User login
Lung Cancer Exposome in U.S. Military Veterans: Study of Environment and Epigenetic Factors on Risk and Survival
Background
The Exposome—the comprehensive accumulation of environmental exposures from birth to death—provides a framework for linking external risk factors to cancer biology. In U.S. veterans, the exposome includes both military-specific exposures (e.g., asbestos, Agent Orange, burn pits) and postservice socioeconomic and environmental factors. These cumulative exposures may drive tumor development and progression via epigenetic mechanisms, though their impact on lung cancer outcomes remain poorly characterized.
Methods
This is a retrospective cohort study of 71 lung cancer subjects (NSCLC and SCLC) from the Jesse Brown VA Medical Center (IRB# 1586320). We assessed the Area Deprivation Index (ADI), Environmental Burden Index (EBI), and occupational exposure in relation to DNA methylation of CDO1, TAC1, SOX17, and HOXA7. Geospatial data were mapped to US census tracts, and standard statistical analysis were conducted.
Results
NSCLC patients exhibited significantly higher methylation levels across all genes. High EBI exposure was associated with lower SOX17 methylation (p = 0.064) and worse overall survival (p = 0.046). In NSCLC patients, occupational exposure predicted a 7.7-fold increased hazard of death (p = 0.027). SOX17 and TAC1 methylation were independently associated with reduced survival (p = 0.037 and 0.0058, respectively). While ADI did not independently predict survival, it correlated with late-stage presentation and reduced HOXA7 methylation.
Conclusions
Exposome factors such as environmental burden and occupational exposure are biologically embedded in lung cancer cell through gene-specific methylation and significantly impact survival. We posit that integrating exposomic and molecular data could enhance lung precision oncology approaches for high-risk veteran populations.
Background
The Exposome—the comprehensive accumulation of environmental exposures from birth to death—provides a framework for linking external risk factors to cancer biology. In U.S. veterans, the exposome includes both military-specific exposures (e.g., asbestos, Agent Orange, burn pits) and postservice socioeconomic and environmental factors. These cumulative exposures may drive tumor development and progression via epigenetic mechanisms, though their impact on lung cancer outcomes remain poorly characterized.
Methods
This is a retrospective cohort study of 71 lung cancer subjects (NSCLC and SCLC) from the Jesse Brown VA Medical Center (IRB# 1586320). We assessed the Area Deprivation Index (ADI), Environmental Burden Index (EBI), and occupational exposure in relation to DNA methylation of CDO1, TAC1, SOX17, and HOXA7. Geospatial data were mapped to US census tracts, and standard statistical analysis were conducted.
Results
NSCLC patients exhibited significantly higher methylation levels across all genes. High EBI exposure was associated with lower SOX17 methylation (p = 0.064) and worse overall survival (p = 0.046). In NSCLC patients, occupational exposure predicted a 7.7-fold increased hazard of death (p = 0.027). SOX17 and TAC1 methylation were independently associated with reduced survival (p = 0.037 and 0.0058, respectively). While ADI did not independently predict survival, it correlated with late-stage presentation and reduced HOXA7 methylation.
Conclusions
Exposome factors such as environmental burden and occupational exposure are biologically embedded in lung cancer cell through gene-specific methylation and significantly impact survival. We posit that integrating exposomic and molecular data could enhance lung precision oncology approaches for high-risk veteran populations.
Background
The Exposome—the comprehensive accumulation of environmental exposures from birth to death—provides a framework for linking external risk factors to cancer biology. In U.S. veterans, the exposome includes both military-specific exposures (e.g., asbestos, Agent Orange, burn pits) and postservice socioeconomic and environmental factors. These cumulative exposures may drive tumor development and progression via epigenetic mechanisms, though their impact on lung cancer outcomes remain poorly characterized.
Methods
This is a retrospective cohort study of 71 lung cancer subjects (NSCLC and SCLC) from the Jesse Brown VA Medical Center (IRB# 1586320). We assessed the Area Deprivation Index (ADI), Environmental Burden Index (EBI), and occupational exposure in relation to DNA methylation of CDO1, TAC1, SOX17, and HOXA7. Geospatial data were mapped to US census tracts, and standard statistical analysis were conducted.
Results
NSCLC patients exhibited significantly higher methylation levels across all genes. High EBI exposure was associated with lower SOX17 methylation (p = 0.064) and worse overall survival (p = 0.046). In NSCLC patients, occupational exposure predicted a 7.7-fold increased hazard of death (p = 0.027). SOX17 and TAC1 methylation were independently associated with reduced survival (p = 0.037 and 0.0058, respectively). While ADI did not independently predict survival, it correlated with late-stage presentation and reduced HOXA7 methylation.
Conclusions
Exposome factors such as environmental burden and occupational exposure are biologically embedded in lung cancer cell through gene-specific methylation and significantly impact survival. We posit that integrating exposomic and molecular data could enhance lung precision oncology approaches for high-risk veteran populations.
Insights Into Veterans’ Motivations and Hesitancies for COVID-19 Vaccine Uptake: A Mixed-Methods Analysis
Insights Into Veterans’ Motivations and Hesitancies for COVID-19 Vaccine Uptake: A Mixed-Methods Analysis
The SARS-CoV-2 virus has resulted in > 778 million reported COVID-19 cases and > 7 million deaths worldwide. 1 About 70% of the eligible US population has completed a primary COVID-19 vaccination series, yet only 17% have received an updated bivalent booster dose.2 These immunization rates fall below the World Health Organization (WHO) target of 70%.3
Early in the pandemic, US Department of Veterans Affairs (VA) vaccination rates ranged from 46% to 71%.4,5 Ensuring a high level of COVID-19 vaccination in the largest integrated US health care system aligns with the VA priority to provide high-quality, evidence-based care to a patient population that is older and has more comorbidities than the overall US population.6-9
Vaccine hesitancy, defined as a “delay in acceptance or refusal of vaccination despite availability of vaccination service,” is a major contributor to suboptimal vaccination rates.10-13 Previous studies used cluster analyses to identify the unique combinations of behavioral and social factors responsible for COVID-19 vaccine hesitancy.10,11 Lack of perceived vaccine effectiveness and low perceived risk of the health consequences from COVID-19 infection were frequently identified in clusters where patients had the lowest intent for vaccination.10,11 Similarly, low trust in health care practitioners (HCPs), government, and pharmaceutical companies diminished intent for vaccination in these clusters.10 These quantitative studies were limited by their exclusive focus on unvaccinated individuals, reliance on self-reported intent, and lack of assessment of a health care system with a COVID-19 vaccine delivery program designed to overcome barriers to health care access, such as the VA.
Prior qualitative studies of vaccine uptake in distinct veteran subgroups (ie, unhoused and in VA facilities with low vaccination rates) demonstrated that overriding medical priorities among the unhoused and vaccine safety concerns were associated with decreased vaccine uptake, and positive perceptions of HCPs and the health care system were associated with increased vaccine uptake.11,12 However, these studies were conducted during periods of greater COVID-19 vaccine availability and acceptance, and prior to booster recommendations.4,12,13
This mixed-methods quality improvement (QI) project assessed the barriers and facilitators of COVID-19 vaccination among veterans receiving primary care at a single VA health care facility. We assessed whether unique patient clusters could be identified based on COVID-19–related and vaccine-related thoughts and feelings and whether cluster membership was associated with COVID-19 vaccination. This analysis also explored how individuals’ beliefs and trust shaped motivations and hesitancies for vaccine uptake in quantitatively derived clusters with varying vaccination rates.
Methods
This QI project was conducted at the VA Pittsburgh Healthcare System (VAPHS), a tertiary care facility serving > 75,000 veterans in Pennsylvania, West Virginia, and Ohio. The VAPHS Institutional Review Board determined this QI study was exempt from review.14-17 Participation was voluntary and had no bearing on VA health care or benefits. Financial support for the project, including key personnel and participant compensation, was provided by VAPHS. We followed the STROBE reporting guideline for cross-sectional studies and the COREQ checklist for qualitative research.18,19
Quantitative Survey
The 32,271 veterans assigned to a VAPHS primary care HCP, effective April 1, 2020, were eligible. To ensure representation of subgroups underrecognized in research and/or QI projects, the sample included all 1980 female patients at VAPHS and a random sample of 500 White and 500 Hispanic and/or non-White men within 4 age categories (< 50, 50-64, 65-84, and > 84 years). For the < 50 years or > 84 years categories, all Hispanic and/or non-White men were included due to small sample sizes.20-22 The nonrandom sampling frame comprised 1708 Hispanic and/or non-White men and 2000 White men. After assigning the 5688 potentially eligible individuals a unique identifier, 31 opted out, resulting in a final sample of 5657 individuals.
The 5657 individuals received a letter requesting their completion of a future questionnaire about COVID-19 infection and vaccines. An electronic Qualtrics questionnaire link was emailed to 3221 individuals; nonresponders received 2 follow-up email reminders. For the 2436 veterans without an email address on file, trained interviewers conducted phone surveys and entered responses. Those patients who completed the questionnaire could enter a drawing to win 1 of 100 cash prizes valued at $100. We collected questionnaire data from July to September 2021.
Questionnaire Items
We constructed a 60-item questionnaire based on prior research on COVID-19 vaccine hesitancy and the WHO Guidebook for Immunization Programs and Implementing Partners.4,23-25 The WHO Guidebook comprises survey items organized within 4 domains reflecting the behavioral and social determinants of vaccination: thoughts and feelings; social processes; motivation and hesitancy; and practical factors.23
Sociodemographic, clinical, and personal characteristics. The survey assessed respondent ethnicity and race and used these data to create a composite race and ethnicity variable. Highest educational level was also attained using 8 response options. The survey also assessed prior COVID-19 infection; prior receipt of vaccines for influenza, pneumonia, tetanus, or shingles; and presence of comorbidities that increase the risk of severe COVID-19 infection. We used administrative data from the VA Corporate Data Warehouse to determine respondent age, sex, geographic residence (urban, rural), and to fill in missing self-reported data on sex (n = 4) and ethnicity and race (n = 12). The survey assessed political views using a 5-point Likert scale (1, very liberal; 5, very conservative) and was collapsed into 3 categories (ie, very conservative or conservative, moderate, very liberal or liberal), with prefer not to answer reported separately
COVID-19 infection and vaccine. We asked veterans if they had ever been infected with COVID-19, whether they had been offered and/or received a COVID-19 vaccine, and type (Pfizer, Moderna, or Johnson & Johnson), and number of doses received. Positive vaccination status was defined as the receipt of ≥ 1 dose of a COVID-19 vaccine approved by the US Food and Drug Administration.
COVID-19 opinions. Respondents were asked about perceived risk of COVID-19 infection and related health outcomes, as well as beliefs about COVID-19 vaccines, using a 4-point Likert scale for all items: (1, not at all concerned; 4, very concerned). Respondents were asked about concerns related to COVID-19 infection and severe illness. They also were asked about vaccine-related short-term adverse effects (AEs) and long-term complications. Respondents were asked how effective they believed COVID-19 vaccines were at preventing infection, serious illness, or death. Unvaccinated and vaccinated veterans were asked similar items, with a qualifier of “before getting vaccinated…” for those who were vaccinated.
Social processes. Respondents were asked to rate their level of trust in various sources of COVID-19 vaccine information using a 4-point Likert scale (1, trust not at all; 4, trust very much). Respondents were asked whether community or religious leaders or close family or friends wanted them to get vaccinated (yes, no, or unsure).
Practical factors. Respondents were asked to rate the logistical difficulty of getting vaccinated or trying to get vaccinated using a 4-point Likert scale (1, not at all; 4, extremely).
Participants
Respondents were asked to participate in a follow-up qualitative interview. Among 293 participants who agreed, we sampled all 86 unvaccinated individuals regardless of cluster assignment, a random sample of 88 individuals in the cluster with the lowest vaccination rate, and all 33 vaccinated individuals in the cluster with the second-lowest vaccination rate. Forty-nine veterans completed qualitative interviews.
Two research staff trained in qualitative research completed telephone interviews, averaging 16.5 minutes (March to May 2022), using semistructured scripts to elicit vaccine-related motivations, hesitancies, or concerns. Interviews were recorded, transcribed, and deidentified. Participants provided written consent for recording and received $50 cash-equivalent compensation for interview completion.
Qualitative Interview Script
The interview script consisted of open-ended questions related to vaccine uptake across WHO domains.23 Both unvaccinated and vaccinated respondents were asked similar questions and customized questions about boosters for the vaccinated subgroup. To assess motivations and hesitancies, respondents were asked how they made their decisions about vaccination and what they considered when deciding. Vaccinated participants were asked about motivations and overcoming concerns. Unvaccinated respondents were asked about reasons for concern. To assess social processes, the interviewers asked participants whose opinion or counsel they trusted when deciding whether to get vaccinated. Questions also focused on positive experiences and vaccination barriers. Vaccinated participants were asked what could have improved their vaccination experiences. Finally, the interviewers asked participants who received a complete primary vaccine series about their motivations and plans related to booster vaccines, and whether information about emerging COVID-19 variants influenced their decisions.
Data Analyses
This analysis used X2 and Fisher exact tests to assess the associations among respondent characteristics, questionnaire responses, vaccination status, and cluster membership. Items phrased similarly were handled in a similar fashion for vaccinated and unvaccinated respondents.
Cluster analysis assessed the possible groupings in responses to the quantitative questionnaire items focused on thoughts and feelings about COVID-19 infection risk and severity, vaccine effectiveness, and vaccine safety. This analysis treated the items’ ordinal response categories as continuous. We performed factor analysis using principal component analysis to explore dimension reduction and account for covariance between items. Two principal components were calculated and applied k-means clustering, determining the number of clusters through agreement from the elbow, gap statistic, and silhouette methods.26 Each cluster was named based on its unique pattern of responses to the items used to define them (eAppendix 1).

Multivariable logistic regression analyses assessed the independent association between cluster membership as the independent measure and vaccination status as the dependent measure, adjusting for respondent sociodemographic and personal characteristics and 2 measures of trust (ie, local VA HCP and the CDC). We selected these trust measures because they represent objective sources of medical information and were independently associated with COVID-19 vaccination status in a logistic regression model comprising all 6 trust items assessed.
This study defined statistical significance as a 2-tailed P value < .05. SAS 9.4 was used for all statistical analyses and Python 3.7.4 and the Scikit-learn package for cluster analyses.27 For qualitative analyses, this study used an inductive thematic approach guided by conventional qualitative content analysis, NVivo 12 Plus for Windows to code and analyze interview transcripts.28,29 We created an initial codebook based on 10 transcripts that were selected for high complexity and represented cluster membership and vaccination status.30,31 After 2 qualitative staff developed the initial codebook, 11 of 49 (22%) transcripts were independently coded by a primary and secondary coder to ensure consistent code application. Both coders reviewed the cocoded transcripts and resolved all discrepancies through negotiated consensus.32 After the cocoding process was complete, the primary coder coded the remaining transcripts. The primary and secondary coder met as needed to review and discuss any questions that arose during the primary coder’s work.
Results
Of 5657 eligible participants, 1208 (21.4%) completed a questionnaire. Overall, 674 (55.8%) were aged < 65 years, 530 (43.9%) were women, 828 (68.5%) were non-Hispanic White, 303 (25.1%) were Black, and 47 (3.9%) were Hispanic, and 1034 (85.6%) were vaccinated (Table 1). Compared to the total sampled population, respondents were more often older, female, and White (eAppendix 2).


Cluster Membership
Four clusters were identified from 1183 (97.9%) participants who provided complete responses to 6 items assessing thoughts and feelings about COVID-19 infection and vaccines (Table 2). Of the 1183 respondents, 375 (31.7%) were Concerned Believers (cluster 1), 336 (28.4%) were Unconcerned Believers (cluster 2), 298 (25.2%) were Concerned Ambivalents (cluster 3), and 174 (14.7%) were Unconcerned Disbelievers (cluster 4). The Concerned Believers were moderately/ very concerned about COVID-19 infection (96.0%) and becoming very ill from infection (94.6%), believed the vaccine was moderately/very effective in preventing COVID-19 infection (100%) and severe illness or death from infection (98.7%), and had slight concern about short-term AEs (92.6%) or long-term complications (92.0%) from the vaccine. The Unconcerned Believers had no/slight concern about COVID-19 infection (76.5%) or becoming very ill (79.2%), believed the vaccine was effective in preventing infection (82.4%) and severe illness and death (83.6%), and had no/slight concern about short-term AEs (94.0%) or long-term complications (87.2%) from the vaccine. The Concerned Ambivalents were moderately/ very concerned about COVID-19 infection (94.3%) and becoming very ill (93.6%), believed the vaccine was moderately/very effective in preventing infection (86.6%) and severe illness or death (86.9%), and were moderately/very concerned about short-term AEs (81.9%) or long-term complications (89.3%) from the vaccine. The Unconcerned Disbelievers had no/slight concern about COVID-19 infection (90.8%) and becoming very ill (88.6%), believed the vaccine was not at all/slightly effective in preventing infection (90.3%) and severe illness or death (87.4%), and were moderately/very concerned about short-term AEs (52.8%) or long-term complications (75.9%) from the vaccine.

Cluster Membership
Respondent age, race and ethnicity, and political viewpoints differed significantly by cluster (P < .001). Compared with the other clusters, the Concerned Believer cluster was older (55.5% age ≥ 65 years vs 16.7%-48.0%) and more frequently reported liberal political views (28.8% vs 4.6%-15.1%). In contrast, the Unconcerned Disbeliever cluster was younger (83.4% age ≤ 64 years vs 44.5%-56.8%) and more frequently reported conservative political views (37.9% vs 17.1%-26.8%) than the other clusters. Whereas the Concerned Ambivalent cluster had the highest proportion of Black (37.7%) and the lowest proportion of White respondents (57.6%), the Unconcerned Disbelievers cluster had the lowest proportion of Black respondents (14.5%) and the highest proportion of White respondents (77.9%). The Unconcerned Disbelievers cluster were significantly less likely to trust COVID-19 vaccine information from any source and to believe those close to them wanted them to get vaccinated.
Association of Cluster Membership and COVID-19 Vaccination
COVID-19 vaccination rates varied more than 3-fold (P < .001) by cluster, with 29.9% of Unconcerned Disbelievers, 93.3% of Concerned Ambivalents, 93.5% of Unconcerned Believers, and 98.9% of Concerned Believers reporting being vaccinated. (Figure). Cluster membership was independently associated with vaccination, with adjusted odds ratios (AORs) of 12.0 (95% CI, 6.1-23.8) for the Concerned Ambivalent, 13.0 (95% CI, 6.9-24.5) for Unconcerned Believer, and 48.6 (95% CI, 15.5-152.1) for Concerned Believer clusters (Table 3). Respondent trust in COVID-19 vaccine information from their VA HCP (AOR 2.1; 95% CI, 1.6-2.8) and the CDC (AOR 1.6; 95% CI, 1.2-2.1) were independently associated with vaccination status, while the remaining respondent sociodemographic or personal characteristics were not.


Qualitative Interview Participants
A 49-participant convenience sample completed interviews, including 30 Concerned Ambivalent, 17 Unconcerned Disbeliever, and 2 Unconcerned Believer respondents cluster. The data were not calculated for Unconcerned Believers due to the small sample size. Interview participants were more likely to be younger, female, non-Hispanic, White, less educated, and more politically conservative than the questionnaire respondents as a whole (Appendix). The vaccination rate for the interview participants was 73.5%, ranging from 29.9% in the Unconcerned Disbeliever to 93.3% in the Concerned Ambivalent cluster. Qualitative themes and participant quotes for Concerned Ambivalent and Unconcerned Disbeliever respondents are in eAppendix 3.
Motivations. Wanting personal protection from becoming infected or severely ill from COVID-19 (63.8%), caregiver wanting to protect others (17.0%), and employment vaccine requirements (14.9%) were frequent motivations for vaccination. Whereas personal protection (90.0%) and protection of others (23.3%) were identified more frequently in the Concerned Ambivalents cluster, employment vaccine requirements (35.3%) were more frequently identified in the Unconcerned Disbelievers cluster.
Hesitancies or concerns. Lack of sufficient information related to rapid vaccine development (55.3%), vaccine AEs (38.3%), and low confidence in vaccine efficacy (23.4%) were frequent concerns or hesitancies about vaccination. Unconcerned Disbelievers expressed higher levels of concern about the vaccine’s rapid development (82.4%), low perceived vaccine efficacy (47.1%), and a lack of trust in governmental vaccine promotion (23.5%) than did the Concerned Ambivalents.
Overcoming concerns. Not wanting to get sick or die from infection coupled with an understanding that vaccine benefits exceed risks (23.4%) and receiving information from a trusted source (10.6%) were common ways of overcoming concerns for vaccination. Although the Unconcerned Disbelievers infrequently identified reasons for overcoming concerns, they identified employment requirements (17.6%) as a reason for vaccination despite concerns. They also identified seeing others with positive vaccine experiences and pressure from family or friends as ways of overcoming concerns (11.8% each).
Social influences. Family members or partners (38.3%), personal opinions (38.3%), and HCPs (23.4%) were frequent social influences for vaccination. Concerned Ambivalents mentioned family members and partners (46.7%), HCPs (26.7%), and friends (20.0%) as common influences, while Unconcerned Disbelievers more frequently relied on their opinion (41.2%) and quoted specific scientifically reputable data sources (17.6%) to guide vaccine decision-making, although it is unclear whether these sources were accessed directly or if this information was obtained indirectly through scientifically unvetted data platforms.
Practical factors. Most participants had positive vaccination experiences (68.1%), determined mainly by the Concerned Ambivalents (90.0%), who were more highly vaccinated. Barriers to vaccination were reported by 9 (19.1%) participants, driven by those in the Concerned Ambivalent cluster (26.7%). Eight (17.0%) participants suggested improvements for vaccination processes, with similar overall reporting frequencies across clusters.
COVID-19 boosters and variants. Wanting continued protection from COVID-19 (36.2%), recommendations from a doctor or trusted source (17.0%), and news about emerging variants (10.6%) were frequent motivations for receiving a vaccine booster (eAppendix 4). These motivations were largely driven by the Concerned Ambivalents, of whom 25 of 30 were booster eligible and 24 received a booster dose. Belief that boosters were unnecessary (8.5%), concerns about efficacy (6.4%), and concerns about AEs (6.4%) were frequently identified hesitancies. These concerns were expressed largely by the Unconcerned Disbelievers, of whom 7 of 17 were booster dose eligible, but only 1 received a dose.
Evolving knowledge about variants was not a major concern overall and did not change existing opinions about the vaccine (36.2%). Concerned Ambivalents believed vaccination provided extra protection against variants (36.7%) and the emergence of variants served as a reminder of the ongoing pandemic (30.0%). In contrast, Unconcerned Disbelievers believed that the threat of variants was overblown (35.3%) and mutations are to be expected (17.6%).
Discussion
This study used a complementary mixed-methods approach to understand the motivations, hesitancies, and social and practical drivers of COVID-19 vaccine uptake among VA beneficiaries. Our quantitative analyses identified 4 distinct clusters based on respondents’ opinions on COVID-19 infection severity and vaccine effectiveness and safety. Veterans in 3 clusters were 12 to 49 times more likely to be vaccinated than those in the remaining cluster, even when controlling for baseline respondent characteristics and level of trust in credible sources of COVID-19 information. The observed vaccination rate of nearly 86% was higher than the contemporaneous national average of 62% for vaccine-eligible individuals, likely reflecting the comprehensive VA vaccine promotion strategies tailored to a patient demographic with a high COVID-19 risk profile.2,10

This cluster analyses demonstrated the importance of thoughts and feelings about COVID-19 infection and vaccination as influential social and behavioral drivers of vaccine uptake. These opinions help explain the strong association between cluster membership and vaccination status in this multivariable modeling. The cluster composition was consistent with findings from studies of nonveteran populations that identified perceived vulnerability to COVID-19 infection, beliefs in vaccine effectiveness, and adherence with protective behaviors during the pandemic as contributors to vaccine uptake.13,33 Qualitative themes showed that personal protection, protecting others, and vaccine mandates were frequent motivators for vaccination. Whereas protection of self and others from COVID-19 infection were more often expressed by the highly vaccinated Concerned Ambivalents, employment and travel vaccine mandates were more often identified by Unconcerned Disbelievers, who had a lower vaccination rate. Among Unconcerned Disbelievers, an employer vaccine requirement was the most frequent qualitative theme for overcoming vaccination concerns.
In addition to cluster membership, our modeling showed that trust in local VA HCPs and the CDC were independently associated with COVID-19 vaccination, which has been found in prior research.20 This qualitative analyses regarding vaccine hesitancy identified trust-related concerns that were more frequently expressed by Unconcerned Disbelievers than Concerned Ambivalents. Concerns included the rapid development of the vaccines potentially limiting the generation of scientifically sound effectiveness and safety data, and potential biases involving the entities promoting vaccine uptake.
Whereas the Concerned Believers, Unconcerned Believers, and Concerned Ambivalents all had high COVID-19 vaccination rates (≥ 93%), the decision-making pathways to vaccine uptake likely differ by their concerns about COVID-19 infection and perceptions of vaccine safety and effectiveness. For example, this mixed-methods analysis consistently showed that people in the Concerned Ambivalent cluster were positively motivated by concerns about COVID-19 infection and severity and beliefs about vaccine effectiveness that were tempered by concerns about vaccine AEs. For this cluster, their frequent thematic expression that the benefits of the vaccine exceed the risks, and the positive social influences of family, friends, and HCPs may explain their high vaccination rate.
Such insights into how the patterns of COVID-19–related thoughts and feelings vary across clusters can be used to design interventions to encourage initial and booster doses of COVID-19 vaccines. For example, messaging that highlights the infectivity and severity of COVID-19 and the potential for persistent negative health outcomes associated with long COVID could reinforce the beliefs of Concerned Believers and Concerned Ambivalents, and such messaging could also be used as a targeted intervention for Unconcerned Believers who expressed fewer concerns about the health consequences of COVID-19.23 Likewise, messaging about the safety profile of COVID-19 vaccines may reduce vaccine hesitancy for Concerned Ambivalents. Importantly, purposeful attention to health equity, community engagement, and involvement of racially diverse HCPs in patient discussions represent successful strategies to increase COVID-19 vaccine uptake among Black individuals, who were disproportionately represented in the Concerned Ambivalent cluster and may possess higher levels of mistrust due to racism experienced within the health care system.24
Our findings suggest that the greatest challenge for overcoming vaccine hesitancy is for individuals in the suboptimally vaccinated (30%) Unconcerned Disbeliever cluster. These individuals had low levels of concern about COVID-19 infection and severity, high levels of concern about vaccine safety, low perceived vaccine effectiveness, and low levels of trust in all information sources about COVID-19. While the Unconcerned Disbelievers cited scientifically reputable data sources, we were unable to verify whether participants accessed these reputable sources of information directly or obtained such information indirectly through potentially biased online sources. Nearly half of this cluster trusted their VA HCP and believed their community or religious leaders would want them to get vaccinated. This qualitative analyses found that Unconcerned Disbelievers relied on personal beliefs for vaccine decision-making more than Concerned Ambivalents. While Unconcerned Disbelievers were less likely to be socially influenced by family, friends, or religious leaders, they still acknowledged some impact from these sources. These findings suggest that addressing vaccine hesitancy among Unconcerned Disbelievers may require a multifaceted approach that respects their reliance on personal research while also leveraging the potential social influences. This approach supports the promising, previously reported practices of harnessing the social influences of HCPs and other community and religious leaders to promote vaccine uptake among Unconcerned Disbelievers.34,35 One evidence-based approach to effectively change patient health care behaviors is through motivational interviewing strategies that use open-ended questions, nonjudgmental interactions, and collaborative decision-making when discussing the risks and benefits of vaccination.21,22
Limitations
This study was conducted at a single VA health care facility and our sampling technique was nonrandom, suggesting that these results may not be generalizable to all veterans or non-VA patient populations. The 21% questionnaire response rate could have introduced selection bias into the respondent sample. All questionnaire data were self-reported, including vaccination status. Finally, the qualitative interviews consisted of a small number of unvaccinated individuals in 2 clusters (ie, Concerned Ambivalents and Unconcerned Disbelievers) and may not have reached thematic saturation in these subgroups.
Conclusions
Quantitative analyses identified 4 clusters based on individual thoughts and feelings about COVID-19 infection and vaccines. Cluster membership and levels of trust in COVID-19 information sources were independently associated with vaccination. Understanding the quantitative patterns of thoughts and beliefs across clusters, enriched by common qualitative themes for vaccine hesitancy, help inform tailored interventions to augment COVID-19 vaccine uptake and highlight the importance of targeted, trust-based communication and culturally sensitive interventions to enhance vaccine uptake across diverse populations.
- World Health Organization. WHO COVID-19 dashboard. Accessed July 18, 2025. https://covid19.who.int/
- Centers for Disease Control and Prevention. COVIDVax- View: Weekly COVID-19 Vaccination Coverage and Intent among Adults. Accessed June 10, 2025. https://www.cdc.gov/covidvaxview/weekly-dashboard/adult-vaccination-coverage.html
- World Health Organization. Strategy to achieve global Covid-19 vaccination by mid-2022. 2021. Accessed April 30, 2025. https://cdn.who.int/media/docs/default-source/immunization/covid-19/strategy-to-achieve-global-covid-19-vaccination-by-mid-2022.pdf
- Jasuja GK, Meterko M, Bradshaw LD, et al. Attitudes and intentions of US veterans regarding COVID-19 vaccination. JAMA Netw Open. 2021;4(11):e2132548. doi:10.1001/jamanetworkopen.2021.32548
- Der-Martirosian C, Steers WN, Northcraft H, Chu K, Dobalian A. Vaccinating veterans for COVID-19 at the U.S. Department of Veterans Affairs. Am J Prev Med. 2022;62(6):e317-e324. doi:10.1016/j.amepre.2021.12.016
- Bloeser K, Lipkowitz-Eaton J. Disproportionate multimorbidity among veterans in middle age. J Public Health (Oxf). 2022;44(1):28-35. doi:10.1093/pubmed/fdab149
- US Department of Veterans Affairs. National Center for Veterans Analysis and Statistics: veteran population. Updated March 26, 2025. Accessed April 30, 2025. https://www.va.gov/vetdata/Veteran_Population.asp
- Olenick M, Flowers M, Diaz VJ. US veterans and their unique issues: enhancing health care professional awareness. Adv Med Educ Pract. 2015;6:635-639. doi:10.2147/AMEP.S89479
- Orkaby AR, Nussbaum L, Ho YL, et al. The burden of frailty among U.S. veterans and its association with mortality, 2002-2012. J Gerontol A Biol Sci Med Sci. 2019;74(8):1257-1264. doi:10.1093/gerona/gly232
- Bass SB, Kelly PJ, Hoadley A, Arroyo Lloret A, Organtini T. Mapping perceptual differences to understand COVID-19 beliefs in those with vaccine hesitancy. J Health Commun. 2022;27(1):49-61. doi:10.1080/10810730.2022.2042627
- Meng L, Masters NB, Lu PJ, et al. Cluster analysis of adults unvaccinated for COVID-19 based on behavioral and social factors, National Immunization Survey-Adult COVID Module, United States. Prev Med. 2023;167:107415. doi:10.1016/j.ypmed.2022.107415
- Gin JL, Balut MD, Dobalian A. COVID-19 vaccination uptake and receptivity among veterans enrolled in homelessness- tailored primary health care clinics: provider trust vs. misinformation. BMC Prim Care. 2024;25(1):24. doi:10.1186/s12875-023-02251-x
- Wilson GM, Ray CE, Kale IO, et al. Age and beliefs about vaccines associated with COVID-19 vaccination among US veterans. Antimicrob Steward Healthc Epidemiol. 2023;3(1):e184. doi:10.1017/ash.2023.446
- VA Pittsburgh Healthcare System (VAPHS). Human Research Protection Program (HRPP) policy for quality assurance/ quality improvement projects. Policy H-013. December 31, 2021. Accessed April 30, 2025. https://www.va.gov/files/2020-11/H-013_QAQI%20Project_revised_updated%20format_clean_508.pdf
- Burkitt KH, Rodriguez KL, Mor MK, et al. Evaluation of a collaborative VA network initiative to reduce racial disparities in blood pressure control among veterans with severe hypertension. Healthc (Amst). 2021;8(suppl 1):100485. doi:10.1016/j.hjdsi.2020.100485
- Sinkowitz-Cochran RL, Burkitt KH, Cuerdon T, et al. The associations between organizational culture and knowledge, attitudes, and practices in a multicenter Veterans Affairs quality improvement initiative to prevent methicillin-resistant Staphylococcus aureus. Am J Infect Control. 2012;40(2):138-143. doi:10.1016/j.ajic.2011.04.332
- Burkitt KH, Sinkowitz-Cochran RL, Obrosky DS, et al. Survey of employee knowledge and attitudes before and after a multicenter Veterans’ Administration quality improvement initiative to reduce nosocomial methicillin-resistant Staphylococcus aureus infections. Am J Infect Control. 2010;38(4):274-282. doi:10.1016/j.ajic.2009.08.019
- STROBE - strengthening the reporting of observational studies in epidemiology. What is STROBE? Accessed April 30, 2025. https://www.strobe-statement.org/
- Tong A, Sainsbury P, Craig J. Consolidated criteria for reporting qualitative research (COREQ): a 32-item checklist for interviews and focus groups. Int J Qual Health Care. 2007;19(6):349-357. doi:10.1093/intqhc/mzm042
- Ward RE, Nguyen XT, Li Y, et al; on behalf of the VA Million Veteran Program. Racial and ethnic disparities in U.S. veteran health characteristics. Int J Environ Res Public Health. 2021;18(5):2411. doi:10.3390/ijerph18052411
- Harrington KM, Nguyen XT, Song RJ, et al; VA Million Veteran Program. Gender differences in demographic and health characteristics of the Million Veteran Program cohort. Womens Health Issues. 2019;29(suppl 1):S56-S66. doi:10.1016/j.whi.2019.04.012
- Washington DL, ed. National Veteran Health Equity Report 2021. Focus on Veterans Health Administration Patient Experience and Health Care Quality. VHA Office of Health Equity; September 2022. Accessed April 30, 2025. https://www.va.gov/healthequity/nvher.asp
- World Health Organization. Data for action: achieving high uptake of COVID-19 vaccines. April 1, 2021. Accessed April 30, 2025. https://www.who.int/publications/i/item/WHO-2019-nCoV-vaccination-demand-planning-2021.1
- Hoffman BL, Boness CL, Chu KH, et al. COVID- 19 vaccine hesitancy, acceptance, and promotion among healthcare workers: a mixed-methods analysis. J Community Health. 2022;47(5):750-758. doi:10.1007/s10900-022-01095-3
- Vasudevan L, Bruening R, Hung A, et al. COVID- 19 vaccination intention and activation among health care system employees: a mixed methods study. Vaccine. 2022;40(35):5141-5152. doi:10.1016/j.vaccine.2022.07.010
- Tibshirani R, Walther G, Hastie T. Estimating the number of clusters in a data set via the gap statistic. J R Stat Soc Series B Stat Methodol. 2001;63(2):411-423. doi:10.1111/1467-9868.00293
- Pedregosa FP, Varoquaux G, Gramfort A, et al. Scikitlearn: machine learning in Python. J Mach Learn Res. 2011;12:2825-2830.
- Proudfoot K. Inductive/deductive hybrid thematic analysis in mixed methods research. J Mix Methods Res. 2022;17(3): 308-326. doi:10.1177/15586898221126816
- Chapman AL, Hadfield M, Chapman CJ. Qualitative research in healthcare: an introduction to grounded theory using thematic analysis. J R Coll Physicians Edinb. 2015;45(3):201-205. doi:10.4997/jrcpe.2015.305
- Grandheim UH, Lundman B. Qualitative content analysis in nursing research: concepts, procedures and measures to achieve trustworthiness. Nurse Educ Today. 2004;24(2):105-112. doi:10.1016/j.nedt.2003.1001
- Sandelowski M. Whatever happened to qualitative description? Res Nurs Health. 2000;23(4):334-340. doi:10.1002/1098-240x(200008)23:4<334::aid-nur9 >3.0.co;2-g
- Garrison DR, Cleveland-Innes M, Koole M, Kappelman J. Revisiting methodological issues in transcript analysis: negotiated coding and reliability. Internet High Educ. 2006;9(1):1-8. doi:10.1016/j.iheduc.2005.11.001
- Wagner AL, Porth JM, Wu Z, Boulton ML, Finlay JM, Kobayashi LC. Vaccine hesitancy during the COVID-19 pandemic: a latent class analysis of middle-aged and older US adults. J Community Health. 2022;47(3):408- 415. doi:10.1007/s10900-022-01064-w
- Syed U, Kapera O, Chandrasekhar A, et al. The role of faith-based organizations in improving vaccination confidence & addressing vaccination disparities to help improve vaccine uptake: a systematic review. Vaccines (Basel). 2023;11(2):449. doi:10.3390/vaccines11020449
- Evans D, Norrbom C, Schmidt S, Powell R, McReynolds J, Sidibe T. Engaging community-based organizations to address barriers in public health programs: lessons learned from COVID-19 vaccine acceptance programs in diverse rural communities. Health Secur. 2023;21(S1):S17-S24. doi:10.1089/hs.2023.0017
The SARS-CoV-2 virus has resulted in > 778 million reported COVID-19 cases and > 7 million deaths worldwide. 1 About 70% of the eligible US population has completed a primary COVID-19 vaccination series, yet only 17% have received an updated bivalent booster dose.2 These immunization rates fall below the World Health Organization (WHO) target of 70%.3
Early in the pandemic, US Department of Veterans Affairs (VA) vaccination rates ranged from 46% to 71%.4,5 Ensuring a high level of COVID-19 vaccination in the largest integrated US health care system aligns with the VA priority to provide high-quality, evidence-based care to a patient population that is older and has more comorbidities than the overall US population.6-9
Vaccine hesitancy, defined as a “delay in acceptance or refusal of vaccination despite availability of vaccination service,” is a major contributor to suboptimal vaccination rates.10-13 Previous studies used cluster analyses to identify the unique combinations of behavioral and social factors responsible for COVID-19 vaccine hesitancy.10,11 Lack of perceived vaccine effectiveness and low perceived risk of the health consequences from COVID-19 infection were frequently identified in clusters where patients had the lowest intent for vaccination.10,11 Similarly, low trust in health care practitioners (HCPs), government, and pharmaceutical companies diminished intent for vaccination in these clusters.10 These quantitative studies were limited by their exclusive focus on unvaccinated individuals, reliance on self-reported intent, and lack of assessment of a health care system with a COVID-19 vaccine delivery program designed to overcome barriers to health care access, such as the VA.
Prior qualitative studies of vaccine uptake in distinct veteran subgroups (ie, unhoused and in VA facilities with low vaccination rates) demonstrated that overriding medical priorities among the unhoused and vaccine safety concerns were associated with decreased vaccine uptake, and positive perceptions of HCPs and the health care system were associated with increased vaccine uptake.11,12 However, these studies were conducted during periods of greater COVID-19 vaccine availability and acceptance, and prior to booster recommendations.4,12,13
This mixed-methods quality improvement (QI) project assessed the barriers and facilitators of COVID-19 vaccination among veterans receiving primary care at a single VA health care facility. We assessed whether unique patient clusters could be identified based on COVID-19–related and vaccine-related thoughts and feelings and whether cluster membership was associated with COVID-19 vaccination. This analysis also explored how individuals’ beliefs and trust shaped motivations and hesitancies for vaccine uptake in quantitatively derived clusters with varying vaccination rates.
Methods
This QI project was conducted at the VA Pittsburgh Healthcare System (VAPHS), a tertiary care facility serving > 75,000 veterans in Pennsylvania, West Virginia, and Ohio. The VAPHS Institutional Review Board determined this QI study was exempt from review.14-17 Participation was voluntary and had no bearing on VA health care or benefits. Financial support for the project, including key personnel and participant compensation, was provided by VAPHS. We followed the STROBE reporting guideline for cross-sectional studies and the COREQ checklist for qualitative research.18,19
Quantitative Survey
The 32,271 veterans assigned to a VAPHS primary care HCP, effective April 1, 2020, were eligible. To ensure representation of subgroups underrecognized in research and/or QI projects, the sample included all 1980 female patients at VAPHS and a random sample of 500 White and 500 Hispanic and/or non-White men within 4 age categories (< 50, 50-64, 65-84, and > 84 years). For the < 50 years or > 84 years categories, all Hispanic and/or non-White men were included due to small sample sizes.20-22 The nonrandom sampling frame comprised 1708 Hispanic and/or non-White men and 2000 White men. After assigning the 5688 potentially eligible individuals a unique identifier, 31 opted out, resulting in a final sample of 5657 individuals.
The 5657 individuals received a letter requesting their completion of a future questionnaire about COVID-19 infection and vaccines. An electronic Qualtrics questionnaire link was emailed to 3221 individuals; nonresponders received 2 follow-up email reminders. For the 2436 veterans without an email address on file, trained interviewers conducted phone surveys and entered responses. Those patients who completed the questionnaire could enter a drawing to win 1 of 100 cash prizes valued at $100. We collected questionnaire data from July to September 2021.
Questionnaire Items
We constructed a 60-item questionnaire based on prior research on COVID-19 vaccine hesitancy and the WHO Guidebook for Immunization Programs and Implementing Partners.4,23-25 The WHO Guidebook comprises survey items organized within 4 domains reflecting the behavioral and social determinants of vaccination: thoughts and feelings; social processes; motivation and hesitancy; and practical factors.23
Sociodemographic, clinical, and personal characteristics. The survey assessed respondent ethnicity and race and used these data to create a composite race and ethnicity variable. Highest educational level was also attained using 8 response options. The survey also assessed prior COVID-19 infection; prior receipt of vaccines for influenza, pneumonia, tetanus, or shingles; and presence of comorbidities that increase the risk of severe COVID-19 infection. We used administrative data from the VA Corporate Data Warehouse to determine respondent age, sex, geographic residence (urban, rural), and to fill in missing self-reported data on sex (n = 4) and ethnicity and race (n = 12). The survey assessed political views using a 5-point Likert scale (1, very liberal; 5, very conservative) and was collapsed into 3 categories (ie, very conservative or conservative, moderate, very liberal or liberal), with prefer not to answer reported separately
COVID-19 infection and vaccine. We asked veterans if they had ever been infected with COVID-19, whether they had been offered and/or received a COVID-19 vaccine, and type (Pfizer, Moderna, or Johnson & Johnson), and number of doses received. Positive vaccination status was defined as the receipt of ≥ 1 dose of a COVID-19 vaccine approved by the US Food and Drug Administration.
COVID-19 opinions. Respondents were asked about perceived risk of COVID-19 infection and related health outcomes, as well as beliefs about COVID-19 vaccines, using a 4-point Likert scale for all items: (1, not at all concerned; 4, very concerned). Respondents were asked about concerns related to COVID-19 infection and severe illness. They also were asked about vaccine-related short-term adverse effects (AEs) and long-term complications. Respondents were asked how effective they believed COVID-19 vaccines were at preventing infection, serious illness, or death. Unvaccinated and vaccinated veterans were asked similar items, with a qualifier of “before getting vaccinated…” for those who were vaccinated.
Social processes. Respondents were asked to rate their level of trust in various sources of COVID-19 vaccine information using a 4-point Likert scale (1, trust not at all; 4, trust very much). Respondents were asked whether community or religious leaders or close family or friends wanted them to get vaccinated (yes, no, or unsure).
Practical factors. Respondents were asked to rate the logistical difficulty of getting vaccinated or trying to get vaccinated using a 4-point Likert scale (1, not at all; 4, extremely).
Participants
Respondents were asked to participate in a follow-up qualitative interview. Among 293 participants who agreed, we sampled all 86 unvaccinated individuals regardless of cluster assignment, a random sample of 88 individuals in the cluster with the lowest vaccination rate, and all 33 vaccinated individuals in the cluster with the second-lowest vaccination rate. Forty-nine veterans completed qualitative interviews.
Two research staff trained in qualitative research completed telephone interviews, averaging 16.5 minutes (March to May 2022), using semistructured scripts to elicit vaccine-related motivations, hesitancies, or concerns. Interviews were recorded, transcribed, and deidentified. Participants provided written consent for recording and received $50 cash-equivalent compensation for interview completion.
Qualitative Interview Script
The interview script consisted of open-ended questions related to vaccine uptake across WHO domains.23 Both unvaccinated and vaccinated respondents were asked similar questions and customized questions about boosters for the vaccinated subgroup. To assess motivations and hesitancies, respondents were asked how they made their decisions about vaccination and what they considered when deciding. Vaccinated participants were asked about motivations and overcoming concerns. Unvaccinated respondents were asked about reasons for concern. To assess social processes, the interviewers asked participants whose opinion or counsel they trusted when deciding whether to get vaccinated. Questions also focused on positive experiences and vaccination barriers. Vaccinated participants were asked what could have improved their vaccination experiences. Finally, the interviewers asked participants who received a complete primary vaccine series about their motivations and plans related to booster vaccines, and whether information about emerging COVID-19 variants influenced their decisions.
Data Analyses
This analysis used X2 and Fisher exact tests to assess the associations among respondent characteristics, questionnaire responses, vaccination status, and cluster membership. Items phrased similarly were handled in a similar fashion for vaccinated and unvaccinated respondents.
Cluster analysis assessed the possible groupings in responses to the quantitative questionnaire items focused on thoughts and feelings about COVID-19 infection risk and severity, vaccine effectiveness, and vaccine safety. This analysis treated the items’ ordinal response categories as continuous. We performed factor analysis using principal component analysis to explore dimension reduction and account for covariance between items. Two principal components were calculated and applied k-means clustering, determining the number of clusters through agreement from the elbow, gap statistic, and silhouette methods.26 Each cluster was named based on its unique pattern of responses to the items used to define them (eAppendix 1).

