein Gold L, Bissonnette R, et al. One-year safety and efficacy of tapinarof cream for the treatment of plaque psoriasis: results from the PSOARING 3 trial. J Am Acad Dermatol. 2022;87:800-806.
  19. Clinical Review Report: Guselkumab (Tremfya) [Internet]. Canadian Agency for Drugs and Technologies in Health; 2018. Accessed September 13, 2024. https://www.ncbi.nlm.nih.gov/books/NBK534047/pdf/Bookshelf_NBK534047.pdf
References
  1. Armstrong AW, Mehta MD, Schupp CW, et al. Psoriasis prevalence in adults in the United States. JAMA Dermatol. 2021;157:940-946.
  2. Parisi R, Iskandar IYK, Kontopantelis E, et al. National, regional, and worldwide epidemiology of psoriasis: systematic analysis and modelling study. BMJ. 2020;369:m1590.
  3. Pilon D, Teeple A, Zhdanava M, et al. The economic burden of psoriasis with high comorbidity among privately insured patients in the United States. J Med Econ. 2019;22:196-203.
  4. Singh S, Taylor C, Kornmehl H, et al. Psoriasis and suicidality: a systematic review and meta-analysis. J Am Acad Dermatol. 2017;77:425-440.e2.
  5. Feldman SR, Goffe B, Rice G, et al. The challenge of managing psoriasis: unmet medical needs and stakeholder perspectives. Am Health Drug Benefits. 2016;9:504-513.
  6. Ford JA, Solomon DH. Challenges in implementing treat-to-target strategies in rheumatology. Rheum Dis Clin North Am. 2019;45:101-112.
  7. Sitbon O, Galiè N. Treat-to-target strategies in pulmonary arterial hypertension: the importance of using multiple goals. Eur Respir Rev. 2010;19:272-278.
  8. Smolen JS, Aletaha D, Bijlsma JW, et al. Treating rheumatoid arthritis to target: recommendations of an international task force. Ann Rheum Dis. 2010;69:631-637.
  9. Wangnoo SK, Sethi B, Sahay RK, et al. Treat-to-target trials in diabetes. Indian J Endocrinol Metab. 2014;18:166-174.
  10. Armstrong AW, Siegel MP, Bagel J, et al. From the Medical Board of the National Psoriasis Foundation: treatment targets for plaque psoriasis. J Am Acad Dermatol. 2017;76:290-298.
  11. Pathirana D, Ormerod AD, Saiag P, et al. European S3-guidelines on the systemic treatment of psoriasis vulgaris. J Eur Acad Dermatol Venereol. 2009;23(Suppl 2):1-70.
  12. Strober BE, van der Walt JM, Armstrong AW, et al. Clinical goals and barriers to effective psoriasis care. Dermatol Ther (Heidelb). 2019; 9:5-18.
  13. Bagel J, Gold LS. Combining topical psoriasis treatment to enhance systemic and phototherapy: a review of the literature. J Drugs Dermatol. 2017;16:1209-1222.
  14. Elmets CA, Korman NJ, Prater EF, et al. Joint AAD-NPF Guidelines of care for the management and treatment of psoriasis with topical therapy and alternative medicine modalities for psoriasis severity measures. J Am Acad Dermatol. 2021;84:432-470.
  15. Stein Gold LF. Topical therapies for psoriasis: improving management strategies and patient adherence. Semin Cutan Med Surg. 2016;35 (2 Suppl 2):S36-S44; quiz S45.
  16. VTAMA® (tapinarof) cream. Prescribing information. Dermavant Sciences; 2022. Accessed September 13, 2024. https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/215272s000lbl.pdf
  17. Lebwohl MG, Stein Gold L, Strober B, et al. Phase 3 trials of tapinarof cream for plaque psoriasis. N Engl J Med. 2021;385:2219-2229 and supplementary appendix.
  18. Strober B, Stein Gold L, Bissonnette R, et al. One-year safety and efficacy of tapinarof cream for the treatment of plaque psoriasis: results from the PSOARING 3 trial. J Am Acad Dermatol. 2022;87:800-806.
  19. Clinical Review Report: Guselkumab (Tremfya) [Internet]. Canadian Agency for Drugs and Technologies in Health; 2018. Accessed September 13, 2024. https://www.ncbi.nlm.nih.gov/books/NBK534047/pdf/Bookshelf_NBK534047.pdf
Issue
Cutis - 114(4)
Issue
Cutis - 114(4)
Page Number
122-127
Page Number
122-127
Publications
Publications
Topics
Article Type
Sections
Inside the Article