Multivariable logistic regression analyses assessed the independent association between cluster membership as the independent measure and vaccination status as the dependent measure, adjusting for respondent sociodemographic and personal characteristics and 2 measures of trust (ie, local VA HCP and the CDC). We selected these trust measures because they represent objective sources of medical information and were independently associated with COVID-19 vaccination status in a logistic regression model comprising all 6 trust items assessed.
This study defined statistical significance as a 2-tailed P value < .05. SAS 9.4 was used for all statistical analyses and Python 3.7.4 and the Scikit-learn package for cluster analyses.27 For qualitative analyses, this study used an inductive thematic approach guided by conventional qualitative content analysis, NVivo 12 Plus for Windows to code and analyze interview transcripts.28,29 We created an initial codebook based on 10 transcripts that were selected for high complexity and represented cluster membership and vaccination status.30,31 After 2 qualitative staff developed the initial codebook, 11 of 49 (22%) transcripts were independently coded by a primary and secondary coder to ensure consistent code application. Both coders reviewed the cocoded transcripts and resolved all discrepancies through negotiated consensus.32 After the cocoding process was complete, the primary coder coded the remaining transcripts. The primary and secondary coder met as needed to review and discuss any questions that arose during the primary coder’s work.
Results
Of 5657 eligible participants, 1208 (21.4%) completed a questionnaire. Overall, 674 (55.8%) were aged < 65 years, 530 (43.9%) were women, 828 (68.5%) were non-Hispanic White, 303 (25.1%) were Black, and 47 (3.9%) were Hispanic, and 1034 (85.6%) were vaccinated (Table 1). Compared to the total sampled population, respondents were more often older, female, and White (eAppendix 2).


Cluster Membership
Four clusters were identified from 1183 (97.9%) participants who provided complete responses to 6 items assessing thoughts and feelings about COVID-19 infection and vaccines (Table 2). Of the 1183 respondents, 375 (31.7%) were Concerned Believers (cluster 1), 336 (28.4%) were Unconcerned Believers (cluster 2), 298 (25.2%) were Concerned Ambivalents (cluster 3), and 174 (14.7%) were Unconcerned Disbelievers (cluster 4). The Concerned Believers were moderately/ very concerned about COVID-19 infection (96.0%) and becoming very ill from infection (94.6%), believed the vaccine was moderately/very effective in preventing COVID-19 infection (100%) and severe illness or death from infection (98.7%), and had slight concern about short-term AEs (92.6%) or long-term complications (92.0%) from the vaccine. The Unconcerned Believers had no/slight concern about COVID-19 infection (76.5%) or becoming very ill (79.2%), believed the vaccine was effective in preventing infection (82.4%) and severe illness and death (83.6%), and had no/slight concern about short-term AEs (94.0%) or long-term complications (87.2%) from the vaccine. The Concerned Ambivalents were moderately/ very concerned about COVID-19 infection (94.3%) and becoming very ill (93.6%), believed the vaccine was moderately/very effective in preventing infection (86.6%) and severe illness or death (86.9%), and were moderately/very concerned about short-term AEs (81.9%) or long-term complications (89.3%) from the vaccine. The Unconcerned Disbelievers had no/slight concern about COVID-19 infection (90.8%) and becoming very ill (88.6%), believed the vaccine was not at all/slightly effective in preventing infection (90.3%) and severe illness or death (87.4%), and were moderately/very concerned about short-term AEs (52.8%) or long-term complications (75.9%) from the vaccine.

Cluster Membership
Respondent age, race and ethnicity, and political viewpoints differed significantly by cluster (P < .001). Compared with the other clusters, the Concerned Believer cluster was older (55.5% age ≥ 65 years vs 16.7%-48.0%) and more frequently reported liberal political views (28.8% vs 4.6%-15.1%). In contrast, the Unconcerned Disbeliever cluster was younger (83.4% age ≤ 64 years vs 44.5%-56.8%) and more frequently reported conservative political views (37.9% vs 17.1%-26.8%) than the other clusters. Whereas the Concerned Ambivalent cluster had the highest proportion of Black (37.7%) and the lowest proportion of White respondents (57.6%), the Unconcerned Disbelievers cluster had the lowest proportion of Black respondents (14.5%) and the highest proportion of White respondents (77.9%). The Unconcerned Disbelievers cluster were significantly less likely to trust COVID-19 vaccine information from any source and to believe those close to them wanted them to get vaccinated.
Association of Cluster Membership and COVID-19 Vaccination
COVID-19 vaccination rates varied more than 3-fold (P < .001) by cluster, with 29.9% of Unconcerned Disbelievers, 93.3% of Concerned Ambivalents, 93.5% of Unconcerned Believers, and 98.9% of Concerned Believers reporting being vaccinated. (Figure). Cluster membership was independently associated with vaccination, with adjusted odds ratios (AORs) of 12.0 (95% CI, 6.1-23.8) for the Concerned Ambivalent, 13.0 (95% CI, 6.9-24.5) for Unconcerned Believer, and 48.6 (95% CI, 15.5-152.1) for Concerned Believer clusters (Table 3). Respondent trust in COVID-19 vaccine information from their VA HCP (AOR 2.1; 95% CI, 1.6-2.8) and the CDC (AOR 1.6; 95% CI, 1.2-2.1) were independently associated with vaccination status, while the remaining respondent sociodemographic or personal characteristics were not.


Qualitative Interview Participants
A 49-participant convenience sample completed interviews, including 30 Concerned Ambivalent, 17 Unconcerned Disbeliever, and 2 Unconcerned Believer respondents cluster. The data were not calculated for Unconcerned Believers due to the small sample size. Interview participants were more likely to be younger, female, non-Hispanic, White, less educated, and more politically conservative than the questionnaire respondents as a whole (Appendix). The vaccination rate for the interview participants was 73.5%, ranging from 29.9% in the Unconcerned Disbeliever to 93.3% in the Concerned Ambivalent cluster. Qualitative themes and participant quotes for Concerned Ambivalent and Unconcerned Disbeliever respondents are in eAppendix 3.
Motivations. Wanting personal protection from becoming infected or severely ill from COVID-19 (63.8%), caregiver wanting to protect others (17.0%), and employment vaccine requirements (14.9%) were frequent motivations for vaccination. Whereas personal protection (90.0%) and protection of others (23.3%) were identified more frequently in the Concerned Ambivalents cluster, employment vaccine requirements (35.3%) were more frequently identified in the Unconcerned Disbelievers cluster.
Hesitancies or concerns. Lack of sufficient information related to rapid vaccine development (55.3%), vaccine AEs (38.3%), and low confidence in vaccine efficacy (23.4%) were frequent concerns or hesitancies about vaccination. Unconcerned Disbelievers expressed higher levels of concern about the vaccine’s rapid development (82.4%), low perceived vaccine efficacy (47.1%), and a lack of trust in governmental vaccine promotion (23.5%) than did the Concerned Ambivalents.
Overcoming concerns. Not wanting to get sick or die from infection coupled with an understanding that vaccine benefits exceed risks (23.4%) and receiving information from a trusted source (10.6%) were common ways of overcoming concerns for vaccination. Although the Unconcerned Disbelievers infrequently identified reasons for overcoming concerns, they identified employment requirements (17.6%) as a reason for vaccination despite concerns. They also identified seeing others with positive vaccine experiences and pressure from family or friends as ways of overcoming concerns (11.8% each).
Social influences. Family members or partners (38.3%), personal opinions (38.3%), and HCPs (23.4%) were frequent social influences for vaccination. Concerned Ambivalents mentioned family members and partners (46.7%), HCPs (26.7%), and friends (20.0%) as common influences, while Unconcerned Disbelievers more frequently relied on their opinion (41.2%) and quoted specific scientifically reputable data sources (17.6%) to guide vaccine decision-making, although it is unclear whether these sources were accessed directly or if this information was obtained indirectly through scientifically unvetted data platforms.
Practical factors. Most participants had positive vaccination experiences (68.1%), determined mainly by the Concerned Ambivalents (90.0%), who were more highly vaccinated. Barriers to vaccination were reported by 9 (19.1%) participants, driven by those in the Concerned Ambivalent cluster (26.7%). Eight (17.0%) participants suggested improvements for vaccination processes, with similar overall reporting frequencies across clusters.
COVID-19 boosters and variants. Wanting continued protection from COVID-19 (36.2%), recommendations from a doctor or trusted source (17.0%), and news about emerging variants (10.6%) were frequent motivations for receiving a vaccine booster (eAppendix 4). These motivations were largely driven by the Concerned Ambivalents, of whom 25 of 30 were booster eligible and 24 received a booster dose. Belief that boosters were unnecessary (8.5%), concerns about efficacy (6.4%), and concerns about AEs (6.4%) were frequently identified hesitancies. These concerns were expressed largely by the Unconcerned Disbelievers, of whom 7 of 17 were booster dose eligible, but only 1 received a dose.
Evolving knowledge about variants was not a major concern overall and did not change existing opinions about the vaccine (36.2%). Concerned Ambivalents believed vaccination provided extra protection against variants (36.7%) and the emergence of variants served as a reminder of the ongoing pandemic (30.0%). In contrast, Unconcerned Disbelievers believed that the threat of variants was overblown (35.3%) and mutations are to be expected (17.6%).
Discussion
This study used a complementary mixed-methods approach to understand the motivations, hesitancies, and social and practical drivers of COVID-19 vaccine uptake among VA beneficiaries. Our quantitative analyses identified 4 distinct clusters based on respondents’ opinions on COVID-19 infection severity and vaccine effectiveness and safety. Veterans in 3 clusters were 12 to 49 times more likely to be vaccinated than those in the remaining cluster, even when controlling for baseline respondent characteristics and level of trust in credible sources of COVID-19 information. The observed vaccination rate of nearly 86% was higher than the contemporaneous national average of 62% for vaccine-eligible individuals, likely reflecting the comprehensive VA vaccine promotion strategies tailored to a patient demographic with a high COVID-19 risk profile.2,10

This cluster analyses demonstrated the importance of thoughts and feelings about COVID-19 infection and vaccination as influential social and behavioral drivers of vaccine uptake. These opinions help explain the strong association between cluster membership and vaccination status in this multivariable modeling. The cluster composition was consistent with findings from studies of nonveteran populations that identified perceived vulnerability to COVID-19 infection, beliefs in vaccine effectiveness, and adherence with protective behaviors during the pandemic as contributors to vaccine uptake.13,33 Qualitative themes showed that personal protection, protecting others, and vaccine mandates were frequent motivators for vaccination. Whereas protection of self and others from COVID-19 infection were more often expressed by the highly vaccinated Concerned Ambivalents, employment and travel vaccine mandates were more often identified by Unconcerned Disbelievers, who had a lower vaccination rate. Among Unconcerned Disbelievers, an employer vaccine requirement was the most frequent qualitative theme for overcoming vaccination concerns.
In addition to cluster membership, our modeling showed that trust in local VA HCPs and the CDC were independently associated with COVID-19 vaccination, which has been found in prior research.20 This qualitative analyses regarding vaccine hesitancy identified trust-related concerns that were more frequently expressed by Unconcerned Disbelievers than Concerned Ambivalents. Concerns included the rapid development of the vaccines potentially limiting the generation of scientifically sound effectiveness and safety data, and potential biases involving the entities promoting vaccine uptake.
Whereas the Concerned Believers, Unconcerned Believers, and Concerned Ambivalents all had high COVID-19 vaccination rates (≥ 93%), the decision-making pathways to vaccine uptake likely differ by their concerns about COVID-19 infection and perceptions of vaccine safety and effectiveness. For example, this mixed-methods analysis consistently showed that people in the Concerned Ambivalent cluster were positively motivated by concerns about COVID-19 infection and severity and beliefs about vaccine effectiveness that were tempered by concerns about vaccine AEs. For this cluster, their frequent thematic expression that the benefits of the vaccine exceed the risks, and the positive social influences of family, friends, and HCPs may explain their high vaccination rate.
Such insights into how the patterns of COVID-19–related thoughts and feelings vary across clusters can be used to design interventions to encourage initial and booster doses of COVID-19 vaccines. For example, messaging that highlights the infectivity and severity of COVID-19 and the potential for persistent negative health outcomes associated with long COVID could reinforce the beliefs of Concerned Believers and Concerned Ambivalents, and such messaging could also be used as a targeted intervention for Unconcerned Believers who expressed fewer concerns about the health consequences of COVID-19.23 Likewise, messaging about the safety profile of COVID-19 vaccines may reduce vaccine hesitancy for Concerned Ambivalents. Importantly, purposeful attention to health equity, community engagement, and involvement of racially diverse HCPs in patient discussions represent successful strategies to increase COVID-19 vaccine uptake among Black individuals, who were disproportionately represented in the Concerned Ambivalent cluster and may possess higher levels of mistrust due to racism experienced within the health care system.24
Our findings suggest that the greatest challenge for overcoming vaccine hesitancy is for individuals in the suboptimally vaccinated (30%) Unconcerned Disbeliever cluster. These individuals had low levels of concern about COVID-19 infection and severity, high levels of concern about vaccine safety, low perceived vaccine effectiveness, and low levels of trust in all information sources about COVID-19. While the Unconcerned Disbelievers cited scientifically reputable data sources, we were unable to verify whether participants accessed these reputable sources of information directly or obtained such information indirectly through potentially biased online sources. Nearly half of this cluster trusted their VA HCP and believed their community or religious leaders would want them to get vaccinated. This qualitative analyses found that Unconcerned Disbelievers relied on personal beliefs for vaccine decision-making more than Concerned Ambivalents. While Unconcerned Disbelievers were less likely to be socially influenced by family, friends, or religious leaders, they still acknowledged some impact from these sources. These findings suggest that addressing vaccine hesitancy among Unconcerned Disbelievers may require a multifaceted approach that respects their reliance on personal research while also leveraging the potential social influences. This approach supports the promising, previously reported practices of harnessing the social influences of HCPs and other community and religious leaders to promote vaccine uptake among Unconcerned Disbelievers.34,35 One evidence-based approach to effectively change patient health care behaviors is through motivational interviewing strategies that use open-ended questions, nonjudgmental interactions, and collaborative decision-making when discussing the risks and benefits of vaccination.21,22
Limitations
This study was conducted at a single VA health care facility and our sampling technique was nonrandom, suggesting that these results may not be generalizable to all veterans or non-VA patient populations. The 21% questionnaire response rate could have introduced selection bias into the respondent sample. All questionnaire data were self-reported, including vaccination status. Finally, the qualitative interviews consisted of a small number of unvaccinated individuals in 2 clusters (ie, Concerned Ambivalents and Unconcerned Disbelievers) and may not have reached thematic saturation in these subgroups.
Conclusions
Quantitative analyses identified 4 clusters based on individual thoughts and feelings about COVID-19 infection and vaccines. Cluster membership and levels of trust in COVID-19 information sources were independently associated with vaccination. Understanding the quantitative patterns of thoughts and beliefs across clusters, enriched by common qualitative themes for vaccine hesitancy, help inform tailored interventions to augment COVID-19 vaccine uptake and highlight the importance of targeted, trust-based communication and culturally sensitive interventions to enhance vaccine uptake across diverse populations.
The SARS-CoV-2 virus has resulted in > 778 million reported COVID-19 cases and > 7 million deaths worldwide. 1 About 70% of the eligible US population has completed a primary COVID-19 vaccination series, yet only 17% have received an updated bivalent booster dose.2 These immunization rates fall below the World Health Organization (WHO) target of 70%.3
Early in the pandemic, US Department of Veterans Affairs (VA) vaccination rates ranged from 46% to 71%.4,5 Ensuring a high level of COVID-19 vaccination in the largest integrated US health care system aligns with the VA priority to provide high-quality, evidence-based care to a patient population that is older and has more comorbidities than the overall US population.6-9
Vaccine hesitancy, defined as a “delay in acceptance or refusal of vaccination despite availability of vaccination service,” is a major contributor to suboptimal vaccination rates.10-13 Previous studies used cluster analyses to identify the unique combinations of behavioral and social factors responsible for COVID-19 vaccine hesitancy.10,11 Lack of perceived vaccine effectiveness and low perceived risk of the health consequences from COVID-19 infection were frequently identified in clusters where patients had the lowest intent for vaccination.10,11 Similarly, low trust in health care practitioners (HCPs), government, and pharmaceutical companies diminished intent for vaccination in these clusters.10 These quantitative studies were limited by their exclusive focus on unvaccinated individuals, reliance on self-reported intent, and lack of assessment of a health care system with a COVID-19 vaccine delivery program designed to overcome barriers to health care access, such as the VA.
Prior qualitative studies of vaccine uptake in distinct veteran subgroups (ie, unhoused and in VA facilities with low vaccination rates) demonstrated that overriding medical priorities among the unhoused and vaccine safety concerns were associated with decreased vaccine uptake, and positive perceptions of HCPs and the health care system were associated with increased vaccine uptake.11,12 However, these studies were conducted during periods of greater COVID-19 vaccine availability and acceptance, and prior to booster recommendations.4,12,13
This mixed-methods quality improvement (QI) project assessed the barriers and facilitators of COVID-19 vaccination among veterans receiving primary care at a single VA health care facility. We assessed whether unique patient clusters could be identified based on COVID-19–related and vaccine-related thoughts and feelings and whether cluster membership was associated with COVID-19 vaccination. This analysis also explored how individuals’ beliefs and trust shaped motivations and hesitancies for vaccine uptake in quantitatively derived clusters with varying vaccination rates.
Methods
This QI project was conducted at the VA Pittsburgh Healthcare System (VAPHS), a tertiary care facility serving > 75,000 veterans in Pennsylvania, West Virginia, and Ohio. The VAPHS Institutional Review Board determined this QI study was exempt from review.14-17 Participation was voluntary and had no bearing on VA health care or benefits. Financial support for the project, including key personnel and participant compensation, was provided by VAPHS. We followed the STROBE reporting guideline for cross-sectional studies and the COREQ checklist for qualitative research.18,19
Quantitative Survey
The 32,271 veterans assigned to a VAPHS primary care HCP, effective April 1, 2020, were eligible. To ensure representation of subgroups underrecognized in research and/or QI projects, the sample included all 1980 female patients at VAPHS and a random sample of 500 White and 500 Hispanic and/or non-White men within 4 age categories (< 50, 50-64, 65-84, and > 84 years). For the < 50 years or > 84 years categories, all Hispanic and/or non-White men were included due to small sample sizes.20-22 The nonrandom sampling frame comprised 1708 Hispanic and/or non-White men and 2000 White men. After assigning the 5688 potentially eligible individuals a unique identifier, 31 opted out, resulting in a final sample of 5657 individuals.
The 5657 individuals received a letter requesting their completion of a future questionnaire about COVID-19 infection and vaccines. An electronic Qualtrics questionnaire link was emailed to 3221 individuals; nonresponders received 2 follow-up email reminders. For the 2436 veterans without an email address on file, trained interviewers conducted phone surveys and entered responses. Those patients who completed the questionnaire could enter a drawing to win 1 of 100 cash prizes valued at $100. We collected questionnaire data from July to September 2021.
Questionnaire Items
We constructed a 60-item questionnaire based on prior research on COVID-19 vaccine hesitancy and the WHO Guidebook for Immunization Programs and Implementing Partners.4,23-25 The WHO Guidebook comprises survey items organized within 4 domains reflecting the behavioral and social determinants of vaccination: thoughts and feelings; social processes; motivation and hesitancy; and practical factors.23
Sociodemographic, clinical, and personal characteristics. The survey assessed respondent ethnicity and race and used these data to create a composite race and ethnicity variable. Highest educational level was also attained using 8 response options. The survey also assessed prior COVID-19 infection; prior receipt of vaccines for influenza, pneumonia, tetanus, or shingles; and presence of comorbidities that increase the risk of severe COVID-19 infection. We used administrative data from the VA Corporate Data Warehouse to determine respondent age, sex, geographic residence (urban, rural), and to fill in missing self-reported data on sex (n = 4) and ethnicity and race (n = 12). The survey assessed political views using a 5-point Likert scale (1, very liberal; 5, very conservative) and was collapsed into 3 categories (ie, very conservative or conservative, moderate, very liberal or liberal), with prefer not to answer reported separately
COVID-19 infection and vaccine. We asked veterans if they had ever been infected with COVID-19, whether they had been offered and/or received a COVID-19 vaccine, and type (Pfizer, Moderna, or Johnson & Johnson), and number of doses received. Positive vaccination status was defined as the receipt of ≥ 1 dose of a COVID-19 vaccine approved by the US Food and Drug Administration.
COVID-19 opinions. Respondents were asked about perceived risk of COVID-19 infection and related health outcomes, as well as beliefs about COVID-19 vaccines, using a 4-point Likert scale for all items: (1, not at all concerned; 4, very concerned). Respondents were asked about concerns related to COVID-19 infection and severe illness. They also were asked about vaccine-related short-term adverse effects (AEs) and long-term complications. Respondents were asked how effective they believed COVID-19 vaccines were at preventing infection, serious illness, or death. Unvaccinated and vaccinated veterans were asked similar items, with a qualifier of “before getting vaccinated…” for those who were vaccinated.
Social processes. Respondents were asked to rate their level of trust in various sources of COVID-19 vaccine information using a 4-point Likert scale (1, trust not at all; 4, trust very much). Respondents were asked whether community or religious leaders or close family or friends wanted them to get vaccinated (yes, no, or unsure).
Practical factors. Respondents were asked to rate the logistical difficulty of getting vaccinated or trying to get vaccinated using a 4-point Likert scale (1, not at all; 4, extremely).
Participants
Respondents were asked to participate in a follow-up qualitative interview. Among 293 participants who agreed, we sampled all 86 unvaccinated individuals regardless of cluster assignment, a random sample of 88 individuals in the cluster with the lowest vaccination rate, and all 33 vaccinated individuals in the cluster with the second-lowest vaccination rate. Forty-nine veterans completed qualitative interviews.
Two research staff trained in qualitative research completed telephone interviews, averaging 16.5 minutes (March to May 2022), using semistructured scripts to elicit vaccine-related motivations, hesitancies, or concerns. Interviews were recorded, transcribed, and deidentified. Participants provided written consent for recording and received $50 cash-equivalent compensation for interview completion.
Qualitative Interview Script
The interview script consisted of open-ended questions related to vaccine uptake across WHO domains.23 Both unvaccinated and vaccinated respondents were asked similar questions and customized questions about boosters for the vaccinated subgroup. To assess motivations and hesitancies, respondents were asked how they made their decisions about vaccination and what they considered when deciding. Vaccinated participants were asked about motivations and overcoming concerns. Unvaccinated respondents were asked about reasons for concern. To assess social processes, the interviewers asked participants whose opinion or counsel they trusted when deciding whether to get vaccinated. Questions also focused on positive experiences and vaccination barriers. Vaccinated participants were asked what could have improved their vaccination experiences. Finally, the interviewers asked participants who received a complete primary vaccine series about their motivations and plans related to booster vaccines, and whether information about emerging COVID-19 variants influenced their decisions.
Data Analyses
This analysis used X2 and Fisher exact tests to assess the associations among respondent characteristics, questionnaire responses, vaccination status, and cluster membership. Items phrased similarly were handled in a similar fashion for vaccinated and unvaccinated respondents.
Cluster analysis assessed the possible groupings in responses to the quantitative questionnaire items focused on thoughts and feelings about COVID-19 infection risk and severity, vaccine effectiveness, and vaccine safety. This analysis treated the items’ ordinal response categories as continuous. We performed factor analysis using principal component analysis to explore dimension reduction and account for covariance between items. Two principal components were calculated and applied k-means clustering, determining the number of clusters through agreement from the elbow, gap statistic, and silhouette methods.26 Each cluster was named based on its unique pattern of responses to the items used to define them (eAppendix 1).

Multivariable logistic regression analyses assessed the independent association between cluster membership as the independent measure and vaccination status as the dependent measure, adjusting for respondent sociodemographic and personal characteristics and 2 measures of trust (ie, local VA HCP and the CDC). We selected these trust measures because they represent objective sources of medical information and were independently associated with COVID-19 vaccination status in a logistic regression model comprising all 6 trust items assessed.
This study defined statistical significance as a 2-tailed P value < .05. SAS 9.4 was used for all statistical analyses and Python 3.7.4 and the Scikit-learn package for cluster analyses.27 For qualitative analyses, this study used an inductive thematic approach guided by conventional qualitative content analysis, NVivo 12 Plus for Windows to code and analyze interview transcripts.28,29 We created an initial codebook based on 10 transcripts that were selected for high complexity and represented cluster membership and vaccination status.30,31 After 2 qualitative staff developed the initial codebook, 11 of 49 (22%) transcripts were independently coded by a primary and secondary coder to ensure consistent code application. Both coders reviewed the cocoded transcripts and resolved all discrepancies through negotiated consensus.32 After the cocoding process was complete, the primary coder coded the remaining transcripts. The primary and secondary coder met as needed to review and discuss any questions that arose during the primary coder’s work.
Results
Of 5657 eligible participants, 1208 (21.4%) completed a questionnaire. Overall, 674 (55.8%) were aged < 65 years, 530 (43.9%) were women, 828 (68.5%) were non-Hispanic White, 303 (25.1%) were Black, and 47 (3.9%) were Hispanic, and 1034 (85.6%) were vaccinated (Table 1). Compared to the total sampled population, respondents were more often older, female, and White (eAppendix 2).


Cluster Membership
Four clusters were identified from 1183 (97.9%) participants who provided complete responses to 6 items assessing thoughts and feelings about COVID-19 infection and vaccines (Table 2). Of the 1183 respondents, 375 (31.7%) were Concerned Believers (cluster 1), 336 (28.4%) were Unconcerned Believers (cluster 2), 298 (25.2%) were Concerned Ambivalents (cluster 3), and 174 (14.7%) were Unconcerned Disbelievers (cluster 4). The Concerned Believers were moderately/ very concerned about COVID-19 infection (96.0%) and becoming very ill from infection (94.6%), believed the vaccine was moderately/very effective in preventing COVID-19 infection (100%) and severe illness or death from infection (98.7%), and had slight concern about short-term AEs (92.6%) or long-term complications (92.0%) from the vaccine. The Unconcerned Believers had no/slight concern about COVID-19 infection (76.5%) or becoming very ill (79.2%), believed the vaccine was effective in preventing infection (82.4%) and severe illness and death (83.6%), and had no/slight concern about short-term AEs (94.0%) or long-term complications (87.2%) from the vaccine. The Concerned Ambivalents were moderately/ very concerned about COVID-19 infection (94.3%) and becoming very ill (93.6%), believed the vaccine was moderately/very effective in preventing infection (86.6%) and severe illness or death (86.9%), and were moderately/very concerned about short-term AEs (81.9%) or long-term complications (89.3%) from the vaccine. The Unconcerned Disbelievers had no/slight concern about COVID-19 infection (90.8%) and becoming very ill (88.6%), believed the vaccine was not at all/slightly effective in preventing infection (90.3%) and severe illness or death (87.4%), and were moderately/very concerned about short-term AEs (52.8%) or long-term complications (75.9%) from the vaccine.

Cluster Membership
Respondent age, race and ethnicity, and political viewpoints differed significantly by cluster (P < .001). Compared with the other clusters, the Concerned Believer cluster was older (55.5% age ≥ 65 years vs 16.7%-48.0%) and more frequently reported liberal political views (28.8% vs 4.6%-15.1%). In contrast, the Unconcerned Disbeliever cluster was younger (83.4% age ≤ 64 years vs 44.5%-56.8%) and more frequently reported conservative political views (37.9% vs 17.1%-26.8%) than the other clusters. Whereas the Concerned Ambivalent cluster had the highest proportion of Black (37.7%) and the lowest proportion of White respondents (57.6%), the Unconcerned Disbelievers cluster had the lowest proportion of Black respondents (14.5%) and the highest proportion of White respondents (77.9%). The Unconcerned Disbelievers cluster were significantly less likely to trust COVID-19 vaccine information from any source and to believe those close to them wanted them to get vaccinated.
Association of Cluster Membership and COVID-19 Vaccination
COVID-19 vaccination rates varied more than 3-fold (P < .001) by cluster, with 29.9% of Unconcerned Disbelievers, 93.3% of Concerned Ambivalents, 93.5% of Unconcerned Believers, and 98.9% of Concerned Believers reporting being vaccinated. (Figure). Cluster membership was independently associated with vaccination, with adjusted odds ratios (AORs) of 12.0 (95% CI, 6.1-23.8) for the Concerned Ambivalent, 13.0 (95% CI, 6.9-24.5) for Unconcerned Believer, and 48.6 (95% CI, 15.5-152.1) for Concerned Believer clusters (Table 3). Respondent trust in COVID-19 vaccine information from their VA HCP (AOR 2.1; 95% CI, 1.6-2.8) and the CDC (AOR 1.6; 95% CI, 1.2-2.1) were independently associated with vaccination status, while the remaining respondent sociodemographic or personal characteristics were not.


Qualitative Interview Participants
A 49-participant convenience sample completed interviews, including 30 Concerned Ambivalent, 17 Unconcerned Disbeliever, and 2 Unconcerned Believer respondents cluster. The data were not calculated for Unconcerned Believers due to the small sample size. Interview participants were more likely to be younger, female, non-Hispanic, White, less educated, and more politically conservative than the questionnaire respondents as a whole (Appendix). The vaccination rate for the interview participants was 73.5%, ranging from 29.9% in the Unconcerned Disbeliever to 93.3% in the Concerned Ambivalent cluster. Qualitative themes and participant quotes for Concerned Ambivalent and Unconcerned Disbeliever respondents are in eAppendix 3.
Motivations. Wanting personal protection from becoming infected or severely ill from COVID-19 (63.8%), caregiver wanting to protect others (17.0%), and employment vaccine requirements (14.9%) were frequent motivations for vaccination. Whereas personal protection (90.0%) and protection of others (23.3%) were identified more frequently in the Concerned Ambivalents cluster, employment vaccine requirements (35.3%) were more frequently identified in the Unconcerned Disbelievers cluster.
Hesitancies or concerns. Lack of sufficient information related to rapid vaccine development (55.3%), vaccine AEs (38.3%), and low confidence in vaccine efficacy (23.4%) were frequent concerns or hesitancies about vaccination. Unconcerned Disbelievers expressed higher levels of concern about the vaccine’s rapid development (82.4%), low perceived vaccine efficacy (47.1%), and a lack of trust in governmental vaccine promotion (23.5%) than did the Concerned Ambivalents.
Overcoming concerns. Not wanting to get sick or die from infection coupled with an understanding that vaccine benefits exceed risks (23.4%) and receiving information from a trusted source (10.6%) were common ways of overcoming concerns for vaccination. Although the Unconcerned Disbelievers infrequently identified reasons for overcoming concerns, they identified employment requirements (17.6%) as a reason for vaccination despite concerns. They also identified seeing others with positive vaccine experiences and pressure from family or friends as ways of overcoming concerns (11.8% each).
Social influences. Family members or partners (38.3%), personal opinions (38.3%), and HCPs (23.4%) were frequent social influences for vaccination. Concerned Ambivalents mentioned family members and partners (46.7%), HCPs (26.7%), and friends (20.0%) as common influences, while Unconcerned Disbelievers more frequently relied on their opinion (41.2%) and quoted specific scientifically reputable data sources (17.6%) to guide vaccine decision-making, although it is unclear whether these sources were accessed directly or if this information was obtained indirectly through scientifically unvetted data platforms.
Practical factors. Most participants had positive vaccination experiences (68.1%), determined mainly by the Concerned Ambivalents (90.0%), who were more highly vaccinated. Barriers to vaccination were reported by 9 (19.1%) participants, driven by those in the Concerned Ambivalent cluster (26.7%). Eight (17.0%) participants suggested improvements for vaccination processes, with similar overall reporting frequencies across clusters.
COVID-19 boosters and variants. Wanting continued protection from COVID-19 (36.2%), recommendations from a doctor or trusted source (17.0%), and news about emerging variants (10.6%) were frequent motivations for receiving a vaccine booster (eAppendix 4). These motivations were largely driven by the Concerned Ambivalents, of whom 25 of 30 were booster eligible and 24 received a booster dose. Belief that boosters were unnecessary (8.5%), concerns about efficacy (6.4%), and concerns about AEs (6.4%) were frequently identified hesitancies. These concerns were expressed largely by the Unconcerned Disbelievers, of whom 7 of 17 were booster dose eligible, but only 1 received a dose.
Evolving knowledge about variants was not a major concern overall and did not change existing opinions about the vaccine (36.2%). Concerned Ambivalents believed vaccination provided extra protection against variants (36.7%) and the emergence of variants served as a reminder of the ongoing pandemic (30.0%). In contrast, Unconcerned Disbelievers believed that the threat of variants was overblown (35.3%) and mutations are to be expected (17.6%).
Discussion
This study used a complementary mixed-methods approach to understand the motivations, hesitancies, and social and practical drivers of COVID-19 vaccine uptake among VA beneficiaries. Our quantitative analyses identified 4 distinct clusters based on respondents’ opinions on COVID-19 infection severity and vaccine effectiveness and safety. Veterans in 3 clusters were 12 to 49 times more likely to be vaccinated than those in the remaining cluster, even when controlling for baseline respondent characteristics and level of trust in credible sources of COVID-19 information. The observed vaccination rate of nearly 86% was higher than the contemporaneous national average of 62% for vaccine-eligible individuals, likely reflecting the comprehensive VA vaccine promotion strategies tailored to a patient demographic with a high COVID-19 risk profile.2,10

This cluster analyses demonstrated the importance of thoughts and feelings about COVID-19 infection and vaccination as influential social and behavioral drivers of vaccine uptake. These opinions help explain the strong association between cluster membership and vaccination status in this multivariable modeling. The cluster composition was consistent with findings from studies of nonveteran populations that identified perceived vulnerability to COVID-19 infection, beliefs in vaccine effectiveness, and adherence with protective behaviors during the pandemic as contributors to vaccine uptake.13,33 Qualitative themes showed that personal protection, protecting others, and vaccine mandates were frequent motivators for vaccination. Whereas protection of self and others from COVID-19 infection were more often expressed by the highly vaccinated Concerned Ambivalents, employment and travel vaccine mandates were more often identified by Unconcerned Disbelievers, who had a lower vaccination rate. Among Unconcerned Disbelievers, an employer vaccine requirement was the most frequent qualitative theme for overcoming vaccination concerns.
In addition to cluster membership, our modeling showed that trust in local VA HCPs and the CDC were independently associated with COVID-19 vaccination, which has been found in prior research.20 This qualitative analyses regarding vaccine hesitancy identified trust-related concerns that were more frequently expressed by Unconcerned Disbelievers than Concerned Ambivalents. Concerns included the rapid development of the vaccines potentially limiting the generation of scientifically sound effectiveness and safety data, and potential biases involving the entities promoting vaccine uptake.
Whereas the Concerned Believers, Unconcerned Believers, and Concerned Ambivalents all had high COVID-19 vaccination rates (≥ 93%), the decision-making pathways to vaccine uptake likely differ by their concerns about COVID-19 infection and perceptions of vaccine safety and effectiveness. For example, this mixed-methods analysis consistently showed that people in the Concerned Ambivalent cluster were positively motivated by concerns about COVID-19 infection and severity and beliefs about vaccine effectiveness that were tempered by concerns about vaccine AEs. For this cluster, their frequent thematic expression that the benefits of the vaccine exceed the risks, and the positive social influences of family, friends, and HCPs may explain their high vaccination rate.
Such insights into how the patterns of COVID-19–related thoughts and feelings vary across clusters can be used to design interventions to encourage initial and booster doses of COVID-19 vaccines. For example, messaging that highlights the infectivity and severity of COVID-19 and the potential for persistent negative health outcomes associated with long COVID could reinforce the beliefs of Concerned Believers and Concerned Ambivalents, and such messaging could also be used as a targeted intervention for Unconcerned Believers who expressed fewer concerns about the health consequences of COVID-19.23 Likewise, messaging about the safety profile of COVID-19 vaccines may reduce vaccine hesitancy for Concerned Ambivalents. Importantly, purposeful attention to health equity, community engagement, and involvement of racially diverse HCPs in patient discussions represent successful strategies to increase COVID-19 vaccine uptake among Black individuals, who were disproportionately represented in the Concerned Ambivalent cluster and may possess higher levels of mistrust due to racism experienced within the health care system.24
Our findings suggest that the greatest challenge for overcoming vaccine hesitancy is for individuals in the suboptimally vaccinated (30%) Unconcerned Disbeliever cluster. These individuals had low levels of concern about COVID-19 infection and severity, high levels of concern about vaccine safety, low perceived vaccine effectiveness, and low levels of trust in all information sources about COVID-19. While the Unconcerned Disbelievers cited scientifically reputable data sources, we were unable to verify whether participants accessed these reputable sources of information directly or obtained such information indirectly through potentially biased online sources. Nearly half of this cluster trusted their VA HCP and believed their community or religious leaders would want them to get vaccinated. This qualitative analyses found that Unconcerned Disbelievers relied on personal beliefs for vaccine decision-making more than Concerned Ambivalents. While Unconcerned Disbelievers were less likely to be socially influenced by family, friends, or religious leaders, they still acknowledged some impact from these sources. These findings suggest that addressing vaccine hesitancy among Unconcerned Disbelievers may require a multifaceted approach that respects their reliance on personal research while also leveraging the potential social influences. This approach supports the promising, previously reported practices of harnessing the social influences of HCPs and other community and religious leaders to promote vaccine uptake among Unconcerned Disbelievers.34,35 One evidence-based approach to effectively change patient health care behaviors is through motivational interviewing strategies that use open-ended questions, nonjudgmental interactions, and collaborative decision-making when discussing the risks and benefits of vaccination.21,22
Limitations
This study was conducted at a single VA health care facility and our sampling technique was nonrandom, suggesting that these results may not be generalizable to all veterans or non-VA patient populations. The 21% questionnaire response rate could have introduced selection bias into the respondent sample. All questionnaire data were self-reported, including vaccination status. Finally, the qualitative interviews consisted of a small number of unvaccinated individuals in 2 clusters (ie, Concerned Ambivalents and Unconcerned Disbelievers) and may not have reached thematic saturation in these subgroups.
Conclusions
Quantitative analyses identified 4 clusters based on individual thoughts and feelings about COVID-19 infection and vaccines. Cluster membership and levels of trust in COVID-19 information sources were independently associated with vaccination. Understanding the quantitative patterns of thoughts and beliefs across clusters, enriched by common qualitative themes for vaccine hesitancy, help inform tailored interventions to augment COVID-19 vaccine uptake and highlight the importance of targeted, trust-based communication and culturally sensitive interventions to enhance vaccine uptake across diverse populations.
- World Health Organization. WHO COVID-19 dashboard. Accessed July 18, 2025. https://covid19.who.int/
- Centers for Disease Control and Prevention. COVIDVax- View: Weekly COVID-19 Vaccination Coverage and Intent among Adults. Accessed June 10, 2025. https://www.cdc.gov/covidvaxview/weekly-dashboard/adult-vaccination-coverage.html
- World Health Organization. Strategy to achieve global Covid-19 vaccination by mid-2022. 2021. Accessed April 30, 2025. https://cdn.who.int/media/docs/default-source/immunization/covid-19/strategy-to-achieve-global-covid-19-vaccination-by-mid-2022.pdf
- Jasuja GK, Meterko M, Bradshaw LD, et al. Attitudes and intentions of US veterans regarding COVID-19 vaccination. JAMA Netw Open. 2021;4(11):e2132548. doi:10.1001/jamanetworkopen.2021.32548
- Der-Martirosian C, Steers WN, Northcraft H, Chu K, Dobalian A. Vaccinating veterans for COVID-19 at the U.S. Department of Veterans Affairs. Am J Prev Med. 2022;62(6):e317-e324. doi:10.1016/j.amepre.2021.12.016
- Bloeser K, Lipkowitz-Eaton J. Disproportionate multimorbidity among veterans in middle age. J Public Health (Oxf). 2022;44(1):28-35. doi:10.1093/pubmed/fdab149
- US Department of Veterans Affairs. National Center for Veterans Analysis and Statistics: veteran population. Updated March 26, 2025. Accessed April 30, 2025. https://www.va.gov/vetdata/Veteran_Population.asp
- Olenick M, Flowers M, Diaz VJ. US veterans and their unique issues: enhancing health care professional awareness. Adv Med Educ Pract. 2015;6:635-639. doi:10.2147/AMEP.S89479
- Orkaby AR, Nussbaum L, Ho YL, et al. The burden of frailty among U.S. veterans and its association with mortality, 2002-2012. J Gerontol A Biol Sci Med Sci. 2019;74(8):1257-1264. doi:10.1093/gerona/gly232
- Bass SB, Kelly PJ, Hoadley A, Arroyo Lloret A, Organtini T. Mapping perceptual differences to understand COVID-19 beliefs in those with vaccine hesitancy. J Health Commun. 2022;27(1):49-61. doi:10.1080/10810730.2022.2042627
- Meng L, Masters NB, Lu PJ, et al. Cluster analysis of adults unvaccinated for COVID-19 based on behavioral and social factors, National Immunization Survey-Adult COVID Module, United States. Prev Med. 2023;167:107415. doi:10.1016/j.ypmed.2022.107415
- Gin JL, Balut MD, Dobalian A. COVID-19 vaccination uptake and receptivity among veterans enrolled in homelessness- tailored primary health care clinics: provider trust vs. misinformation. BMC Prim Care. 2024;25(1):24. doi:10.1186/s12875-023-02251-x
- Wilson GM, Ray CE, Kale IO, et al. Age and beliefs about vaccines associated with COVID-19 vaccination among US veterans. Antimicrob Steward Healthc Epidemiol. 2023;3(1):e184. doi:10.1017/ash.2023.446
- VA Pittsburgh Healthcare System (VAPHS). Human Research Protection Program (HRPP) policy for quality assurance/ quality improvement projects. Policy H-013. December 31, 2021. Accessed April 30, 2025. https://www.va.gov/files/2020-11/H-013_QAQI%20Project_revised_updated%20format_clean_508.pdf
- Burkitt KH, Rodriguez KL, Mor MK, et al. Evaluation of a collaborative VA network initiative to reduce racial disparities in blood pressure control among veterans with severe hypertension. Healthc (Amst). 2021;8(suppl 1):100485. doi:10.1016/j.hjdsi.2020.100485
- Sinkowitz-Cochran RL, Burkitt KH, Cuerdon T, et al. The associations between organizational culture and knowledge, attitudes, and practices in a multicenter Veterans Affairs quality improvement initiative to prevent methicillin-resistant Staphylococcus aureus. Am J Infect Control. 2012;40(2):138-143. doi:10.1016/j.ajic.2011.04.332
- Burkitt KH, Sinkowitz-Cochran RL, Obrosky DS, et al. Survey of employee knowledge and attitudes before and after a multicenter Veterans’ Administration quality improvement initiative to reduce nosocomial methicillin-resistant Staphylococcus aureus infections. Am J Infect Control. 2010;38(4):274-282. doi:10.1016/j.ajic.2009.08.019
- STROBE - strengthening the reporting of observational studies in epidemiology. What is STROBE? Accessed April 30, 2025. https://www.strobe-statement.org/
- Tong A, Sainsbury P, Craig J. Consolidated criteria for reporting qualitative research (COREQ): a 32-item checklist for interviews and focus groups. Int J Qual Health Care. 2007;19(6):349-357. doi:10.1093/intqhc/mzm042
- Ward RE, Nguyen XT, Li Y, et al; on behalf of the VA Million Veteran Program. Racial and ethnic disparities in U.S. veteran health characteristics. Int J Environ Res Public Health. 2021;18(5):2411. doi:10.3390/ijerph18052411
- Harrington KM, Nguyen XT, Song RJ, et al; VA Million Veteran Program. Gender differences in demographic and health characteristics of the Million Veteran Program cohort. Womens Health Issues. 2019;29(suppl 1):S56-S66. doi:10.1016/j.whi.2019.04.012
- Washington DL, ed. National Veteran Health Equity Report 2021. Focus on Veterans Health Administration Patient Experience and Health Care Quality. VHA Office of Health Equity; September 2022. Accessed April 30, 2025. https://www.va.gov/healthequity/nvher.asp
- World Health Organization. Data for action: achieving high uptake of COVID-19 vaccines. April 1, 2021. Accessed April 30, 2025. https://www.who.int/publications/i/item/WHO-2019-nCoV-vaccination-demand-planning-2021.1
- Hoffman BL, Boness CL, Chu KH, et al. COVID- 19 vaccine hesitancy, acceptance, and promotion among healthcare workers: a mixed-methods analysis. J Community Health. 2022;47(5):750-758. doi:10.1007/s10900-022-01095-3
- Vasudevan L, Bruening R, Hung A, et al. COVID- 19 vaccination intention and activation among health care system employees: a mixed methods study. Vaccine. 2022;40(35):5141-5152. doi:10.1016/j.vaccine.2022.07.010
- Tibshirani R, Walther G, Hastie T. Estimating the number of clusters in a data set via the gap statistic. J R Stat Soc Series B Stat Methodol. 2001;63(2):411-423. doi:10.1111/1467-9868.00293
- Pedregosa FP, Varoquaux G, Gramfort A, et al. Scikitlearn: machine learning in Python. J Mach Learn Res. 2011;12:2825-2830.
- Proudfoot K. Inductive/deductive hybrid thematic analysis in mixed methods research. J Mix Methods Res. 2022;17(3): 308-326. doi:10.1177/15586898221126816
- Chapman AL, Hadfield M, Chapman CJ. Qualitative research in healthcare: an introduction to grounded theory using thematic analysis. J R Coll Physicians Edinb. 2015;45(3):201-205. doi:10.4997/jrcpe.2015.305
- Grandheim UH, Lundman B. Qualitative content analysis in nursing research: concepts, procedures and measures to achieve trustworthiness. Nurse Educ Today. 2004;24(2):105-112. doi:10.1016/j.nedt.2003.1001
- Sandelowski M. Whatever happened to qualitative description? Res Nurs Health. 2000;23(4):334-340. doi:10.1002/1098-240x(200008)23:4<334::aid-nur9 >3.0.co;2-g
- Garrison DR, Cleveland-Innes M, Koole M, Kappelman J. Revisiting methodological issues in transcript analysis: negotiated coding and reliability. Internet High Educ. 2006;9(1):1-8. doi:10.1016/j.iheduc.2005.11.001
- Wagner AL, Porth JM, Wu Z, Boulton ML, Finlay JM, Kobayashi LC. Vaccine hesitancy during the COVID-19 pandemic: a latent class analysis of middle-aged and older US adults. J Community Health. 2022;47(3):408- 415. doi:10.1007/s10900-022-01064-w
- Syed U, Kapera O, Chandrasekhar A, et al. The role of faith-based organizations in improving vaccination confidence & addressing vaccination disparities to help improve vaccine uptake: a systematic review. Vaccines (Basel). 2023;11(2):449. doi:10.3390/vaccines11020449
- Evans D, Norrbom C, Schmidt S, Powell R, McReynolds J, Sidibe T. Engaging community-based organizations to address barriers in public health programs: lessons learned from COVID-19 vaccine acceptance programs in diverse rural communities. Health Secur. 2023;21(S1):S17-S24. doi:10.1089/hs.2023.0017
- World Health Organization. WHO COVID-19 dashboard. Accessed July 18, 2025. https://covid19.who.int/
- Centers for Disease Control and Prevention. COVIDVax- View: Weekly COVID-19 Vaccination Coverage and Intent among Adults. Accessed June 10, 2025. https://www.cdc.gov/covidvaxview/weekly-dashboard/adult-vaccination-coverage.html
- World Health Organization. Strategy to achieve global Covid-19 vaccination by mid-2022. 2021. Accessed April 30, 2025. https://cdn.who.int/media/docs/default-source/immunization/covid-19/strategy-to-achieve-global-covid-19-vaccination-by-mid-2022.pdf
- Jasuja GK, Meterko M, Bradshaw LD, et al. Attitudes and intentions of US veterans regarding COVID-19 vaccination. JAMA Netw Open. 2021;4(11):e2132548. doi:10.1001/jamanetworkopen.2021.32548
- Der-Martirosian C, Steers WN, Northcraft H, Chu K, Dobalian A. Vaccinating veterans for COVID-19 at the U.S. Department of Veterans Affairs. Am J Prev Med. 2022;62(6):e317-e324. doi:10.1016/j.amepre.2021.12.016
- Bloeser K, Lipkowitz-Eaton J. Disproportionate multimorbidity among veterans in middle age. J Public Health (Oxf). 2022;44(1):28-35. doi:10.1093/pubmed/fdab149
- US Department of Veterans Affairs. National Center for Veterans Analysis and Statistics: veteran population. Updated March 26, 2025. Accessed April 30, 2025. https://www.va.gov/vetdata/Veteran_Population.asp
- Olenick M, Flowers M, Diaz VJ. US veterans and their unique issues: enhancing health care professional awareness. Adv Med Educ Pract. 2015;6:635-639. doi:10.2147/AMEP.S89479
- Orkaby AR, Nussbaum L, Ho YL, et al. The burden of frailty among U.S. veterans and its association with mortality, 2002-2012. J Gerontol A Biol Sci Med Sci. 2019;74(8):1257-1264. doi:10.1093/gerona/gly232
- Bass SB, Kelly PJ, Hoadley A, Arroyo Lloret A, Organtini T. Mapping perceptual differences to understand COVID-19 beliefs in those with vaccine hesitancy. J Health Commun. 2022;27(1):49-61. doi:10.1080/10810730.2022.2042627
- Meng L, Masters NB, Lu PJ, et al. Cluster analysis of adults unvaccinated for COVID-19 based on behavioral and social factors, National Immunization Survey-Adult COVID Module, United States. Prev Med. 2023;167:107415. doi:10.1016/j.ypmed.2022.107415
- Gin JL, Balut MD, Dobalian A. COVID-19 vaccination uptake and receptivity among veterans enrolled in homelessness- tailored primary health care clinics: provider trust vs. misinformation. BMC Prim Care. 2024;25(1):24. doi:10.1186/s12875-023-02251-x
- Wilson GM, Ray CE, Kale IO, et al. Age and beliefs about vaccines associated with COVID-19 vaccination among US veterans. Antimicrob Steward Healthc Epidemiol. 2023;3(1):e184. doi:10.1017/ash.2023.446
- VA Pittsburgh Healthcare System (VAPHS). Human Research Protection Program (HRPP) policy for quality assurance/ quality improvement projects. Policy H-013. December 31, 2021. Accessed April 30, 2025. https://www.va.gov/files/2020-11/H-013_QAQI%20Project_revised_updated%20format_clean_508.pdf
- Burkitt KH, Rodriguez KL, Mor MK, et al. Evaluation of a collaborative VA network initiative to reduce racial disparities in blood pressure control among veterans with severe hypertension. Healthc (Amst). 2021;8(suppl 1):100485. doi:10.1016/j.hjdsi.2020.100485
- Sinkowitz-Cochran RL, Burkitt KH, Cuerdon T, et al. The associations between organizational culture and knowledge, attitudes, and practices in a multicenter Veterans Affairs quality improvement initiative to prevent methicillin-resistant Staphylococcus aureus. Am J Infect Control. 2012;40(2):138-143. doi:10.1016/j.ajic.2011.04.332
- Burkitt KH, Sinkowitz-Cochran RL, Obrosky DS, et al. Survey of employee knowledge and attitudes before and after a multicenter Veterans’ Administration quality improvement initiative to reduce nosocomial methicillin-resistant Staphylococcus aureus infections. Am J Infect Control. 2010;38(4):274-282. doi:10.1016/j.ajic.2009.08.019
- STROBE - strengthening the reporting of observational studies in epidemiology. What is STROBE? Accessed April 30, 2025. https://www.strobe-statement.org/
- Tong A, Sainsbury P, Craig J. Consolidated criteria for reporting qualitative research (COREQ): a 32-item checklist for interviews and focus groups. Int J Qual Health Care. 2007;19(6):349-357. doi:10.1093/intqhc/mzm042
- Ward RE, Nguyen XT, Li Y, et al; on behalf of the VA Million Veteran Program. Racial and ethnic disparities in U.S. veteran health characteristics. Int J Environ Res Public Health. 2021;18(5):2411. doi:10.3390/ijerph18052411
- Harrington KM, Nguyen XT, Song RJ, et al; VA Million Veteran Program. Gender differences in demographic and health characteristics of the Million Veteran Program cohort. Womens Health Issues. 2019;29(suppl 1):S56-S66. doi:10.1016/j.whi.2019.04.012
- Washington DL, ed. National Veteran Health Equity Report 2021. Focus on Veterans Health Administration Patient Experience and Health Care Quality. VHA Office of Health Equity; September 2022. Accessed April 30, 2025. https://www.va.gov/healthequity/nvher.asp
- World Health Organization. Data for action: achieving high uptake of COVID-19 vaccines. April 1, 2021. Accessed April 30, 2025. https://www.who.int/publications/i/item/WHO-2019-nCoV-vaccination-demand-planning-2021.1
- Hoffman BL, Boness CL, Chu KH, et al. COVID- 19 vaccine hesitancy, acceptance, and promotion among healthcare workers: a mixed-methods analysis. J Community Health. 2022;47(5):750-758. doi:10.1007/s10900-022-01095-3
- Vasudevan L, Bruening R, Hung A, et al. COVID- 19 vaccination intention and activation among health care system employees: a mixed methods study. Vaccine. 2022;40(35):5141-5152. doi:10.1016/j.vaccine.2022.07.010
- Tibshirani R, Walther G, Hastie T. Estimating the number of clusters in a data set via the gap statistic. J R Stat Soc Series B Stat Methodol. 2001;63(2):411-423. doi:10.1111/1467-9868.00293
- Pedregosa FP, Varoquaux G, Gramfort A, et al. Scikitlearn: machine learning in Python. J Mach Learn Res. 2011;12:2825-2830.
- Proudfoot K. Inductive/deductive hybrid thematic analysis in mixed methods research. J Mix Methods Res. 2022;17(3): 308-326. doi:10.1177/15586898221126816
- Chapman AL, Hadfield M, Chapman CJ. Qualitative research in healthcare: an introduction to grounded theory using thematic analysis. J R Coll Physicians Edinb. 2015;45(3):201-205. doi:10.4997/jrcpe.2015.305
- Grandheim UH, Lundman B. Qualitative content analysis in nursing research: concepts, procedures and measures to achieve trustworthiness. Nurse Educ Today. 2004;24(2):105-112. doi:10.1016/j.nedt.2003.1001
- Sandelowski M. Whatever happened to qualitative description? Res Nurs Health. 2000;23(4):334-340. doi:10.1002/1098-240x(200008)23:4<334::aid-nur9 >3.0.co;2-g
- Garrison DR, Cleveland-Innes M, Koole M, Kappelman J. Revisiting methodological issues in transcript analysis: negotiated coding and reliability. Internet High Educ. 2006;9(1):1-8. doi:10.1016/j.iheduc.2005.11.001
- Wagner AL, Porth JM, Wu Z, Boulton ML, Finlay JM, Kobayashi LC. Vaccine hesitancy during the COVID-19 pandemic: a latent class analysis of middle-aged and older US adults. J Community Health. 2022;47(3):408- 415. doi:10.1007/s10900-022-01064-w
- Syed U, Kapera O, Chandrasekhar A, et al. The role of faith-based organizations in improving vaccination confidence & addressing vaccination disparities to help improve vaccine uptake: a systematic review. Vaccines (Basel). 2023;11(2):449. doi:10.3390/vaccines11020449
- Evans D, Norrbom C, Schmidt S, Powell R, McReynolds J, Sidibe T. Engaging community-based organizations to address barriers in public health programs: lessons learned from COVID-19 vaccine acceptance programs in diverse rural communities. Health Secur. 2023;21(S1):S17-S24. doi:10.1089/hs.2023.0017
Insights Into Veterans’ Motivations and Hesitancies for COVID-19 Vaccine Uptake: A Mixed-Methods Analysis
Insights Into Veterans’ Motivations and Hesitancies for COVID-19 Vaccine Uptake: A Mixed-Methods Analysis


Radiation and Medical Oncology Perspectives on Oligometastatic Disease Treatment
Radiation and Medical Oncology Perspectives on Oligometastatic Disease Treatment
The treatment of metastatic solid tumors has been based historically on systemic therapies, with the goal of delaying progression and extend life as long as possible, with tolerable treatment-related adverse events. Some exceptions were made for local treatment with surgery or radiotherapy (RT), often for patients with a single metastasis. A 1939 report describes a patient with renal adenocarcinoma and a solitary lung metastasis who underwent RT to the lung lesion after nephrectomy and subsequently partial lobectomy after the metastatic lesion progressed. The authors argued that if a metastasis appears solitary and accessible, it is plausible to remove it in addition to the primary growth.1
In 1995 Hellman and Weichselbaum proposed oligometastatic disease (OMD). They reasoned that malignancy exists along a spectrum from localized disease to widely disseminated disease, with OMD existing in between with a still-restricted tumor metastatic capacity. Appropriately selected patients with OMD may be candidates for prolonged disease-free survival or cure with the addition of local therapy to systemic therapy.2
The EORTC 4004 phase 2 randomized control trial (RCT) analyzed radiofrequency ablation (RFA) for colorectal liver metastases with systemic therapy vs systemic therapy alone for patients with ≤ 9 liver lesions.3 Systemic therapyconsisted of 5-FU/leucovorin/oxaliplatin, with bevacizumab added to the regimen 3.5 years into the study, per updated standard- of-care. This trial was the first to demonstrate the benefit of aggressive local treatment vs system treatment alone for OMD with a progression-free survival (PFS) benefit (16.8 vs 9.9 months; hazard ratio [HR], 0.63; P = .03) and overall survival (OS) benefit (45.3 vs 40.5 months; HR, 0.74; P = .02) with the addition of local treatment with RFA.
Since the presentations of the SABR-COMET phase 2 RCT and another study by Gomez et al at the American Society for Radiation Oncology (ASTRO) 2018 annual meeting, the paradigm for offering local RT for OMD has rapidly evolved. Both studies found PFS and OS benefits of RT for patients with OMD.4,5 Additional RCTs have since demonstrated that for properly selected patients with OMD, aggressive local RT improved PFS and OS.6-9 These small studies have led to larger RCTs to better understand who benefits from local consolidative treatment, particularly RT.10,11
There is a large degree of heterogeneity in how oncologists define and approach OMD treatment. The 2020 European Society for Radiotherapy and Oncology (ESTRO) and ASTRO consensus guidelines defined the OMD state as 1 to 5 metastatic lesions for which all metastatic sites are safely treatable.12 The purpose of this study was to evaluate perceptions and practice patterns among radiation oncologists and medical oncologists regarding the use of local RT for OMD across the Veterans Health Administration (VHA).
Methods
A 12-question survey was developed by the VHA Palliative Radiotherapy Task Force using the ESTRO-ASTRO consensus guidelines to define OMD. The survey was emailed to the VHA radiation oncology and medical oncology listservs on August 1, 2023. These listservs consist of physicians in these specialties either directly employed by the VHA or serve in its facilities as contractors. The original response closure date was August 11, 2023, but it was extended to August 18, 2023, to increase responses. No incentives were offered to respondents. Two email reminders were sent to the medical oncology listserv and 3 to the radiation oncology listserv. Descriptive statistics and X2 tests were used for data analysis. The impact of specialty and presence of an on-site department of radiation oncology were reviewed. This project was approved by the VHA National Oncology Program and National Radiation Oncology Program.
Results
The survey was sent to 125 radiation oncologists and 515 medical oncologists and 106 were completed for a 16.6% response rate. There were 59 (55.7%) radiation oncologist responses and 47 (44.3%) medical oncologist responses. Most (96.2%) respondents were board-certified, and 84 (79.2%) were affiliated with an academic center. Not every respondent answered every question (Table).

All respondents (n = 105) indicated there is a potential benefit of high-dose RT for appropriately selected cases. Ninety-four oncologists (88.7%) believed that RT for OMD contributes to cure (88.1% of radiation oncologists, 89.4% of medical oncologists; P = .84) for appropriately selected cases. Some respondents who did not believe RT for OMD contributes to cure added comments about other perceived benefits, such as local disease control for palliation, delaying systemic therapy with its associated toxicities, and prolongation of disease-free survival or OS. A higher percentage of respondents with academic affiliations believed high-dose RT contributes to cure, although this difference did not reach statistical significance (Figure 1).

Fifty-five respondents (51.9%; 55.2% radiation oncologists vs 50.0% medical oncologists; P = .60) responded that local RT for OMD treatment should not be limited by primary tumor type. Of respondents who responded that OMD treatment should be limited based on the type of primary tumor, many provided comments that argued there was a benefit for non-small cell lung cancer (NSCLC), prostate adenocarcinoma (PCa), and colorectal cancer.
The definition of how many metastatic lesions qualify as OMD varied. A total of 48.6% of respondents defined OMD as ≤ 3 lesions and 42.9% answered ≤ 5 lesions. A majority of radiation oncologists (55.2%) classified ≤ 5 lesions as OMD, whereas a majority of medical oncologists (66.0%) considered ≤ 3 lesions as OMD (P = .006) (Figure 2).

Thirty-six medical oncologists (76.6%) report having an on-site department of radiation oncology (Figure 3). This subgroup was more likely to consider local RT potentially curative compared with their medical oncology peers without on-site radiation oncology (94.4% vs 72.7%; P = .04).

Case Management
The 3 clinical cases demonstrated the heterogeneity of management approaches for OMD. The first described a man aged 65 years with PCa and 2 asymptomatic pelvic bone metastases. Ninety-three respondents (90.3%) recommended RT at the primary site and 74.8% recommended RT to both the primary site and metastatic foci. Sixty-three respondents (67.7%) recommended a STAMPEDE-compatible dose, and 30 (32.3%) recommended a definitive dose.
The second clinical case was a 60-year-old man with a cT1N2M1 NSCLC, with a solitary metastatic focus to the left iliac wing. Fifty-eight respondents (54.7%) recommended upfront systemic chemotherapy and the option of local therapy to the chest and metastatic focus after initial chemotherapy; 28 respondents (26.4%) recommended upfront chemoradiation to the chest and definitive radiation to the left iliac wing metastasis.
The third clinical case described a male aged 70 years with a history of a treated base of tongue squamous cell carcinoma, with a solitary metastatic focus within the right lung. Respondents could pick multiple treatment options and 85 (81.7%) favored upfront definitive local therapy with surgery or stereotactic body radiotherapy (SBRT), rather than upfront chemotherapy, with future consideration for local treatment. About half of respondents (51.8%) recommended SBRT and 41.2% would let the patient decide between surgery or SBRT. Additionally, 39.6% included in their patient counselling that the treatment may be for curative intent.
Discussion
The use of local treatment to increased PFS, OS, or even cure treatment for OMD has become more accepted since the 2018 ASTRO meeting.4,5 Palma et al analyzed a controlled primary malignancy of any histology and ≤ 5 metastatic lesions, with all lesions amenable to SBRT.4 With a median follow-up of 51 months when comparing the standard-of-care (SOC) arm and the SBRT arm, the 5-year PFS was not reached and the 5-year OS rates were 17.7% and 42.3% (P = .006), respectively. In the SBRT arm, about 1 in 5 patients survived > 5 years without a recurrence or disease progression, vs 0 patients in the control arm. There was a 29% rate of grade 2 or higher toxicity in the SBRT arm, including 3 deaths that were likely due to treatment. Subsequent trials, such as the phase 3 SABR-COMET-3 (1-3 metastases), phase 3 SABR-COMET-10 (4-10 metastases), and phase 1 ARREST (> 10 metastases) trials, have been specifically designed to minimize treatment-related toxicities.13-15
Gomez et al analyzed patients at 3 sites with a controlled NSCLC primary tumor and ≤ 3 metastases.5 At a follow-up of 38.8 months, the PFS was 4.4 months in the SOC arm vs 14.2 months in the RT and/or surgery local treatment arm (P = .02). There was also an OS benefit of 17.0 vs 41.2 months (P = .02), respectively.
Several RCTs soon followed that demonstrated improved PFS and OS with local radiotherapy for OMD; however, total metastatic ablation of the foci is necessary to attain these PFS and OS benefits.6-9 Still, an oncologic benefit has yet to be proven. The randomized NRGBR002 study phase 2/3 trial for oligometastatic breast cancer included patients with ≤ 4 extracranial metastases and controlled primary disease to metastasis-directed therapy (SBRT and/ or surgical resection) and systemic therapy vs systemic therapy alone.10 The study did not demonstrate improved PFS or OS at 3 years. However, for most breast cancers, especially with the rapid advancements in systemic therapy that have been achieved, longer follow-up may be necessary to detect a significant difference.
The prospective single-arm phase 2 SABR-5 trial retrospectively demonstrated important lessons about the timing of SBRT and systemic therapy.11 This study included patients with ≤ 5 metastases of any histology, and they received SBRT to all lesions. SABR-5 retrospectively compared patients who received upfront systemic therapy followed by SBRT vs another cohort that first received SBRT and did not receive systemic therapy until there was disease progression. Patients with oligo-progression were excluded, as it demonstrated systemic drug resistance. At a median follow-up time of 34 months, delayed systemic treatment was associated with shorter PFS (23 vs 34 months, respectively; P = .001), but not worse 3-year OS (80% vs 85%, respectively; P = .66). In addition, the delayed systemic treatment arm demonstrated a reduced risk of grade 2 or higher SBRT-related toxicity (odds ratio, 0.35; P < .001).
Similarly, the STOMP phase 2 trial analyzed the role of metastasis-directed therapy (MDT) in delaying initiation of androgen deprivation therapy (ADT) in a randomized phase 2 trial.16 This study included patients with asymptomatic PCa with a biochemical recurrence after primary treatment, 1 to 3 extracranial metastatic lesions, and serum testosterone levels > 50 ng/mL. Sixty-two patients were randomized 1:1 to either MDT (SBRT or surgery) of all lesions or surveillance. The 5-year ADT-free survival was 34% for MDT vs 8% for surveillance (P = .06).
VHA Radiation Oncology
The VHA has 138 departments of medical oncology, but only 41 departments of radiation oncology. Compared with medical oncologists without an on-site radiation oncology department, those with on-site departments were more likely to believe that local RT was potentially curative (94.4% vs 72.7%, respectively; P = .04). This finding suggests that a cancer center that includes both specialties has closer collaboration, which results in greater inclination to embrace local RT for OMD, as it has demonstrated PFS and OS benefits.
The radiation and medical oncologists surveyed had statistically significant differences in response by specialty regarding the maximal number of lesions still believed to constitute OMD. Most radiation oncologists classified ≤ 5 lesions as OMD, whereas most medical oncologists classified ≤ 3 lesions as OMD. This difference is not unexpected. There is no universally agreed-upon definition of OMD, and criteria differ across studies.
While the SABR-COMET trial did include ≤ 5 metastatic lesions, it was a phase 2 RCT, making subgroup analysis difficult. Ongoing phase 3 trials that are more specific in the number of metastases, comparing 1 to 3 vs 4 to 10 metastases (SABR-COMET-3 and SABR-COMET-10, respectively).13,14 There is even an ongoing phase 1 trial (ARREST) studying the potential benefits of treating (“restraining”) > 10 metastases, if dosimetrically feasible.15 Within the VHA, VA STARPORT is investigating MDT for recurrent or de novo hormone-sensitive metastatic PCa.17 The ongoing HALT phase 2/3 trial focuses on patients with actionable mutations to help determine management of oligo-progression in mutation-positive NSCLC.18
There was no significant difference by specialty in who responded that offering local RT for OMD treatment should not be limited by histology (55.2% of radiation oncologists and 50.0% of medical oncologists; P = .60). Oncologists could make the argument that some histologies (eg, pancreatic adenocarcinomas) have such poor prognoses that local RT would not meaningfully affect oncologic outcomes, while potentially adding toxicity, whereas others could point to improved systemic therapy regimens and the low toxicity rates with careful hypofractionation regimens. Of note, the 41-patient phase 2 EXTEND trial for pancreatic ductal adenocarcinoma suggested an oncologic benefit to MDT, with far better PFS and no grade ≥ 3 toxicities related to MDT.19 About half of respondents for each specialty believed the primary histology should affect the decision. Further clarification may emerge from phase 3 trials.
Of note, a 2023 study of 44 radiation and medical oncologists at 2 Harvard Medical School-affiliated hospitals found that for synchronous OMD, 50.0% of medical oncologists and 5.3% (P < .01) of radiation oncologists recommended systemic treatment, suggesting a greater divergence in approach than found in this study.20
Limitations
The response rate of 17.0% raised a potential for selection bias, but this rate is expected for a nonincentivized medical survey. A study by the American Board of Internal Medicine with 11 surveys and 6 weekly email contacts only generated a 23.7% response rate, while another study among physicians demonstrated a 4.5% response rate for email-based contact and 11.8% for mail-based contact.21,22 We could have asked participants questions regarding demographics and geography to ensure the survey represented a diverse sample of the medical community, although additional questions would likely suppress the response rate. Additional data collection about respondents may elucidate the rationale for differences in their responses, especially between the specialties. In a planned subsequent survey in several years, the question on the number of lesions that qualifies as OMD may be amended to reflect the context and dosimetry for the maximal number of metastases constituting OMD; the joint ESTRO-ASTRO consensus defined OMD as 1 to 5 metastatic lesions, but in which all metastatic sites must be safely treatable.12 Also, fewer example cases could be included to simplify the survey and boost response rates. A future survey may ask about the timing of SBRT and systemic therapy, and whether SBRT can safely delay systemic therapy.
Conclusions
Survey results demonstrated significant confidence among both radiation oncologists and medical oncologists that local RT for OMD improves outcomes, which is encouraging and a reflection of the recent evidence-based paradigm shift in viewing metastatic disease as a spectrum. However, there is a difference between radiation oncologists and medical oncologists in how they define OMD, and preferred treatment of the sample cases presented revealed nuanced differences by specialty. Close collaboration with radiation oncologists influences the belief of medical oncologists in the beneficial role of RT for OMD. As more phase 3 data for OMD local treatments emerge, additional investigation is needed on how beliefs and practice patterns evolve among radiation and medical oncologists.
- Barney JD, Churchill EJ. Adenocarcinoma of the kidney with metastasis to the lung. J Urology. 1939.
- Hellman S, Weichselbaum RR. Oligometastases. J Clin Oncol. 1995;13(1):8-10. doi:10.1200/JCO.1995.13.1.8
- Ruers T, Punt C, Van Coevorden F, et al. Radiofrequency ablation combined with systemic treatment versus systemic treatment alone in patients with non-resectable colorectal liver metastases: a randomized EORTC Intergroup phase II study (EORTC 4004). Ann Oncol. 2012;23(10):2619-2626. doi:10.1093/annonc/mds053
- Palma DA, Olson R, Harrow S, et al. Stereotactic ablative radiotherapy for the comprehensive treatment of oligometastatic cancers: long-term results of the SABR-COMET phase II randomized trial. J Clin Oncol. 2020;38(25):2830- 2838. doi:10.1200/JCO.20.00818
- Gomez DR, Tang C, Zhang J, et al. Local consolidative therapy vs. maintenance therapy or observation for patients with oligometastatic non-small-cell lung cancer: long-term results of a multi-institutional, phase II, randomized study. J Clin Oncol. 2019;37(18):1558-1565. doi:10.1200/JCO.19.00201
- Iyengar P, Wardak Z, Gerber DE, et al. Consolidative radiotherapy for limited metastatic non-small-cell lung cancer: a phase 2 randomized clinical trial. JAMA Oncol. 2018;4(1):e173501. doi:10.1001/jamaoncol.2017.3501
- Phillips R, Shi WY, Deek M, et al. Outcomes of observation vs stereotactic ablative radiation for oligometastatic prostate cancer: the ORIOLE phase 2 randomized clinical trial. JAMA Oncol. 2020;6(5):650-659. doi:10.1001/jamaoncol.2020.0147
- Wang XS, Bai YF, Verma V, et al. Randomized trial of first-line tyrosine kinase inhibitor with or without radiotherapy for synchronous oligometastatic EGFR-mutated NSCLC. J Natl Cancer Inst 2023;115(6):742-748. doi:10.1093/jnci/djac015
- Tang C, Sherry AD, Haymaker C, et al. Addition of metastasis- directed therapy to intermittent hormone therapy for oligometastatic prostate cancer (EXTEND): a multicenter, randomized phase II trial. Am Soc Radiat Oncol Annu Meet. 2023;9(6):825-834. doi:10.1001/jamaoncol.2023.0161
- Chmura SJ, Winter KA, Woodward WA, et al. NRG-BR002: a phase IIR/III trial of standard of care systemic therapy with or without stereotactic body radiotherapy (SBRT) and/or surgical resection (SR) for newly oligometastatic breast cancer (NCT02364557). J Clin Oncol. 2022;40:1007. doi:10.1200/JCO.2022.40.16_suppl.1007
- Baker S, Lechner L, Liu M, et al. Upfront versus delayed systemic therapy in patients with oligometastatic cancer treated with SABR in the phase 2 SABR-5 trial. Int J Radiat Oncol Biol Phys. 2024;118(5):1497-1506. doi:10.1016/j.ijrobp.2023.11.007
- Lievens Y, Guckenberger M, Gomez D, et al. Defining oligometastatic disease from a radiation oncology perspective: an ESTRO-ASTRO consensus document. Radiother Oncol. 2020;148:157-166. doi:10.1016/j.radonc.2020.04.003
- Olson R, Mathews L, Liu M, et al. Stereotactic ablative radiotherapy for the comprehensive treatment of 1-3 oligometastatic tumors (SABR-COMET-3): study protocol for a randomized phase III trial. BMC Cancer. 2020;20(1):380. doi:10.1186/s12885-020-06876-4
- Palma DA, Olson R, Harrow S, et al. Stereotactic ablative radiotherapy for the comprehensive treatment of 4-10 oligometastatic tumors (SABR-COMET-10): study protocol for a randomized phase III trial. BMC Cancer. 2019;19(1):816. doi:10.1186/s12885-019-5977-6
- Bauman GS, Corkum MT, Fakir H, et al. Ablative radiation therapy to restrain everything safely treatable (ARREST): study protocol for a phase I trial treating polymetastatic cancer with stereotactic radiotherapy. BMC Cancer. 2021;21(1):405. doi:10.1186/s12885-021-08136-5
- Ost P, Reynders D, Decaestecker K, et al. Surveillance or metastasis-directed therapy for oligometastatic prostate cancer recurrence (STOMP): five-year results of a randomized phase II trial. J Clin Oncol. 2020;38:suppl.
- Solanki AA, Campbell D, Carlson K, et al. Veterans Affairs seamless phase II/III randomized trial of standard systemic therapy with or without PET-directed local therapy for oligometastatic prostate cancer (VA STARPORT). J Clin Oncol. 2024;42:16.
- McDonald F, Guckenberger M, Popat S. EP08.03-005 HALT – Targeted therapy with or without dose-intensified radiotherapy in oligo-progressive disease in oncogene addicted lung tumours. J Thor Oncol. 2022;17:S492.
- Ludmir EB, Sherry AD, Fellman BM, et al. Addition of metastasis- directed therapy to systemic therapy for oligometastatic pancreatic ductal adenocarcinoma (EXTEND): a multicenter, randomized phase II trial. J Clin Oncol. 2024;42(32):3795-3805. doi:10.1200/JCO.24.00081
- Cho HL, Balboni T, Christ SB, et al. Is oligometastatic cancer curable? A survey of oncologist perspectives, decision making, and communication. Adv Radiat Oncol. 2023;8(5):101221. doi:10.1016/j.adro.2023.101221
- Barnhart BJ, Reddy SG, Arnold GK. Remind me again: physician response to web surveys: the effect of email reminders across 11 opinion survey efforts at the American Board of Internal Medicine from 2017 to 2019. Eval Health Prof. 2021;44(3):245-259. doi:10.1177/01632787211019445
- Murphy CC, Lee SJC, Geiger AM, et al. A randomized trial of mail and email recruitment strategies for a physician survey on clinical trial accrual. BMC Med Res Methodol. 2020;20(1):123. doi:10.1186/s12874-020-01014-x
The treatment of metastatic solid tumors has been based historically on systemic therapies, with the goal of delaying progression and extend life as long as possible, with tolerable treatment-related adverse events. Some exceptions were made for local treatment with surgery or radiotherapy (RT), often for patients with a single metastasis. A 1939 report describes a patient with renal adenocarcinoma and a solitary lung metastasis who underwent RT to the lung lesion after nephrectomy and subsequently partial lobectomy after the metastatic lesion progressed. The authors argued that if a metastasis appears solitary and accessible, it is plausible to remove it in addition to the primary growth.1
In 1995 Hellman and Weichselbaum proposed oligometastatic disease (OMD). They reasoned that malignancy exists along a spectrum from localized disease to widely disseminated disease, with OMD existing in between with a still-restricted tumor metastatic capacity. Appropriately selected patients with OMD may be candidates for prolonged disease-free survival or cure with the addition of local therapy to systemic therapy.2
The EORTC 4004 phase 2 randomized control trial (RCT) analyzed radiofrequency ablation (RFA) for colorectal liver metastases with systemic therapy vs systemic therapy alone for patients with ≤ 9 liver lesions.3 Systemic therapyconsisted of 5-FU/leucovorin/oxaliplatin, with bevacizumab added to the regimen 3.5 years into the study, per updated standard- of-care. This trial was the first to demonstrate the benefit of aggressive local treatment vs system treatment alone for OMD with a progression-free survival (PFS) benefit (16.8 vs 9.9 months; hazard ratio [HR], 0.63; P = .03) and overall survival (OS) benefit (45.3 vs 40.5 months; HR, 0.74; P = .02) with the addition of local treatment with RFA.
Since the presentations of the SABR-COMET phase 2 RCT and another study by Gomez et al at the American Society for Radiation Oncology (ASTRO) 2018 annual meeting, the paradigm for offering local RT for OMD has rapidly evolved. Both studies found PFS and OS benefits of RT for patients with OMD.4,5 Additional RCTs have since demonstrated that for properly selected patients with OMD, aggressive local RT improved PFS and OS.6-9 These small studies have led to larger RCTs to better understand who benefits from local consolidative treatment, particularly RT.10,11
There is a large degree of heterogeneity in how oncologists define and approach OMD treatment. The 2020 European Society for Radiotherapy and Oncology (ESTRO) and ASTRO consensus guidelines defined the OMD state as 1 to 5 metastatic lesions for which all metastatic sites are safely treatable.12 The purpose of this study was to evaluate perceptions and practice patterns among radiation oncologists and medical oncologists regarding the use of local RT for OMD across the Veterans Health Administration (VHA).
Methods
A 12-question survey was developed by the VHA Palliative Radiotherapy Task Force using the ESTRO-ASTRO consensus guidelines to define OMD. The survey was emailed to the VHA radiation oncology and medical oncology listservs on August 1, 2023. These listservs consist of physicians in these specialties either directly employed by the VHA or serve in its facilities as contractors. The original response closure date was August 11, 2023, but it was extended to August 18, 2023, to increase responses. No incentives were offered to respondents. Two email reminders were sent to the medical oncology listserv and 3 to the radiation oncology listserv. Descriptive statistics and X2 tests were used for data analysis. The impact of specialty and presence of an on-site department of radiation oncology were reviewed. This project was approved by the VHA National Oncology Program and National Radiation Oncology Program.
Results
The survey was sent to 125 radiation oncologists and 515 medical oncologists and 106 were completed for a 16.6% response rate. There were 59 (55.7%) radiation oncologist responses and 47 (44.3%) medical oncologist responses. Most (96.2%) respondents were board-certified, and 84 (79.2%) were affiliated with an academic center. Not every respondent answered every question (Table).

All respondents (n = 105) indicated there is a potential benefit of high-dose RT for appropriately selected cases. Ninety-four oncologists (88.7%) believed that RT for OMD contributes to cure (88.1% of radiation oncologists, 89.4% of medical oncologists; P = .84) for appropriately selected cases. Some respondents who did not believe RT for OMD contributes to cure added comments about other perceived benefits, such as local disease control for palliation, delaying systemic therapy with its associated toxicities, and prolongation of disease-free survival or OS. A higher percentage of respondents with academic affiliations believed high-dose RT contributes to cure, although this difference did not reach statistical significance (Figure 1).

Fifty-five respondents (51.9%; 55.2% radiation oncologists vs 50.0% medical oncologists; P = .60) responded that local RT for OMD treatment should not be limited by primary tumor type. Of respondents who responded that OMD treatment should be limited based on the type of primary tumor, many provided comments that argued there was a benefit for non-small cell lung cancer (NSCLC), prostate adenocarcinoma (PCa), and colorectal cancer.
The definition of how many metastatic lesions qualify as OMD varied. A total of 48.6% of respondents defined OMD as ≤ 3 lesions and 42.9% answered ≤ 5 lesions. A majority of radiation oncologists (55.2%) classified ≤ 5 lesions as OMD, whereas a majority of medical oncologists (66.0%) considered ≤ 3 lesions as OMD (P = .006) (Figure 2).

Thirty-six medical oncologists (76.6%) report having an on-site department of radiation oncology (Figure 3). This subgroup was more likely to consider local RT potentially curative compared with their medical oncology peers without on-site radiation oncology (94.4% vs 72.7%; P = .04).