 

Practice Points

  • In clinical practice, many patients with psoriasis do not achieve treatment targets set forth by the National Psoriasis Foundation and the European Academy of Dermatology and Venereology, and topical treatments alone generally are insufficient in achieving treatment goals for psoriasis.
  • Tapinarof cream 1% is a nonsteroidal aryl hydrocarbon receptor agonist approved by the US Food and Drug Administration for the treatment of plaque psoriasis in adults; it also is being studied for the treatment of plaque psoriasis in children 2 years and older.
  • Tapinarof cream 1% is an effective topical treatment option for patients with plaque psoriasis of any severity, with no limitations on treatment duration, total extent of use, or application sites, including intertriginous skin and sensitive areas.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media
Media Files

Pediatric Melanoma Outcomes by Race and Socioeconomic Factors

Article Type
Changed
Mon, 10/07/2024 - 10:35
Display Headline
Pediatric Melanoma Outcomes by Race and Socioeconomic Factors

To the Editor:

Skin cancers are extremely common worldwide. Malignant melanomas comprise approximately 1 in 5 of these cancers. Exposure to UV radiation is postulated to be responsible for a global rise in melanoma cases over the past 50 years.1 Pediatric melanoma is a particularly rare condition that affects approximately 6 in every 1 million children.2 Melanoma incidence in children ranges by age, increasing by approximately 10-fold from age 1 to 4 years to age 15 to 19 years. Tumor ulceration is a feature more commonly seen among children younger than 10 years and is associated with worse outcomes. Tumor thickness and ulceration strongly predict sentinel lymph node metastases among children, which also is associated with a poor prognosis.3

A recent study evaluating stage IV melanoma survival rates in adolescents and young adults (AYAs) vs older adults found that survival is much worse among AYAs. Thicker tumors and public health insurance also were associated with worse survival rates for AYAs, while early detection was associated with better survival rates.4

Health disparities and their role in the prognosis of pediatric melanoma is another important factor. One study analyzed this relationship at the state level using Texas Cancer Registry data (1995-2009).5 Patients’ socioeconomic status (SES) and driving distance to the nearest pediatric cancer care center were included in the analysis. Hispanic children were found to be 3 times more likely to present with advanced disease than non-Hispanic White children. Although SES and distance to the nearest treatment center were not found to affect the melanoma stage at presentation, Hispanic ethnicity or being in the lowest SES quartile were correlated with a higher mortality risk.5

When considering specific subtypes of melanoma, acral lentiginous melanoma (ALM) is known to develop in patients with skin of color. A 2023 study by Holman et al6 reported that the percentage of melanomas that were ALMs ranged from 0.8% in non-Hispanic White individuals to 19.1% in Hispanic Black, American Indian/Alaska Native, and Asian/Pacific Islander individuals. However, ALM is rare in children. In a pooled cohort study with patient information retrieved from the nationwide Dutch Pathology Registry, only 1 child and 1 adolescent were found to have ALM across a total of 514 patients.7 We sought to analyze pediatric melanoma outcomes based on race and other barriers to appropriate care.

We conducted a search of the Surveillance, Epidemiology, and End Results (SEER) database from January 1995 to December 2016 for patients aged 21 years and younger with a primary melanoma diagnosis. The primary outcome was the 5-year survival rate. County-level SES variables were used to calculate a prosperity index. Kaplan-Meier analysis and Cox proportional hazards model were used to compare 5-year survival rates among the different racial/ethnic groups.

A sample of 2742 patients was identified during the study period and followed for 5 years. Eighty-two percent were White, 6% Hispanic, 2% Asian, 1% Black, and 5% classified as other/unknown race (data were missing for 4%). The cohort was predominantly female (61%). White patients were more likely to present with localized disease than any other race/ethnicity (83% vs 65% in Hispanic, 60% in Asian/Pacific Islander, and 45% in Black patients [P<.05]).