Case Management
The 3 clinical cases demonstrated the heterogeneity of management approaches for OMD. The first described a man aged 65 years with PCa and 2 asymptomatic pelvic bone metastases. Ninety-three respondents (90.3%) recommended RT at the primary site and 74.8% recommended RT to both the primary site and metastatic foci. Sixty-three respondents (67.7%) recommended a STAMPEDE-compatible dose, and 30 (32.3%) recommended a definitive dose.
The second clinical case was a 60-year-old man with a cT1N2M1 NSCLC, with a solitary metastatic focus to the left iliac wing. Fifty-eight respondents (54.7%) recommended upfront systemic chemotherapy and the option of local therapy to the chest and metastatic focus after initial chemotherapy; 28 respondents (26.4%) recommended upfront chemoradiation to the chest and definitive radiation to the left iliac wing metastasis.
The third clinical case described a male aged 70 years with a history of a treated base of tongue squamous cell carcinoma, with a solitary metastatic focus within the right lung. Respondents could pick multiple treatment options and 85 (81.7%) favored upfront definitive local therapy with surgery or stereotactic body radiotherapy (SBRT), rather than upfront chemotherapy, with future consideration for local treatment. About half of respondents (51.8%) recommended SBRT and 41.2% would let the patient decide between surgery or SBRT. Additionally, 39.6% included in their patient counselling that the treatment may be for curative intent.
Discussion
The use of local treatment to increased PFS, OS, or even cure treatment for OMD has become more accepted since the 2018 ASTRO meeting.4,5 Palma et al analyzed a controlled primary malignancy of any histology and ≤ 5 metastatic lesions, with all lesions amenable to SBRT.4 With a median follow-up of 51 months when comparing the standard-of-care (SOC) arm and the SBRT arm, the 5-year PFS was not reached and the 5-year OS rates were 17.7% and 42.3% (P = .006), respectively. In the SBRT arm, about 1 in 5 patients survived > 5 years without a recurrence or disease progression, vs 0 patients in the control arm. There was a 29% rate of grade 2 or higher toxicity in the SBRT arm, including 3 deaths that were likely due to treatment. Subsequent trials, such as the phase 3 SABR-COMET-3 (1-3 metastases), phase 3 SABR-COMET-10 (4-10 metastases), and phase 1 ARREST (> 10 metastases) trials, have been specifically designed to minimize treatment-related toxicities.13-15
Gomez et al analyzed patients at 3 sites with a controlled NSCLC primary tumor and ≤ 3 metastases.5 At a follow-up of 38.8 months, the PFS was 4.4 months in the SOC arm vs 14.2 months in the RT and/or surgery local treatment arm (P = .02). There was also an OS benefit of 17.0 vs 41.2 months (P = .02), respectively.
Several RCTs soon followed that demonstrated improved PFS and OS with local radiotherapy for OMD; however, total metastatic ablation of the foci is necessary to attain these PFS and OS benefits.6-9 Still, an oncologic benefit has yet to be proven. The randomized NRGBR002 study phase 2/3 trial for oligometastatic breast cancer included patients with ≤ 4 extracranial metastases and controlled primary disease to metastasis-directed therapy (SBRT and/ or surgical resection) and systemic therapy vs systemic therapy alone.10 The study did not demonstrate improved PFS or OS at 3 years. However, for most breast cancers, especially with the rapid advancements in systemic therapy that have been achieved, longer follow-up may be necessary to detect a significant difference.
The prospective single-arm phase 2 SABR-5 trial retrospectively demonstrated important lessons about the timing of SBRT and systemic therapy.11 This study included patients with ≤ 5 metastases of any histology, and they received SBRT to all lesions. SABR-5 retrospectively compared patients who received upfront systemic therapy followed by SBRT vs another cohort that first received SBRT and did not receive systemic therapy until there was disease progression. Patients with oligo-progression were excluded, as it demonstrated systemic drug resistance. At a median follow-up time of 34 months, delayed systemic treatment was associated with shorter PFS (23 vs 34 months, respectively; P = .001), but not worse 3-year OS (80% vs 85%, respectively; P = .66). In addition, the delayed systemic treatment arm demonstrated a reduced risk of grade 2 or higher SBRT-related toxicity (odds ratio, 0.35; P < .001).
Similarly, the STOMP phase 2 trial analyzed the role of metastasis-directed therapy (MDT) in delaying initiation of androgen deprivation therapy (ADT) in a randomized phase 2 trial.16 This study included patients with asymptomatic PCa with a biochemical recurrence after primary treatment, 1 to 3 extracranial metastatic lesions, and serum testosterone levels > 50 ng/mL. Sixty-two patients were randomized 1:1 to either MDT (SBRT or surgery) of all lesions or surveillance. The 5-year ADT-free survival was 34% for MDT vs 8% for surveillance (P = .06).
VHA Radiation Oncology
The VHA has 138 departments of medical oncology, but only 41 departments of radiation oncology. Compared with medical oncologists without an on-site radiation oncology department, those with on-site departments were more likely to believe that local RT was potentially curative (94.4% vs 72.7%, respectively; P = .04). This finding suggests that a cancer center that includes both specialties has closer collaboration, which results in greater inclination to embrace local RT for OMD, as it has demonstrated PFS and OS benefits.
The radiation and medical oncologists surveyed had statistically significant differences in response by specialty regarding the maximal number of lesions still believed to constitute OMD. Most radiation oncologists classified ≤ 5 lesions as OMD, whereas most medical oncologists classified ≤ 3 lesions as OMD. This difference is not unexpected. There is no universally agreed-upon definition of OMD, and criteria differ across studies.
While the SABR-COMET trial did include ≤ 5 metastatic lesions, it was a phase 2 RCT, making subgroup analysis difficult. Ongoing phase 3 trials that are more specific in the number of metastases, comparing 1 to 3 vs 4 to 10 metastases (SABR-COMET-3 and SABR-COMET-10, respectively).13,14 There is even an ongoing phase 1 trial (ARREST) studying the potential benefits of treating (“restraining”) > 10 metastases, if dosimetrically feasible.15 Within the VHA, VA STARPORT is investigating MDT for recurrent or de novo hormone-sensitive metastatic PCa.17 The ongoing HALT phase 2/3 trial focuses on patients with actionable mutations to help determine management of oligo-progression in mutation-positive NSCLC.18
There was no significant difference by specialty in who responded that offering local RT for OMD treatment should not be limited by histology (55.2% of radiation oncologists and 50.0% of medical oncologists; P = .60). Oncologists could make the argument that some histologies (eg, pancreatic adenocarcinomas) have such poor prognoses that local RT would not meaningfully affect oncologic outcomes, while potentially adding toxicity, whereas others could point to improved systemic therapy regimens and the low toxicity rates with careful hypofractionation regimens. Of note, the 41-patient phase 2 EXTEND trial for pancreatic ductal adenocarcinoma suggested an oncologic benefit to MDT, with far better PFS and no grade ≥ 3 toxicities related to MDT.19 About half of respondents for each specialty believed the primary histology should affect the decision. Further clarification may emerge from phase 3 trials.
Of note, a 2023 study of 44 radiation and medical oncologists at 2 Harvard Medical School-affiliated hospitals found that for synchronous OMD, 50.0% of medical oncologists and 5.3% (P < .01) of radiation oncologists recommended systemic treatment, suggesting a greater divergence in approach than found in this study.20
Limitations
The response rate of 17.0% raised a potential for selection bias, but this rate is expected for a nonincentivized medical survey. A study by the American Board of Internal Medicine with 11 surveys and 6 weekly email contacts only generated a 23.7% response rate, while another study among physicians demonstrated a 4.5% response rate for email-based contact and 11.8% for mail-based contact.21,22 We could have asked participants questions regarding demographics and geography to ensure the survey represented a diverse sample of the medical community, although additional questions would likely suppress the response rate. Additional data collection about respondents may elucidate the rationale for differences in their responses, especially between the specialties. In a planned subsequent survey in several years, the question on the number of lesions that qualifies as OMD may be amended to reflect the context and dosimetry for the maximal number of metastases constituting OMD; the joint ESTRO-ASTRO consensus defined OMD as 1 to 5 metastatic lesions, but in which all metastatic sites must be safely treatable.12 Also, fewer example cases could be included to simplify the survey and boost response rates. A future survey may ask about the timing of SBRT and systemic therapy, and whether SBRT can safely delay systemic therapy.
Conclusions
Survey results demonstrated significant confidence among both radiation oncologists and medical oncologists that local RT for OMD improves outcomes, which is encouraging and a reflection of the recent evidence-based paradigm shift in viewing metastatic disease as a spectrum. However, there is a difference between radiation oncologists and medical oncologists in how they define OMD, and preferred treatment of the sample cases presented revealed nuanced differences by specialty. Close collaboration with radiation oncologists influences the belief of medical oncologists in the beneficial role of RT for OMD. As more phase 3 data for OMD local treatments emerge, additional investigation is needed on how beliefs and practice patterns evolve among radiation and medical oncologists.
The treatment of metastatic solid tumors has been based historically on systemic therapies, with the goal of delaying progression and extend life as long as possible, with tolerable treatment-related adverse events. Some exceptions were made for local treatment with surgery or radiotherapy (RT), often for patients with a single metastasis. A 1939 report describes a patient with renal adenocarcinoma and a solitary lung metastasis who underwent RT to the lung lesion after nephrectomy and subsequently partial lobectomy after the metastatic lesion progressed. The authors argued that if a metastasis appears solitary and accessible, it is plausible to remove it in addition to the primary growth.1
In 1995 Hellman and Weichselbaum proposed oligometastatic disease (OMD). They reasoned that malignancy exists along a spectrum from localized disease to widely disseminated disease, with OMD existing in between with a still-restricted tumor metastatic capacity. Appropriately selected patients with OMD may be candidates for prolonged disease-free survival or cure with the addition of local therapy to systemic therapy.2
The EORTC 4004 phase 2 randomized control trial (RCT) analyzed radiofrequency ablation (RFA) for colorectal liver metastases with systemic therapy vs systemic therapy alone for patients with ≤ 9 liver lesions.3 Systemic therapyconsisted of 5-FU/leucovorin/oxaliplatin, with bevacizumab added to the regimen 3.5 years into the study, per updated standard- of-care. This trial was the first to demonstrate the benefit of aggressive local treatment vs system treatment alone for OMD with a progression-free survival (PFS) benefit (16.8 vs 9.9 months; hazard ratio [HR], 0.63; P = .03) and overall survival (OS) benefit (45.3 vs 40.5 months; HR, 0.74; P = .02) with the addition of local treatment with RFA.
Since the presentations of the SABR-COMET phase 2 RCT and another study by Gomez et al at the American Society for Radiation Oncology (ASTRO) 2018 annual meeting, the paradigm for offering local RT for OMD has rapidly evolved. Both studies found PFS and OS benefits of RT for patients with OMD.4,5 Additional RCTs have since demonstrated that for properly selected patients with OMD, aggressive local RT improved PFS and OS.6-9 These small studies have led to larger RCTs to better understand who benefits from local consolidative treatment, particularly RT.10,11
There is a large degree of heterogeneity in how oncologists define and approach OMD treatment. The 2020 European Society for Radiotherapy and Oncology (ESTRO) and ASTRO consensus guidelines defined the OMD state as 1 to 5 metastatic lesions for which all metastatic sites are safely treatable.12 The purpose of this study was to evaluate perceptions and practice patterns among radiation oncologists and medical oncologists regarding the use of local RT for OMD across the Veterans Health Administration (VHA).
Methods
A 12-question survey was developed by the VHA Palliative Radiotherapy Task Force using the ESTRO-ASTRO consensus guidelines to define OMD. The survey was emailed to the VHA radiation oncology and medical oncology listservs on August 1, 2023. These listservs consist of physicians in these specialties either directly employed by the VHA or serve in its facilities as contractors. The original response closure date was August 11, 2023, but it was extended to August 18, 2023, to increase responses. No incentives were offered to respondents. Two email reminders were sent to the medical oncology listserv and 3 to the radiation oncology listserv. Descriptive statistics and X2 tests were used for data analysis. The impact of specialty and presence of an on-site department of radiation oncology were reviewed. This project was approved by the VHA National Oncology Program and National Radiation Oncology Program.
Results
The survey was sent to 125 radiation oncologists and 515 medical oncologists and 106 were completed for a 16.6% response rate. There were 59 (55.7%) radiation oncologist responses and 47 (44.3%) medical oncologist responses. Most (96.2%) respondents were board-certified, and 84 (79.2%) were affiliated with an academic center. Not every respondent answered every question (Table).

All respondents (n = 105) indicated there is a potential benefit of high-dose RT for appropriately selected cases. Ninety-four oncologists (88.7%) believed that RT for OMD contributes to cure (88.1% of radiation oncologists, 89.4% of medical oncologists; P = .84) for appropriately selected cases. Some respondents who did not believe RT for OMD contributes to cure added comments about other perceived benefits, such as local disease control for palliation, delaying systemic therapy with its associated toxicities, and prolongation of disease-free survival or OS. A higher percentage of respondents with academic affiliations believed high-dose RT contributes to cure, although this difference did not reach statistical significance (Figure 1).

Fifty-five respondents (51.9%; 55.2% radiation oncologists vs 50.0% medical oncologists; P = .60) responded that local RT for OMD treatment should not be limited by primary tumor type. Of respondents who responded that OMD treatment should be limited based on the type of primary tumor, many provided comments that argued there was a benefit for non-small cell lung cancer (NSCLC), prostate adenocarcinoma (PCa), and colorectal cancer.
The definition of how many metastatic lesions qualify as OMD varied. A total of 48.6% of respondents defined OMD as ≤ 3 lesions and 42.9% answered ≤ 5 lesions. A majority of radiation oncologists (55.2%) classified ≤ 5 lesions as OMD, whereas a majority of medical oncologists (66.0%) considered ≤ 3 lesions as OMD (P = .006) (Figure 2).

Thirty-six medical oncologists (76.6%) report having an on-site department of radiation oncology (Figure 3). This subgroup was more likely to consider local RT potentially curative compared with their medical oncology peers without on-site radiation oncology (94.4% vs 72.7%; P = .04).

Case Management
The 3 clinical cases demonstrated the heterogeneity of management approaches for OMD. The first described a man aged 65 years with PCa and 2 asymptomatic pelvic bone metastases. Ninety-three respondents (90.3%) recommended RT at the primary site and 74.8% recommended RT to both the primary site and metastatic foci. Sixty-three respondents (67.7%) recommended a STAMPEDE-compatible dose, and 30 (32.3%) recommended a definitive dose.
The second clinical case was a 60-year-old man with a cT1N2M1 NSCLC, with a solitary metastatic focus to the left iliac wing. Fifty-eight respondents (54.7%) recommended upfront systemic chemotherapy and the option of local therapy to the chest and metastatic focus after initial chemotherapy; 28 respondents (26.4%) recommended upfront chemoradiation to the chest and definitive radiation to the left iliac wing metastasis.
The third clinical case described a male aged 70 years with a history of a treated base of tongue squamous cell carcinoma, with a solitary metastatic focus within the right lung. Respondents could pick multiple treatment options and 85 (81.7%) favored upfront definitive local therapy with surgery or stereotactic body radiotherapy (SBRT), rather than upfront chemotherapy, with future consideration for local treatment. About half of respondents (51.8%) recommended SBRT and 41.2% would let the patient decide between surgery or SBRT. Additionally, 39.6% included in their patient counselling that the treatment may be for curative intent.
Discussion
The use of local treatment to increased PFS, OS, or even cure treatment for OMD has become more accepted since the 2018 ASTRO meeting.4,5 Palma et al analyzed a controlled primary malignancy of any histology and ≤ 5 metastatic lesions, with all lesions amenable to SBRT.4 With a median follow-up of 51 months when comparing the standard-of-care (SOC) arm and the SBRT arm, the 5-year PFS was not reached and the 5-year OS rates were 17.7% and 42.3% (P = .006), respectively. In the SBRT arm, about 1 in 5 patients survived > 5 years without a recurrence or disease progression, vs 0 patients in the control arm. There was a 29% rate of grade 2 or higher toxicity in the SBRT arm, including 3 deaths that were likely due to treatment. Subsequent trials, such as the phase 3 SABR-COMET-3 (1-3 metastases), phase 3 SABR-COMET-10 (4-10 metastases), and phase 1 ARREST (> 10 metastases) trials, have been specifically designed to minimize treatment-related toxicities.13-15
Gomez et al analyzed patients at 3 sites with a controlled NSCLC primary tumor and ≤ 3 metastases.5 At a follow-up of 38.8 months, the PFS was 4.4 months in the SOC arm vs 14.2 months in the RT and/or surgery local treatment arm (P = .02). There was also an OS benefit of 17.0 vs 41.2 months (P = .02), respectively.
Several RCTs soon followed that demonstrated improved PFS and OS with local radiotherapy for OMD; however, total metastatic ablation of the foci is necessary to attain these PFS and OS benefits.6-9 Still, an oncologic benefit has yet to be proven. The randomized NRGBR002 study phase 2/3 trial for oligometastatic breast cancer included patients with ≤ 4 extracranial metastases and controlled primary disease to metastasis-directed therapy (SBRT and/ or surgical resection) and systemic therapy vs systemic therapy alone.10 The study did not demonstrate improved PFS or OS at 3 years. However, for most breast cancers, especially with the rapid advancements in systemic therapy that have been achieved, longer follow-up may be necessary to detect a significant difference.
The prospective single-arm phase 2 SABR-5 trial retrospectively demonstrated important lessons about the timing of SBRT and systemic therapy.11 This study included patients with ≤ 5 metastases of any histology, and they received SBRT to all lesions. SABR-5 retrospectively compared patients who received upfront systemic therapy followed by SBRT vs another cohort that first received SBRT and did not receive systemic therapy until there was disease progression. Patients with oligo-progression were excluded, as it demonstrated systemic drug resistance. At a median follow-up time of 34 months, delayed systemic treatment was associated with shorter PFS (23 vs 34 months, respectively; P = .001), but not worse 3-year OS (80% vs 85%, respectively; P = .66). In addition, the delayed systemic treatment arm demonstrated a reduced risk of grade 2 or higher SBRT-related toxicity (odds ratio, 0.35; P < .001).
Similarly, the STOMP phase 2 trial analyzed the role of metastasis-directed therapy (MDT) in delaying initiation of androgen deprivation therapy (ADT) in a randomized phase 2 trial.16 This study included patients with asymptomatic PCa with a biochemical recurrence after primary treatment, 1 to 3 extracranial metastatic lesions, and serum testosterone levels > 50 ng/mL. Sixty-two patients were randomized 1:1 to either MDT (SBRT or surgery) of all lesions or surveillance. The 5-year ADT-free survival was 34% for MDT vs 8% for surveillance (P = .06).
VHA Radiation Oncology
The VHA has 138 departments of medical oncology, but only 41 departments of radiation oncology. Compared with medical oncologists without an on-site radiation oncology department, those with on-site departments were more likely to believe that local RT was potentially curative (94.4% vs 72.7%, respectively; P = .04). This finding suggests that a cancer center that includes both specialties has closer collaboration, which results in greater inclination to embrace local RT for OMD, as it has demonstrated PFS and OS benefits.
The radiation and medical oncologists surveyed had statistically significant differences in response by specialty regarding the maximal number of lesions still believed to constitute OMD. Most radiation oncologists classified ≤ 5 lesions as OMD, whereas most medical oncologists classified ≤ 3 lesions as OMD. This difference is not unexpected. There is no universally agreed-upon definition of OMD, and criteria differ across studies.
While the SABR-COMET trial did include ≤ 5 metastatic lesions, it was a phase 2 RCT, making subgroup analysis difficult. Ongoing phase 3 trials that are more specific in the number of metastases, comparing 1 to 3 vs 4 to 10 metastases (SABR-COMET-3 and SABR-COMET-10, respectively).13,14 There is even an ongoing phase 1 trial (ARREST) studying the potential benefits of treating (“restraining”) > 10 metastases, if dosimetrically feasible.15 Within the VHA, VA STARPORT is investigating MDT for recurrent or de novo hormone-sensitive metastatic PCa.17 The ongoing HALT phase 2/3 trial focuses on patients with actionable mutations to help determine management of oligo-progression in mutation-positive NSCLC.18
There was no significant difference by specialty in who responded that offering local RT for OMD treatment should not be limited by histology (55.2% of radiation oncologists and 50.0% of medical oncologists; P = .60). Oncologists could make the argument that some histologies (eg, pancreatic adenocarcinomas) have such poor prognoses that local RT would not meaningfully affect oncologic outcomes, while potentially adding toxicity, whereas others could point to improved systemic therapy regimens and the low toxicity rates with careful hypofractionation regimens. Of note, the 41-patient phase 2 EXTEND trial for pancreatic ductal adenocarcinoma suggested an oncologic benefit to MDT, with far better PFS and no grade ≥ 3 toxicities related to MDT.19 About half of respondents for each specialty believed the primary histology should affect the decision. Further clarification may emerge from phase 3 trials.
Of note, a 2023 study of 44 radiation and medical oncologists at 2 Harvard Medical School-affiliated hospitals found that for synchronous OMD, 50.0% of medical oncologists and 5.3% (P < .01) of radiation oncologists recommended systemic treatment, suggesting a greater divergence in approach than found in this study.20
Limitations
The response rate of 17.0% raised a potential for selection bias, but this rate is expected for a nonincentivized medical survey. A study by the American Board of Internal Medicine with 11 surveys and 6 weekly email contacts only generated a 23.7% response rate, while another study among physicians demonstrated a 4.5% response rate for email-based contact and 11.8% for mail-based contact.21,22 We could have asked participants questions regarding demographics and geography to ensure the survey represented a diverse sample of the medical community, although additional questions would likely suppress the response rate. Additional data collection about respondents may elucidate the rationale for differences in their responses, especially between the specialties. In a planned subsequent survey in several years, the question on the number of lesions that qualifies as OMD may be amended to reflect the context and dosimetry for the maximal number of metastases constituting OMD; the joint ESTRO-ASTRO consensus defined OMD as 1 to 5 metastatic lesions, but in which all metastatic sites must be safely treatable.12 Also, fewer example cases could be included to simplify the survey and boost response rates. A future survey may ask about the timing of SBRT and systemic therapy, and whether SBRT can safely delay systemic therapy.
Conclusions
Survey results demonstrated significant confidence among both radiation oncologists and medical oncologists that local RT for OMD improves outcomes, which is encouraging and a reflection of the recent evidence-based paradigm shift in viewing metastatic disease as a spectrum. However, there is a difference between radiation oncologists and medical oncologists in how they define OMD, and preferred treatment of the sample cases presented revealed nuanced differences by specialty. Close collaboration with radiation oncologists influences the belief of medical oncologists in the beneficial role of RT for OMD. As more phase 3 data for OMD local treatments emerge, additional investigation is needed on how beliefs and practice patterns evolve among radiation and medical oncologists.
- Barney JD, Churchill EJ. Adenocarcinoma of the kidney with metastasis to the lung. J Urology. 1939.
- Hellman S, Weichselbaum RR. Oligometastases. J Clin Oncol. 1995;13(1):8-10. doi:10.1200/JCO.1995.13.1.8
- Ruers T, Punt C, Van Coevorden F, et al. Radiofrequency ablation combined with systemic treatment versus systemic treatment alone in patients with non-resectable colorectal liver metastases: a randomized EORTC Intergroup phase II study (EORTC 4004). Ann Oncol. 2012;23(10):2619-2626. doi:10.1093/annonc/mds053
- Palma DA, Olson R, Harrow S, et al. Stereotactic ablative radiotherapy for the comprehensive treatment of oligometastatic cancers: long-term results of the SABR-COMET phase II randomized trial. J Clin Oncol. 2020;38(25):2830- 2838. doi:10.1200/JCO.20.00818
- Gomez DR, Tang C, Zhang J, et al. Local consolidative therapy vs. maintenance therapy or observation for patients with oligometastatic non-small-cell lung cancer: long-term results of a multi-institutional, phase II, randomized study. J Clin Oncol. 2019;37(18):1558-1565. doi:10.1200/JCO.19.00201
- Iyengar P, Wardak Z, Gerber DE, et al. Consolidative radiotherapy for limited metastatic non-small-cell lung cancer: a phase 2 randomized clinical trial. JAMA Oncol. 2018;4(1):e173501. doi:10.1001/jamaoncol.2017.3501
- Phillips R, Shi WY, Deek M, et al. Outcomes of observation vs stereotactic ablative radiation for oligometastatic prostate cancer: the ORIOLE phase 2 randomized clinical trial. JAMA Oncol. 2020;6(5):650-659. doi:10.1001/jamaoncol.2020.0147
- Wang XS, Bai YF, Verma V, et al. Randomized trial of first-line tyrosine kinase inhibitor with or without radiotherapy for synchronous oligometastatic EGFR-mutated NSCLC. J Natl Cancer Inst 2023;115(6):742-748. doi:10.1093/jnci/djac015
- Tang C, Sherry AD, Haymaker C, et al. Addition of metastasis- directed therapy to intermittent hormone therapy for oligometastatic prostate cancer (EXTEND): a multicenter, randomized phase II trial. Am Soc Radiat Oncol Annu Meet. 2023;9(6):825-834. doi:10.1001/jamaoncol.2023.0161
- Chmura SJ, Winter KA, Woodward WA, et al. NRG-BR002: a phase IIR/III trial of standard of care systemic therapy with or without stereotactic body radiotherapy (SBRT) and/or surgical resection (SR) for newly oligometastatic breast cancer (NCT02364557). J Clin Oncol. 2022;40:1007. doi:10.1200/JCO.2022.40.16_suppl.1007
- Baker S, Lechner L, Liu M, et al. Upfront versus delayed systemic therapy in patients with oligometastatic cancer treated with SABR in the phase 2 SABR-5 trial. Int J Radiat Oncol Biol Phys. 2024;118(5):1497-1506. doi:10.1016/j.ijrobp.2023.11.007
- Lievens Y, Guckenberger M, Gomez D, et al. Defining oligometastatic disease from a radiation oncology perspective: an ESTRO-ASTRO consensus document. Radiother Oncol. 2020;148:157-166. doi:10.1016/j.radonc.2020.04.003
- Olson R, Mathews L, Liu M, et al. Stereotactic ablative radiotherapy for the comprehensive treatment of 1-3 oligometastatic tumors (SABR-COMET-3): study protocol for a randomized phase III trial. BMC Cancer. 2020;20(1):380. doi:10.1186/s12885-020-06876-4
- Palma DA, Olson R, Harrow S, et al. Stereotactic ablative radiotherapy for the comprehensive treatment of 4-10 oligometastatic tumors (SABR-COMET-10): study protocol for a randomized phase III trial. BMC Cancer. 2019;19(1):816. doi:10.1186/s12885-019-5977-6
- Bauman GS, Corkum MT, Fakir H, et al. Ablative radiation therapy to restrain everything safely treatable (ARREST): study protocol for a phase I trial treating polymetastatic cancer with stereotactic radiotherapy. BMC Cancer. 2021;21(1):405. doi:10.1186/s12885-021-08136-5
- Ost P, Reynders D, Decaestecker K, et al. Surveillance or metastasis-directed therapy for oligometastatic prostate cancer recurrence (STOMP): five-year results of a randomized phase II trial. J Clin Oncol. 2020;38:suppl.
- Solanki AA, Campbell D, Carlson K, et al. Veterans Affairs seamless phase II/III randomized trial of standard systemic therapy with or without PET-directed local therapy for oligometastatic prostate cancer (VA STARPORT). J Clin Oncol. 2024;42:16.
- McDonald F, Guckenberger M, Popat S. EP08.03-005 HALT – Targeted therapy with or without dose-intensified radiotherapy in oligo-progressive disease in oncogene addicted lung tumours. J Thor Oncol. 2022;17:S492.
- Ludmir EB, Sherry AD, Fellman BM, et al. Addition of metastasis- directed therapy to systemic therapy for oligometastatic pancreatic ductal adenocarcinoma (EXTEND): a multicenter, randomized phase II trial. J Clin Oncol. 2024;42(32):3795-3805. doi:10.1200/JCO.24.00081
- Cho HL, Balboni T, Christ SB, et al. Is oligometastatic cancer curable? A survey of oncologist perspectives, decision making, and communication. Adv Radiat Oncol. 2023;8(5):101221. doi:10.1016/j.adro.2023.101221
- Barnhart BJ, Reddy SG, Arnold GK. Remind me again: physician response to web surveys: the effect of email reminders across 11 opinion survey efforts at the American Board of Internal Medicine from 2017 to 2019. Eval Health Prof. 2021;44(3):245-259. doi:10.1177/01632787211019445
- Murphy CC, Lee SJC, Geiger AM, et al. A randomized trial of mail and email recruitment strategies for a physician survey on clinical trial accrual. BMC Med Res Methodol. 2020;20(1):123. doi:10.1186/s12874-020-01014-x
- Barney JD, Churchill EJ. Adenocarcinoma of the kidney with metastasis to the lung. J Urology. 1939.
- Hellman S, Weichselbaum RR. Oligometastases. J Clin Oncol. 1995;13(1):8-10. doi:10.1200/JCO.1995.13.1.8
- Ruers T, Punt C, Van Coevorden F, et al. Radiofrequency ablation combined with systemic treatment versus systemic treatment alone in patients with non-resectable colorectal liver metastases: a randomized EORTC Intergroup phase II study (EORTC 4004). Ann Oncol. 2012;23(10):2619-2626. doi:10.1093/annonc/mds053
- Palma DA, Olson R, Harrow S, et al. Stereotactic ablative radiotherapy for the comprehensive treatment of oligometastatic cancers: long-term results of the SABR-COMET phase II randomized trial. J Clin Oncol. 2020;38(25):2830- 2838. doi:10.1200/JCO.20.00818
- Gomez DR, Tang C, Zhang J, et al. Local consolidative therapy vs. maintenance therapy or observation for patients with oligometastatic non-small-cell lung cancer: long-term results of a multi-institutional, phase II, randomized study. J Clin Oncol. 2019;37(18):1558-1565. doi:10.1200/JCO.19.00201
- Iyengar P, Wardak Z, Gerber DE, et al. Consolidative radiotherapy for limited metastatic non-small-cell lung cancer: a phase 2 randomized clinical trial. JAMA Oncol. 2018;4(1):e173501. doi:10.1001/jamaoncol.2017.3501
- Phillips R, Shi WY, Deek M, et al. Outcomes of observation vs stereotactic ablative radiation for oligometastatic prostate cancer: the ORIOLE phase 2 randomized clinical trial. JAMA Oncol. 2020;6(5):650-659. doi:10.1001/jamaoncol.2020.0147
- Wang XS, Bai YF, Verma V, et al. Randomized trial of first-line tyrosine kinase inhibitor with or without radiotherapy for synchronous oligometastatic EGFR-mutated NSCLC. J Natl Cancer Inst 2023;115(6):742-748. doi:10.1093/jnci/djac015
- Tang C, Sherry AD, Haymaker C, et al. Addition of metastasis- directed therapy to intermittent hormone therapy for oligometastatic prostate cancer (EXTEND): a multicenter, randomized phase II trial. Am Soc Radiat Oncol Annu Meet. 2023;9(6):825-834. doi:10.1001/jamaoncol.2023.0161
- Chmura SJ, Winter KA, Woodward WA, et al. NRG-BR002: a phase IIR/III trial of standard of care systemic therapy with or without stereotactic body radiotherapy (SBRT) and/or surgical resection (SR) for newly oligometastatic breast cancer (NCT02364557). J Clin Oncol. 2022;40:1007. doi:10.1200/JCO.2022.40.16_suppl.1007
- Baker S, Lechner L, Liu M, et al. Upfront versus delayed systemic therapy in patients with oligometastatic cancer treated with SABR in the phase 2 SABR-5 trial. Int J Radiat Oncol Biol Phys. 2024;118(5):1497-1506. doi:10.1016/j.ijrobp.2023.11.007
- Lievens Y, Guckenberger M, Gomez D, et al. Defining oligometastatic disease from a radiation oncology perspective: an ESTRO-ASTRO consensus document. Radiother Oncol. 2020;148:157-166. doi:10.1016/j.radonc.2020.04.003
- Olson R, Mathews L, Liu M, et al. Stereotactic ablative radiotherapy for the comprehensive treatment of 1-3 oligometastatic tumors (SABR-COMET-3): study protocol for a randomized phase III trial. BMC Cancer. 2020;20(1):380. doi:10.1186/s12885-020-06876-4
- Palma DA, Olson R, Harrow S, et al. Stereotactic ablative radiotherapy for the comprehensive treatment of 4-10 oligometastatic tumors (SABR-COMET-10): study protocol for a randomized phase III trial. BMC Cancer. 2019;19(1):816. doi:10.1186/s12885-019-5977-6
- Bauman GS, Corkum MT, Fakir H, et al. Ablative radiation therapy to restrain everything safely treatable (ARREST): study protocol for a phase I trial treating polymetastatic cancer with stereotactic radiotherapy. BMC Cancer. 2021;21(1):405. doi:10.1186/s12885-021-08136-5
- Ost P, Reynders D, Decaestecker K, et al. Surveillance or metastasis-directed therapy for oligometastatic prostate cancer recurrence (STOMP): five-year results of a randomized phase II trial. J Clin Oncol. 2020;38:suppl.
- Solanki AA, Campbell D, Carlson K, et al. Veterans Affairs seamless phase II/III randomized trial of standard systemic therapy with or without PET-directed local therapy for oligometastatic prostate cancer (VA STARPORT). J Clin Oncol. 2024;42:16.
- McDonald F, Guckenberger M, Popat S. EP08.03-005 HALT – Targeted therapy with or without dose-intensified radiotherapy in oligo-progressive disease in oncogene addicted lung tumours. J Thor Oncol. 2022;17:S492.
- Ludmir EB, Sherry AD, Fellman BM, et al. Addition of metastasis- directed therapy to systemic therapy for oligometastatic pancreatic ductal adenocarcinoma (EXTEND): a multicenter, randomized phase II trial. J Clin Oncol. 2024;42(32):3795-3805. doi:10.1200/JCO.24.00081
- Cho HL, Balboni T, Christ SB, et al. Is oligometastatic cancer curable? A survey of oncologist perspectives, decision making, and communication. Adv Radiat Oncol. 2023;8(5):101221. doi:10.1016/j.adro.2023.101221
- Barnhart BJ, Reddy SG, Arnold GK. Remind me again: physician response to web surveys: the effect of email reminders across 11 opinion survey efforts at the American Board of Internal Medicine from 2017 to 2019. Eval Health Prof. 2021;44(3):245-259. doi:10.1177/01632787211019445
- Murphy CC, Lee SJC, Geiger AM, et al. A randomized trial of mail and email recruitment strategies for a physician survey on clinical trial accrual. BMC Med Res Methodol. 2020;20(1):123. doi:10.1186/s12874-020-01014-x
Radiation and Medical Oncology Perspectives on Oligometastatic Disease Treatment
Radiation and Medical Oncology Perspectives on Oligometastatic Disease Treatment
Assessing the Impact of Antidepressants on Cancer Treatment: A Retrospective Analysis of 14 Antineoplastic Agents
Assessing the Impact of Antidepressants on Cancer Treatment: A Retrospective Analysis of 14 Antineoplastic Agents
Cancer patients experience depression at rates > 5 times that of the general population.1-11 Despite an increase in palliative care use, depression rates continued to rise.2-4 Between 5% to 16% of outpatients, 4% to 14% of inpatients, and up to 49% of patients receiving palliative care experience depression.5 This issue also impacts families and caregivers.1 A 2021 meta-analysis found that 23% of active military personnel and 20% of veterans experience depression.11
Antidepressants approved by the US Food and Drug Administration (FDA) target the serotonin, norepinephrine, or dopamine systems and include boxed warnings about an increased risk of suicidal thoughts in adults aged 18 to 24 years.12,13 These medications are categorized into several classes: monoamine oxidase inhibitors (MAOIs), tricyclic antidepressants (TCAs), tetracyclic antidepressants (TeCAs), norepinephrine-dopamine reuptake inhibitors (NDRIs), selective serotonin reuptake inhibitors (SSRIs), serotonin-norepinephrine reuptake inhibitors (SNRIs), serotonin receptor modulators (SRMs), serotonin-melatonin receptor antagonists (SMRAs), and N—methyl-D-aspartate receptor antagonists (NMDARAs).14,15 The first FDA-approved antidepressants, iproniazid (an MAOI) and imipramine (a TCA) laid the foundation for the development of newer classes like SSRIs and SNRIs.15-17
Older antidepressants such as MAOIs and TCAs are used less due to their adverse effects (AEs) and drug interactions. MAOIs, such as iproniazid, selegiline, moclobemide, tranylcypromine, isocarboxazid, and phenelzine, have numerous AEs and drug interactions, making them unsuitable for first- or second-line treatment of depression.14,18-21 TCAs such as doxepin, amitriptyline, nortriptyline, imipramine, desipramine, clomipramine, trimipramine, protriptyline, maprotiline, and amoxapine have a narrow therapeutic index requiring careful monitoring for signs of toxicity such as QRS widening, tremors, or confusion. Despite the issues, TCAs are generally classified as second-line agents for major depressive disorder (MDD). TCAs have off-label uses for migraine prophylaxis, treatment of obsessive-compulsive disorder (OCD), insomnia, and chronic pain management first-line.14,22-29
Newer antidepressants, including TeCAs and NDRIs, are typically more effective, but also come with safety concerns. TeCAs like mirtazapine interact with several medications, including MAOIs, serotonin-increasing drugs, alcohol, cannabidiol, and marijuana. Mirtazapine is FDA-approved for the treatment of moderate to severe depression in adults. It is also used off-label to treat insomnia, panic disorder, posttraumatic stress disorder (PTSD), generalized anxiety disorder (GAD), social anxiety disorder (SAD), headaches, and migraines. Compared to other antidepressants, mirtazapine is effective for all stages of depression and addresses a broad range of related symptoms.14,30-34 NDRIs, such as bupropion, also interact with various medications, including MAOIs, other antidepressants, stimulants, and alcohol. Bupropion is FDA-approved for smoking cessation and to treat depression and SAD. It is also used off-label for depression- related bipolar disorder or sexual dysfunction, attention-deficit/hyperactivity disorder (ADHD), and obesity.14,35-42
SSRIs, SNRIs, and SRMs should be used with caution. SSRIs such as sertraline, citalopram, escitalopram, fluoxetine, paroxetine, and fluvoxamine are first-line treatments for depression and various psychiatric disorders due to their safety and efficacy. Common AEs of SSRIs include sexual dysfunction, sleep disturbances, weight changes, and gastrointestinal (GI) issues. SSRIs can prolong the QT interval, posing a risk of life-threatening arrhythmia, and may interact with other medications, necessitating treatment adjustments. The FDA approved SSRIs for MDD, GAD, bulimia nervosa, bipolar depression, OCD, panic disorder, premenstrual dysphoric disorder, treatment-resistant depression, PTSD, and SAD. Off-label uses include binge eating disorder, body dysmorphic disorder, fibromyalgia, premature ejaculation, paraphilias, autism, Raynaud phenomenon, and vasomotor symptoms associated with menopause. Among SSRIs, sertraline and escitalopram are noted for their effectiveness and tolerability.14,43-53
SNRIs, including duloxetine, venlafaxine, desvenlafaxine, milnacipran, and levomilnacipran, may increase bleeding risk, especially when taken with blood thinners. They can also elevate blood pressure, which may worsen if combined with stimulants. SNRIs may interact with other medications that affect serotonin levels, increasing the risk of serotonin syndrome when taken with triptans, pain medications, or other antidepressants.14 Desvenlafaxine has been approved by the FDA (but not by the European Medicines Agency).54-56 Duloxetine is FDA-approved for the treatment of depression, neuropathic pain, anxiety disorders, fibromyalgia, and musculoskeletal disorders. It is used off-label to treat chemotherapy-induced peripheral neuropathy and stress urinary incontinence.57-61 Venlafaxine is FDA-approved for depression, SAD, and panic disorder, and is prescribed off-label to treat ADHD, neuropathy, fibromyalgia, cataplexy, and PTSD, either alone or in combination with other medications.62,63 Milnacipran is not approved for MDD; levomilnacipran received approval in 2013.64
SRMs such as trazodone, nefazodone, vilazodone, and vortioxetine also function as serotonin reuptake inhibitors.14,15 Trazodone is FDA-approved for MDD. It has been used off-label to treat anxiety, Alzheimer disease, substance misuse, bulimia nervosa, insomnia, fibromyalgia, and PTSD when first-line SSRIs are ineffective. A notable AE of trazodone is orthostatic hypotension, which can lead to dizziness and increase the risk of falls, especially in geriatric patients.65-70 Nefazodone was discontinued in Europe in 2003 due to rare cases of liver toxicity but remains available in the US.71-74 Vilazodone and vortioxetine are FDA-approved.
The latest classes of antidepressants include SMRAs and NMDARAs.14 Agomelatine, an SMRA, was approved in Europe in 2009 but rejected by the FDA in 2011 due to liver toxicity.75 NMDARAs like esketamine and a combination of dextromethorphan and bupropion received FDA approval in 2019 and 2022, respectively.76,77
This retrospective study analyzes noncancer drugs used during systemic chemotherapy based on a dataset of 14 antineoplastic agents. It sought to identify the most dispensed noncancer drug groups, discuss findings, compare patients with and without antidepressant prescriptions, and examine trends in antidepressant use from 2002 to 2023. This analysis expands on prior research.78-81
Methods
The Walter Reed National Military Medical Center Institutional Review Board approved the study protocol and ensured compliance with the Health Insurance Portability and Accountability Act as an exempt protocol. The Joint Pathology Center (JPC) of the US Department of Defense (DoD) Cancer Registry Program and Military Health System (MHS) data experts from the Comprehensive Ambulatory/Professional Encounter Record (CAPER) and Pharmacy Data Transaction Service (PDTS) provided data for the analysis.
Data Sources
The JPC DoD Cancer Registry Program contains data from 1998 to 2024. CAPER and PDTS are part of the MHS Data Repository/Management Analysis and Reporting Tool database. Each observation in CAPER represents an ambulatory encounter at a military treatment facility (MTF). CAPER records are available from 2003 to 2024. PDTS records are available from 2002 to 2004. Each observation in PDTS represents a prescription filled for an MHS beneficiary, excluding those filled at international civilian pharmacies and inpatient pharmacy prescriptions.
This cross-sectional analysis requested data extraction for specific cancer drugs from the DoD Cancer Registry, focusing on treatment details, diagnosis dates, patient demographics, and physicians’ comments on AEs. After identifying patients, CAPER was used to identify additional health conditions. PDTS was used to compile a list of prescription medications filled during systemic cancer treatment or < 2 years postdiagnosis.
The 2016 Surveillance, Epidemiology, and End Results Program Coding and Staging Manual and International Classification of Diseases for Oncology, 3rd edition, 1st revision, were used to decode disease and cancer types.82,83 Data sorting and analysis were performed using Microsoft Excel. The percentage for the total was calculated by using the number of patients or data available within the subgroup divided by the total number of patients or data variables. To compare the mean number of dispensed antidepressants to those without antidepressants, a 2-tailed, 2-sample z test was used to calculate the P value and determine statistical significance (P < .05) using socscistatistics.com.
Data were extracted 3 times between 2021 and 2023. The initial 2021 protocol focused on erlotinib and gefitinib. A modified protocol in 2022 added paclitaxel, cisplatin, docetaxel, pemetrexed, and crizotinib; further modification in 2023 included 8 new antineoplastic agents and 2 anticoagulants. Sotorasib has not been prescribed in the MHS, and JPC lacks records for noncancer drugs. The 2023 dataset comprised 2210 patients with cancer treated with 14 antineoplastic agents; 2104 had documented diagnoses and 2113 had recorded prescriptions. Data for erlotinib, gefitinib, and paclitaxel have been published previously.78,79
Results
Of 2113 patients with recorded prescriptions, 1297 patients (61.4%) received 109 cancer drugs, including 96 antineoplastics, 7 disease-modifying antirheumatic agents, 4 biologic response modifiers, and 2 calcitonin gene-related peptides. Fourteen antineoplastic agents had complete data from JPC, while others were noted for combination therapies or treatment switches from the PDTS (Table 1). Seventy-six cancer drugs were prescribed with antidepressants in 489 patients (eAppendix).

The JPC provided 2242 entries for 2210 patients, ranging in age from 2 months to 88 years (mean, 56 years), documenting treatment from September 1988 to January 2023. Thirty-two patients had duplicate entries due to multiple cancer locations or occurrences. Of the 2242 patients, 1541 (68.7%) were aged > 50 years, 975 patients (43.5%) had cancers that were stage III or IV, and 1267 (56.5%) had cancers that were stage 0, I, II, or not applicable/unknown. There were 51 different types of cancer: breast, lung, testicular, endometrial, and ovarian were most common (n ≥ 100 patients). Forty-two cancer types were documented among 750 patients prescribed antidepressants (Table 2).

The CAPER database recorded 8882 unique diagnoses for 2104 patients, while PDTS noted 1089 unique prescriptions within 273 therapeutic codes for 2113 patients. Nine therapeutic codes (opiate agonists, adrenals, cathartics-laxatives, nonsteroidal anti-inflammatory agents, antihistamines for GI conditions, 5-HT3 receptor antagonists, analgesics and antipyretic miscellanea, antineoplastic agents, and proton-pump inhibitors) and 8 drugs (dexamethasone, prochlorperazine, ondansetron, docusate, acetaminophen, ibuprofen, oxycodone, and polyethylene glycol 3350) were associated with > 1000 patients (≥ 50%). Patients had between 1 and 275 unique health conditions and filled 1 to 108 prescriptions. The mean (SD) number of diagnoses and prescriptions was 50 (28) and 29 (12), respectively. Of the 273 therapeutic codes, 30 groups were analyzed, with others categorized into miscellaneous groups such as lotions, vaccines, and devices. Significant differences in mean number of prescriptions were found for patients taking antidepressants compared to those not (P < .05), except for anticonvulsants and antipsychotics (P = .12 and .09, respectively) (Table 3).

Antidepressants
Of the 2113 patients with recorded prescriptions, 750 (35.5%) were dispensed 17 different antidepressants. Among these 17 antidepressants, 183 (8.7%) patients received duloxetine, 158 (7.5%) received venlafaxine, 118 (5.6%) received trazodone, and 107 (5.1%) received sertraline (Figure 1, Table 4). Of the 750 patients, 509 (67.9%) received 1 antidepressant, 168 (22.4%) received 2, 60 (8.0%) received 3, and 13 (1.7%) received > 3. Combinations varied, but only duloxetine and trazodone were prescribed to > 10 patients.



Antidepressants were prescribed annually at an overall mean (SD) rate of 23% (5%) from 2003 to 2022 (Figure 2). Patients on antidepressants during systemic therapy had a greater number of diagnosed medical conditions and received more prescription medications compared to those not taking antidepressants (P < .001) (Figure 3). The 745 patients taking antidepressants in CAPER data had between 1 and 275 diagnosed medical issues, with a mean (SD) of 55 (31) vs a range of 1 to 209 and a mean (SD) of 46 (26) for the 1359 patients not taking antidepressants. The 750 patients on antidepressants in PDTS data had between 8 and 108 prescriptions dispensed, with a mean (SD) of 32 (12), vs a range of 1 to 65 prescriptions and a mean (SD) of 29 (12) for 1363 patients not taking antidepressants.


Discussion
The JPC DoD Cancer Registry includes information on cancer types, stages, treatment regimens, and physicians’ notes, while noncancer drugs are sourced from the PDTS database. The pharmacy uses a different documentation system, leading to varied classifications.
Database reliance has its drawbacks. For example, megestrol is coded as a cancer drug, although it’s primarily used for endometrial or gynecologic cancers. Many drugs have multiple therapeutic codes assigned to them, including 10 antineoplastic agents: diclofenac, Bacillus Calmette-Guérin (BCG), megestrol acetate, tamoxifen, anastrozole, letrozole, leuprolide, goserelin, degarelix, and fluorouracil. Diclofenac, BCG, and mitomycin have been repurposed for cancer treatment.84-87 From 2003 to 2023, diclofenac was prescribed to 350 patients for mild-to-moderate pain, with only 2 patients receiving it for cancer in 2018. FDA-approved for bladder cancer in 1990, BCG was prescribed for cancer treatment for 1 patient in 2021 after being used for vaccines between 2003 and 2018. Tamoxifen, used for hormone receptor-positive breast cancer from 2004 to 2017 with 53 patients, switched to estrogen agonist-antagonists from 2017 to 2023 with 123 patients. Only a few of the 168 patients were prescribed tamoxifen using both codes.88-91 Anastrozole and letrozole were coded as antiestrogens for 7 and 18 patients, respectively, while leuprolide and goserelin were coded as gonadotropins for 59 and 18 patients. Degarelix was coded as antigonadotropins, fluorouracil as skin and mucous membrane agents miscellaneous, and megestrol acetate as progestins for 7, 6, and 3 patients, respectively. Duloxetine was given to 186 patients, primarily for depression from 2005 to 2023, with 7 patients treated for fibromyalgia from 2022 to 2023.
Antidepressants Observed
Tables 1 and 5 provide insight into the FDA approval of 14 antineoplastics and antidepressants and their CYP metabolic pathways.92-122 In Table 4, the most prescribed antidepressant classes are SNRIs, SRMs, SSRIs, TeCAs, NDRIs, and TCAs. This trend highlights a preference for newer medications with weak CYP inhibition. A total of 349 patients were prescribed SSRIs, 343 SNRIs, 119 SRMs, 109 TCAs, 83 TeCAs, and 79 NDRIs. MAOIs, SMRAs, and NMDARAs were not observed in this dataset. While there are instances of dextromethorphan-bupropion and sertraline-escitalopram being dispensed together, it remains unclear whether these were NMDARA combinations.
Among the 14 specific antineoplastic agents, 10 are metabolized by CYP isoenzymes, primarily CYP3A4. Duloxetine neither inhibits nor is metabolized by CYP3A4, a reason it is often recommended, following venlafaxine.
Both duloxetine and venlafaxine are used off-label for chemotherapy-induced peripheral neuropathy related to paclitaxel and docetaxel. According to the CYP metabolized pathway, duloxetine tends to have more favorable DDIs than venlafaxine. In PDTS data, 371 patients were treated with paclitaxel and 180 with docetaxel, with respective antidepressant prescriptions of 156 and 70. Of the 156 patients dispensed paclitaxel, 62 (40%) were dispensed with duloxetine compared to 43 (28%) with venlafaxine. Of the 70 patients dispensed docetaxel, 23 (33%) received duloxetine vs 24 (34%) with venlafaxine.
Of 85 patients prescribed duloxetine, 75 received it with either paclitaxel or docetaxel (5 received both). Five patients had documented AEs (1 neuropathy related). Of 67 patients prescribed venlafaxine, 66 received it with either paclitaxel or docetaxel. Two patients had documented AEs (1 was neuropathy related, the same patient who received duloxetine). Of the 687 patients treated with paclitaxel and 337 with docetaxel in all databases, 4 experienced neuropathic AEs from both medications.79
Antidepressants can increase the risk of bleeding, especially when combined with blood thinners, and may elevate blood pressure, particularly alongside stimulants. Of the 554 patients prescribed 9 different anticoagulants, enoxaparin, apixaban, and rivaroxaban were the most common (each > 100 patients). Among these, 201 patients (36%) received both anticoagulants and antidepressants: duloxetine for 64 patients, venlafaxine for 30, trazodone for 35, and sertraline for 26. There were no data available to assess bleeding rates related to the evaluation of DDIs between these medication classes.
Antidepressants can be prescribed for erectile dysfunction. Of the 148 patients prescribed an antidepressant for erectile dysfunction, duloxetine, trazodone, and mirtazapine were the most common. Antidepressant preferences varied by cancer type. Duloxetine was the only antidepressant used for all types of cancer. Venlafaxine, duloxetine, trazodone, sertraline, and escitalopram were the most prescribed antidepressants for breast cancer, while duloxetine, mirtazapine, citalopram, sertraline, and trazodone were the most prescribed for lung cancer. Sertraline, duloxetine, trazodone, amitriptyline, and escitalopram were most common for testicular cancer. Duloxetine, venlafaxine, trazodone, amitriptyline, and sertraline were the most prescribed for endometrial cancer, while duloxetine, venlafaxine, amitriptyline, citalopram, and sertraline were most prescribed for ovarian cancer.
The broadness of International Statistical Classification of Diseases, Tenth Revision codes made it challenging to identify nondepression diagnoses in the analyzed population. However, if all antidepressants were prescribed to treat depression, service members with cancer exhibited a higher depression rate (35%) than the general population (25%). Of 2104 patients, 191 (9.1%) had mood disorders, and 706 (33.6%) had mental disorders: 346 (49.0%) had 1 diagnosis, and 360 (51.0%) had multiple diagnoses. The percentage of diagnoses varied yearly, with notable drops in 2003, 2007, 2011, 2014, and 2018, and peaks in 2006, 2008, 2013, 2017, and 2022. This fluctuation was influenced by events like the establishment of PDTS in 2002, the 2008 economic recession, a hospital relocation in 2011, the 2014 Ebola outbreak, and the COVID-19 pandemic. Although the number of patients receiving antidepressants increased from 2019 to 2022, the overall percentage of patients receiving them did not significantly change from 2003 to 2022, aligning with previous research.5,125
Many medications have potential uses beyond what is detailed in the prescribing information. Antidepressants can relieve pain, while pain medications may help with depression. Opioids were once thought to effectively treat depression, but this perspective has changed with a greater understanding of their risks, including misuse.126-131 Pain is a severe and often unbearable AE of cancer. Of 2113 patients, 92% received opioids; 34% received both opioids and antidepressants; 2% received only antidepressants; and 7% received neither. This study didn’t clarify whether those on opioids alone recognized their depression or if those on both were aware of their dependence. While SSRIs are generally not addictive, they can lead to physical dependence, and any medication can be abused if not managed properly.132-134
Conclusions
This retrospective study analyzes data from antineoplastic agents used in systemic cancer treatment between 1988 and 2023, with a particular focus on the use of antidepressants. Data on antidepressant prescriptions are incomplete and specific to these agents, which means the findings cannot be generalized to all antidepressants. Hence, the results indicate that patients taking antidepressants had more diagnosed health issues and received more medications compared to patients who were not on these drugs.
This study underscores the need for further research into the effects of antidepressants on cancer treatment, utilizing all data from the DoD Cancer Registry. Future research should explore DDIs between antidepressants and other cancer and noncancer medications, as this study did not assess AE documentation, unlike in studies involving erlotinib, gefitinib, and paclitaxel.78,79 Further investigation is needed to evaluate the impact of discontinuing antidepressant use during cancer treatment. This comprehensive overview provides insights for clinicians to help them make informed decisions regarding the prescription of antidepressants in the context of cancer treatment.
- National Cancer Institute. Depression (PDQ)-Health Professional Version. Updated July 25, 2024. Accessed April 4, 2025. https://www.cancer.gov/about-cancer/coping/feelings/depression-hp-pdq
- Krebber AM, Buffart LM, Kleijn G, et al. Prevalence of depression in cancer patients: a meta-analysis of diagnostic interviews and self-report instruments. Psychooncology. 2014;23(2):121-130. doi:10.1002/pon.3409
- Xiang X, An R, Gehlert S. Trends in antidepressant use among U.S. cancer survivors, 1999-2012. Psychiatr Serv. 2015;66(6):564. doi:10.1176/appi.ps.201500007
- Hartung TJ, Brähler E, Faller H, et al. The risk of being depressed is significantly higher in cancer patients than in the general population: Prevalence and severity of depressive symptoms across major cancer types. Eur J Cancer. 2017;72:46-53. doi:10.1016/j.ejca.2016.11.017
- Walker J, Holm Hansen C, Martin P, et al. Prevalence of depression in adults with cancer: a systematic review. Ann Oncol. 2013;24(4):895-900. doi:10.1093/annonc/mds575
- Pitman A, Suleman S, Hyde N, Hodgkiss A. Depression and anxiety in patients with cancer. BMJ. 2018;361:k1415. doi:10.1136/bmj.k1415
- Kisely S, Alotiby MKN, Protani MM, Soole R, Arnautovska U, Siskind D. Breast cancer treatment disparities in patients with severe mental illness: a systematic review and meta-analysis. Psychooncology. 2023;32(5):651-662. doi:10.1002/pon.6120
- Massie MJ. Prevalence of depression in patients with cancer. J Natl Cancer Inst Monogr. 2004;(32):57-71. doi:10.1093/jncimonographs/lgh014
- Rodin G, Katz M, Lloyd N, Green E, Mackay JA, Wong RK. Treatment of depression in cancer patients. Curr Oncol. 2007;14(5):180-188. doi:10.3747/co.2007.146
- Wilson KG, Chochinov HM, Skirko MG, et al. Depression and anxiety disorders in palliative cancer care. J Pain Symptom Manage. 2007;33(2):118-129. doi:10.1016/j.jpainsymman.2006.07.016
- Moradi Y, Dowran B, Sepandi M. The global prevalence of depression, suicide ideation, and attempts in the military forces: a systematic review and meta-analysis of cross sectional studies. BMC Psychiatry. 2021;21(1):510. doi:10.1186/s12888-021-03526-2
- Lu CY, Zhang F, Lakoma MD, et al. Changes in antidepressant use by young people and suicidal behavior after FDA warnings and media coverage: quasi-experimental study. BMJ. 2014;348:g3596. doi:10.1136/bmj.g3596
- Friedman RA. Antidepressants’ black-box warning--10 years later. N Engl J Med. 2014;371(18):1666-1668. doi:10.1056/NEJMp1408480
- Sheffler ZM, Patel P, Abdijadid S. Antidepressants. In: StatPearls. StatPearls Publishing. Updated May 26, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK538182/
- Miller JJ. Antidepressants, Part 1: 100 Years and Counting. Accessed April 4, 2025. Psychiatric Times. https:// www.psychiatrictimes.com/view/antidepressants-part-1-100-years-and-counting
- Hillhouse TM, Porter JH. A brief history of the development of antidepressant drugs: from monoamines to glutamate. Exp Clin Psychopharmacol. 2015;23(1):1-21. doi:10.1037/a0038550
- Mitchell PB, Mitchell MS. The management of depression. Part 2. The place of the new antidepressants. Aust Fam Physician. 1994;23(9):1771-1781.
- Sabri MA, Saber-Ayad MM. MAO Inhibitors. In: Stat- Pearls. StatPearls Publishing. Updated June 5, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK557395/
- Sub Laban T, Saadabadi A. Monoamine Oxidase Inhibitors (MAOI). In: StatPearls. StatPearls Publishing. Updated July 17, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK539848/
- Fiedorowicz JG, Swartz KL. The role of monoamine oxidase inhibitors in current psychiatric practice. J Psychiatr Pract. 2004;10(4):239-248. doi:10.1097/00131746-200407000-00005
- Flockhart DA. Dietary restrictions and drug interactions with monoamine oxidase inhibitors: an update. J Clin Psychiatry. 2012;73 Suppl 1:17-24. doi:10.4088/JCP.11096su1c.03
- Moraczewski J, Awosika AO, Aedma KK. Tricyclic Antidepressants. In: StatPearls. StatPearls Publishing. Updated August 17, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK557791/
- Almasi A, Patel P, Meza CE. Doxepin. In: StatPearls. StatPearls Publishing. Updated February 14, 2024. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK542306/
- Thour A, Marwaha R. Amitriptyline. In: StatPearls. Stat- Pearls Publishing. Updated July 18, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK537225/
- Radley DC, Finkelstein SN, Stafford RS. Off-label prescribing among office-based physicians. Arch Intern Med. 2006;166(9):1021-1026. doi:10.1001/archinte.166.9.1021
- Tesfaye S, Sloan G, Petrie J, et al. Comparison of amitriptyline supplemented with pregabalin, pregabalin supplemented with amitriptyline, and duloxetine supplemented with pregabalin for the treatment of diabetic peripheral neuropathic pain (OPTION-DM): a multicentre, double-blind, randomised crossover trial. Lancet. 2022;400(10353):680- 690. doi:10.1016/S0140-6736(22)01472-6
- Farag HM, Yunusa I, Goswami H, Sultan I, Doucette JA, Eguale T. Comparison of amitriptyline and US Food and Drug Administration-approved treatments for fibromyalgia: a systematic review and network metaanalysis. JAMA Netw Open. 2022;5(5):e2212939. doi:10.1001/jamanetworkopen.2022.12939
- Merwar G, Gibbons JR, Hosseini SA, Saadabadi A. Nortriptyline. In: StatPearls. StatPearls Publishing. Updated June 5, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK482214/
- Fayez R, Gupta V. Imipramine. In: StatPearls. StatPearls Publishing. Updated May 22, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK557656/
- Jilani TN, Gibbons JR, Faizy RM, Saadabadi A. Mirtazapine. In: StatPearls. StatPearls Publishing. Updated August 28, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK519059/
- Nutt DJ. Tolerability and safety aspects of mirtazapine. Hum Psychopharmacol. 2002;17 Suppl 1:S37-S41. doi:10.1002/hup.388
- Gandotra K, Chen P, Jaskiw GE, Konicki PE, Strohl KP. Effective treatment of insomnia with mirtazapine attenuates concomitant suicidal ideation. J Clin Sleep Med. 2018;14(5):901-902. doi:10.5664/jcsm.7142
- Anttila SA, Leinonen EV. A review of the pharmacological and clinical profile of mirtazapine. CNS Drug Rev. 2001;7(3):249-264. doi:10.1111/j.1527-3458.2001.tb00198.x
- Wang SM, Han C, Bahk WM, et al. Addressing the side effects of contemporary antidepressant drugs: a comprehensive review. Chonnam Med J. 2018;54(2):101-112. doi:10.4068/cmj.2018.54.2.101
- Huecker MR, Smiley A, Saadabadi A. Bupropion. In: StatPearls. StatPearls Publishing. Updated April 9, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK470212/
- Hsieh MT, Tseng PT, Wu YC, et al. Effects of different pharmacologic smoking cessation treatments on body weight changes and success rates in patients with nicotine dependence: a network meta-analysis. Obes Rev. 2019;20(6):895- 905. doi:10.1111/obr.12835
- Hankosky ER, Bush HM, Dwoskin LP, et al. Retrospective analysis of health claims to evaluate pharmacotherapies with potential for repurposing: association of bupropion and stimulant use disorder remission. AMIA Annu Symp Proc. 2018;2018:1292-1299.
- Livingstone-Banks J, Norris E, Hartmann-Boyce J, West R, Jarvis M, Hajek P. Relapse prevention interventions for smoking cessation. Cochrane Database Syst Rev. 2019; 2(2):CD003999. doi:10.1002/14651858.CD003999.pub5
- Clayton AH, Kingsberg SA, Goldstein I. Evaluation and management of hypoactive sexual desire disorder. Sex Med. 2018;6(2):59-74. doi:10.1016/j.esxm.2018.01.004
- Verbeeck W, Bekkering GE, Van den Noortgate W, Kramers C. Bupropion for attention deficit hyperactivity disorder (ADHD) in adults. Cochrane Database Syst Rev. 2017;10(10):CD009504. doi:10.1002/14651858.CD009504.pub2
- Ng QX. A systematic review of the use of bupropion for attention-deficit/hyperactivity disorder in children and adolescents. J Child Adolesc Psychopharmacol. 2017;27(2):112-116. doi:10.1089/cap.2016.0124
- Fava M, Rush AJ, Thase ME, et al. 15 years of clinical experience with bupropion HCl: from bupropion to bupropion SR to bupropion XL. Prim Care Companion J Clin Psychiatry. 2005;7(3):106-113. doi:10.4088/pcc.v07n0305
- Chu A, Wadhwa R. Selective Serotonin Reuptake Inhibitors. In: StatPearls. StatPearls Publishing. Updated May 1, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK554406/
- Singh HK, Saadabadi A. Sertraline. In: StatPearls. Stat- Pearls Publishing. Updated February 13, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK547689/
- MacQueen G, Born L, Steiner M. The selective serotonin reuptake inhibitor sertraline: its profile and use in psychiatric disorders. CNS Drug Rev. 2001;7(1):1-24. doi:10.1111/j.1527-3458.2001.tb00188.x
- Cipriani A, Furukawa TA, Salanti G, et al. Comparative efficacy and acceptability of 12 new-generation antidepressants: a multiple-treatments meta-analysis. Lancet. 2009;373(9665):746-758. doi:10.1016/S0140-6736(09)60046-5
- Nelson JC. The STAR*D study: a four-course meal that leaves us wanting more. Am J Psychiatry. 2006;163(11):1864-1866. doi:10.1176/ajp.2006.163.11.1864
- Sharbaf Shoar N, Fariba KA, Padhy RK. Citalopram. In: StatPearls. StatPearls Publishing. Updated November 7, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK482222/
- Landy K, Rosani A, Estevez R. Escitalopram. In: Stat- Pearls. StatPearls Publishing. Updated November 10, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK557734/
- Cavanah LR, Ray P, Goldhirsh JL, Huey LY, Piper BJ. Rise of escitalopram and the fall of citalopram. medRxiv. Preprint published online May 8, 2023. doi:10.1101/2023.05.07.23289632
- Sohel AJ, Shutter MC, Patel P, Molla M. Fluoxetine. In: StatPearls. StatPearls Publishing. Updated July 4, 2022. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK459223/
- Wong DT, Perry KW, Bymaster FP. Case history: the discovery of fluoxetine hydrochloride (Prozac). Nat Rev Drug Discov. 2005;4(9):764-774. doi:10.1038/nrd1821
- Shrestha P, Fariba KA, Abdijadid S. Paroxetine. In: Stat- Pearls. StatPearls Publishing. Updated July 17, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK526022/
- Naseeruddin R, Rosani A, Marwaha R. Desvenlafaxine. In: StatPearls. StatPearls Publishing. Updated July 10, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK534829
- Lieberman DZ, Massey SH. Desvenlafaxine in major depressive disorder: an evidence-based review of its place in therapy. Core Evid. 2010;4:67-82. doi:10.2147/ce.s5998
- Withdrawal assessment report for Ellefore (desvenlafaxine). European Medicine Agency. 2009. Accessed April 4, 2025. https://www.ema.europa.eu/en/documents/withdrawal-report/withdrawal-assessment-report-ellefore_en.pdf
- Dhaliwal JS, Spurling BC, Molla M. Duloxetine. In: Stat- Pearls. Statpearls Publishing. Updated May 29, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK549806/
- Hershman DL, Lacchetti C, Dworkin RH, et al. Prevention and management of chemotherapy-induced peripheral neuropathy in survivors of adult cancers: American Society of Clinical Oncology clinical practice guideline. J Clin Oncol. 2014;32(18):1941-1967. doi:10.1200/JCO.2013.54.0914
- Sommer C, Häuser W, Alten R, et al. Medikamentöse Therapie des Fibromyalgiesyndroms. Systematische Übersicht und Metaanalyse [Drug therapy of fibromyalgia syndrome. Systematic review, meta-analysis and guideline]. Schmerz. 2012;26(3):297-310. doi:10.1007/s00482-012-1172-2
- Bril V, England J, Franklin GM, et al. Evidence-based guideline: treatment of painful diabetic neuropathy: report of the American Academy of Neurology, the American Association of Neuromuscular and Electrodiagnostic Medicine, and the American Academy of Physical Medicine and Rehabilitation. Neurology. 2011;76(20):1758-1765. doi:10.1212/WNL.0b013e3182166ebe
- Attal N, Cruccu G, Baron R, et al. EFNS guidelines on the pharmacological treatment of neuropathic pain: 2010 revision. Eur J Neurol. 2010;17(9):1113-e88. doi:10.1111/j.1468-1331.2010.02999.x
- Singh D, Saadabadi A. Venlafaxine. In: StatPearls. StatePearls Publishing. Updated February 26, 2024. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK535363/
- Sertraline and venlafaxine: new indication. Prevention of recurrent depression: no advance. Prescrire Int. 2005;14(75):19-20.
- Bruno A, Morabito P, S p i n a E , M u s c a t ello MRl. The role of levomilnacipran in the management of major depressive disorder: a comprehensive review. Curr Neuro pharmacol. 2016;14(2):191-199. doi:10.2174/1570159x14666151117122458
- Shin JJ, Saadabadi A. Trazodone. In: StatPearls. StatPearls Publishing. Updated February 29, 2024. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK470560/
- Khouzam HR. A review of trazodone use in psychiatric and medical conditions. Postgrad Med. 2017;129(1):140-148. doi:10.1080/00325481.2017.1249265
- Smales ET, Edwards BA, Deyoung PN, et al. Trazodone effects on obstructive sleep apnea and non-REM arousal threshold. Ann Am Thorac Soc. 2015;12(5):758-764. doi:10.1513/AnnalsATS.201408-399OC
- Eckert DJ, Malhotra A, Wellman A, White DP. Trazodone increases the respiratory arousal threshold in patients with obstructive sleep apnea and a low arousal threshold. Sleep. 2014;37(4):811-819. doi:10.5665/sleep.3596
- Schatzberg AF, Nemeroff CB, eds. The American Psychiat ric Association Publishing Textbook of Psychopharmacology. 4th ed. American Psychiatric Publishing; 2009.
- Ruxton K, Woodman RJ, Mangoni AA. Drugs with anticholinergic effects and cognitive impairment, falls and allcause mortality in older adults: a systematic review and meta-analysis. Br J Clin Pharmacol. 2015;80(2):209-220. doi:10.1111/bcp.12617
- Nefazodone. In: LiverTox: Clinical and Research Information on Drug-Induced Liver Injury. National Institute of Diabetes and Digestive and Kidney Diseases. Updated March 6, 2020. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK548179/
- Drugs of Current Interest: Nefazodone. WHO Pharmaceuticals Newsletter:(1). 2003(1):7. https://web.archive.org/web/20150403165029/http:/apps.who.int/medicinedocs/en/d/Js4944e/3.html
- Choi S. Nefazodone (serzone) withdrawn because of hepatotoxicity. CMAJ. 2003;169(11):1187.
- Teva Nefazodone Statement. News release. Teva USA. December 20, 2021. Accessed April 4, 2025. https:// www.tevausa.com/news-and-media/press-releases/teva-nefazodone-statement/
- Levitan MN, Papelbaum M, Nardi AE. Profile of agomelatine and its potential in the treatment of generalized anxiety disorder. Neuropsychiatr Dis Treat. 2015;11:1149- 1155. doi:10.2147/NDT.S67470
- Fu DJ, Ionescu DF, Li X, et al. Esketamine nasal spray for rapid reduction of major depressive disorder symptoms in patients who have active suicidal ideation with intent: double-blind, randomized study (ASPIRE I). J Clin Psychiatry. 2020;81(3):19m13191. doi:10.4088/JCP.19m13191
- Iosifescu DV, Jones A, O’Gorman C, et al. Efficacy and safety of AXS-05 (dextromethorphan-bupropion) in patients with major depressive disorder: a phase 3 randomized clinical trial (GEMINI). J Clin Psychiatry. 2022;83(4): 21m14345. doi:10.4088/JCP.21m14345
- Luong TT, Powers CN, Reinhardt BJ, et al. Retrospective evaluation of drug-drug interactions with erlotinib and gefitinib use in the Military Health System. Fed Pract. 2023;40(Suppl 3):S24-S34. doi:10.12788/fp.0401
- Luong TT, Shou KJ, Reinhard BJ, Kigelman OF, Greenfield KM. Paclitaxel drug-drug interactions in the Military Health System. Fed Pract. 2024;41(Suppl 3):S70-S82. doi:10.12788/fp.0499
- Luong TT, Powers CN, Reinhardt BJ, Weina PJ. Preclinical drug-drug interactions (DDIs) of gefitinib with/without losartan and selective serotonin reuptake inhibitors (SSRIs): citalopram, fluoxetine, fluvoxamine, paroxetine, sertraline, and venlafaxine. Curr Res Pharmacol Drug Discov. 2022;3:100112. doi:10.1016/j.crphar.2022.100112
- Luong TT, McAnulty MJ, Evers DL, Reinhardt BJ, Weina PJ. Pre-clinical drug-drug interaction (DDI) of gefitinib or erlotinib with cytochrome P450 (CYP) inhibiting drugs, fluoxetine and/or losartan. Curr Res Toxicol. 2021;2:217- 224. doi:10.1016/j.crtox.2021.05.006,
- Adamo M, Dickie L, Ruhl J. SEER program coding and staging manual 2016. National Cancer Institute; 2016. Accessed April 4, 2025. https://seer.cancer.gov/archive/manuals/2016/SPCSM_2016_maindoc.pdf
- World Health Organization. International classification of diseases for oncology (ICD-O). 3rd ed, 1st revision. World Health Organization; 2013. Accessed April 4, 2025. https://apps.who.int/iris/handle/10665/96612
- Simon S. FDA approves Jelmyto (mitomycin gel) for urothelial cancer. American Cancer Society. April 20, 2020. Accessed April 4, 2025. https://www.cancer.org/cancer/latest-news/fda-approves-jelmyto-mitomycin-gel-for-urothelial-cancer.html
- Pantziarka P, Sukhatme V, Bouche G, Meheus L, Sukhatme VP. Repurposing drugs in oncology (ReDO)- diclofenac as an anti-cancer agent. Ecancermedicalscience. 2016;10:610. doi:10.3332/ecancer.2016.610
- Choi S, Kim S, Park J, Lee SE, Kim C, Kang D. Diclofenac: a nonsteroidal anti-inflammatory drug inducing cancer cell death by inhibiting microtubule polymerization and autophagy flux. Antioxidants (Basel). 2022;11(5):1009. doi:10.3390/antiox11051009
- Tontonoz M. The ABCs of BCG: oldest approved immunotherapy gets new explanation. Memorial Sloan Kettering Cancer Center. July 17, 2020. Accessed April 4, 2025. https://www.mskcc.org/news/oldest-approved-immunotherapy-gets-new-explanation
- Jordan VC. Tamoxifen as the first targeted long-term adjuvant therapy for breast cancer. Endocr Relat Cancer. 2014;21(3):R235-R246. doi:10.1530/ERC-14-0092
- Cole MP, Jones CT, Todd ID. A new anti-oestrogenic agent in late breast cancer. An early clinical appraisal of ICI46474. Br J Cancer. 1971;25(2):270-275. doi:10.1038/bjc.1971.33
- Jordan VC. Tamoxifen: a most unlikely pioneering medicine. Nat Rev Drug Discov. 2003;2(3):205-213. doi:10.1038/nrd1031
- Maximov PY, McDaniel RE, Jordan VC. Tamoxifen: Pioneering Medicine in Breast Cancer. Springer Basel; 2013. Accessed April 4, 2025. https://link.springer.com/book/10.1007/978-3-0348-0664-0
- Taxol (paclitaxel). Prescribing information. Bristol-Myers Squibb Company; 2011. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2011/020262s049lbl.pdf
- Abraxane (paclitaxel). Prescribing information. Celgene Corporation; 2020. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/021660s047lbl.pdf
- Tarceva (erlotinib). Prescribing information. OSI Pharmaceuticals, LLC; 2016. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2016/021743s025lbl.pdf
- Docetaxel. Prescribing information. Sichuan Hyiyu Pharmaceutical Co.; 2022. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/215813s000lbl.pdf
- Alimta (pemetrexed). Prescribing information. Teva Pharmaceuticals; 2022. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/208419s004lbl.pdf
- Tagrisso (osimertinib). Prescribing information. Astra- Zeneca Pharmaceuticals; 2020. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/208065s021lbl.pdf
- Iressa (gefitinib). Prescribing information. AstraZeneca Pharmaceuticals; 2018. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/206995s003lbl.pdf
- Kadcyla (ado-trastuzumab emtansine). Prescribing information. Genentech, Inc.; 2013. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2013/125427lbl.pdf
- Alecensa (alectinib). Prescribing information. Genetech, Inc.; 2017. Accessed April 4, 2025. https:// www.accessdata.fda.gov/drugsatfda_docs/label/2017/208434s003lbl.pdf
- Xalkori (crizotinib). Prescribing information. Pfizer Laboratories; 2022. Accessed April 4, 2025. https:// www.accessdata.fda.gov/drugsatfda_docs/label/2022/202570s033lbl.pdf
- Lorbrena (lorlatinib). Prescribing information. Pfizer Laboratories; 2018. Accessed April 14, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/210868s000lbl.pdf
- Alunbrig (brigatinib). Prescribing information. Takeda Pharmaceutical Company; 2020. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/208772s008lbl.pdf
- Rozlytrek (entrectinib). Prescribing information. Genentech, Inc.; 2019. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/212725s000lbl.pdf
- Herceptin (trastuzumab). Prescribing information. Genentech, Inc.; 2010. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2010/103792s5250lbl.pdf
- Cybalta (duloxetine). Prescribing information. Eli Lilly and Company; 2017. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/021427s049lbl.pdf
- Effexor XR (venlafaxine). Prescribing information. Pfizer Wyeth Pharmaceuticals Inc; 2022. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/020699s112lbl.pdf
- Desyrel (trazodone hydrochloride). Prescribing information. Pragma Pharmaceuticals; 2017. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/018207s032lbl.pdf
- Sertraline hydrochloride. Prescribing information. Almatica Pharma LLC; 2021. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2021/215133s000lbl.pdf
- Remeron (mirtazapine). Prescribing information. Merck & Co. Inc; 2020. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/020415s029,%20021208s019lbl.pdf
- Celexa (citalopram). Prescribing information. Allergan USA Inc; 2022. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/020822s041lbl.pdf
- information. GlaxoSmithKline; 2019. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/020358s066lbl.pdf
- Amitriptyline hydrochloride tablet. Prescribing information. Quality Care Products LLC; 2022. Accessed April 4, 2025. https://www.accessdata.fda.gov/spl/data/0f12f50f-7087-46e7-a2e6-356b4c566c9f/0f12f50f-7087-46e7-a2e6-356b4c566c9f.xml
- Lexapro (escitalopram). Prescribing information. AbbVie Inc; 2023. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2023/021323s055,021365s039lbl.pdf
- Fluoxetine. Prescribing information. Edgemont Pharmaceutical, LLC; 2017. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/202133s004s005lbl.pdf
- Paxil (paroxetine). Prescribing Information. Apotex Inc; 2021. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2021/020031s077lbl.pdf
- Pamelor (nortriptyline HCl). Prescribing information. Mallinckrodt, Inc; 2012. Accessed April 4, 2025. https:// www.accessdata.fda.gov/drugsatfda_docs/label/2012/018012s029,018013s061lbl.pdf
- Silenor (doxepin). Prescribing information. Currax Pharmaceuticals; 2020. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/022036s006lbl.pdf
- Tofranil-PM (imipramine pamote). Prescribing information. Mallinckrodt, Inc; 2014. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2014/017090s078lbl.pdf
- Norpramin (desipramine hydrochloride). Prescribing information. Sanofi-aventis U.S. LLC; 2014. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2014/014399s069lbl.pdf
- Khedezla (desvenlafaxine). Prescribing information. Osmotical Pharmaceutical US LLC; 2019. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/204683s006lbl.pdf
- Nefazodone hydrochloride. Prescribing information. Bryant Ranch Prepack; 2022. Accessed April 4, 2025. https://www.accessdata.fda.gov/spl/data/0bd4c34a-4f43-4c84-8b98-1d074cba97d5/0bd4c34a-4f43-4c84-8b98-1d074cba97d5.xml
- Grassi L, Nanni MG, Rodin G, Li M, Caruso R. The use of antidepressants in oncology: a review and practical tips for oncologists. Ann Oncol. 2018;29(1):101-111. doi:10.1093/annonc/mdx526
- Lee E, Park Y, Li D, Rodriguez-Fuguet A, Wang X, Zhang WC. Antidepressant use and lung cancer risk and survival: a meta-analysis of observational studies. Cancer Res Commun. 2023;3(6):1013-1025. doi:10.1158/2767-9764.CRC-23-0003
- Olfson M, Marcus SC. National patterns in antidepressant medication treatment. Arch Gen Psychiatry. 2009;66(8):848 -856. doi:10.1001/archgenpsychiatry.2009.81
- Grattan A, Sullivan MD, Saunders KW, Campbell CI, Von Korff MR. Depression and prescription opioid misuse among chronic opioid therapy recipients with no history of substance abuse. Ann Fam Med. 2012;10(4):304-311. doi:10.1370/afm.1371
- Cowan DT, Wilson-Barnett J, Griffiths P, Allan LG. A survey of chronic noncancer pain patients prescribed opioid analgesics. Pain Med. 2003;4(4):340-351. doi:10.1111/j.1526-4637.2003.03038.x
- Breckenridge J, Clark JD. Patient characteristics associated with opioid versus nonsteroidal anti-inflammatory drug management of chronic low back pain. J Pain. 2003;4(6):344-350. doi:10.1016/s1526-5900(03)00638-2
- Edlund MJ, Martin BC, Devries A, Fan MY, Braden JB, Sullivan MD. Trends in use of opioids for chronic noncancer pain among individuals with mental health and substance use disorders: the TROUP study. Clin J Pain. 2010;26(1):1-8. doi:10.1097/AJP.0b013e3181b99f35
- Sullivan MD, Edlund MJ, Fan MY, DeVries A, Braden JB, Martin BC. Risks for possible and probable opioid misuse among recipients of chronic opioid therapy in commercial and medicaid insurance plans: the TROUP study. Pain. 2010;150(2):332-339. doi:10.1016/j.pain.2010.05.020
- Dunn KM, Saunders KW, Rutter CM, et al. Opioid prescriptions for chronic pain and overdose: a cohort study. Ann Intern Med. 2010;152(2):85-92. doi:10.7326/0003-4819-152-2-201001190-00006
- Haddad P. Do antidepressants have any potential to cause addiction? J Psychopharmacol. 1999;13(3):300- 307. doi:10.1177/026988119901300321
- Lakeview Health Staff. America’s most abused antidepressants. Lakeview Health. January 24, 2004. Accessed April 4, 2025. https://www.lakeviewhealth.com/blog/us-most-abused-antidepressants/
- Greenhouse Treatment Center Editorial Staff. Addiction to antidepressants: is it possible? America Addiction Centers: Greenhouse Treatment Center. Updated April 23, 2024. Accessed April 4, 2025. https://greenhousetreatment.com/prescription-medication/antidepressants/
Cancer patients experience depression at rates > 5 times that of the general population.1-11 Despite an increase in palliative care use, depression rates continued to rise.2-4 Between 5% to 16% of outpatients, 4% to 14% of inpatients, and up to 49% of patients receiving palliative care experience depression.5 This issue also impacts families and caregivers.1 A 2021 meta-analysis found that 23% of active military personnel and 20% of veterans experience depression.11
Antidepressants approved by the US Food and Drug Administration (FDA) target the serotonin, norepinephrine, or dopamine systems and include boxed warnings about an increased risk of suicidal thoughts in adults aged 18 to 24 years.12,13 These medications are categorized into several classes: monoamine oxidase inhibitors (MAOIs), tricyclic antidepressants (TCAs), tetracyclic antidepressants (TeCAs), norepinephrine-dopamine reuptake inhibitors (NDRIs), selective serotonin reuptake inhibitors (SSRIs), serotonin-norepinephrine reuptake inhibitors (SNRIs), serotonin receptor modulators (SRMs), serotonin-melatonin receptor antagonists (SMRAs), and N—methyl-D-aspartate receptor antagonists (NMDARAs).14,15 The first FDA-approved antidepressants, iproniazid (an MAOI) and imipramine (a TCA) laid the foundation for the development of newer classes like SSRIs and SNRIs.15-17
Older antidepressants such as MAOIs and TCAs are used less due to their adverse effects (AEs) and drug interactions. MAOIs, such as iproniazid, selegiline, moclobemide, tranylcypromine, isocarboxazid, and phenelzine, have numerous AEs and drug interactions, making them unsuitable for first- or second-line treatment of depression.14,18-21 TCAs such as doxepin, amitriptyline, nortriptyline, imipramine, desipramine, clomipramine, trimipramine, protriptyline, maprotiline, and amoxapine have a narrow therapeutic index requiring careful monitoring for signs of toxicity such as QRS widening, tremors, or confusion. Despite the issues, TCAs are generally classified as second-line agents for major depressive disorder (MDD). TCAs have off-label uses for migraine prophylaxis, treatment of obsessive-compulsive disorder (OCD), insomnia, and chronic pain management first-line.14,22-29
Newer antidepressants, including TeCAs and NDRIs, are typically more effective, but also come with safety concerns. TeCAs like mirtazapine interact with several medications, including MAOIs, serotonin-increasing drugs, alcohol, cannabidiol, and marijuana. Mirtazapine is FDA-approved for the treatment of moderate to severe depression in adults. It is also used off-label to treat insomnia, panic disorder, posttraumatic stress disorder (PTSD), generalized anxiety disorder (GAD), social anxiety disorder (SAD), headaches, and migraines. Compared to other antidepressants, mirtazapine is effective for all stages of depression and addresses a broad range of related symptoms.14,30-34 NDRIs, such as bupropion, also interact with various medications, including MAOIs, other antidepressants, stimulants, and alcohol. Bupropion is FDA-approved for smoking cessation and to treat depression and SAD. It is also used off-label for depression- related bipolar disorder or sexual dysfunction, attention-deficit/hyperactivity disorder (ADHD), and obesity.14,35-42
SSRIs, SNRIs, and SRMs should be used with caution. SSRIs such as sertraline, citalopram, escitalopram, fluoxetine, paroxetine, and fluvoxamine are first-line treatments for depression and various psychiatric disorders due to their safety and efficacy. Common AEs of SSRIs include sexual dysfunction, sleep disturbances, weight changes, and gastrointestinal (GI) issues. SSRIs can prolong the QT interval, posing a risk of life-threatening arrhythmia, and may interact with other medications, necessitating treatment adjustments. The FDA approved SSRIs for MDD, GAD, bulimia nervosa, bipolar depression, OCD, panic disorder, premenstrual dysphoric disorder, treatment-resistant depression, PTSD, and SAD. Off-label uses include binge eating disorder, body dysmorphic disorder, fibromyalgia, premature ejaculation, paraphilias, autism, Raynaud phenomenon, and vasomotor symptoms associated with menopause. Among SSRIs, sertraline and escitalopram are noted for their effectiveness and tolerability.14,43-53
SNRIs, including duloxetine, venlafaxine, desvenlafaxine, milnacipran, and levomilnacipran, may increase bleeding risk, especially when taken with blood thinners. They can also elevate blood pressure, which may worsen if combined with stimulants. SNRIs may interact with other medications that affect serotonin levels, increasing the risk of serotonin syndrome when taken with triptans, pain medications, or other antidepressants.14 Desvenlafaxine has been approved by the FDA (but not by the European Medicines Agency).54-56 Duloxetine is FDA-approved for the treatment of depression, neuropathic pain, anxiety disorders, fibromyalgia, and musculoskeletal disorders. It is used off-label to treat chemotherapy-induced peripheral neuropathy and stress urinary incontinence.57-61 Venlafaxine is FDA-approved for depression, SAD, and panic disorder, and is prescribed off-label to treat ADHD, neuropathy, fibromyalgia, cataplexy, and PTSD, either alone or in combination with other medications.62,63 Milnacipran is not approved for MDD; levomilnacipran received approval in 2013.64
SRMs such as trazodone, nefazodone, vilazodone, and vortioxetine also function as serotonin reuptake inhibitors.14,15 Trazodone is FDA-approved for MDD. It has been used off-label to treat anxiety, Alzheimer disease, substance misuse, bulimia nervosa, insomnia, fibromyalgia, and PTSD when first-line SSRIs are ineffective. A notable AE of trazodone is orthostatic hypotension, which can lead to dizziness and increase the risk of falls, especially in geriatric patients.65-70 Nefazodone was discontinued in Europe in 2003 due to rare cases of liver toxicity but remains available in the US.71-74 Vilazodone and vortioxetine are FDA-approved.
The latest classes of antidepressants include SMRAs and NMDARAs.14 Agomelatine, an SMRA, was approved in Europe in 2009 but rejected by the FDA in 2011 due to liver toxicity.75 NMDARAs like esketamine and a combination of dextromethorphan and bupropion received FDA approval in 2019 and 2022, respectively.76,77
This retrospective study analyzes noncancer drugs used during systemic chemotherapy based on a dataset of 14 antineoplastic agents. It sought to identify the most dispensed noncancer drug groups, discuss findings, compare patients with and without antidepressant prescriptions, and examine trends in antidepressant use from 2002 to 2023. This analysis expands on prior research.78-81
Methods
The Walter Reed National Military Medical Center Institutional Review Board approved the study protocol and ensured compliance with the Health Insurance Portability and Accountability Act as an exempt protocol. The Joint Pathology Center (JPC) of the US Department of Defense (DoD) Cancer Registry Program and Military Health System (MHS) data experts from the Comprehensive Ambulatory/Professional Encounter Record (CAPER) and Pharmacy Data Transaction Service (PDTS) provided data for the analysis.
Data Sources
The JPC DoD Cancer Registry Program contains data from 1998 to 2024. CAPER and PDTS are part of the MHS Data Repository/Management Analysis and Reporting Tool database. Each observation in CAPER represents an ambulatory encounter at a military treatment facility (MTF). CAPER records are available from 2003 to 2024. PDTS records are available from 2002 to 2004. Each observation in PDTS represents a prescription filled for an MHS beneficiary, excluding those filled at international civilian pharmacies and inpatient pharmacy prescriptions.
This cross-sectional analysis requested data extraction for specific cancer drugs from the DoD Cancer Registry, focusing on treatment details, diagnosis dates, patient demographics, and physicians’ comments on AEs. After identifying patients, CAPER was used to identify additional health conditions. PDTS was used to compile a list of prescription medications filled during systemic cancer treatment or < 2 years postdiagnosis.
The 2016 Surveillance, Epidemiology, and End Results Program Coding and Staging Manual and International Classification of Diseases for Oncology, 3rd edition, 1st revision, were used to decode disease and cancer types.82,83 Data sorting and analysis were performed using Microsoft Excel. The percentage for the total was calculated by using the number of patients or data available within the subgroup divided by the total number of patients or data variables. To compare the mean number of dispensed antidepressants to those without antidepressants, a 2-tailed, 2-sample z test was used to calculate the P value and determine statistical significance (P < .05) using socscistatistics.com.
Data were extracted 3 times between 2021 and 2023. The initial 2021 protocol focused on erlotinib and gefitinib. A modified protocol in 2022 added paclitaxel, cisplatin, docetaxel, pemetrexed, and crizotinib; further modification in 2023 included 8 new antineoplastic agents and 2 anticoagulants. Sotorasib has not been prescribed in the MHS, and JPC lacks records for noncancer drugs. The 2023 dataset comprised 2210 patients with cancer treated with 14 antineoplastic agents; 2104 had documented diagnoses and 2113 had recorded prescriptions. Data for erlotinib, gefitinib, and paclitaxel have been published previously.78,79
Results
Of 2113 patients with recorded prescriptions, 1297 patients (61.4%) received 109 cancer drugs, including 96 antineoplastics, 7 disease-modifying antirheumatic agents, 4 biologic response modifiers, and 2 calcitonin gene-related peptides. Fourteen antineoplastic agents had complete data from JPC, while others were noted for combination therapies or treatment switches from the PDTS (Table 1). Seventy-six cancer drugs were prescribed with antidepressants in 489 patients (eAppendix).