Black and Hispanic patients had the worst 5-year survival rates on bivariate analysis. On multivariate analysis, this finding remained significant for Hispanic patients when compared with White patients (hazard ratio, 2.37 [P<.05]). Increasing age, male sex, advanced stage at diagnosis, and failure to receive surgery were associated with increased odds of mortality.

Patients with regionalized and disseminated disease had increased odds of mortality (6.16 and 64.45, respectively; P<.05) compared with patients with localized disease. Socioeconomic status and urbanization were not found to influence 5-year survival rates.

Pediatric melanoma often presents a clinical challenge with special considerations. Pediatric-specific predisposing risk factors for melanoma and an atypical clinical presentation are some of the major concerns that necessitate a tailored approach to this malignancy, especially among different age groups, skin types, and racial and socioeconomic groups.5

Standard ABCDE criteria often are inadequate for accurate detection of pediatric melanomas. Initial lesions often manifest as raised, red, amelanotic lesions mimicking pyogenic granulomas. Lesions tend to be very small (<6 mm in diameter) and can be uniform in color, thereby making the melanoma more difficult to detect compared to the characteristic findings in adults.5 Bleeding or ulceration often can be a warning sign during physical examination.

With regard to incidence, pediatric melanoma is relatively rare. Since the 1970s, the incidence of pediatric melanoma has been increasing; however, a recent analysis of the SEER database showed a decreasing trend from 2000 to 2010.4

Our analysis of the SEER data showed an increased risk for pediatric melanoma in older adolescents. In addition, the incidence of pediatric melanoma was higher in females of all racial groups except Asian/Pacific Islander individuals. However, SES was not found to significantly influence the 5-year survival rate in pediatric melanoma.

White pediatric patients were more likely to present with localized disease compared with other races. Pediatric melanoma patients with regional disease had a 6-fold increase in mortality rate vs those with localized disease; those with disseminated disease had a 65-fold higher risk. Consistent with this, Black and Hispanic patients had the worst 5-year survival rates on bivariate analysis.

These findings suggest a relationship between race, melanoma spread, and disease severity. Patient education programs need to be directed specifically to minority groups to improve their knowledge on evolving skin lesions and sun protection practices. Physicians also need to have heightened suspicion and better knowledge of the unique traits of pediatric melanoma.5

Given the considerable influence these disparities can have on melanoma outcomes, further research is needed to characterize outcomes based on race and determine obstacles to appropriate care. Improved public outreach initiatives that accommodate specific cultural barriers (eg, language, traditional patterns of behavior) also are required to improve current circumstances.

References
  1. Arnold M, Singh D, Laversanne M, et al. Global burden of cutaneous melanoma in 2020 and projections to 2040. JAMA Dermatol. 2022;158:495-503.
  2. McCormack L, Hawryluk EB. Pediatric melanoma update. G Ital Dermatol Venereol. 2018;153:707-715.
  3. Saiyed FK, Hamilton EC, Austin MT. Pediatric melanoma: incidence, treatment, and prognosis. Pediatric Health Med Ther. 2017;8:39-45.
  4. Wojcik KY, Hawkins M, Anderson-Mellies A, et al. Melanoma survival by age group: population-based disparities for adolescent and young adult patients by stage, tumor thickness, and insurance type. J Am Acad Dermatol. 2023;88:831-840.
  5. Hamilton EC, Nguyen HT, Chang YC, et al. Health disparities influence childhood melanoma stage at diagnosis and outcome. J Pediatr. 2016;175:182-187.
  6. Holman DM, King JB, White A, et al. Acral lentiginous melanoma incidence by sex, race, ethnicity, and stage in the United States, 2010-2019. Prev Med. 2023;175:107692. doi:10.1016/j.ypmed.2023.107692
  7. El Sharouni MA, Rawson RV, Potter AJ, et al. Melanomas in children and adolescents: clinicopathologic features and survival outcomes. J Am Acad Dermatol. 2023;88:609-616. doi:10.1016/j.jaad.2022.08.067
Article PDF
Author and Disclosure Information

From Howard University, Washington, DC. Drs. Ahuja, Atoba, Tahmazian and Khushbakht are from the College of Medicine, and Dr. Nnorom is from the Department of Surgery.