The JPC provided 2242 entries for 2210 patients, ranging in age from 2 months to 88 years (mean, 56 years), documenting treatment from September 1988 to January 2023. Thirty-two patients had duplicate entries due to multiple cancer locations or occurrences. Of the 2242 patients, 1541 (68.7%) were aged > 50 years, 975 patients (43.5%) had cancers that were stage III or IV, and 1267 (56.5%) had cancers that were stage 0, I, II, or not applicable/unknown. There were 51 different types of cancer: breast, lung, testicular, endometrial, and ovarian were most common (n ≥ 100 patients). Forty-two cancer types were documented among 750 patients prescribed antidepressants (Table 2).

The CAPER database recorded 8882 unique diagnoses for 2104 patients, while PDTS noted 1089 unique prescriptions within 273 therapeutic codes for 2113 patients. Nine therapeutic codes (opiate agonists, adrenals, cathartics-laxatives, nonsteroidal anti-inflammatory agents, antihistamines for GI conditions, 5-HT3 receptor antagonists, analgesics and antipyretic miscellanea, antineoplastic agents, and proton-pump inhibitors) and 8 drugs (dexamethasone, prochlorperazine, ondansetron, docusate, acetaminophen, ibuprofen, oxycodone, and polyethylene glycol 3350) were associated with > 1000 patients (≥ 50%). Patients had between 1 and 275 unique health conditions and filled 1 to 108 prescriptions. The mean (SD) number of diagnoses and prescriptions was 50 (28) and 29 (12), respectively. Of the 273 therapeutic codes, 30 groups were analyzed, with others categorized into miscellaneous groups such as lotions, vaccines, and devices. Significant differences in mean number of prescriptions were found for patients taking antidepressants compared to those not (P < .05), except for anticonvulsants and antipsychotics (P = .12 and .09, respectively) (Table 3).

Antidepressants
Of the 2113 patients with recorded prescriptions, 750 (35.5%) were dispensed 17 different antidepressants. Among these 17 antidepressants, 183 (8.7%) patients received duloxetine, 158 (7.5%) received venlafaxine, 118 (5.6%) received trazodone, and 107 (5.1%) received sertraline (Figure 1, Table 4). Of the 750 patients, 509 (67.9%) received 1 antidepressant, 168 (22.4%) received 2, 60 (8.0%) received 3, and 13 (1.7%) received > 3. Combinations varied, but only duloxetine and trazodone were prescribed to > 10 patients.



Antidepressants were prescribed annually at an overall mean (SD) rate of 23% (5%) from 2003 to 2022 (Figure 2). Patients on antidepressants during systemic therapy had a greater number of diagnosed medical conditions and received more prescription medications compared to those not taking antidepressants (P < .001) (Figure 3). The 745 patients taking antidepressants in CAPER data had between 1 and 275 diagnosed medical issues, with a mean (SD) of 55 (31) vs a range of 1 to 209 and a mean (SD) of 46 (26) for the 1359 patients not taking antidepressants. The 750 patients on antidepressants in PDTS data had between 8 and 108 prescriptions dispensed, with a mean (SD) of 32 (12), vs a range of 1 to 65 prescriptions and a mean (SD) of 29 (12) for 1363 patients not taking antidepressants.


Discussion
The JPC DoD Cancer Registry includes information on cancer types, stages, treatment regimens, and physicians’ notes, while noncancer drugs are sourced from the PDTS database. The pharmacy uses a different documentation system, leading to varied classifications.
Database reliance has its drawbacks. For example, megestrol is coded as a cancer drug, although it’s primarily used for endometrial or gynecologic cancers. Many drugs have multiple therapeutic codes assigned to them, including 10 antineoplastic agents: diclofenac, Bacillus Calmette-Guérin (BCG), megestrol acetate, tamoxifen, anastrozole, letrozole, leuprolide, goserelin, degarelix, and fluorouracil. Diclofenac, BCG, and mitomycin have been repurposed for cancer treatment.84-87 From 2003 to 2023, diclofenac was prescribed to 350 patients for mild-to-moderate pain, with only 2 patients receiving it for cancer in 2018. FDA-approved for bladder cancer in 1990, BCG was prescribed for cancer treatment for 1 patient in 2021 after being used for vaccines between 2003 and 2018. Tamoxifen, used for hormone receptor-positive breast cancer from 2004 to 2017 with 53 patients, switched to estrogen agonist-antagonists from 2017 to 2023 with 123 patients. Only a few of the 168 patients were prescribed tamoxifen using both codes.88-91 Anastrozole and letrozole were coded as antiestrogens for 7 and 18 patients, respectively, while leuprolide and goserelin were coded as gonadotropins for 59 and 18 patients. Degarelix was coded as antigonadotropins, fluorouracil as skin and mucous membrane agents miscellaneous, and megestrol acetate as progestins for 7, 6, and 3 patients, respectively. Duloxetine was given to 186 patients, primarily for depression from 2005 to 2023, with 7 patients treated for fibromyalgia from 2022 to 2023.
Antidepressants Observed
Tables 1 and 5 provide insight into the FDA approval of 14 antineoplastics and antidepressants and their CYP metabolic pathways.92-122 In Table 4, the most prescribed antidepressant classes are SNRIs, SRMs, SSRIs, TeCAs, NDRIs, and TCAs. This trend highlights a preference for newer medications with weak CYP inhibition. A total of 349 patients were prescribed SSRIs, 343 SNRIs, 119 SRMs, 109 TCAs, 83 TeCAs, and 79 NDRIs. MAOIs, SMRAs, and NMDARAs were not observed in this dataset. While there are instances of dextromethorphan-bupropion and sertraline-escitalopram being dispensed together, it remains unclear whether these were NMDARA combinations.
Among the 14 specific antineoplastic agents, 10 are metabolized by CYP isoenzymes, primarily CYP3A4. Duloxetine neither inhibits nor is metabolized by CYP3A4, a reason it is often recommended, following venlafaxine.
Both duloxetine and venlafaxine are used off-label for chemotherapy-induced peripheral neuropathy related to paclitaxel and docetaxel. According to the CYP metabolized pathway, duloxetine tends to have more favorable DDIs than venlafaxine. In PDTS data, 371 patients were treated with paclitaxel and 180 with docetaxel, with respective antidepressant prescriptions of 156 and 70. Of the 156 patients dispensed paclitaxel, 62 (40%) were dispensed with duloxetine compared to 43 (28%) with venlafaxine. Of the 70 patients dispensed docetaxel, 23 (33%) received duloxetine vs 24 (34%) with venlafaxine.
Of 85 patients prescribed duloxetine, 75 received it with either paclitaxel or docetaxel (5 received both). Five patients had documented AEs (1 neuropathy related). Of 67 patients prescribed venlafaxine, 66 received it with either paclitaxel or docetaxel. Two patients had documented AEs (1 was neuropathy related, the same patient who received duloxetine). Of the 687 patients treated with paclitaxel and 337 with docetaxel in all databases, 4 experienced neuropathic AEs from both medications.79
Antidepressants can increase the risk of bleeding, especially when combined with blood thinners, and may elevate blood pressure, particularly alongside stimulants. Of the 554 patients prescribed 9 different anticoagulants, enoxaparin, apixaban, and rivaroxaban were the most common (each > 100 patients). Among these, 201 patients (36%) received both anticoagulants and antidepressants: duloxetine for 64 patients, venlafaxine for 30, trazodone for 35, and sertraline for 26. There were no data available to assess bleeding rates related to the evaluation of DDIs between these medication classes.
Antidepressants can be prescribed for erectile dysfunction. Of the 148 patients prescribed an antidepressant for erectile dysfunction, duloxetine, trazodone, and mirtazapine were the most common. Antidepressant preferences varied by cancer type. Duloxetine was the only antidepressant used for all types of cancer. Venlafaxine, duloxetine, trazodone, sertraline, and escitalopram were the most prescribed antidepressants for breast cancer, while duloxetine, mirtazapine, citalopram, sertraline, and trazodone were the most prescribed for lung cancer. Sertraline, duloxetine, trazodone, amitriptyline, and escitalopram were most common for testicular cancer. Duloxetine, venlafaxine, trazodone, amitriptyline, and sertraline were the most prescribed for endometrial cancer, while duloxetine, venlafaxine, amitriptyline, citalopram, and sertraline were most prescribed for ovarian cancer.
The broadness of International Statistical Classification of Diseases, Tenth Revision codes made it challenging to identify nondepression diagnoses in the analyzed population. However, if all antidepressants were prescribed to treat depression, service members with cancer exhibited a higher depression rate (35%) than the general population (25%). Of 2104 patients, 191 (9.1%) had mood disorders, and 706 (33.6%) had mental disorders: 346 (49.0%) had 1 diagnosis, and 360 (51.0%) had multiple diagnoses. The percentage of diagnoses varied yearly, with notable drops in 2003, 2007, 2011, 2014, and 2018, and peaks in 2006, 2008, 2013, 2017, and 2022. This fluctuation was influenced by events like the establishment of PDTS in 2002, the 2008 economic recession, a hospital relocation in 2011, the 2014 Ebola outbreak, and the COVID-19 pandemic. Although the number of patients receiving antidepressants increased from 2019 to 2022, the overall percentage of patients receiving them did not significantly change from 2003 to 2022, aligning with previous research.5,125
Many medications have potential uses beyond what is detailed in the prescribing information. Antidepressants can relieve pain, while pain medications may help with depression. Opioids were once thought to effectively treat depression, but this perspective has changed with a greater understanding of their risks, including misuse.126-131 Pain is a severe and often unbearable AE of cancer. Of 2113 patients, 92% received opioids; 34% received both opioids and antidepressants; 2% received only antidepressants; and 7% received neither. This study didn’t clarify whether those on opioids alone recognized their depression or if those on both were aware of their dependence. While SSRIs are generally not addictive, they can lead to physical dependence, and any medication can be abused if not managed properly.132-134
Conclusions
This retrospective study analyzes data from antineoplastic agents used in systemic cancer treatment between 1988 and 2023, with a particular focus on the use of antidepressants. Data on antidepressant prescriptions are incomplete and specific to these agents, which means the findings cannot be generalized to all antidepressants. Hence, the results indicate that patients taking antidepressants had more diagnosed health issues and received more medications compared to patients who were not on these drugs.
This study underscores the need for further research into the effects of antidepressants on cancer treatment, utilizing all data from the DoD Cancer Registry. Future research should explore DDIs between antidepressants and other cancer and noncancer medications, as this study did not assess AE documentation, unlike in studies involving erlotinib, gefitinib, and paclitaxel.78,79 Further investigation is needed to evaluate the impact of discontinuing antidepressant use during cancer treatment. This comprehensive overview provides insights for clinicians to help them make informed decisions regarding the prescription of antidepressants in the context of cancer treatment.
Cancer patients experience depression at rates > 5 times that of the general population.1-11 Despite an increase in palliative care use, depression rates continued to rise.2-4 Between 5% to 16% of outpatients, 4% to 14% of inpatients, and up to 49% of patients receiving palliative care experience depression.5 This issue also impacts families and caregivers.1 A 2021 meta-analysis found that 23% of active military personnel and 20% of veterans experience depression.11
Antidepressants approved by the US Food and Drug Administration (FDA) target the serotonin, norepinephrine, or dopamine systems and include boxed warnings about an increased risk of suicidal thoughts in adults aged 18 to 24 years.12,13 These medications are categorized into several classes: monoamine oxidase inhibitors (MAOIs), tricyclic antidepressants (TCAs), tetracyclic antidepressants (TeCAs), norepinephrine-dopamine reuptake inhibitors (NDRIs), selective serotonin reuptake inhibitors (SSRIs), serotonin-norepinephrine reuptake inhibitors (SNRIs), serotonin receptor modulators (SRMs), serotonin-melatonin receptor antagonists (SMRAs), and N—methyl-D-aspartate receptor antagonists (NMDARAs).14,15 The first FDA-approved antidepressants, iproniazid (an MAOI) and imipramine (a TCA) laid the foundation for the development of newer classes like SSRIs and SNRIs.15-17
Older antidepressants such as MAOIs and TCAs are used less due to their adverse effects (AEs) and drug interactions. MAOIs, such as iproniazid, selegiline, moclobemide, tranylcypromine, isocarboxazid, and phenelzine, have numerous AEs and drug interactions, making them unsuitable for first- or second-line treatment of depression.14,18-21 TCAs such as doxepin, amitriptyline, nortriptyline, imipramine, desipramine, clomipramine, trimipramine, protriptyline, maprotiline, and amoxapine have a narrow therapeutic index requiring careful monitoring for signs of toxicity such as QRS widening, tremors, or confusion. Despite the issues, TCAs are generally classified as second-line agents for major depressive disorder (MDD). TCAs have off-label uses for migraine prophylaxis, treatment of obsessive-compulsive disorder (OCD), insomnia, and chronic pain management first-line.14,22-29
Newer antidepressants, including TeCAs and NDRIs, are typically more effective, but also come with safety concerns. TeCAs like mirtazapine interact with several medications, including MAOIs, serotonin-increasing drugs, alcohol, cannabidiol, and marijuana. Mirtazapine is FDA-approved for the treatment of moderate to severe depression in adults. It is also used off-label to treat insomnia, panic disorder, posttraumatic stress disorder (PTSD), generalized anxiety disorder (GAD), social anxiety disorder (SAD), headaches, and migraines. Compared to other antidepressants, mirtazapine is effective for all stages of depression and addresses a broad range of related symptoms.14,30-34 NDRIs, such as bupropion, also interact with various medications, including MAOIs, other antidepressants, stimulants, and alcohol. Bupropion is FDA-approved for smoking cessation and to treat depression and SAD. It is also used off-label for depression- related bipolar disorder or sexual dysfunction, attention-deficit/hyperactivity disorder (ADHD), and obesity.14,35-42
SSRIs, SNRIs, and SRMs should be used with caution. SSRIs such as sertraline, citalopram, escitalopram, fluoxetine, paroxetine, and fluvoxamine are first-line treatments for depression and various psychiatric disorders due to their safety and efficacy. Common AEs of SSRIs include sexual dysfunction, sleep disturbances, weight changes, and gastrointestinal (GI) issues. SSRIs can prolong the QT interval, posing a risk of life-threatening arrhythmia, and may interact with other medications, necessitating treatment adjustments. The FDA approved SSRIs for MDD, GAD, bulimia nervosa, bipolar depression, OCD, panic disorder, premenstrual dysphoric disorder, treatment-resistant depression, PTSD, and SAD. Off-label uses include binge eating disorder, body dysmorphic disorder, fibromyalgia, premature ejaculation, paraphilias, autism, Raynaud phenomenon, and vasomotor symptoms associated with menopause. Among SSRIs, sertraline and escitalopram are noted for their effectiveness and tolerability.14,43-53
SNRIs, including duloxetine, venlafaxine, desvenlafaxine, milnacipran, and levomilnacipran, may increase bleeding risk, especially when taken with blood thinners. They can also elevate blood pressure, which may worsen if combined with stimulants. SNRIs may interact with other medications that affect serotonin levels, increasing the risk of serotonin syndrome when taken with triptans, pain medications, or other antidepressants.14 Desvenlafaxine has been approved by the FDA (but not by the European Medicines Agency).54-56 Duloxetine is FDA-approved for the treatment of depression, neuropathic pain, anxiety disorders, fibromyalgia, and musculoskeletal disorders. It is used off-label to treat chemotherapy-induced peripheral neuropathy and stress urinary incontinence.57-61 Venlafaxine is FDA-approved for depression, SAD, and panic disorder, and is prescribed off-label to treat ADHD, neuropathy, fibromyalgia, cataplexy, and PTSD, either alone or in combination with other medications.62,63 Milnacipran is not approved for MDD; levomilnacipran received approval in 2013.64
SRMs such as trazodone, nefazodone, vilazodone, and vortioxetine also function as serotonin reuptake inhibitors.14,15 Trazodone is FDA-approved for MDD. It has been used off-label to treat anxiety, Alzheimer disease, substance misuse, bulimia nervosa, insomnia, fibromyalgia, and PTSD when first-line SSRIs are ineffective. A notable AE of trazodone is orthostatic hypotension, which can lead to dizziness and increase the risk of falls, especially in geriatric patients.65-70 Nefazodone was discontinued in Europe in 2003 due to rare cases of liver toxicity but remains available in the US.71-74 Vilazodone and vortioxetine are FDA-approved.
The latest classes of antidepressants include SMRAs and NMDARAs.14 Agomelatine, an SMRA, was approved in Europe in 2009 but rejected by the FDA in 2011 due to liver toxicity.75 NMDARAs like esketamine and a combination of dextromethorphan and bupropion received FDA approval in 2019 and 2022, respectively.76,77
This retrospective study analyzes noncancer drugs used during systemic chemotherapy based on a dataset of 14 antineoplastic agents. It sought to identify the most dispensed noncancer drug groups, discuss findings, compare patients with and without antidepressant prescriptions, and examine trends in antidepressant use from 2002 to 2023. This analysis expands on prior research.78-81
Methods
The Walter Reed National Military Medical Center Institutional Review Board approved the study protocol and ensured compliance with the Health Insurance Portability and Accountability Act as an exempt protocol. The Joint Pathology Center (JPC) of the US Department of Defense (DoD) Cancer Registry Program and Military Health System (MHS) data experts from the Comprehensive Ambulatory/Professional Encounter Record (CAPER) and Pharmacy Data Transaction Service (PDTS) provided data for the analysis.
Data Sources
The JPC DoD Cancer Registry Program contains data from 1998 to 2024. CAPER and PDTS are part of the MHS Data Repository/Management Analysis and Reporting Tool database. Each observation in CAPER represents an ambulatory encounter at a military treatment facility (MTF). CAPER records are available from 2003 to 2024. PDTS records are available from 2002 to 2004. Each observation in PDTS represents a prescription filled for an MHS beneficiary, excluding those filled at international civilian pharmacies and inpatient pharmacy prescriptions.
This cross-sectional analysis requested data extraction for specific cancer drugs from the DoD Cancer Registry, focusing on treatment details, diagnosis dates, patient demographics, and physicians’ comments on AEs. After identifying patients, CAPER was used to identify additional health conditions. PDTS was used to compile a list of prescription medications filled during systemic cancer treatment or < 2 years postdiagnosis.
The 2016 Surveillance, Epidemiology, and End Results Program Coding and Staging Manual and International Classification of Diseases for Oncology, 3rd edition, 1st revision, were used to decode disease and cancer types.82,83 Data sorting and analysis were performed using Microsoft Excel. The percentage for the total was calculated by using the number of patients or data available within the subgroup divided by the total number of patients or data variables. To compare the mean number of dispensed antidepressants to those without antidepressants, a 2-tailed, 2-sample z test was used to calculate the P value and determine statistical significance (P < .05) using socscistatistics.com.
Data were extracted 3 times between 2021 and 2023. The initial 2021 protocol focused on erlotinib and gefitinib. A modified protocol in 2022 added paclitaxel, cisplatin, docetaxel, pemetrexed, and crizotinib; further modification in 2023 included 8 new antineoplastic agents and 2 anticoagulants. Sotorasib has not been prescribed in the MHS, and JPC lacks records for noncancer drugs. The 2023 dataset comprised 2210 patients with cancer treated with 14 antineoplastic agents; 2104 had documented diagnoses and 2113 had recorded prescriptions. Data for erlotinib, gefitinib, and paclitaxel have been published previously.78,79
Results
Of 2113 patients with recorded prescriptions, 1297 patients (61.4%) received 109 cancer drugs, including 96 antineoplastics, 7 disease-modifying antirheumatic agents, 4 biologic response modifiers, and 2 calcitonin gene-related peptides. Fourteen antineoplastic agents had complete data from JPC, while others were noted for combination therapies or treatment switches from the PDTS (Table 1). Seventy-six cancer drugs were prescribed with antidepressants in 489 patients (eAppendix).

The JPC provided 2242 entries for 2210 patients, ranging in age from 2 months to 88 years (mean, 56 years), documenting treatment from September 1988 to January 2023. Thirty-two patients had duplicate entries due to multiple cancer locations or occurrences. Of the 2242 patients, 1541 (68.7%) were aged > 50 years, 975 patients (43.5%) had cancers that were stage III or IV, and 1267 (56.5%) had cancers that were stage 0, I, II, or not applicable/unknown. There were 51 different types of cancer: breast, lung, testicular, endometrial, and ovarian were most common (n ≥ 100 patients). Forty-two cancer types were documented among 750 patients prescribed antidepressants (Table 2).

The CAPER database recorded 8882 unique diagnoses for 2104 patients, while PDTS noted 1089 unique prescriptions within 273 therapeutic codes for 2113 patients. Nine therapeutic codes (opiate agonists, adrenals, cathartics-laxatives, nonsteroidal anti-inflammatory agents, antihistamines for GI conditions, 5-HT3 receptor antagonists, analgesics and antipyretic miscellanea, antineoplastic agents, and proton-pump inhibitors) and 8 drugs (dexamethasone, prochlorperazine, ondansetron, docusate, acetaminophen, ibuprofen, oxycodone, and polyethylene glycol 3350) were associated with > 1000 patients (≥ 50%). Patients had between 1 and 275 unique health conditions and filled 1 to 108 prescriptions. The mean (SD) number of diagnoses and prescriptions was 50 (28) and 29 (12), respectively. Of the 273 therapeutic codes, 30 groups were analyzed, with others categorized into miscellaneous groups such as lotions, vaccines, and devices. Significant differences in mean number of prescriptions were found for patients taking antidepressants compared to those not (P < .05), except for anticonvulsants and antipsychotics (P = .12 and .09, respectively) (Table 3).

Antidepressants
Of the 2113 patients with recorded prescriptions, 750 (35.5%) were dispensed 17 different antidepressants. Among these 17 antidepressants, 183 (8.7%) patients received duloxetine, 158 (7.5%) received venlafaxine, 118 (5.6%) received trazodone, and 107 (5.1%) received sertraline (Figure 1, Table 4). Of the 750 patients, 509 (67.9%) received 1 antidepressant, 168 (22.4%) received 2, 60 (8.0%) received 3, and 13 (1.7%) received > 3. Combinations varied, but only duloxetine and trazodone were prescribed to > 10 patients.



Antidepressants were prescribed annually at an overall mean (SD) rate of 23% (5%) from 2003 to 2022 (Figure 2). Patients on antidepressants during systemic therapy had a greater number of diagnosed medical conditions and received more prescription medications compared to those not taking antidepressants (P < .001) (Figure 3). The 745 patients taking antidepressants in CAPER data had between 1 and 275 diagnosed medical issues, with a mean (SD) of 55 (31) vs a range of 1 to 209 and a mean (SD) of 46 (26) for the 1359 patients not taking antidepressants. The 750 patients on antidepressants in PDTS data had between 8 and 108 prescriptions dispensed, with a mean (SD) of 32 (12), vs a range of 1 to 65 prescriptions and a mean (SD) of 29 (12) for 1363 patients not taking antidepressants.


Discussion
The JPC DoD Cancer Registry includes information on cancer types, stages, treatment regimens, and physicians’ notes, while noncancer drugs are sourced from the PDTS database. The pharmacy uses a different documentation system, leading to varied classifications.
Database reliance has its drawbacks. For example, megestrol is coded as a cancer drug, although it’s primarily used for endometrial or gynecologic cancers. Many drugs have multiple therapeutic codes assigned to them, including 10 antineoplastic agents: diclofenac, Bacillus Calmette-Guérin (BCG), megestrol acetate, tamoxifen, anastrozole, letrozole, leuprolide, goserelin, degarelix, and fluorouracil. Diclofenac, BCG, and mitomycin have been repurposed for cancer treatment.84-87 From 2003 to 2023, diclofenac was prescribed to 350 patients for mild-to-moderate pain, with only 2 patients receiving it for cancer in 2018. FDA-approved for bladder cancer in 1990, BCG was prescribed for cancer treatment for 1 patient in 2021 after being used for vaccines between 2003 and 2018. Tamoxifen, used for hormone receptor-positive breast cancer from 2004 to 2017 with 53 patients, switched to estrogen agonist-antagonists from 2017 to 2023 with 123 patients. Only a few of the 168 patients were prescribed tamoxifen using both codes.88-91 Anastrozole and letrozole were coded as antiestrogens for 7 and 18 patients, respectively, while leuprolide and goserelin were coded as gonadotropins for 59 and 18 patients. Degarelix was coded as antigonadotropins, fluorouracil as skin and mucous membrane agents miscellaneous, and megestrol acetate as progestins for 7, 6, and 3 patients, respectively. Duloxetine was given to 186 patients, primarily for depression from 2005 to 2023, with 7 patients treated for fibromyalgia from 2022 to 2023.
Antidepressants Observed
Tables 1 and 5 provide insight into the FDA approval of 14 antineoplastics and antidepressants and their CYP metabolic pathways.92-122 In Table 4, the most prescribed antidepressant classes are SNRIs, SRMs, SSRIs, TeCAs, NDRIs, and TCAs. This trend highlights a preference for newer medications with weak CYP inhibition. A total of 349 patients were prescribed SSRIs, 343 SNRIs, 119 SRMs, 109 TCAs, 83 TeCAs, and 79 NDRIs. MAOIs, SMRAs, and NMDARAs were not observed in this dataset. While there are instances of dextromethorphan-bupropion and sertraline-escitalopram being dispensed together, it remains unclear whether these were NMDARA combinations.
Among the 14 specific antineoplastic agents, 10 are metabolized by CYP isoenzymes, primarily CYP3A4. Duloxetine neither inhibits nor is metabolized by CYP3A4, a reason it is often recommended, following venlafaxine.
Both duloxetine and venlafaxine are used off-label for chemotherapy-induced peripheral neuropathy related to paclitaxel and docetaxel. According to the CYP metabolized pathway, duloxetine tends to have more favorable DDIs than venlafaxine. In PDTS data, 371 patients were treated with paclitaxel and 180 with docetaxel, with respective antidepressant prescriptions of 156 and 70. Of the 156 patients dispensed paclitaxel, 62 (40%) were dispensed with duloxetine compared to 43 (28%) with venlafaxine. Of the 70 patients dispensed docetaxel, 23 (33%) received duloxetine vs 24 (34%) with venlafaxine.
Of 85 patients prescribed duloxetine, 75 received it with either paclitaxel or docetaxel (5 received both). Five patients had documented AEs (1 neuropathy related). Of 67 patients prescribed venlafaxine, 66 received it with either paclitaxel or docetaxel. Two patients had documented AEs (1 was neuropathy related, the same patient who received duloxetine). Of the 687 patients treated with paclitaxel and 337 with docetaxel in all databases, 4 experienced neuropathic AEs from both medications.79
Antidepressants can increase the risk of bleeding, especially when combined with blood thinners, and may elevate blood pressure, particularly alongside stimulants. Of the 554 patients prescribed 9 different anticoagulants, enoxaparin, apixaban, and rivaroxaban were the most common (each > 100 patients). Among these, 201 patients (36%) received both anticoagulants and antidepressants: duloxetine for 64 patients, venlafaxine for 30, trazodone for 35, and sertraline for 26. There were no data available to assess bleeding rates related to the evaluation of DDIs between these medication classes.
Antidepressants can be prescribed for erectile dysfunction. Of the 148 patients prescribed an antidepressant for erectile dysfunction, duloxetine, trazodone, and mirtazapine were the most common. Antidepressant preferences varied by cancer type. Duloxetine was the only antidepressant used for all types of cancer. Venlafaxine, duloxetine, trazodone, sertraline, and escitalopram were the most prescribed antidepressants for breast cancer, while duloxetine, mirtazapine, citalopram, sertraline, and trazodone were the most prescribed for lung cancer. Sertraline, duloxetine, trazodone, amitriptyline, and escitalopram were most common for testicular cancer. Duloxetine, venlafaxine, trazodone, amitriptyline, and sertraline were the most prescribed for endometrial cancer, while duloxetine, venlafaxine, amitriptyline, citalopram, and sertraline were most prescribed for ovarian cancer.
The broadness of International Statistical Classification of Diseases, Tenth Revision codes made it challenging to identify nondepression diagnoses in the analyzed population. However, if all antidepressants were prescribed to treat depression, service members with cancer exhibited a higher depression rate (35%) than the general population (25%). Of 2104 patients, 191 (9.1%) had mood disorders, and 706 (33.6%) had mental disorders: 346 (49.0%) had 1 diagnosis, and 360 (51.0%) had multiple diagnoses. The percentage of diagnoses varied yearly, with notable drops in 2003, 2007, 2011, 2014, and 2018, and peaks in 2006, 2008, 2013, 2017, and 2022. This fluctuation was influenced by events like the establishment of PDTS in 2002, the 2008 economic recession, a hospital relocation in 2011, the 2014 Ebola outbreak, and the COVID-19 pandemic. Although the number of patients receiving antidepressants increased from 2019 to 2022, the overall percentage of patients receiving them did not significantly change from 2003 to 2022, aligning with previous research.5,125
Many medications have potential uses beyond what is detailed in the prescribing information. Antidepressants can relieve pain, while pain medications may help with depression. Opioids were once thought to effectively treat depression, but this perspective has changed with a greater understanding of their risks, including misuse.126-131 Pain is a severe and often unbearable AE of cancer. Of 2113 patients, 92% received opioids; 34% received both opioids and antidepressants; 2% received only antidepressants; and 7% received neither. This study didn’t clarify whether those on opioids alone recognized their depression or if those on both were aware of their dependence. While SSRIs are generally not addictive, they can lead to physical dependence, and any medication can be abused if not managed properly.132-134
Conclusions
This retrospective study analyzes data from antineoplastic agents used in systemic cancer treatment between 1988 and 2023, with a particular focus on the use of antidepressants. Data on antidepressant prescriptions are incomplete and specific to these agents, which means the findings cannot be generalized to all antidepressants. Hence, the results indicate that patients taking antidepressants had more diagnosed health issues and received more medications compared to patients who were not on these drugs.
This study underscores the need for further research into the effects of antidepressants on cancer treatment, utilizing all data from the DoD Cancer Registry. Future research should explore DDIs between antidepressants and other cancer and noncancer medications, as this study did not assess AE documentation, unlike in studies involving erlotinib, gefitinib, and paclitaxel.78,79 Further investigation is needed to evaluate the impact of discontinuing antidepressant use during cancer treatment. This comprehensive overview provides insights for clinicians to help them make informed decisions regarding the prescription of antidepressants in the context of cancer treatment.
- National Cancer Institute. Depression (PDQ)-Health Professional Version. Updated July 25, 2024. Accessed April 4, 2025. https://www.cancer.gov/about-cancer/coping/feelings/depression-hp-pdq
- Krebber AM, Buffart LM, Kleijn G, et al. Prevalence of depression in cancer patients: a meta-analysis of diagnostic interviews and self-report instruments. Psychooncology. 2014;23(2):121-130. doi:10.1002/pon.3409
- Xiang X, An R, Gehlert S. Trends in antidepressant use among U.S. cancer survivors, 1999-2012. Psychiatr Serv. 2015;66(6):564. doi:10.1176/appi.ps.201500007
- Hartung TJ, Brähler E, Faller H, et al. The risk of being depressed is significantly higher in cancer patients than in the general population: Prevalence and severity of depressive symptoms across major cancer types. Eur J Cancer. 2017;72:46-53. doi:10.1016/j.ejca.2016.11.017
- Walker J, Holm Hansen C, Martin P, et al. Prevalence of depression in adults with cancer: a systematic review. Ann Oncol. 2013;24(4):895-900. doi:10.1093/annonc/mds575
- Pitman A, Suleman S, Hyde N, Hodgkiss A. Depression and anxiety in patients with cancer. BMJ. 2018;361:k1415. doi:10.1136/bmj.k1415
- Kisely S, Alotiby MKN, Protani MM, Soole R, Arnautovska U, Siskind D. Breast cancer treatment disparities in patients with severe mental illness: a systematic review and meta-analysis. Psychooncology. 2023;32(5):651-662. doi:10.1002/pon.6120
- Massie MJ. Prevalence of depression in patients with cancer. J Natl Cancer Inst Monogr. 2004;(32):57-71. doi:10.1093/jncimonographs/lgh014
- Rodin G, Katz M, Lloyd N, Green E, Mackay JA, Wong RK. Treatment of depression in cancer patients. Curr Oncol. 2007;14(5):180-188. doi:10.3747/co.2007.146
- Wilson KG, Chochinov HM, Skirko MG, et al. Depression and anxiety disorders in palliative cancer care. J Pain Symptom Manage. 2007;33(2):118-129. doi:10.1016/j.jpainsymman.2006.07.016
- Moradi Y, Dowran B, Sepandi M. The global prevalence of depression, suicide ideation, and attempts in the military forces: a systematic review and meta-analysis of cross sectional studies. BMC Psychiatry. 2021;21(1):510. doi:10.1186/s12888-021-03526-2
- Lu CY, Zhang F, Lakoma MD, et al. Changes in antidepressant use by young people and suicidal behavior after FDA warnings and media coverage: quasi-experimental study. BMJ. 2014;348:g3596. doi:10.1136/bmj.g3596
- Friedman RA. Antidepressants’ black-box warning--10 years later. N Engl J Med. 2014;371(18):1666-1668. doi:10.1056/NEJMp1408480
- Sheffler ZM, Patel P, Abdijadid S. Antidepressants. In: StatPearls. StatPearls Publishing. Updated May 26, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK538182/
- Miller JJ. Antidepressants, Part 1: 100 Years and Counting. Accessed April 4, 2025. Psychiatric Times. https:// www.psychiatrictimes.com/view/antidepressants-part-1-100-years-and-counting
- Hillhouse TM, Porter JH. A brief history of the development of antidepressant drugs: from monoamines to glutamate. Exp Clin Psychopharmacol. 2015;23(1):1-21. doi:10.1037/a0038550
- Mitchell PB, Mitchell MS. The management of depression. Part 2. The place of the new antidepressants. Aust Fam Physician. 1994;23(9):1771-1781.
- Sabri MA, Saber-Ayad MM. MAO Inhibitors. In: Stat- Pearls. StatPearls Publishing. Updated June 5, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK557395/
- Sub Laban T, Saadabadi A. Monoamine Oxidase Inhibitors (MAOI). In: StatPearls. StatPearls Publishing. Updated July 17, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK539848/
- Fiedorowicz JG, Swartz KL. The role of monoamine oxidase inhibitors in current psychiatric practice. J Psychiatr Pract. 2004;10(4):239-248. doi:10.1097/00131746-200407000-00005
- Flockhart DA. Dietary restrictions and drug interactions with monoamine oxidase inhibitors: an update. J Clin Psychiatry. 2012;73 Suppl 1:17-24. doi:10.4088/JCP.11096su1c.03
- Moraczewski J, Awosika AO, Aedma KK. Tricyclic Antidepressants. In: StatPearls. StatPearls Publishing. Updated August 17, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK557791/
- Almasi A, Patel P, Meza CE. Doxepin. In: StatPearls. StatPearls Publishing. Updated February 14, 2024. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK542306/
- Thour A, Marwaha R. Amitriptyline. In: StatPearls. Stat- Pearls Publishing. Updated July 18, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK537225/
- Radley DC, Finkelstein SN, Stafford RS. Off-label prescribing among office-based physicians. Arch Intern Med. 2006;166(9):1021-1026. doi:10.1001/archinte.166.9.1021
- Tesfaye S, Sloan G, Petrie J, et al. Comparison of amitriptyline supplemented with pregabalin, pregabalin supplemented with amitriptyline, and duloxetine supplemented with pregabalin for the treatment of diabetic peripheral neuropathic pain (OPTION-DM): a multicentre, double-blind, randomised crossover trial. Lancet. 2022;400(10353):680- 690. doi:10.1016/S0140-6736(22)01472-6
- Farag HM, Yunusa I, Goswami H, Sultan I, Doucette JA, Eguale T. Comparison of amitriptyline and US Food and Drug Administration-approved treatments for fibromyalgia: a systematic review and network metaanalysis. JAMA Netw Open. 2022;5(5):e2212939. doi:10.1001/jamanetworkopen.2022.12939
- Merwar G, Gibbons JR, Hosseini SA, Saadabadi A. Nortriptyline. In: StatPearls. StatPearls Publishing. Updated June 5, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK482214/
- Fayez R, Gupta V. Imipramine. In: StatPearls. StatPearls Publishing. Updated May 22, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK557656/
- Jilani TN, Gibbons JR, Faizy RM, Saadabadi A. Mirtazapine. In: StatPearls. StatPearls Publishing. Updated August 28, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK519059/
- Nutt DJ. Tolerability and safety aspects of mirtazapine. Hum Psychopharmacol. 2002;17 Suppl 1:S37-S41. doi:10.1002/hup.388
- Gandotra K, Chen P, Jaskiw GE, Konicki PE, Strohl KP. Effective treatment of insomnia with mirtazapine attenuates concomitant suicidal ideation. J Clin Sleep Med. 2018;14(5):901-902. doi:10.5664/jcsm.7142
- Anttila SA, Leinonen EV. A review of the pharmacological and clinical profile of mirtazapine. CNS Drug Rev. 2001;7(3):249-264. doi:10.1111/j.1527-3458.2001.tb00198.x
- Wang SM, Han C, Bahk WM, et al. Addressing the side effects of contemporary antidepressant drugs: a comprehensive review. Chonnam Med J. 2018;54(2):101-112. doi:10.4068/cmj.2018.54.2.101
- Huecker MR, Smiley A, Saadabadi A. Bupropion. In: StatPearls. StatPearls Publishing. Updated April 9, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK470212/
- Hsieh MT, Tseng PT, Wu YC, et al. Effects of different pharmacologic smoking cessation treatments on body weight changes and success rates in patients with nicotine dependence: a network meta-analysis. Obes Rev. 2019;20(6):895- 905. doi:10.1111/obr.12835
- Hankosky ER, Bush HM, Dwoskin LP, et al. Retrospective analysis of health claims to evaluate pharmacotherapies with potential for repurposing: association of bupropion and stimulant use disorder remission. AMIA Annu Symp Proc. 2018;2018:1292-1299.
- Livingstone-Banks J, Norris E, Hartmann-Boyce J, West R, Jarvis M, Hajek P. Relapse prevention interventions for smoking cessation. Cochrane Database Syst Rev. 2019; 2(2):CD003999. doi:10.1002/14651858.CD003999.pub5
- Clayton AH, Kingsberg SA, Goldstein I. Evaluation and management of hypoactive sexual desire disorder. Sex Med. 2018;6(2):59-74. doi:10.1016/j.esxm.2018.01.004
- Verbeeck W, Bekkering GE, Van den Noortgate W, Kramers C. Bupropion for attention deficit hyperactivity disorder (ADHD) in adults. Cochrane Database Syst Rev. 2017;10(10):CD009504. doi:10.1002/14651858.CD009504.pub2
- Ng QX. A systematic review of the use of bupropion for attention-deficit/hyperactivity disorder in children and adolescents. J Child Adolesc Psychopharmacol. 2017;27(2):112-116. doi:10.1089/cap.2016.0124
- Fava M, Rush AJ, Thase ME, et al. 15 years of clinical experience with bupropion HCl: from bupropion to bupropion SR to bupropion XL. Prim Care Companion J Clin Psychiatry. 2005;7(3):106-113. doi:10.4088/pcc.v07n0305
- Chu A, Wadhwa R. Selective Serotonin Reuptake Inhibitors. In: StatPearls. StatPearls Publishing. Updated May 1, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK554406/
- Singh HK, Saadabadi A. Sertraline. In: StatPearls. Stat- Pearls Publishing. Updated February 13, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK547689/
- MacQueen G, Born L, Steiner M. The selective serotonin reuptake inhibitor sertraline: its profile and use in psychiatric disorders. CNS Drug Rev. 2001;7(1):1-24. doi:10.1111/j.1527-3458.2001.tb00188.x
- Cipriani A, Furukawa TA, Salanti G, et al. Comparative efficacy and acceptability of 12 new-generation antidepressants: a multiple-treatments meta-analysis. Lancet. 2009;373(9665):746-758. doi:10.1016/S0140-6736(09)60046-5
- Nelson JC. The STAR*D study: a four-course meal that leaves us wanting more. Am J Psychiatry. 2006;163(11):1864-1866. doi:10.1176/ajp.2006.163.11.1864
- Sharbaf Shoar N, Fariba KA, Padhy RK. Citalopram. In: StatPearls. StatPearls Publishing. Updated November 7, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK482222/
- Landy K, Rosani A, Estevez R. Escitalopram. In: Stat- Pearls. StatPearls Publishing. Updated November 10, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK557734/
- Cavanah LR, Ray P, Goldhirsh JL, Huey LY, Piper BJ. Rise of escitalopram and the fall of citalopram. medRxiv. Preprint published online May 8, 2023. doi:10.1101/2023.05.07.23289632
- Sohel AJ, Shutter MC, Patel P, Molla M. Fluoxetine. In: StatPearls. StatPearls Publishing. Updated July 4, 2022. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK459223/
- Wong DT, Perry KW, Bymaster FP. Case history: the discovery of fluoxetine hydrochloride (Prozac). Nat Rev Drug Discov. 2005;4(9):764-774. doi:10.1038/nrd1821
- Shrestha P, Fariba KA, Abdijadid S. Paroxetine. In: Stat- Pearls. StatPearls Publishing. Updated July 17, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK526022/
- Naseeruddin R, Rosani A, Marwaha R. Desvenlafaxine. In: StatPearls. StatPearls Publishing. Updated July 10, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK534829
- Lieberman DZ, Massey SH. Desvenlafaxine in major depressive disorder: an evidence-based review of its place in therapy. Core Evid. 2010;4:67-82. doi:10.2147/ce.s5998
- Withdrawal assessment report for Ellefore (desvenlafaxine). European Medicine Agency. 2009. Accessed April 4, 2025. https://www.ema.europa.eu/en/documents/withdrawal-report/withdrawal-assessment-report-ellefore_en.pdf
- Dhaliwal JS, Spurling BC, Molla M. Duloxetine. In: Stat- Pearls. Statpearls Publishing. Updated May 29, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK549806/
- Hershman DL, Lacchetti C, Dworkin RH, et al. Prevention and management of chemotherapy-induced peripheral neuropathy in survivors of adult cancers: American Society of Clinical Oncology clinical practice guideline. J Clin Oncol. 2014;32(18):1941-1967. doi:10.1200/JCO.2013.54.0914
- Sommer C, Häuser W, Alten R, et al. Medikamentöse Therapie des Fibromyalgiesyndroms. Systematische Übersicht und Metaanalyse [Drug therapy of fibromyalgia syndrome. Systematic review, meta-analysis and guideline]. Schmerz. 2012;26(3):297-310. doi:10.1007/s00482-012-1172-2
- Bril V, England J, Franklin GM, et al. Evidence-based guideline: treatment of painful diabetic neuropathy: report of the American Academy of Neurology, the American Association of Neuromuscular and Electrodiagnostic Medicine, and the American Academy of Physical Medicine and Rehabilitation. Neurology. 2011;76(20):1758-1765. doi:10.1212/WNL.0b013e3182166ebe
- Attal N, Cruccu G, Baron R, et al. EFNS guidelines on the pharmacological treatment of neuropathic pain: 2010 revision. Eur J Neurol. 2010;17(9):1113-e88. doi:10.1111/j.1468-1331.2010.02999.x
- Singh D, Saadabadi A. Venlafaxine. In: StatPearls. StatePearls Publishing. Updated February 26, 2024. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK535363/
- Sertraline and venlafaxine: new indication. Prevention of recurrent depression: no advance. Prescrire Int. 2005;14(75):19-20.
- Bruno A, Morabito P, S p i n a E , M u s c a t ello MRl. The role of levomilnacipran in the management of major depressive disorder: a comprehensive review. Curr Neuro pharmacol. 2016;14(2):191-199. doi:10.2174/1570159x14666151117122458
- Shin JJ, Saadabadi A. Trazodone. In: StatPearls. StatPearls Publishing. Updated February 29, 2024. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK470560/
- Khouzam HR. A review of trazodone use in psychiatric and medical conditions. Postgrad Med. 2017;129(1):140-148. doi:10.1080/00325481.2017.1249265
- Smales ET, Edwards BA, Deyoung PN, et al. Trazodone effects on obstructive sleep apnea and non-REM arousal threshold. Ann Am Thorac Soc. 2015;12(5):758-764. doi:10.1513/AnnalsATS.201408-399OC
- Eckert DJ, Malhotra A, Wellman A, White DP. Trazodone increases the respiratory arousal threshold in patients with obstructive sleep apnea and a low arousal threshold. Sleep. 2014;37(4):811-819. doi:10.5665/sleep.3596
- Schatzberg AF, Nemeroff CB, eds. The American Psychiat ric Association Publishing Textbook of Psychopharmacology. 4th ed. American Psychiatric Publishing; 2009.
- Ruxton K, Woodman RJ, Mangoni AA. Drugs with anticholinergic effects and cognitive impairment, falls and allcause mortality in older adults: a systematic review and meta-analysis. Br J Clin Pharmacol. 2015;80(2):209-220. doi:10.1111/bcp.12617
- Nefazodone. In: LiverTox: Clinical and Research Information on Drug-Induced Liver Injury. National Institute of Diabetes and Digestive and Kidney Diseases. Updated March 6, 2020. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK548179/
- Drugs of Current Interest: Nefazodone. WHO Pharmaceuticals Newsletter:(1). 2003(1):7. https://web.archive.org/web/20150403165029/http:/apps.who.int/medicinedocs/en/d/Js4944e/3.html
- Choi S. Nefazodone (serzone) withdrawn because of hepatotoxicity. CMAJ. 2003;169(11):1187.
- Teva Nefazodone Statement. News release. Teva USA. December 20, 2021. Accessed April 4, 2025. https:// www.tevausa.com/news-and-media/press-releases/teva-nefazodone-statement/
- Levitan MN, Papelbaum M, Nardi AE. Profile of agomelatine and its potential in the treatment of generalized anxiety disorder. Neuropsychiatr Dis Treat. 2015;11:1149- 1155. doi:10.2147/NDT.S67470
- Fu DJ, Ionescu DF, Li X, et al. Esketamine nasal spray for rapid reduction of major depressive disorder symptoms in patients who have active suicidal ideation with intent: double-blind, randomized study (ASPIRE I). J Clin Psychiatry. 2020;81(3):19m13191. doi:10.4088/JCP.19m13191
- Iosifescu DV, Jones A, O’Gorman C, et al. Efficacy and safety of AXS-05 (dextromethorphan-bupropion) in patients with major depressive disorder: a phase 3 randomized clinical trial (GEMINI). J Clin Psychiatry. 2022;83(4): 21m14345. doi:10.4088/JCP.21m14345
- Luong TT, Powers CN, Reinhardt BJ, et al. Retrospective evaluation of drug-drug interactions with erlotinib and gefitinib use in the Military Health System. Fed Pract. 2023;40(Suppl 3):S24-S34. doi:10.12788/fp.0401
- Luong TT, Shou KJ, Reinhard BJ, Kigelman OF, Greenfield KM. Paclitaxel drug-drug interactions in the Military Health System. Fed Pract. 2024;41(Suppl 3):S70-S82. doi:10.12788/fp.0499
- Luong TT, Powers CN, Reinhardt BJ, Weina PJ. Preclinical drug-drug interactions (DDIs) of gefitinib with/without losartan and selective serotonin reuptake inhibitors (SSRIs): citalopram, fluoxetine, fluvoxamine, paroxetine, sertraline, and venlafaxine. Curr Res Pharmacol Drug Discov. 2022;3:100112. doi:10.1016/j.crphar.2022.100112
- Luong TT, McAnulty MJ, Evers DL, Reinhardt BJ, Weina PJ. Pre-clinical drug-drug interaction (DDI) of gefitinib or erlotinib with cytochrome P450 (CYP) inhibiting drugs, fluoxetine and/or losartan. Curr Res Toxicol. 2021;2:217- 224. doi:10.1016/j.crtox.2021.05.006,
- Adamo M, Dickie L, Ruhl J. SEER program coding and staging manual 2016. National Cancer Institute; 2016. Accessed April 4, 2025. https://seer.cancer.gov/archive/manuals/2016/SPCSM_2016_maindoc.pdf
- World Health Organization. International classification of diseases for oncology (ICD-O). 3rd ed, 1st revision. World Health Organization; 2013. Accessed April 4, 2025. https://apps.who.int/iris/handle/10665/96612
- Simon S. FDA approves Jelmyto (mitomycin gel) for urothelial cancer. American Cancer Society. April 20, 2020. Accessed April 4, 2025. https://www.cancer.org/cancer/latest-news/fda-approves-jelmyto-mitomycin-gel-for-urothelial-cancer.html
- Pantziarka P, Sukhatme V, Bouche G, Meheus L, Sukhatme VP. Repurposing drugs in oncology (ReDO)- diclofenac as an anti-cancer agent. Ecancermedicalscience. 2016;10:610. doi:10.3332/ecancer.2016.610
- Choi S, Kim S, Park J, Lee SE, Kim C, Kang D. Diclofenac: a nonsteroidal anti-inflammatory drug inducing cancer cell death by inhibiting microtubule polymerization and autophagy flux. Antioxidants (Basel). 2022;11(5):1009. doi:10.3390/antiox11051009
- Tontonoz M. The ABCs of BCG: oldest approved immunotherapy gets new explanation. Memorial Sloan Kettering Cancer Center. July 17, 2020. Accessed April 4, 2025. https://www.mskcc.org/news/oldest-approved-immunotherapy-gets-new-explanation
- Jordan VC. Tamoxifen as the first targeted long-term adjuvant therapy for breast cancer. Endocr Relat Cancer. 2014;21(3):R235-R246. doi:10.1530/ERC-14-0092
- Cole MP, Jones CT, Todd ID. A new anti-oestrogenic agent in late breast cancer. An early clinical appraisal of ICI46474. Br J Cancer. 1971;25(2):270-275. doi:10.1038/bjc.1971.33
- Jordan VC. Tamoxifen: a most unlikely pioneering medicine. Nat Rev Drug Discov. 2003;2(3):205-213. doi:10.1038/nrd1031
- Maximov PY, McDaniel RE, Jordan VC. Tamoxifen: Pioneering Medicine in Breast Cancer. Springer Basel; 2013. Accessed April 4, 2025. https://link.springer.com/book/10.1007/978-3-0348-0664-0
- Taxol (paclitaxel). Prescribing information. Bristol-Myers Squibb Company; 2011. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2011/020262s049lbl.pdf
- Abraxane (paclitaxel). Prescribing information. Celgene Corporation; 2020. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/021660s047lbl.pdf
- Tarceva (erlotinib). Prescribing information. OSI Pharmaceuticals, LLC; 2016. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2016/021743s025lbl.pdf
- Docetaxel. Prescribing information. Sichuan Hyiyu Pharmaceutical Co.; 2022. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/215813s000lbl.pdf
- Alimta (pemetrexed). Prescribing information. Teva Pharmaceuticals; 2022. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/208419s004lbl.pdf
- Tagrisso (osimertinib). Prescribing information. Astra- Zeneca Pharmaceuticals; 2020. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/208065s021lbl.pdf
- Iressa (gefitinib). Prescribing information. AstraZeneca Pharmaceuticals; 2018. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/206995s003lbl.pdf
- Kadcyla (ado-trastuzumab emtansine). Prescribing information. Genentech, Inc.; 2013. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2013/125427lbl.pdf
- Alecensa (alectinib). Prescribing information. Genetech, Inc.; 2017. Accessed April 4, 2025. https:// www.accessdata.fda.gov/drugsatfda_docs/label/2017/208434s003lbl.pdf
- Xalkori (crizotinib). Prescribing information. Pfizer Laboratories; 2022. Accessed April 4, 2025. https:// www.accessdata.fda.gov/drugsatfda_docs/label/2022/202570s033lbl.pdf
- Lorbrena (lorlatinib). Prescribing information. Pfizer Laboratories; 2018. Accessed April 14, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/210868s000lbl.pdf
- Alunbrig (brigatinib). Prescribing information. Takeda Pharmaceutical Company; 2020. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/208772s008lbl.pdf
- Rozlytrek (entrectinib). Prescribing information. Genentech, Inc.; 2019. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/212725s000lbl.pdf
- Herceptin (trastuzumab). Prescribing information. Genentech, Inc.; 2010. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2010/103792s5250lbl.pdf
- Cybalta (duloxetine). Prescribing information. Eli Lilly and Company; 2017. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/021427s049lbl.pdf
- Effexor XR (venlafaxine). Prescribing information. Pfizer Wyeth Pharmaceuticals Inc; 2022. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/020699s112lbl.pdf
- Desyrel (trazodone hydrochloride). Prescribing information. Pragma Pharmaceuticals; 2017. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/018207s032lbl.pdf
- Sertraline hydrochloride. Prescribing information. Almatica Pharma LLC; 2021. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2021/215133s000lbl.pdf
- Remeron (mirtazapine). Prescribing information. Merck & Co. Inc; 2020. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/020415s029,%20021208s019lbl.pdf
- Celexa (citalopram). Prescribing information. Allergan USA Inc; 2022. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/020822s041lbl.pdf
- information. GlaxoSmithKline; 2019. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/020358s066lbl.pdf
- Amitriptyline hydrochloride tablet. Prescribing information. Quality Care Products LLC; 2022. Accessed April 4, 2025. https://www.accessdata.fda.gov/spl/data/0f12f50f-7087-46e7-a2e6-356b4c566c9f/0f12f50f-7087-46e7-a2e6-356b4c566c9f.xml
- Lexapro (escitalopram). Prescribing information. AbbVie Inc; 2023. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2023/021323s055,021365s039lbl.pdf
- Fluoxetine. Prescribing information. Edgemont Pharmaceutical, LLC; 2017. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/202133s004s005lbl.pdf
- Paxil (paroxetine). Prescribing Information. Apotex Inc; 2021. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2021/020031s077lbl.pdf
- Pamelor (nortriptyline HCl). Prescribing information. Mallinckrodt, Inc; 2012. Accessed April 4, 2025. https:// www.accessdata.fda.gov/drugsatfda_docs/label/2012/018012s029,018013s061lbl.pdf
- Silenor (doxepin). Prescribing information. Currax Pharmaceuticals; 2020. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/022036s006lbl.pdf
- Tofranil-PM (imipramine pamote). Prescribing information. Mallinckrodt, Inc; 2014. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2014/017090s078lbl.pdf
- Norpramin (desipramine hydrochloride). Prescribing information. Sanofi-aventis U.S. LLC; 2014. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2014/014399s069lbl.pdf
- Khedezla (desvenlafaxine). Prescribing information. Osmotical Pharmaceutical US LLC; 2019. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/204683s006lbl.pdf
- Nefazodone hydrochloride. Prescribing information. Bryant Ranch Prepack; 2022. Accessed April 4, 2025. https://www.accessdata.fda.gov/spl/data/0bd4c34a-4f43-4c84-8b98-1d074cba97d5/0bd4c34a-4f43-4c84-8b98-1d074cba97d5.xml
- Grassi L, Nanni MG, Rodin G, Li M, Caruso R. The use of antidepressants in oncology: a review and practical tips for oncologists. Ann Oncol. 2018;29(1):101-111. doi:10.1093/annonc/mdx526
- Lee E, Park Y, Li D, Rodriguez-Fuguet A, Wang X, Zhang WC. Antidepressant use and lung cancer risk and survival: a meta-analysis of observational studies. Cancer Res Commun. 2023;3(6):1013-1025. doi:10.1158/2767-9764.CRC-23-0003
- Olfson M, Marcus SC. National patterns in antidepressant medication treatment. Arch Gen Psychiatry. 2009;66(8):848 -856. doi:10.1001/archgenpsychiatry.2009.81
- Grattan A, Sullivan MD, Saunders KW, Campbell CI, Von Korff MR. Depression and prescription opioid misuse among chronic opioid therapy recipients with no history of substance abuse. Ann Fam Med. 2012;10(4):304-311. doi:10.1370/afm.1371
- Cowan DT, Wilson-Barnett J, Griffiths P, Allan LG. A survey of chronic noncancer pain patients prescribed opioid analgesics. Pain Med. 2003;4(4):340-351. doi:10.1111/j.1526-4637.2003.03038.x
- Breckenridge J, Clark JD. Patient characteristics associated with opioid versus nonsteroidal anti-inflammatory drug management of chronic low back pain. J Pain. 2003;4(6):344-350. doi:10.1016/s1526-5900(03)00638-2
- Edlund MJ, Martin BC, Devries A, Fan MY, Braden JB, Sullivan MD. Trends in use of opioids for chronic noncancer pain among individuals with mental health and substance use disorders: the TROUP study. Clin J Pain. 2010;26(1):1-8. doi:10.1097/AJP.0b013e3181b99f35
- Sullivan MD, Edlund MJ, Fan MY, DeVries A, Braden JB, Martin BC. Risks for possible and probable opioid misuse among recipients of chronic opioid therapy in commercial and medicaid insurance plans: the TROUP study. Pain. 2010;150(2):332-339. doi:10.1016/j.pain.2010.05.020
- Dunn KM, Saunders KW, Rutter CM, et al. Opioid prescriptions for chronic pain and overdose: a cohort study. Ann Intern Med. 2010;152(2):85-92. doi:10.7326/0003-4819-152-2-201001190-00006
- Haddad P. Do antidepressants have any potential to cause addiction? J Psychopharmacol. 1999;13(3):300- 307. doi:10.1177/026988119901300321
- Lakeview Health Staff. America’s most abused antidepressants. Lakeview Health. January 24, 2004. Accessed April 4, 2025. https://www.lakeviewhealth.com/blog/us-most-abused-antidepressants/
- Greenhouse Treatment Center Editorial Staff. Addiction to antidepressants: is it possible? America Addiction Centers: Greenhouse Treatment Center. Updated April 23, 2024. Accessed April 4, 2025. https://greenhousetreatment.com/prescription-medication/antidepressants/
- National Cancer Institute. Depression (PDQ)-Health Professional Version. Updated July 25, 2024. Accessed April 4, 2025. https://www.cancer.gov/about-cancer/coping/feelings/depression-hp-pdq
- Krebber AM, Buffart LM, Kleijn G, et al. Prevalence of depression in cancer patients: a meta-analysis of diagnostic interviews and self-report instruments. Psychooncology. 2014;23(2):121-130. doi:10.1002/pon.3409
- Xiang X, An R, Gehlert S. Trends in antidepressant use among U.S. cancer survivors, 1999-2012. Psychiatr Serv. 2015;66(6):564. doi:10.1176/appi.ps.201500007
- Hartung TJ, Brähler E, Faller H, et al. The risk of being depressed is significantly higher in cancer patients than in the general population: Prevalence and severity of depressive symptoms across major cancer types. Eur J Cancer. 2017;72:46-53. doi:10.1016/j.ejca.2016.11.017
- Walker J, Holm Hansen C, Martin P, et al. Prevalence of depression in adults with cancer: a systematic review. Ann Oncol. 2013;24(4):895-900. doi:10.1093/annonc/mds575
- Pitman A, Suleman S, Hyde N, Hodgkiss A. Depression and anxiety in patients with cancer. BMJ. 2018;361:k1415. doi:10.1136/bmj.k1415
- Kisely S, Alotiby MKN, Protani MM, Soole R, Arnautovska U, Siskind D. Breast cancer treatment disparities in patients with severe mental illness: a systematic review and meta-analysis. Psychooncology. 2023;32(5):651-662. doi:10.1002/pon.6120
- Massie MJ. Prevalence of depression in patients with cancer. J Natl Cancer Inst Monogr. 2004;(32):57-71. doi:10.1093/jncimonographs/lgh014
- Rodin G, Katz M, Lloyd N, Green E, Mackay JA, Wong RK. Treatment of depression in cancer patients. Curr Oncol. 2007;14(5):180-188. doi:10.3747/co.2007.146
- Wilson KG, Chochinov HM, Skirko MG, et al. Depression and anxiety disorders in palliative cancer care. J Pain Symptom Manage. 2007;33(2):118-129. doi:10.1016/j.jpainsymman.2006.07.016
- Moradi Y, Dowran B, Sepandi M. The global prevalence of depression, suicide ideation, and attempts in the military forces: a systematic review and meta-analysis of cross sectional studies. BMC Psychiatry. 2021;21(1):510. doi:10.1186/s12888-021-03526-2
- Lu CY, Zhang F, Lakoma MD, et al. Changes in antidepressant use by young people and suicidal behavior after FDA warnings and media coverage: quasi-experimental study. BMJ. 2014;348:g3596. doi:10.1136/bmj.g3596
- Friedman RA. Antidepressants’ black-box warning--10 years later. N Engl J Med. 2014;371(18):1666-1668. doi:10.1056/NEJMp1408480
- Sheffler ZM, Patel P, Abdijadid S. Antidepressants. In: StatPearls. StatPearls Publishing. Updated May 26, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK538182/
- Miller JJ. Antidepressants, Part 1: 100 Years and Counting. Accessed April 4, 2025. Psychiatric Times. https:// www.psychiatrictimes.com/view/antidepressants-part-1-100-years-and-counting
- Hillhouse TM, Porter JH. A brief history of the development of antidepressant drugs: from monoamines to glutamate. Exp Clin Psychopharmacol. 2015;23(1):1-21. doi:10.1037/a0038550
- Mitchell PB, Mitchell MS. The management of depression. Part 2. The place of the new antidepressants. Aust Fam Physician. 1994;23(9):1771-1781.
- Sabri MA, Saber-Ayad MM. MAO Inhibitors. In: Stat- Pearls. StatPearls Publishing. Updated June 5, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK557395/
- Sub Laban T, Saadabadi A. Monoamine Oxidase Inhibitors (MAOI). In: StatPearls. StatPearls Publishing. Updated July 17, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK539848/
- Fiedorowicz JG, Swartz KL. The role of monoamine oxidase inhibitors in current psychiatric practice. J Psychiatr Pract. 2004;10(4):239-248. doi:10.1097/00131746-200407000-00005
- Flockhart DA. Dietary restrictions and drug interactions with monoamine oxidase inhibitors: an update. J Clin Psychiatry. 2012;73 Suppl 1:17-24. doi:10.4088/JCP.11096su1c.03
- Moraczewski J, Awosika AO, Aedma KK. Tricyclic Antidepressants. In: StatPearls. StatPearls Publishing. Updated August 17, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK557791/
- Almasi A, Patel P, Meza CE. Doxepin. In: StatPearls. StatPearls Publishing. Updated February 14, 2024. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK542306/
- Thour A, Marwaha R. Amitriptyline. In: StatPearls. Stat- Pearls Publishing. Updated July 18, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK537225/
- Radley DC, Finkelstein SN, Stafford RS. Off-label prescribing among office-based physicians. Arch Intern Med. 2006;166(9):1021-1026. doi:10.1001/archinte.166.9.1021
- Tesfaye S, Sloan G, Petrie J, et al. Comparison of amitriptyline supplemented with pregabalin, pregabalin supplemented with amitriptyline, and duloxetine supplemented with pregabalin for the treatment of diabetic peripheral neuropathic pain (OPTION-DM): a multicentre, double-blind, randomised crossover trial. Lancet. 2022;400(10353):680- 690. doi:10.1016/S0140-6736(22)01472-6
- Farag HM, Yunusa I, Goswami H, Sultan I, Doucette JA, Eguale T. Comparison of amitriptyline and US Food and Drug Administration-approved treatments for fibromyalgia: a systematic review and network metaanalysis. JAMA Netw Open. 2022;5(5):e2212939. doi:10.1001/jamanetworkopen.2022.12939
- Merwar G, Gibbons JR, Hosseini SA, Saadabadi A. Nortriptyline. In: StatPearls. StatPearls Publishing. Updated June 5, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK482214/
- Fayez R, Gupta V. Imipramine. In: StatPearls. StatPearls Publishing. Updated May 22, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK557656/
- Jilani TN, Gibbons JR, Faizy RM, Saadabadi A. Mirtazapine. In: StatPearls. StatPearls Publishing. Updated August 28, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK519059/
- Nutt DJ. Tolerability and safety aspects of mirtazapine. Hum Psychopharmacol. 2002;17 Suppl 1:S37-S41. doi:10.1002/hup.388
- Gandotra K, Chen P, Jaskiw GE, Konicki PE, Strohl KP. Effective treatment of insomnia with mirtazapine attenuates concomitant suicidal ideation. J Clin Sleep Med. 2018;14(5):901-902. doi:10.5664/jcsm.7142
- Anttila SA, Leinonen EV. A review of the pharmacological and clinical profile of mirtazapine. CNS Drug Rev. 2001;7(3):249-264. doi:10.1111/j.1527-3458.2001.tb00198.x
- Wang SM, Han C, Bahk WM, et al. Addressing the side effects of contemporary antidepressant drugs: a comprehensive review. Chonnam Med J. 2018;54(2):101-112. doi:10.4068/cmj.2018.54.2.101
- Huecker MR, Smiley A, Saadabadi A. Bupropion. In: StatPearls. StatPearls Publishing. Updated April 9, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK470212/
- Hsieh MT, Tseng PT, Wu YC, et al. Effects of different pharmacologic smoking cessation treatments on body weight changes and success rates in patients with nicotine dependence: a network meta-analysis. Obes Rev. 2019;20(6):895- 905. doi:10.1111/obr.12835
- Hankosky ER, Bush HM, Dwoskin LP, et al. Retrospective analysis of health claims to evaluate pharmacotherapies with potential for repurposing: association of bupropion and stimulant use disorder remission. AMIA Annu Symp Proc. 2018;2018:1292-1299.
- Livingstone-Banks J, Norris E, Hartmann-Boyce J, West R, Jarvis M, Hajek P. Relapse prevention interventions for smoking cessation. Cochrane Database Syst Rev. 2019; 2(2):CD003999. doi:10.1002/14651858.CD003999.pub5
- Clayton AH, Kingsberg SA, Goldstein I. Evaluation and management of hypoactive sexual desire disorder. Sex Med. 2018;6(2):59-74. doi:10.1016/j.esxm.2018.01.004
- Verbeeck W, Bekkering GE, Van den Noortgate W, Kramers C. Bupropion for attention deficit hyperactivity disorder (ADHD) in adults. Cochrane Database Syst Rev. 2017;10(10):CD009504. doi:10.1002/14651858.CD009504.pub2
- Ng QX. A systematic review of the use of bupropion for attention-deficit/hyperactivity disorder in children and adolescents. J Child Adolesc Psychopharmacol. 2017;27(2):112-116. doi:10.1089/cap.2016.0124
- Fava M, Rush AJ, Thase ME, et al. 15 years of clinical experience with bupropion HCl: from bupropion to bupropion SR to bupropion XL. Prim Care Companion J Clin Psychiatry. 2005;7(3):106-113. doi:10.4088/pcc.v07n0305
- Chu A, Wadhwa R. Selective Serotonin Reuptake Inhibitors. In: StatPearls. StatPearls Publishing. Updated May 1, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK554406/
- Singh HK, Saadabadi A. Sertraline. In: StatPearls. Stat- Pearls Publishing. Updated February 13, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK547689/
- MacQueen G, Born L, Steiner M. The selective serotonin reuptake inhibitor sertraline: its profile and use in psychiatric disorders. CNS Drug Rev. 2001;7(1):1-24. doi:10.1111/j.1527-3458.2001.tb00188.x
- Cipriani A, Furukawa TA, Salanti G, et al. Comparative efficacy and acceptability of 12 new-generation antidepressants: a multiple-treatments meta-analysis. Lancet. 2009;373(9665):746-758. doi:10.1016/S0140-6736(09)60046-5
- Nelson JC. The STAR*D study: a four-course meal that leaves us wanting more. Am J Psychiatry. 2006;163(11):1864-1866. doi:10.1176/ajp.2006.163.11.1864
- Sharbaf Shoar N, Fariba KA, Padhy RK. Citalopram. In: StatPearls. StatPearls Publishing. Updated November 7, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK482222/
- Landy K, Rosani A, Estevez R. Escitalopram. In: Stat- Pearls. StatPearls Publishing. Updated November 10, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK557734/
- Cavanah LR, Ray P, Goldhirsh JL, Huey LY, Piper BJ. Rise of escitalopram and the fall of citalopram. medRxiv. Preprint published online May 8, 2023. doi:10.1101/2023.05.07.23289632
- Sohel AJ, Shutter MC, Patel P, Molla M. Fluoxetine. In: StatPearls. StatPearls Publishing. Updated July 4, 2022. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK459223/
- Wong DT, Perry KW, Bymaster FP. Case history: the discovery of fluoxetine hydrochloride (Prozac). Nat Rev Drug Discov. 2005;4(9):764-774. doi:10.1038/nrd1821
- Shrestha P, Fariba KA, Abdijadid S. Paroxetine. In: Stat- Pearls. StatPearls Publishing. Updated July 17, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK526022/
- Naseeruddin R, Rosani A, Marwaha R. Desvenlafaxine. In: StatPearls. StatPearls Publishing. Updated July 10, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK534829
- Lieberman DZ, Massey SH. Desvenlafaxine in major depressive disorder: an evidence-based review of its place in therapy. Core Evid. 2010;4:67-82. doi:10.2147/ce.s5998
- Withdrawal assessment report for Ellefore (desvenlafaxine). European Medicine Agency. 2009. Accessed April 4, 2025. https://www.ema.europa.eu/en/documents/withdrawal-report/withdrawal-assessment-report-ellefore_en.pdf
- Dhaliwal JS, Spurling BC, Molla M. Duloxetine. In: Stat- Pearls. Statpearls Publishing. Updated May 29, 2023. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK549806/
- Hershman DL, Lacchetti C, Dworkin RH, et al. Prevention and management of chemotherapy-induced peripheral neuropathy in survivors of adult cancers: American Society of Clinical Oncology clinical practice guideline. J Clin Oncol. 2014;32(18):1941-1967. doi:10.1200/JCO.2013.54.0914
- Sommer C, Häuser W, Alten R, et al. Medikamentöse Therapie des Fibromyalgiesyndroms. Systematische Übersicht und Metaanalyse [Drug therapy of fibromyalgia syndrome. Systematic review, meta-analysis and guideline]. Schmerz. 2012;26(3):297-310. doi:10.1007/s00482-012-1172-2
- Bril V, England J, Franklin GM, et al. Evidence-based guideline: treatment of painful diabetic neuropathy: report of the American Academy of Neurology, the American Association of Neuromuscular and Electrodiagnostic Medicine, and the American Academy of Physical Medicine and Rehabilitation. Neurology. 2011;76(20):1758-1765. doi:10.1212/WNL.0b013e3182166ebe
- Attal N, Cruccu G, Baron R, et al. EFNS guidelines on the pharmacological treatment of neuropathic pain: 2010 revision. Eur J Neurol. 2010;17(9):1113-e88. doi:10.1111/j.1468-1331.2010.02999.x
- Singh D, Saadabadi A. Venlafaxine. In: StatPearls. StatePearls Publishing. Updated February 26, 2024. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK535363/
- Sertraline and venlafaxine: new indication. Prevention of recurrent depression: no advance. Prescrire Int. 2005;14(75):19-20.
- Bruno A, Morabito P, S p i n a E , M u s c a t ello MRl. The role of levomilnacipran in the management of major depressive disorder: a comprehensive review. Curr Neuro pharmacol. 2016;14(2):191-199. doi:10.2174/1570159x14666151117122458
- Shin JJ, Saadabadi A. Trazodone. In: StatPearls. StatPearls Publishing. Updated February 29, 2024. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK470560/
- Khouzam HR. A review of trazodone use in psychiatric and medical conditions. Postgrad Med. 2017;129(1):140-148. doi:10.1080/00325481.2017.1249265
- Smales ET, Edwards BA, Deyoung PN, et al. Trazodone effects on obstructive sleep apnea and non-REM arousal threshold. Ann Am Thorac Soc. 2015;12(5):758-764. doi:10.1513/AnnalsATS.201408-399OC
- Eckert DJ, Malhotra A, Wellman A, White DP. Trazodone increases the respiratory arousal threshold in patients with obstructive sleep apnea and a low arousal threshold. Sleep. 2014;37(4):811-819. doi:10.5665/sleep.3596
- Schatzberg AF, Nemeroff CB, eds. The American Psychiat ric Association Publishing Textbook of Psychopharmacology. 4th ed. American Psychiatric Publishing; 2009.
- Ruxton K, Woodman RJ, Mangoni AA. Drugs with anticholinergic effects and cognitive impairment, falls and allcause mortality in older adults: a systematic review and meta-analysis. Br J Clin Pharmacol. 2015;80(2):209-220. doi:10.1111/bcp.12617
- Nefazodone. In: LiverTox: Clinical and Research Information on Drug-Induced Liver Injury. National Institute of Diabetes and Digestive and Kidney Diseases. Updated March 6, 2020. Accessed April 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK548179/
- Drugs of Current Interest: Nefazodone. WHO Pharmaceuticals Newsletter:(1). 2003(1):7. https://web.archive.org/web/20150403165029/http:/apps.who.int/medicinedocs/en/d/Js4944e/3.html
- Choi S. Nefazodone (serzone) withdrawn because of hepatotoxicity. CMAJ. 2003;169(11):1187.
- Teva Nefazodone Statement. News release. Teva USA. December 20, 2021. Accessed April 4, 2025. https:// www.tevausa.com/news-and-media/press-releases/teva-nefazodone-statement/
- Levitan MN, Papelbaum M, Nardi AE. Profile of agomelatine and its potential in the treatment of generalized anxiety disorder. Neuropsychiatr Dis Treat. 2015;11:1149- 1155. doi:10.2147/NDT.S67470
- Fu DJ, Ionescu DF, Li X, et al. Esketamine nasal spray for rapid reduction of major depressive disorder symptoms in patients who have active suicidal ideation with intent: double-blind, randomized study (ASPIRE I). J Clin Psychiatry. 2020;81(3):19m13191. doi:10.4088/JCP.19m13191
- Iosifescu DV, Jones A, O’Gorman C, et al. Efficacy and safety of AXS-05 (dextromethorphan-bupropion) in patients with major depressive disorder: a phase 3 randomized clinical trial (GEMINI). J Clin Psychiatry. 2022;83(4): 21m14345. doi:10.4088/JCP.21m14345
- Luong TT, Powers CN, Reinhardt BJ, et al. Retrospective evaluation of drug-drug interactions with erlotinib and gefitinib use in the Military Health System. Fed Pract. 2023;40(Suppl 3):S24-S34. doi:10.12788/fp.0401
- Luong TT, Shou KJ, Reinhard BJ, Kigelman OF, Greenfield KM. Paclitaxel drug-drug interactions in the Military Health System. Fed Pract. 2024;41(Suppl 3):S70-S82. doi:10.12788/fp.0499
- Luong TT, Powers CN, Reinhardt BJ, Weina PJ. Preclinical drug-drug interactions (DDIs) of gefitinib with/without losartan and selective serotonin reuptake inhibitors (SSRIs): citalopram, fluoxetine, fluvoxamine, paroxetine, sertraline, and venlafaxine. Curr Res Pharmacol Drug Discov. 2022;3:100112. doi:10.1016/j.crphar.2022.100112
- Luong TT, McAnulty MJ, Evers DL, Reinhardt BJ, Weina PJ. Pre-clinical drug-drug interaction (DDI) of gefitinib or erlotinib with cytochrome P450 (CYP) inhibiting drugs, fluoxetine and/or losartan. Curr Res Toxicol. 2021;2:217- 224. doi:10.1016/j.crtox.2021.05.006,
- Adamo M, Dickie L, Ruhl J. SEER program coding and staging manual 2016. National Cancer Institute; 2016. Accessed April 4, 2025. https://seer.cancer.gov/archive/manuals/2016/SPCSM_2016_maindoc.pdf
- World Health Organization. International classification of diseases for oncology (ICD-O). 3rd ed, 1st revision. World Health Organization; 2013. Accessed April 4, 2025. https://apps.who.int/iris/handle/10665/96612
- Simon S. FDA approves Jelmyto (mitomycin gel) for urothelial cancer. American Cancer Society. April 20, 2020. Accessed April 4, 2025. https://www.cancer.org/cancer/latest-news/fda-approves-jelmyto-mitomycin-gel-for-urothelial-cancer.html
- Pantziarka P, Sukhatme V, Bouche G, Meheus L, Sukhatme VP. Repurposing drugs in oncology (ReDO)- diclofenac as an anti-cancer agent. Ecancermedicalscience. 2016;10:610. doi:10.3332/ecancer.2016.610
- Choi S, Kim S, Park J, Lee SE, Kim C, Kang D. Diclofenac: a nonsteroidal anti-inflammatory drug inducing cancer cell death by inhibiting microtubule polymerization and autophagy flux. Antioxidants (Basel). 2022;11(5):1009. doi:10.3390/antiox11051009
- Tontonoz M. The ABCs of BCG: oldest approved immunotherapy gets new explanation. Memorial Sloan Kettering Cancer Center. July 17, 2020. Accessed April 4, 2025. https://www.mskcc.org/news/oldest-approved-immunotherapy-gets-new-explanation
- Jordan VC. Tamoxifen as the first targeted long-term adjuvant therapy for breast cancer. Endocr Relat Cancer. 2014;21(3):R235-R246. doi:10.1530/ERC-14-0092
- Cole MP, Jones CT, Todd ID. A new anti-oestrogenic agent in late breast cancer. An early clinical appraisal of ICI46474. Br J Cancer. 1971;25(2):270-275. doi:10.1038/bjc.1971.33
- Jordan VC. Tamoxifen: a most unlikely pioneering medicine. Nat Rev Drug Discov. 2003;2(3):205-213. doi:10.1038/nrd1031
- Maximov PY, McDaniel RE, Jordan VC. Tamoxifen: Pioneering Medicine in Breast Cancer. Springer Basel; 2013. Accessed April 4, 2025. https://link.springer.com/book/10.1007/978-3-0348-0664-0
- Taxol (paclitaxel). Prescribing information. Bristol-Myers Squibb Company; 2011. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2011/020262s049lbl.pdf
- Abraxane (paclitaxel). Prescribing information. Celgene Corporation; 2020. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/021660s047lbl.pdf
- Tarceva (erlotinib). Prescribing information. OSI Pharmaceuticals, LLC; 2016. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2016/021743s025lbl.pdf
- Docetaxel. Prescribing information. Sichuan Hyiyu Pharmaceutical Co.; 2022. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/215813s000lbl.pdf
- Alimta (pemetrexed). Prescribing information. Teva Pharmaceuticals; 2022. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/208419s004lbl.pdf
- Tagrisso (osimertinib). Prescribing information. Astra- Zeneca Pharmaceuticals; 2020. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/208065s021lbl.pdf
- Iressa (gefitinib). Prescribing information. AstraZeneca Pharmaceuticals; 2018. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/206995s003lbl.pdf
- Kadcyla (ado-trastuzumab emtansine). Prescribing information. Genentech, Inc.; 2013. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2013/125427lbl.pdf
- Alecensa (alectinib). Prescribing information. Genetech, Inc.; 2017. Accessed April 4, 2025. https:// www.accessdata.fda.gov/drugsatfda_docs/label/2017/208434s003lbl.pdf
- Xalkori (crizotinib). Prescribing information. Pfizer Laboratories; 2022. Accessed April 4, 2025. https:// www.accessdata.fda.gov/drugsatfda_docs/label/2022/202570s033lbl.pdf
- Lorbrena (lorlatinib). Prescribing information. Pfizer Laboratories; 2018. Accessed April 14, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/210868s000lbl.pdf
- Alunbrig (brigatinib). Prescribing information. Takeda Pharmaceutical Company; 2020. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/208772s008lbl.pdf
- Rozlytrek (entrectinib). Prescribing information. Genentech, Inc.; 2019. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/212725s000lbl.pdf
- Herceptin (trastuzumab). Prescribing information. Genentech, Inc.; 2010. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2010/103792s5250lbl.pdf
- Cybalta (duloxetine). Prescribing information. Eli Lilly and Company; 2017. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/021427s049lbl.pdf
- Effexor XR (venlafaxine). Prescribing information. Pfizer Wyeth Pharmaceuticals Inc; 2022. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/020699s112lbl.pdf
- Desyrel (trazodone hydrochloride). Prescribing information. Pragma Pharmaceuticals; 2017. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/018207s032lbl.pdf
- Sertraline hydrochloride. Prescribing information. Almatica Pharma LLC; 2021. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2021/215133s000lbl.pdf
- Remeron (mirtazapine). Prescribing information. Merck & Co. Inc; 2020. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/020415s029,%20021208s019lbl.pdf
- Celexa (citalopram). Prescribing information. Allergan USA Inc; 2022. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/020822s041lbl.pdf
- information. GlaxoSmithKline; 2019. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/020358s066lbl.pdf
- Amitriptyline hydrochloride tablet. Prescribing information. Quality Care Products LLC; 2022. Accessed April 4, 2025. https://www.accessdata.fda.gov/spl/data/0f12f50f-7087-46e7-a2e6-356b4c566c9f/0f12f50f-7087-46e7-a2e6-356b4c566c9f.xml
- Lexapro (escitalopram). Prescribing information. AbbVie Inc; 2023. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2023/021323s055,021365s039lbl.pdf
- Fluoxetine. Prescribing information. Edgemont Pharmaceutical, LLC; 2017. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/202133s004s005lbl.pdf
- Paxil (paroxetine). Prescribing Information. Apotex Inc; 2021. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2021/020031s077lbl.pdf
- Pamelor (nortriptyline HCl). Prescribing information. Mallinckrodt, Inc; 2012. Accessed April 4, 2025. https:// www.accessdata.fda.gov/drugsatfda_docs/label/2012/018012s029,018013s061lbl.pdf
- Silenor (doxepin). Prescribing information. Currax Pharmaceuticals; 2020. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/022036s006lbl.pdf
- Tofranil-PM (imipramine pamote). Prescribing information. Mallinckrodt, Inc; 2014. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2014/017090s078lbl.pdf
- Norpramin (desipramine hydrochloride). Prescribing information. Sanofi-aventis U.S. LLC; 2014. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2014/014399s069lbl.pdf
- Khedezla (desvenlafaxine). Prescribing information. Osmotical Pharmaceutical US LLC; 2019. Accessed April 4, 2025. https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/204683s006lbl.pdf
- Nefazodone hydrochloride. Prescribing information. Bryant Ranch Prepack; 2022. Accessed April 4, 2025. https://www.accessdata.fda.gov/spl/data/0bd4c34a-4f43-4c84-8b98-1d074cba97d5/0bd4c34a-4f43-4c84-8b98-1d074cba97d5.xml
- Grassi L, Nanni MG, Rodin G, Li M, Caruso R. The use of antidepressants in oncology: a review and practical tips for oncologists. Ann Oncol. 2018;29(1):101-111. doi:10.1093/annonc/mdx526
- Lee E, Park Y, Li D, Rodriguez-Fuguet A, Wang X, Zhang WC. Antidepressant use and lung cancer risk and survival: a meta-analysis of observational studies. Cancer Res Commun. 2023;3(6):1013-1025. doi:10.1158/2767-9764.CRC-23-0003
- Olfson M, Marcus SC. National patterns in antidepressant medication treatment. Arch Gen Psychiatry. 2009;66(8):848 -856. doi:10.1001/archgenpsychiatry.2009.81
- Grattan A, Sullivan MD, Saunders KW, Campbell CI, Von Korff MR. Depression and prescription opioid misuse among chronic opioid therapy recipients with no history of substance abuse. Ann Fam Med. 2012;10(4):304-311. doi:10.1370/afm.1371
- Cowan DT, Wilson-Barnett J, Griffiths P, Allan LG. A survey of chronic noncancer pain patients prescribed opioid analgesics. Pain Med. 2003;4(4):340-351. doi:10.1111/j.1526-4637.2003.03038.x
- Breckenridge J, Clark JD. Patient characteristics associated with opioid versus nonsteroidal anti-inflammatory drug management of chronic low back pain. J Pain. 2003;4(6):344-350. doi:10.1016/s1526-5900(03)00638-2
- Edlund MJ, Martin BC, Devries A, Fan MY, Braden JB, Sullivan MD. Trends in use of opioids for chronic noncancer pain among individuals with mental health and substance use disorders: the TROUP study. Clin J Pain. 2010;26(1):1-8. doi:10.1097/AJP.0b013e3181b99f35
- Sullivan MD, Edlund MJ, Fan MY, DeVries A, Braden JB, Martin BC. Risks for possible and probable opioid misuse among recipients of chronic opioid therapy in commercial and medicaid insurance plans: the TROUP study. Pain. 2010;150(2):332-339. doi:10.1016/j.pain.2010.05.020
- Dunn KM, Saunders KW, Rutter CM, et al. Opioid prescriptions for chronic pain and overdose: a cohort study. Ann Intern Med. 2010;152(2):85-92. doi:10.7326/0003-4819-152-2-201001190-00006
- Haddad P. Do antidepressants have any potential to cause addiction? J Psychopharmacol. 1999;13(3):300- 307. doi:10.1177/026988119901300321
- Lakeview Health Staff. America’s most abused antidepressants. Lakeview Health. January 24, 2004. Accessed April 4, 2025. https://www.lakeviewhealth.com/blog/us-most-abused-antidepressants/
- Greenhouse Treatment Center Editorial Staff. Addiction to antidepressants: is it possible? America Addiction Centers: Greenhouse Treatment Center. Updated April 23, 2024. Accessed April 4, 2025. https://greenhousetreatment.com/prescription-medication/antidepressants/
Assessing the Impact of Antidepressants on Cancer Treatment: A Retrospective Analysis of 14 Antineoplastic Agents
Assessing the Impact of Antidepressants on Cancer Treatment: A Retrospective Analysis of 14 Antineoplastic Agents
Associations Between Prescreening Dietary Patterns and Longitudinal Colonoscopy Outcomes in Veterans
Associations Between Prescreening Dietary Patterns and Longitudinal Colonoscopy Outcomes in Veterans
Screening for colorectal cancer (CRC) with colonoscopy enables the identification and removal of CRC precursors (colonic adenomas) and has been associated with reduced risk of CRC incidence and mortality.1-3 Furthermore, there is consensus that diet and lifestyle may be associated with forestalling CRC pathogenesis at the intermediate adenoma stages.4-7 However, studies have shown that US veterans have poorer diet quality and a higher risk for neoplasia compared with nonveterans, reinforcing the need for tailored clinical approaches.8,9 Combining screening with conversations about modifiable environmental and lifestyle risk factors, such as poor diet, is a highly relevant and possibly easily leveraged prevention for those at high risk. However, there is limited evidence for any particular dietary patterns or dietary features that are most important over time.7
Several dietary components have been shown to be associated with CRC risk,10 either as potentially chemopreventive (fiber, fruits and vegetables,11 dairy,12 supplemental vitamin D,13 calcium,14 and multivitamins15) or carcinogenic (red meat16 and alcohol17). Previous studies of veterans have similarly shown that higher intake of fiber and vitamin D reduced risk, and red meat is associated with an increased risk for finding CRC precursors during colonoscopy.18 However, these dietary categories are often analyzed in isolation. Studying healthy dietary patterns in aggregate may be more clinically relevant and easier to implement for prevention of CRC and its precursors.19-21 Healthy dietary patterns, such as the US Dietary Guidelines for Americans represented by the Healthy Eating Index (HEI), the Mediterranean diet (MD), and the Dietary Approaches to Stop Hypertension (DASH) diet, have been associated with lower risk for chronic disease.22-24 Despite the extant literature, no known studies have compared these dietary patterns for associations with risk of CRC precursor or CRC development among US veterans undergoing long-term screening and follow-up after a baseline colonoscopy.
The objective of this study was to test for associations between baseline scores of healthy dietary patterns and the most severe colonoscopy findings (MSCFs) over ≥ 10 years following a baseline screening colonoscopy in veterans.
Methods
Participants in the Cooperative Studies Program (CSP) #380 cohort study included 3121 asymptomatic veterans aged 50 to 75 years at baseline who had consented to initial screening colonoscopy between 1994 and 1997, with subsequent follow-up and surveillance.25 Prior to their colonoscopy, all participants completed a baseline study survey that included questions about cancer risk factors including family history of CRC, diet, physical activity, and medication use.
Included in this cross-sectional analysis were data from a sample of veteran participants of the CSP #380 cohort with 1 baseline colonoscopy, follow-up surveillance through 2009, a cancer risk factor survey collected at baseline, and complete demographic and clinical indicator data. Excluded from the analysis were 67 participants with insufficient responses to the dietary food frequency questionnaire (FFQ) and 31 participants with missing body mass index (BMI), 3023 veterans.
Measures
MSCF. The outcome of interest in this study was the MSCF recorded across all participant colonoscopies during the study period. MSCF was categorized as either (1) no neoplasia; (2) < 2 nonadvanced adenomas, including small adenomas (diameter < 10 mm) with tubular histology; or (3) advanced neoplasia (AN), which is characterized by adenomas > 10 mm in diameter, with villous histology, with high-grade dysplasia, or CRC.
Dietary patterns. Dietary pattern scores representing dietary quality and calculated based on recommendations of the US Dietary Guidelines for Americans using the HEI, MD, and DASH diets were independent variables.26-28 These 3 dietary patterns were chosen for their hypothesized relationship with CRC risk, but each weighs food categories differently (Appendix 1).22-24,29 Dietary pattern scores were calculated using the CSP #380 self-reported responses to 129 baseline survey questions adapted from a well-established and previously validated semiquantitative FFQ.30 The form was administered by mail twice to a sample of 127 participants at baseline and at 1 year. During this interval, men completed 1-week diet records twice, spaced about 6 months apart. Mean values for intake of most nutrients assessed by the 2 methods were similar. Intraclass correlation coefficients for the baseline and 1-year FFQ-assessed nutrient intakes that ranged from 0.47 for vitamin E (without supplements) to 0.80 for vitamin C (with supplements). Correlation coefficients between the energy-adjusted nutrient intakes were measured by diet records and the 1-year FFQ, which asked about diet during the year encompassing the diet records. Higher raw and percent scores indicated better alignment with recommendations from each respective dietary pattern. Percent scores were calculated as a standardizing method and used in analyses for ease of comparing the dietary patterns. Scoring can be found in Appendix 2.