The authors have no relevant financial disclosures to report.

Acknowledgments—Coauthor Lori Wilson, MD, died on October 14, 2022. The authors would like to thank Anjali Ahuja (Centreville, Virginia) for her help with critically revising the manuscript for important intellectual content.

Correspondence: Geeta Ahuja, MD, 2041 Georgia Ave NW, Washington, DC 20060 ([email protected]).Cutis. 2024 October;114(4):110-111. doi:10.12788/cutis.1110

Publications
Topics
Page Number
110-111
Sections
Author and Disclosure Information

From Howard University, Washington, DC. Drs. Ahuja, Atoba, Tahmazian and Khushbakht are from the College of Medicine, and Dr. Nnorom is from the Department of Surgery.

The authors have no relevant financial disclosures to report.

Acknowledgments—Coauthor Lori Wilson, MD, died on October 14, 2022. The authors would like to thank Anjali Ahuja (Centreville, Virginia) for her help with critically revising the manuscript for important intellectual content.

Correspondence: Geeta Ahuja, MD, 2041 Georgia Ave NW, Washington, DC 20060 ([email protected]).Cutis. 2024 October;114(4):110-111. doi:10.12788/cutis.1110

Author and Disclosure Information

From Howard University, Washington, DC. Drs. Ahuja, Atoba, Tahmazian and Khushbakht are from the College of Medicine, and Dr. Nnorom is from the Department of Surgery.

The authors have no relevant financial disclosures to report.

Acknowledgments—Coauthor Lori Wilson, MD, died on October 14, 2022. The authors would like to thank Anjali Ahuja (Centreville, Virginia) for her help with critically revising the manuscript for important intellectual content.

Correspondence: Geeta Ahuja, MD, 2041 Georgia Ave NW, Washington, DC 20060 ([email protected]).Cutis. 2024 October;114(4):110-111. doi:10.12788/cutis.1110

Article PDF
Article PDF

To the Editor:

Skin cancers are extremely common worldwide. Malignant melanomas comprise approximately 1 in 5 of these cancers. Exposure to UV radiation is postulated to be responsible for a global rise in melanoma cases over the past 50 years.1 Pediatric melanoma is a particularly rare condition that affects approximately 6 in every 1 million children.2 Melanoma incidence in children ranges by age, increasing by approximately 10-fold from age 1 to 4 years to age 15 to 19 years. Tumor ulceration is a feature more commonly seen among children younger than 10 years and is associated with worse outcomes. Tumor thickness and ulceration strongly predict sentinel lymph node metastases among children, which also is associated with a poor prognosis.3

A recent study evaluating stage IV melanoma survival rates in adolescents and young adults (AYAs) vs older adults found that survival is much worse among AYAs. Thicker tumors and public health insurance also were associated with worse survival rates for AYAs, while early detection was associated with better survival rates.4

Health disparities and their role in the prognosis of pediatric melanoma is another important factor. One study analyzed this relationship at the state level using Texas Cancer Registry data (1995-2009).5 Patients’ socioeconomic status (SES) and driving distance to the nearest pediatric cancer care center were included in the analysis. Hispanic children were found to be 3 times more likely to present with advanced disease than non-Hispanic White children. Although SES and distance to the nearest treatment center were not found to affect the melanoma stage at presentation, Hispanic ethnicity or being in the lowest SES quartile were correlated with a higher mortality risk.5

When considering specific subtypes of melanoma, acral lentiginous melanoma (ALM) is known to develop in patients with skin of color. A 2023 study by Holman et al6 reported that the percentage of melanomas that were ALMs ranged from 0.8% in non-Hispanic White individuals to 19.1% in Hispanic Black, American Indian/Alaska Native, and Asian/Pacific Islander individuals. However, ALM is rare in children. In a pooled cohort study with patient information retrieved from the nationwide Dutch Pathology Registry, only 1 child and 1 adolescent were found to have ALM across a total of 514 patients.7 We sought to analyze pediatric melanoma outcomes based on race and other barriers to appropriate care.

We conducted a search of the Surveillance, Epidemiology, and End Results (SEER) database from January 1995 to December 2016 for patients aged 21 years and younger with a primary melanoma diagnosis. The primary outcome was the 5-year survival rate. County-level SES variables were used to calculate a prosperity index. Kaplan-Meier analysis and Cox proportional hazards model were used to compare 5-year survival rates among the different racial/ethnic groups.

A sample of 2742 patients was identified during the study period and followed for 5 years. Eighty-two percent were White, 6% Hispanic, 2% Asian, 1% Black, and 5% classified as other/unknown race (data were missing for 4%). The cohort was predominantly female (61%). White patients were more likely to present with localized disease than any other race/ethnicity (83% vs 65% in Hispanic, 60% in Asian/Pacific Islander, and 45% in Black patients [P<.05]).

Black and Hispanic patients had the worst 5-year survival rates on bivariate analysis. On multivariate analysis, this finding remained significant for Hispanic patients when compared with White patients (hazard ratio, 2.37 [P<.05]). Increasing age, male sex, advanced stage at diagnosis, and failure to receive surgery were associated with increased odds of mortality.

Patients with regionalized and disseminated disease had increased odds of mortality (6.16 and 64.45, respectively; P<.05) compared with patients with localized disease. Socioeconomic status and urbanization were not found to influence 5-year survival rates.

Pediatric melanoma often presents a clinical challenge with special considerations. Pediatric-specific predisposing risk factors for melanoma and an atypical clinical presentation are some of the major concerns that necessitate a tailored approach to this malignancy, especially among different age groups, skin types, and racial and socioeconomic groups.5

Standard ABCDE criteria often are inadequate for accurate detection of pediatric melanomas. Initial lesions often manifest as raised, red, amelanotic lesions mimicking pyogenic granulomas. Lesions tend to be very small (<6 mm in diameter) and can be uniform in color, thereby making the melanoma more difficult to detect compared to the characteristic findings in adults.5 Bleeding or ulceration often can be a warning sign during physical examination.

With regard to incidence, pediatric melanoma is relatively rare. Since the 1970s, the incidence of pediatric melanoma has been increasing; however, a recent analysis of the SEER database showed a decreasing trend from 2000 to 2010.4

Our analysis of the SEER data showed an increased risk for pediatric melanoma in older adolescents. In addition, the incidence of pediatric melanoma was higher in females of all racial groups except Asian/Pacific Islander individuals. However, SES was not found to significantly influence the 5-year survival rate in pediatric melanoma.

White pediatric patients were more likely to present with localized disease compared with other races. Pediatric melanoma patients with regional disease had a 6-fold increase in mortality rate vs those with localized disease; those with disseminated disease had a 65-fold higher risk. Consistent with this, Black and Hispanic patients had the worst 5-year survival rates on bivariate analysis.

These findings suggest a relationship between race, melanoma spread, and disease severity. Patient education programs need to be directed specifically to minority groups to improve their knowledge on evolving skin lesions and sun protection practices. Physicians also need to have heightened suspicion and better knowledge of the unique traits of pediatric melanoma.5

Given the considerable influence these disparities can have on melanoma outcomes, further research is needed to characterize outcomes based on race and determine obstacles to appropriate care. Improved public outreach initiatives that accommodate specific cultural barriers (eg, language, traditional patterns of behavior) also are required to improve current circumstances.

To the Editor:

Skin cancers are extremely common worldwide. Malignant melanomas comprise approximately 1 in 5 of these cancers. Exposure to UV radiation is postulated to be responsible for a global rise in melanoma cases over the past 50 years.1 Pediatric melanoma is a particularly rare condition that affects approximately 6 in every 1 million children.2 Melanoma incidence in children ranges by age, increasing by approximately 10-fold from age 1 to 4 years to age 15 to 19 years. Tumor ulceration is a feature more commonly seen among children younger than 10 years and is associated with worse outcomes. Tumor thickness and ulceration strongly predict sentinel lymph node metastases among children, which also is associated with a poor prognosis.3

A recent study evaluating stage IV melanoma survival rates in adolescents and young adults (AYAs) vs older adults found that survival is much worse among AYAs. Thicker tumors and public health insurance also were associated with worse survival rates for AYAs, while early detection was associated with better survival rates.4

Health disparities and their role in the prognosis of pediatric melanoma is another important factor. One study analyzed this relationship at the state level using Texas Cancer Registry data (1995-2009).5 Patients’ socioeconomic status (SES) and driving distance to the nearest pediatric cancer care center were included in the analysis. Hispanic children were found to be 3 times more likely to present with advanced disease than non-Hispanic White children. Although SES and distance to the nearest treatment center were not found to affect the melanoma stage at presentation, Hispanic ethnicity or being in the lowest SES quartile were correlated with a higher mortality risk.5

When considering specific subtypes of melanoma, acral lentiginous melanoma (ALM) is known to develop in patients with skin of color. A 2023 study by Holman et al6 reported that the percentage of melanomas that were ALMs ranged from 0.8% in non-Hispanic White individuals to 19.1% in Hispanic Black, American Indian/Alaska Native, and Asian/Pacific Islander individuals. However, ALM is rare in children. In a pooled cohort study with patient information retrieved from the nationwide Dutch Pathology Registry, only 1 child and 1 adolescent were found to have ALM across a total of 514 patients.7 We sought to analyze pediatric melanoma outcomes based on race and other barriers to appropriate care.

We conducted a search of the Surveillance, Epidemiology, and End Results (SEER) database from January 1995 to December 2016 for patients aged 21 years and younger with a primary melanoma diagnosis. The primary outcome was the 5-year survival rate. County-level SES variables were used to calculate a prosperity index. Kaplan-Meier analysis and Cox proportional hazards model were used to compare 5-year survival rates among the different racial/ethnic groups.

A sample of 2742 patients was identified during the study period and followed for 5 years. Eighty-two percent were White, 6% Hispanic, 2% Asian, 1% Black, and 5% classified as other/unknown race (data were missing for 4%). The cohort was predominantly female (61%). White patients were more likely to present with localized disease than any other race/ethnicity (83% vs 65% in Hispanic, 60% in Asian/Pacific Islander, and 45% in Black patients [P<.05]).

Black and Hispanic patients had the worst 5-year survival rates on bivariate analysis. On multivariate analysis, this finding remained significant for Hispanic patients when compared with White patients (hazard ratio, 2.37 [P<.05]). Increasing age, male sex, advanced stage at diagnosis, and failure to receive surgery were associated with increased odds of mortality.

Patients with regionalized and disseminated disease had increased odds of mortality (6.16 and 64.45, respectively; P<.05) compared with patients with localized disease. Socioeconomic status and urbanization were not found to influence 5-year survival rates.

Pediatric melanoma often presents a clinical challenge with special considerations. Pediatric-specific predisposing risk factors for melanoma and an atypical clinical presentation are some of the major concerns that necessitate a tailored approach to this malignancy, especially among different age groups, skin types, and racial and socioeconomic groups.5

Standard ABCDE criteria often are inadequate for accurate detection of pediatric melanomas. Initial lesions often manifest as raised, red, amelanotic lesions mimicking pyogenic granulomas. Lesions tend to be very small (<6 mm in diameter) and can be uniform in color, thereby making the melanoma more difficult to detect compared to the characteristic findings in adults.5 Bleeding or ulceration often can be a warning sign during physical examination.

With regard to incidence, pediatric melanoma is relatively rare. Since the 1970s, the incidence of pediatric melanoma has been increasing; however, a recent analysis of the SEER database showed a decreasing trend from 2000 to 2010.4

Our analysis of the SEER data showed an increased risk for pediatric melanoma in older adolescents. In addition, the incidence of pediatric melanoma was higher in females of all racial groups except Asian/Pacific Islander individuals. However, SES was not found to significantly influence the 5-year survival rate in pediatric melanoma.

White pediatric patients were more likely to present with localized disease compared with other races. Pediatric melanoma patients with regional disease had a 6-fold increase in mortality rate vs those with localized disease; those with disseminated disease had a 65-fold higher risk. Consistent with this, Black and Hispanic patients had the worst 5-year survival rates on bivariate analysis.

These findings suggest a relationship between race, melanoma spread, and disease severity. Patient education programs need to be directed specifically to minority groups to improve their knowledge on evolving skin lesions and sun protection practices. Physicians also need to have heightened suspicion and better knowledge of the unique traits of pediatric melanoma.5

Given the considerable influence these disparities can have on melanoma outcomes, further research is needed to characterize outcomes based on race and determine obstacles to appropriate care. Improved public outreach initiatives that accommodate specific cultural barriers (eg, language, traditional patterns of behavior) also are required to improve current circumstances.

References
  1. Arnold M, Singh D, Laversanne M, et al. Global burden of cutaneous melanoma in 2020 and projections to 2040. JAMA Dermatol. 2022;158:495-503.
  2. McCormack L, Hawryluk EB. Pediatric melanoma update. G Ital Dermatol Venereol. 2018;153:707-715.
  3. Saiyed FK, Hamilton EC, Austin MT. Pediatric melanoma: incidence, treatment, and prognosis. Pediatric Health Med Ther. 2017;8:39-45.
  4. Wojcik KY, Hawkins M, Anderson-Mellies A, et al. Melanoma survival by age group: population-based disparities for adolescent and young adult patients by stage, tumor thickness, and insurance type. J Am Acad Dermatol. 2023;88:831-840.
  5. Hamilton EC, Nguyen HT, Chang YC, et al. Health disparities influence childhood melanoma stage at diagnosis and outcome. J Pediatr. 2016;175:182-187.
  6. Holman DM, King JB, White A, et al. Acral lentiginous melanoma incidence by sex, race, ethnicity, and stage in the United States, 2010-2019. Prev Med. 2023;175:107692. doi:10.1016/j.ypmed.2023.107692
  7. El Sharouni MA, Rawson RV, Potter AJ, et al. Melanomas in children and adolescents: clinicopathologic features and survival outcomes. J Am Acad Dermatol. 2023;88:609-616. doi:10.1016/j.jaad.2022.08.067
References
  1. Arnold M, Singh D, Laversanne M, et al. Global burden of cutaneous melanoma in 2020 and projections to 2040. JAMA Dermatol. 2022;158:495-503.
  2. McCormack L, Hawryluk EB. Pediatric melanoma update. G Ital Dermatol Venereol. 2018;153:707-715.
  3. Saiyed FK, Hamilton EC, Austin MT. Pediatric melanoma: incidence, treatment, and prognosis. Pediatric Health Med Ther. 2017;8:39-45.
  4. Wojcik KY, Hawkins M, Anderson-Mellies A, et al. Melanoma survival by age group: population-based disparities for adolescent and young adult patients by stage, tumor thickness, and insurance type. J Am Acad Dermatol. 2023;88:831-840.
  5. Hamilton EC, Nguyen HT, Chang YC, et al. Health disparities influence childhood melanoma stage at diagnosis and outcome. J Pediatr. 2016;175:182-187.
  6. Holman DM, King JB, White A, et al. Acral lentiginous melanoma incidence by sex, race, ethnicity, and stage in the United States, 2010-2019. Prev Med. 2023;175:107692. doi:10.1016/j.ypmed.2023.107692
  7. El Sharouni MA, Rawson RV, Potter AJ, et al. Melanomas in children and adolescents: clinicopathologic features and survival outcomes. J Am Acad Dermatol. 2023;88:609-616. doi:10.1016/j.jaad.2022.08.067
Page Number
110-111
Page Number
110-111
Publications
Publications
Topics
Article Type
Display Headline
Pediatric Melanoma Outcomes by Race and Socioeconomic Factors
Display Headline
Pediatric Melanoma Outcomes by Race and Socioeconomic Factors
Sections
Inside the Article

Practice Points

  • Pediatric melanoma is a unique clinical entity with a different clinical presentation than in adults.
  • Thicker tumors and disseminated disease are associated with a worse prognosis, and these factors are more commonly seen in Black and Hispanic patients.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media