Demographic characteristics and clinical indicators. Demographic characteristics included age categories, sex, and race/ethnicity. Clinical indicators included BMI, the number of comorbid conditions used to calculate the Charlson Comorbidity Index, family history of CRC in first-degree relatives, number of follow-up colonoscopies across the study period, and food-based vitamin D intake.31 These variables were chosen for their applicability found in previous CSP #380 cohort studies.18,32,33 Self-reported race and ethnicity were collapsed due to small numbers in some groups. The authors acknowledge these are distinct concepts and the variable has limited utility other than for controlling for systemic racism in the model.
Statistical Analyses
Descriptive statistics were used to describe distributional assumptions for all variables, including demographics, clinical indicators, colonoscopy results, and dietary patterns. Pairwise correlations between the total dietary pattern scores and food category scores were calculated with Pearson correlation (r).
Multinomial logistic regression models were created using SAS procedure LOGISTIC with the outcome of the categorical MSCF (no neoplasia, nonadvanced adenoma, or AN).34 A model was created for each independent predictor variable of interest (ie, the HEI, MD, or DASH percentage-standardized dietary pattern score and each food category comprising each dietary pattern score). All models were adjusted for age, sex, race/ethnicity, BMI, number of comorbidities, family history of CRC, number of follow-up colonoscopies, and estimated daily food-derived vitamin D intake. The demographic and clinical indicators were included in the models as they are known to be associated with CRC risk.18 The number of colonoscopies was included to control for surveillance intensity presuming risk for AN is reduced as polyps are removed. Because colonoscopy findings from an initial screening have unique clinical implications compared with follow- up and surveillance, MSCF was observed in 2 ways in sensitivity analyses: (1) baseline and (2) aggregate follow-up and surveillance only, excluding baseline findings.
Adjusted odds ratios (aORs) and 95% CIs for each of the MSCF outcomes with a reference finding of no neoplasia for the models are presented. We chose not to adjust for multiple comparisons across the different dietary patterns given the correlation between dietary pattern total and category scores but did adjust for multiple comparisons for dietary categories within each dietary pattern. Tests for statistical significance used α= .05 for the dietary pattern total scores and P values for the dietary category scores for each dietary pattern controlled for false discovery rate using the MULTTEST SAS procedure.35 All data manipulations and analyses were performed using SAS version 9.4.
Results
The study included 3023 patients. All were aged 50 to 75 years, 2923 (96.7%) were male and 2532 (83.8%) were non-Hispanic White (Table 1). Most participants were overweight or obese (n = 2535 [83.8%]), 2024 (67.0%) had ≤ 2 comorbidities, and 2602 (86.1%) had no family history of CRC. The MSCF for 1628 patients (53.9%) was no neoplasia, 966 patients (32.0%) was nonadvanced adenoma, and 429 participants (14.2%) had AN.

Mean percent scores were 58.5% for HEI, 38.2% for MD, and 63.1% for the DASH diet, with higher percentages indicating greater alignment with the recommendations for each diet (Table 2). All 3 dietary patterns scores standardized to percentages were strongly and significantly correlated in pairwise comparisons: HEI:MD, r = 0.62 (P < .001); HEI:DASH, r = 0.60 (P < .001); and MD:DASH, r = 0.72 (P < .001). Likewise, food category scores were significantly correlated across dietary patterns. For example, whole grain and fiber values from each dietary score were strongly correlated in pairwise comparisons: HEI Whole Grain:MD Grain, r = 0.64 (P < .001); HEI Whole Grain:DASH Fiber, r = 0.71 (P < .001); and MD Grain:DASH Fiber, r = 0.70 (P < .001).

Associations between individual participants' dietary pattern scores and the outcome of their pooled MSCF from baseline screening and ≥ 10 years of surveillance are presented in Table 3. For each single-point increases in dietary pattern scores (reflecting better dietary quality), aORs for nonadvanced adenoma vs no neoplasia were slightly lower but not statistically significantly: HEI, aOR, 1.00 (95% CI, 0.99-1.01); MD, aOR, 0.98 (95% CI, 0.94-1.02); and DASH, aOR, 0.99 (95% CI, 0.99-1.00). aORs for AN vs no neoplasia were slightly lower for each dietary pattern assessed, and only the MD and DASH scores were significantly different from 1.00: HEI, aOR, 1.00 (95% CI, 0.99-1.01); MD, aOR, 0.95 (95% CI, 0.90-1.00); and DASH, aOR, 0.99 (95% CI, 0.98-1.00).

We observed lower odds for nonadvanced adenoma and AN among all these dietary patterns when there was greater alignment with the recommended intake of whole grains and fiber. In separate models conducted using food categories comprising the dietary patterns as independent variables and after correcting for multiple tests, higher scores for the HEI Refined Grain category were associated with higher odds for nonadvanced adenoma (aOR, 1.03 [95% CI, 1.01-1.05]; P = .01) and AN (aOR, 1.05 [95% CI, 1.02-1.08]; P < .001). Higher scores for the HEI Whole Grain category were associated with lower odds for nonadvanced adenoma (aOR, 0.97 [95% CI, 0.95-0.99]; P = .01) and AN (aOR, 0.96 [95% CI, 0.93-0.99]; P = .01). Higher scores for the MD Grain category were significantly associated with lower odds for nonadvanced adenoma (aOR, 0.44 [95% CI, 0.26-0.75]; P = .002) and AN (aOR, 0.29 [95% CI, 0.14-0.62]; P = .001). The DASH Grains category also was significantly associated with lower odds for AN (aOR, 0.86 [95% CI, 0.78-0.95]; P = .002).
Discussion
In this study of 3023 veterans undergoing first-time screening colonoscopy and ≥ 10 years of surveillance, we found that healthy dietary patterns, as assessed by the MD and DASH diet, were significantly associated with lower risk of AN. Additionally, we identified lower odds for AN and nonadvanced adenoma compared with no neoplasia for higher grain scores for all the dietary patterns studied. Other food categories that comprise the dietary pattern scores had mixed associations with the MSCF outcomes. Several other studies have examined associations between dietary patterns and risk for CRC but to our knowledge, no studies have explored these associations among US veterans.
These results also indicate study participants had better than average (based on a 50% threshold) dietary quality according to the HEI and DASH diet scoring methods we used, but poor dietary quality according to the MD scoring method. The mean HEI scores for the present study were higher than a US Department of Agriculture study by Dong et al that compared dietary quality between veterans and nonveterans using the HEI, for which veterans’ expected HEI score was 45.6 of 100.8 This could be explained by the fact that the participants needed to be healthy to be eligible and those with healthier behaviors overall may have self-selected into the study due to motivation for screening during a time when screening was not yet commonplace. 36 Similarly, participants of the present study had higher adherence to the DASH diet (63.1%) than adolescents with diabetes in a study by Günther et al. Conversely, firefighters who were coached to use a Mediterranean-style dietary pattern and dietary had higher adherence to MD than did participants in this study.27
A closer examination of specific food category component scores that comprise the 3 distinct dietary patterns revealed mixed results from the multinomial modeling, which may have to do with the guideline thresholds used to calculate the dietary scores. When analyzed separately in the logistic regression models for their associations with nonadvanced adenomas and AN compared with no neoplasia, higher MD and DASH fruit scores (but not HEI fruit scores) were found to be significant. Other studies have had mixed findings when attempting to test for associations of fruit intake with adenoma recurrence.10,37
This study had some unexpected findings. Vegetable intake was not associated with nonadvanced adenomas or AN risk. Studies of food categories have consistently found vegetable (specifically cruciferous ones) intake to be linked with lower odds for cancers.38 Likewise, the red meat category, which was only a unique food category in the MD score, was not associated with nonadvanced adenomas or AN. Despite consistent literature suggesting higher intake of red meat and processed meats increases CRC risk, in 2019 the Nutritional Recommendations Consortium indicated that the evidence was weak.39,40 This study showed higher DASH diet scores for low-fat dairy, which were maximized when participants reported at least 50% of their dairy servings per day as being low-fat, had lower odds for AN. Yet, the MD scores for low-fat dairy had no association with either outcome; their calculation was based on total number of servings per week. This difference in findings suggests the fat intake ratio may be more relevant to CRC risk than intake quantity.
The literature is mixed regarding fatty acid intake and CRC risk, which may be relevant to both dairy and meat intake. One systematic review and meta-analysis found dietary fat and types of fatty acid intake had no association with CRC risk.41 However, a more recent meta-analysis that assessed both dietary intake and plasma levels of fatty acids did find some statistically significant differences for various types of fatty acids and CRC risk.42
The findings in the present study that grain intake is associated with lower odds for more severe colonoscopy findings among veterans are notable.43 Lieberman et al, using the CSP #380 data, found that cereal fiber intake was associated with a lower odds for AN compared with hyperplastic polyps (OR, 0.98 [95% CI, 0.96- 1.00]).18 Similarly, Hullings et al determined that older adults in the highest quintile of cereal fiber intake had significantly lower odds of CRC than those in lower odds for CRC when compared with lowest quintile (OR, 0.89 [95% CI, 0.83- 0.96]; P < .001).44 These findings support existing guidance that prioritizes whole grains as a key source of dietary fiber for CRC prevention.
A recent literature review on fiber, fat, and CRC risk suggested a consensus regarding one protective mechanism: dietary fiber from grains modulates the gut microbiota by promoting butyrate synthesis.45 Butyrate is a short-chain fatty acid that supports energy production in colonocytes and has tumor-suppressing properties.46 Our findings suggest there could be more to learn about the relationship between butyrate production and reduction of CRC risk through metabolomic studies that use measurements of plasma butyrate. These studies may examine associations between not just a singular food or food category, but rather food patterns that include fruits, vegetables, nuts and seeds, and whole grains known to promote butyrate production and plasma butyrate.47
Improved understanding of mechanisms and risk-modifying lifestyle factors such as dietary patterns may enhance prevention strategies. Identifying the collective chemopreventive characteristics of a specific dietary pattern (eg, MD) will be helpful to clinicians and health care staff to promote healthy eating to reduce cancer risk. More studies are needed to understand whether such promotion is more clinically applicable and effective for patients, as compared with eating more or less of specific foods (eg, more whole grains, less red meat). Furthermore, considering important environmental factors collectively beyond dietary patterns may offer a way to better tailor screening and implement a variety of lifestyle interventions. In the literature, this is often referred to as a teachable moment when patients’ attentions are captured and may position them to be more receptive to guidance.48
Limitations
This study has several important limitations and leaves opportunities for future studies that explore the role of dietary patterns and AN or CRC risk. First, the FFQ data used to calculate dietary pattern scores used in analysis were only captured at baseline, and there are nearly 3 decades across the study period. However, it is widely assumed that the diets of older adults, like those included in this study, remain stable over time which is appropriate given our sample population was aged 50 to 75 years when the baseline FFQ data were collected.49-51 Additionally, while the HEI is a well-documented, standard scoring method for dietary quality, there are multitudes of dietary pattern scoring approaches for MD and DASH.23,52,53 Finally, findings from this study using the sample of veterans may not be generalizable to a broader population. Future longitudinal studies that test for a clinically significant change threshold are warranted.
Conclusion
Results of this study suggest future research should further explore the effects of dietary patterns, particularly intake of specific food groups in combination, as opposed to individual nutrients or food items, on AN and CRC risk. Possible studies might explore these dietary patterns for their mechanistic role in altering the microbiome metabolism, which may influence CRC outcomes or include diet in a more comprehensive, holistic risk score that could be used to predict colonic neoplasia risk or in intervention studies that assess the effects of dietary changes on long-term CRC prevention. We suggest there are differences in people’s dietary intake patterns that might be important to consider when implementing tailored approaches to CRC risk mitigation.
- Zauber AG, Winawer SJ, O’Brien MJ, et al. Colonoscopic polypectomy and long-term prevention of colorectalcancer deaths. N Engl J Med. 2012;366(8):687-696. doi:10.1056/NEJMoa1100370
- Nishihara R, Wu K, Lochhead P, et al. Long-term colorectal-cancer incidence and mortality after lower endoscopy. N Engl J Med. 2013;369(12):1095-1105. doi:10.1056/NEJMoa1301969
- Bretthauer M, Løberg M, Wieszczy P, et al. Effect of colonoscopy screening on risks of colorectal cancer and related death. N Engl J Med. 2022;387(17):1547-1556. doi:10.1056/NEJMoa2208375
- Cottet V, Bonithon-Kopp C, Kronborg O, et al. Dietary patterns and the risk of colorectal adenoma recurrence in a European intervention trial. Eur J Cancer Prev. 2005;14(1):21.
- Miller PE, Lesko SM, Muscat JE, Lazarus P, Hartman TJ. Dietary patterns and colorectal adenoma and cancer risk: a review of the epidemiological evidence. Nutr Cancer. 2010;62(4):413-424. doi:10.1080/01635580903407114
- Godos J, Bella F, Torrisi A, Sciacca S, Galvano F, Grosso G. Dietary patterns and risk of colorectal adenoma: a systematic review and meta-analysis of observational studies. J Hum Nutr Diet Off J Br Diet Assoc. 2016;29(6):757-767. doi:10.1111/jhn.12395
- Haggar FA, Boushey RP. Colorectal cancer epidemiology: incidence, mortality, survival, and risk factors. Clin Colon Rectal Surg. 2009;22(4):191-197. doi:10.1055/s-0029-1242458
- Dong D, Stewart H, Carlson AC. An Examination of Veterans’ Diet Quality. U.S. Department of Agriculture, Economic Research Service; 2019:32.
- El-Halabi MM, Rex DK, Saito A, Eckert GJ, Kahi CJ. Defining adenoma detection rate benchmarks in average-risk male veterans. Gastrointest Endosc. 2019;89(1):137-143. doi:10.1016/j.gie.2018.08.021
- Alberts DS, Hess LM, eds. Fundamentals of Cancer Prevention. Springer International Publishing; 2019. doi:10.1007/978-3-030-15935-1
- Dahm CC, Keogh RH, Spencer EA, et al. Dietary fiber and colorectal cancer risk: a nested case-control study using food diaries. J Natl Cancer Inst. 2010;102(9):614-626. doi:10.1093/jnci/djq092
- Aune D, Lau R, Chan DSM, et al. Dairy products and colorectal cancer risk: a systematic review and metaanalysis of cohort studies. Ann Oncol. 2012;23(1):37-45. doi:10.1093/annonc/mdr269
- Lee JE, Li H, Chan AT, et al. Circulating levels of vitamin D and colon and rectal cancer: the Physicians’ Health Study and a meta-analysis of prospective studies. Cancer Prev Res Phila Pa. 2011;4(5):735-743. doi:10.1158/1940-6207.CAPR-10-0289
- Carroll C, Cooper K, Papaioannou D, Hind D, Pilgrim H, Tappenden P. Supplemental calcium in the chemoprevention of colorectal cancer: a systematic review and meta-analysis. Clin Ther. 2010;32(5):789-803. doi:10.1016/j.clinthera.2010.04.024
- Park Y, Spiegelman D, Hunter DJ, et al. Intakes of vitamins A, C, and E and use of multiple vitamin supplements and risk of colon cancer: a pooled analysis of prospective cohort studies. Cancer Causes Control CCC. 2010;21(11):1745- 1757. doi:10.1007/s10552-010-9549-y
- Alexander DD, Weed DL, Miller PE, Mohamed MA. Red meat and colorectal cancer: a quantitative update on the state of the epidemiologic science. J Am Coll Nutr. 2015;34(6):521-543. doi:10.1080/07315724.2014.992553
- Park SY, Wilkens LR, Setiawan VW, Monroe KR, Haiman CA, Le Marchand L. Alcohol intake and colorectal cancer risk in the multiethnic cohort study. Am J Epidemiol. 2019;188(1):67-76. doi:10.1093/aje/kwy208
- Lieberman DA. Risk Factors for advanced colonic neoplasia and hyperplastic polyps in asymptomatic individuals. JAMA. 2003;290(22):2959. doi:10.1001/jama.290.22.2959
- Archambault AN, Jeon J, Lin Y, et al. Risk stratification for early-onset colorectal cancer using a combination of genetic and environmental risk scores: an international multi-center study. J Natl Cancer Inst. 2022;114(4):528-539. doi:10.1093/jnci/djac003
- Carr PR, Weigl K, Edelmann D, et al. Estimation of absolute risk of colorectal cancer based on healthy lifestyle, genetic risk, and colonoscopy status in a populationbased study. Gastroenterology. 2020;159(1):129-138.e9. doi:10.1053/j.gastro.2020.03.016
- Sullivan BA, Qin X, Miller C, et al. Screening colonoscopy findings are associated with noncolorectal cancer mortality. Clin Transl Gastroenterol. 2022;13(4):e00479. doi:10.14309/ctg.0000000000000479
- Erben V, Carr PR, Holleczek B, Stegmaier C, Hoffmeister M, Brenner H. Dietary patterns and risk of advanced colorectal neoplasms: A large population based screening study in Germany. Prev Med. 2018;111:101-109. doi:10.1016/j.ypmed.2018.02.025
- Donovan MG, Selmin OI, Doetschman TC, Romagnolo DF. Mediterranean diet: prevention of colorectal cancer. Front Nutr. 2017;4:59. doi:10.3389/fnut.2017.00059
- Mohseni R, Mohseni F, Alizadeh S, Abbasi S. The Association of Dietary Approaches to Stop Hypertension (DASH) diet with the risk of colorectal cancer: a meta-analysis of observational studies.Nutr Cancer. 2020;72(5):778-790. doi:10.1080/01635581.2019.1651880
- Lieberman DA, Weiss DG, Bond JH, Ahnen DJ, Garewal H, Chejfec G. Use of colonoscopy to screen asymptomatic adults for colorectal cancer. Veterans Affairs Cooperative Study Group 380. N Engl J Med. 2000;343(3):162-168. doi:10.1056/NEJM200007203430301
- Developing the Healthy Eating Index (HEI) | EGRP/ DCCPS/NCI/NIH. Accessed July 22, 2025. https://epi.grants.cancer.gov/hei/developing.html#2015c
- Reeve E, Piccici F, Feairheller DL. Validation of a Mediterranean diet scoring system for intervention based research. J Nutr Med Diet Care. 2021;7(1):053. doi:10.23937/2572-3278/1510053
- Günther AL, Liese AD, Bell RA, et al. ASSOCIATION BETWEEN THE DIETARY APPROACHES TO HYPERTENSION (DASH) DIET AND HYPERTENSION IN YOUTH WITH DIABETES. Hypertens Dallas Tex 1979. 2009;53(1):6-12. doi:10.1161/HYPERTENSIONAHA.108.116665
- Buckland G, Agudo A, Luján L, et al. Adherence to a Mediterranean diet and risk of gastric adenocarcinoma within the European Prospective Investigation into Cancer and Nutrition (EPIC) cohort study. Am J Clin Nutr. 2010;91(2):381- 390. doi:10.3945/ajcn.2009.28209
- Rimm EB, Giovannucci EL, Stampfer MJ, Colditz GA, Litin LB, Willett WC. Reproducibility and validity of an expanded self-administered semiquantitative food frequency questionnaire among male health professionals. Am J Epidemiol. 1992;135(10):1114-1126. doi:10.1093/oxfordjournals.aje.a116211
- Charlson ME, Pompei P, Ales KL, MacKenzie CR. A new method of classifying prognostic comorbidity in longitudinal studies: development and validation. J Chronic Dis. 1987;40(5):373-383. doi:10.1016/0021-9681(87)90171-8
- Lieberman DA, Weiss DG, Harford WV, et al. Fiveyear colon surveillance after screening colonoscopy. Gastroenterology. 2007;133(4):1077-1085. doi:10.1053/j.gastro.2007.07.006
- Lieberman D, Sullivan BA, Hauser ER, et al. Baseline colonoscopy findings associated with 10-year outcomes in a screening cohort undergoing colonoscopy surveillance. Gastroenterology. 2020;158(4):862-874.e8. doi:10.1053/j.gastro.2019.07.052
- PROC LOGISTIC: PROC LOGISTIC Statement : SAS/STAT(R) 9.22 User’s Guide. Accessed July 22, 2025. https://support.sas.com/documentation/cdl/en/statug/63347/HTML/default/viewer.htm#statug_logistic_sect004.htm
- PROC MULTTEST: PROC MULTTEST Statement : SAS/ STAT(R) 9.22 User’s Guide. Accessed July 22, 2025. https://support.sas.com/documentation/cdl/en/statug/63347/HTML/default/viewer.htm#statug_multtest_sect005.htm
- Elston DM. Participation bias, self-selection bias, and response bias. J Am Acad Dermatol. Published online June 18, 2021. doi:10.1016/j.jaad.2021.06.025
- Sansbury LB, Wanke K, Albert PS, et al. The effect of strict adherence to a high-fiber, high-fruit and -vegetable, and low-fat eating pattern on adenoma recurrence. Am J Epidemiol. 2009;170(5):576-584. doi:10.1093/aje/kwp169
- Borgas P, Gonzalez G, Veselkov K, Mirnezami R. Phytochemically rich dietary components and the risk of colorectal cancer: A systematic review and meta-analysis of observational studies. World J Clin Oncol. 2021;12(6):482- 499. doi:10.5306/wjco.v12.i6.482
- Papadimitriou N, Markozannes G, Kanellopoulou A, et al. An umbrella review of the evidence associating diet and cancer risk at 11 anatomical sites. Nat Commun. 2021;12(1):4579. doi:10.1038/s41467-021-24861-8
- Johnston BC, Zeraatkar D, Han MA, et al. Unprocessed red meat and processed meat consumption: dietary guideline recommendations from the nutritional recommendations (NutriRECS) Consortium. Ann Intern Med. 2019;171(10):756-764. doi:10.7326/M19-1621
- Kim M, Park K. Dietary fat intake and risk of colorectal cancer: a systematic review and meta-analysis of prospective studies. Nutrients. 2018;10(12):1963. doi:10.3390/nu10121963
- Lu Y, Li D, Wang L, et al. Comprehensive investigation on associations between dietary intake and blood levels of fatty acids and colorectal cancer risk. Nutrients. 2023;15(3):730. doi:10.3390/nu15030730
- Gherasim A, Arhire LI, Ni.a O, Popa AD, Graur M, Mihalache L. The relationship between lifestyle components and dietary patterns. Proc Nutr Soc. 2020;79(3):311-323. doi:10.1017/S0029665120006898
- Hullings AG, Sinha R, Liao LM, Freedman ND, Graubard BI, Loftfield E. Whole grain and dietary fiber intake and risk of colorectal cancer in the NIH-AARP Diet and Health Study cohort. Am J Clin Nutr. 2020;112(3):603- 612. doi:10.1093/ajcn/nqaa161
- Ocvirk S, Wilson AS, Appolonia CN, Thomas TK, O’Keefe SJD. Fiber, fat, and colorectal cancer: new insight into modifiable dietary risk factors. Curr Gastroenterol Rep. 2019;21(11):62. doi:10.1007/s11894-019-0725-2
- O’Keefe SJD. Diet, microorganisms and their metabolites, and colon cancer. Nat Rev Gastroenterol Hepatol. 2016;13(12):691-706. doi:10.1038/nrgastro.2016.165
- The health benefits and side effects of Butyrate Cleveland Clinic. July 11, 2022. Accessed July 22, 2025. https://health.clevelandclinic.org/butyrate-benefits/
- Knudsen MD, Wang L, Wang K, et al. Changes in lifestyle factors after endoscopic screening: a prospective study in the United States. Clin Gastroenterol Hepatol Off ClinPract J Am Gastroenterol Assoc. 2022;20(6):e1240-e1249. doi:10.1016/j.cgh.2021.07.014
- Thorpe MG, Milte CM, Crawford D, McNaughton SA. Education and lifestyle predict change in dietary patterns and diet quality of adults 55 years and over. Nutr J. 2019;18(1):67. doi:10.1186/s12937-019-0495-6
- Chapman K, Ogden J. How do people change their diet?: an exploration into mechanisms of dietary change. J Health Psychol. 2009;14(8):1229-1242. doi:10.1177/1359105309342289
- Djoussé L, Petrone AB, Weir NL, et al. Repeated versus single measurement of plasma omega-3 fatty acids and risk of heart failure. Eur J Nutr. 2014;53(6):1403-1408. doi:10.1007/s00394-013-0642-3
- Bach-Faig A, Berry EM, Lairon D, et al. Mediterranean diet pyramid today. Science and cultural updates. Public Health Nutr. 2011;14(12A):2274-2284. doi:10.1017/S1368980011002515
- Miller PE, Cross AJ, Subar AF, et al. Comparison of 4 established DASH diet indexes: examining associations of index scores and colorectal cancer123. Am J Clin Nutr. 2013;98(3):794-803. doi:10.3945/ajcn.113.063602
- Krebs-Smith SM, Pannucci TE, Subar AF, et al. Update of the Healthy Eating Index: HEI-2015. J Acad Nutr Diet. 2018;118(9):1591-1602. doi:10.1016/j.jand.2018.05.021
- P.R. Pehrsson, Cutrufelli RL, Gebhardt SE, et al. USDA Database for the Added Sugars Content of Selected Foods. USDA; 2005. www.ars.usda.gov/nutrientdata
Screening for colorectal cancer (CRC) with colonoscopy enables the identification and removal of CRC precursors (colonic adenomas) and has been associated with reduced risk of CRC incidence and mortality.1-3 Furthermore, there is consensus that diet and lifestyle may be associated with forestalling CRC pathogenesis at the intermediate adenoma stages.4-7 However, studies have shown that US veterans have poorer diet quality and a higher risk for neoplasia compared with nonveterans, reinforcing the need for tailored clinical approaches.8,9 Combining screening with conversations about modifiable environmental and lifestyle risk factors, such as poor diet, is a highly relevant and possibly easily leveraged prevention for those at high risk. However, there is limited evidence for any particular dietary patterns or dietary features that are most important over time.7
Several dietary components have been shown to be associated with CRC risk,10 either as potentially chemopreventive (fiber, fruits and vegetables,11 dairy,12 supplemental vitamin D,13 calcium,14 and multivitamins15) or carcinogenic (red meat16 and alcohol17). Previous studies of veterans have similarly shown that higher intake of fiber and vitamin D reduced risk, and red meat is associated with an increased risk for finding CRC precursors during colonoscopy.18 However, these dietary categories are often analyzed in isolation. Studying healthy dietary patterns in aggregate may be more clinically relevant and easier to implement for prevention of CRC and its precursors.19-21 Healthy dietary patterns, such as the US Dietary Guidelines for Americans represented by the Healthy Eating Index (HEI), the Mediterranean diet (MD), and the Dietary Approaches to Stop Hypertension (DASH) diet, have been associated with lower risk for chronic disease.22-24 Despite the extant literature, no known studies have compared these dietary patterns for associations with risk of CRC precursor or CRC development among US veterans undergoing long-term screening and follow-up after a baseline colonoscopy.
The objective of this study was to test for associations between baseline scores of healthy dietary patterns and the most severe colonoscopy findings (MSCFs) over ≥ 10 years following a baseline screening colonoscopy in veterans.
Methods
Participants in the Cooperative Studies Program (CSP) #380 cohort study included 3121 asymptomatic veterans aged 50 to 75 years at baseline who had consented to initial screening colonoscopy between 1994 and 1997, with subsequent follow-up and surveillance.25 Prior to their colonoscopy, all participants completed a baseline study survey that included questions about cancer risk factors including family history of CRC, diet, physical activity, and medication use.
Included in this cross-sectional analysis were data from a sample of veteran participants of the CSP #380 cohort with 1 baseline colonoscopy, follow-up surveillance through 2009, a cancer risk factor survey collected at baseline, and complete demographic and clinical indicator data. Excluded from the analysis were 67 participants with insufficient responses to the dietary food frequency questionnaire (FFQ) and 31 participants with missing body mass index (BMI), 3023 veterans.
Measures
MSCF. The outcome of interest in this study was the MSCF recorded across all participant colonoscopies during the study period. MSCF was categorized as either (1) no neoplasia; (2) < 2 nonadvanced adenomas, including small adenomas (diameter < 10 mm) with tubular histology; or (3) advanced neoplasia (AN), which is characterized by adenomas > 10 mm in diameter, with villous histology, with high-grade dysplasia, or CRC.
Dietary patterns. Dietary pattern scores representing dietary quality and calculated based on recommendations of the US Dietary Guidelines for Americans using the HEI, MD, and DASH diets were independent variables.26-28 These 3 dietary patterns were chosen for their hypothesized relationship with CRC risk, but each weighs food categories differently (Appendix 1).22-24,29 Dietary pattern scores were calculated using the CSP #380 self-reported responses to 129 baseline survey questions adapted from a well-established and previously validated semiquantitative FFQ.30 The form was administered by mail twice to a sample of 127 participants at baseline and at 1 year. During this interval, men completed 1-week diet records twice, spaced about 6 months apart. Mean values for intake of most nutrients assessed by the 2 methods were similar. Intraclass correlation coefficients for the baseline and 1-year FFQ-assessed nutrient intakes that ranged from 0.47 for vitamin E (without supplements) to 0.80 for vitamin C (with supplements). Correlation coefficients between the energy-adjusted nutrient intakes were measured by diet records and the 1-year FFQ, which asked about diet during the year encompassing the diet records. Higher raw and percent scores indicated better alignment with recommendations from each respective dietary pattern. Percent scores were calculated as a standardizing method and used in analyses for ease of comparing the dietary patterns. Scoring can be found in Appendix 2.


Demographic characteristics and clinical indicators. Demographic characteristics included age categories, sex, and race/ethnicity. Clinical indicators included BMI, the number of comorbid conditions used to calculate the Charlson Comorbidity Index, family history of CRC in first-degree relatives, number of follow-up colonoscopies across the study period, and food-based vitamin D intake.31 These variables were chosen for their applicability found in previous CSP #380 cohort studies.18,32,33 Self-reported race and ethnicity were collapsed due to small numbers in some groups. The authors acknowledge these are distinct concepts and the variable has limited utility other than for controlling for systemic racism in the model.
Statistical Analyses
Descriptive statistics were used to describe distributional assumptions for all variables, including demographics, clinical indicators, colonoscopy results, and dietary patterns. Pairwise correlations between the total dietary pattern scores and food category scores were calculated with Pearson correlation (r).
Multinomial logistic regression models were created using SAS procedure LOGISTIC with the outcome of the categorical MSCF (no neoplasia, nonadvanced adenoma, or AN).34 A model was created for each independent predictor variable of interest (ie, the HEI, MD, or DASH percentage-standardized dietary pattern score and each food category comprising each dietary pattern score). All models were adjusted for age, sex, race/ethnicity, BMI, number of comorbidities, family history of CRC, number of follow-up colonoscopies, and estimated daily food-derived vitamin D intake. The demographic and clinical indicators were included in the models as they are known to be associated with CRC risk.18 The number of colonoscopies was included to control for surveillance intensity presuming risk for AN is reduced as polyps are removed. Because colonoscopy findings from an initial screening have unique clinical implications compared with follow- up and surveillance, MSCF was observed in 2 ways in sensitivity analyses: (1) baseline and (2) aggregate follow-up and surveillance only, excluding baseline findings.
Adjusted odds ratios (aORs) and 95% CIs for each of the MSCF outcomes with a reference finding of no neoplasia for the models are presented. We chose not to adjust for multiple comparisons across the different dietary patterns given the correlation between dietary pattern total and category scores but did adjust for multiple comparisons for dietary categories within each dietary pattern. Tests for statistical significance used α= .05 for the dietary pattern total scores and P values for the dietary category scores for each dietary pattern controlled for false discovery rate using the MULTTEST SAS procedure.35 All data manipulations and analyses were performed using SAS version 9.4.
Results
The study included 3023 patients. All were aged 50 to 75 years, 2923 (96.7%) were male and 2532 (83.8%) were non-Hispanic White (Table 1). Most participants were overweight or obese (n = 2535 [83.8%]), 2024 (67.0%) had ≤ 2 comorbidities, and 2602 (86.1%) had no family history of CRC. The MSCF for 1628 patients (53.9%) was no neoplasia, 966 patients (32.0%) was nonadvanced adenoma, and 429 participants (14.2%) had AN.

Mean percent scores were 58.5% for HEI, 38.2% for MD, and 63.1% for the DASH diet, with higher percentages indicating greater alignment with the recommendations for each diet (Table 2). All 3 dietary patterns scores standardized to percentages were strongly and significantly correlated in pairwise comparisons: HEI:MD, r = 0.62 (P < .001); HEI:DASH, r = 0.60 (P < .001); and MD:DASH, r = 0.72 (P < .001). Likewise, food category scores were significantly correlated across dietary patterns. For example, whole grain and fiber values from each dietary score were strongly correlated in pairwise comparisons: HEI Whole Grain:MD Grain, r = 0.64 (P < .001); HEI Whole Grain:DASH Fiber, r = 0.71 (P < .001); and MD Grain:DASH Fiber, r = 0.70 (P < .001).

Associations between individual participants' dietary pattern scores and the outcome of their pooled MSCF from baseline screening and ≥ 10 years of surveillance are presented in Table 3. For each single-point increases in dietary pattern scores (reflecting better dietary quality), aORs for nonadvanced adenoma vs no neoplasia were slightly lower but not statistically significantly: HEI, aOR, 1.00 (95% CI, 0.99-1.01); MD, aOR, 0.98 (95% CI, 0.94-1.02); and DASH, aOR, 0.99 (95% CI, 0.99-1.00). aORs for AN vs no neoplasia were slightly lower for each dietary pattern assessed, and only the MD and DASH scores were significantly different from 1.00: HEI, aOR, 1.00 (95% CI, 0.99-1.01); MD, aOR, 0.95 (95% CI, 0.90-1.00); and DASH, aOR, 0.99 (95% CI, 0.98-1.00).

We observed lower odds for nonadvanced adenoma and AN among all these dietary patterns when there was greater alignment with the recommended intake of whole grains and fiber. In separate models conducted using food categories comprising the dietary patterns as independent variables and after correcting for multiple tests, higher scores for the HEI Refined Grain category were associated with higher odds for nonadvanced adenoma (aOR, 1.03 [95% CI, 1.01-1.05]; P = .01) and AN (aOR, 1.05 [95% CI, 1.02-1.08]; P < .001). Higher scores for the HEI Whole Grain category were associated with lower odds for nonadvanced adenoma (aOR, 0.97 [95% CI, 0.95-0.99]; P = .01) and AN (aOR, 0.96 [95% CI, 0.93-0.99]; P = .01). Higher scores for the MD Grain category were significantly associated with lower odds for nonadvanced adenoma (aOR, 0.44 [95% CI, 0.26-0.75]; P = .002) and AN (aOR, 0.29 [95% CI, 0.14-0.62]; P = .001). The DASH Grains category also was significantly associated with lower odds for AN (aOR, 0.86 [95% CI, 0.78-0.95]; P = .002).
Discussion
In this study of 3023 veterans undergoing first-time screening colonoscopy and ≥ 10 years of surveillance, we found that healthy dietary patterns, as assessed by the MD and DASH diet, were significantly associated with lower risk of AN. Additionally, we identified lower odds for AN and nonadvanced adenoma compared with no neoplasia for higher grain scores for all the dietary patterns studied. Other food categories that comprise the dietary pattern scores had mixed associations with the MSCF outcomes. Several other studies have examined associations between dietary patterns and risk for CRC but to our knowledge, no studies have explored these associations among US veterans.
These results also indicate study participants had better than average (based on a 50% threshold) dietary quality according to the HEI and DASH diet scoring methods we used, but poor dietary quality according to the MD scoring method. The mean HEI scores for the present study were higher than a US Department of Agriculture study by Dong et al that compared dietary quality between veterans and nonveterans using the HEI, for which veterans’ expected HEI score was 45.6 of 100.8 This could be explained by the fact that the participants needed to be healthy to be eligible and those with healthier behaviors overall may have self-selected into the study due to motivation for screening during a time when screening was not yet commonplace. 36 Similarly, participants of the present study had higher adherence to the DASH diet (63.1%) than adolescents with diabetes in a study by Günther et al. Conversely, firefighters who were coached to use a Mediterranean-style dietary pattern and dietary had higher adherence to MD than did participants in this study.27
A closer examination of specific food category component scores that comprise the 3 distinct dietary patterns revealed mixed results from the multinomial modeling, which may have to do with the guideline thresholds used to calculate the dietary scores. When analyzed separately in the logistic regression models for their associations with nonadvanced adenomas and AN compared with no neoplasia, higher MD and DASH fruit scores (but not HEI fruit scores) were found to be significant. Other studies have had mixed findings when attempting to test for associations of fruit intake with adenoma recurrence.10,37
This study had some unexpected findings. Vegetable intake was not associated with nonadvanced adenomas or AN risk. Studies of food categories have consistently found vegetable (specifically cruciferous ones) intake to be linked with lower odds for cancers.38 Likewise, the red meat category, which was only a unique food category in the MD score, was not associated with nonadvanced adenomas or AN. Despite consistent literature suggesting higher intake of red meat and processed meats increases CRC risk, in 2019 the Nutritional Recommendations Consortium indicated that the evidence was weak.39,40 This study showed higher DASH diet scores for low-fat dairy, which were maximized when participants reported at least 50% of their dairy servings per day as being low-fat, had lower odds for AN. Yet, the MD scores for low-fat dairy had no association with either outcome; their calculation was based on total number of servings per week. This difference in findings suggests the fat intake ratio may be more relevant to CRC risk than intake quantity.
The literature is mixed regarding fatty acid intake and CRC risk, which may be relevant to both dairy and meat intake. One systematic review and meta-analysis found dietary fat and types of fatty acid intake had no association with CRC risk.41 However, a more recent meta-analysis that assessed both dietary intake and plasma levels of fatty acids did find some statistically significant differences for various types of fatty acids and CRC risk.42
The findings in the present study that grain intake is associated with lower odds for more severe colonoscopy findings among veterans are notable.43 Lieberman et al, using the CSP #380 data, found that cereal fiber intake was associated with a lower odds for AN compared with hyperplastic polyps (OR, 0.98 [95% CI, 0.96- 1.00]).18 Similarly, Hullings et al determined that older adults in the highest quintile of cereal fiber intake had significantly lower odds of CRC than those in lower odds for CRC when compared with lowest quintile (OR, 0.89 [95% CI, 0.83- 0.96]; P < .001).44 These findings support existing guidance that prioritizes whole grains as a key source of dietary fiber for CRC prevention.
A recent literature review on fiber, fat, and CRC risk suggested a consensus regarding one protective mechanism: dietary fiber from grains modulates the gut microbiota by promoting butyrate synthesis.45 Butyrate is a short-chain fatty acid that supports energy production in colonocytes and has tumor-suppressing properties.46 Our findings suggest there could be more to learn about the relationship between butyrate production and reduction of CRC risk through metabolomic studies that use measurements of plasma butyrate. These studies may examine associations between not just a singular food or food category, but rather food patterns that include fruits, vegetables, nuts and seeds, and whole grains known to promote butyrate production and plasma butyrate.47
Improved understanding of mechanisms and risk-modifying lifestyle factors such as dietary patterns may enhance prevention strategies. Identifying the collective chemopreventive characteristics of a specific dietary pattern (eg, MD) will be helpful to clinicians and health care staff to promote healthy eating to reduce cancer risk. More studies are needed to understand whether such promotion is more clinically applicable and effective for patients, as compared with eating more or less of specific foods (eg, more whole grains, less red meat). Furthermore, considering important environmental factors collectively beyond dietary patterns may offer a way to better tailor screening and implement a variety of lifestyle interventions. In the literature, this is often referred to as a teachable moment when patients’ attentions are captured and may position them to be more receptive to guidance.48
Limitations
This study has several important limitations and leaves opportunities for future studies that explore the role of dietary patterns and AN or CRC risk. First, the FFQ data used to calculate dietary pattern scores used in analysis were only captured at baseline, and there are nearly 3 decades across the study period. However, it is widely assumed that the diets of older adults, like those included in this study, remain stable over time which is appropriate given our sample population was aged 50 to 75 years when the baseline FFQ data were collected.49-51 Additionally, while the HEI is a well-documented, standard scoring method for dietary quality, there are multitudes of dietary pattern scoring approaches for MD and DASH.23,52,53 Finally, findings from this study using the sample of veterans may not be generalizable to a broader population. Future longitudinal studies that test for a clinically significant change threshold are warranted.
Conclusion
Results of this study suggest future research should further explore the effects of dietary patterns, particularly intake of specific food groups in combination, as opposed to individual nutrients or food items, on AN and CRC risk. Possible studies might explore these dietary patterns for their mechanistic role in altering the microbiome metabolism, which may influence CRC outcomes or include diet in a more comprehensive, holistic risk score that could be used to predict colonic neoplasia risk or in intervention studies that assess the effects of dietary changes on long-term CRC prevention. We suggest there are differences in people’s dietary intake patterns that might be important to consider when implementing tailored approaches to CRC risk mitigation.
Screening for colorectal cancer (CRC) with colonoscopy enables the identification and removal of CRC precursors (colonic adenomas) and has been associated with reduced risk of CRC incidence and mortality.1-3 Furthermore, there is consensus that diet and lifestyle may be associated with forestalling CRC pathogenesis at the intermediate adenoma stages.4-7 However, studies have shown that US veterans have poorer diet quality and a higher risk for neoplasia compared with nonveterans, reinforcing the need for tailored clinical approaches.8,9 Combining screening with conversations about modifiable environmental and lifestyle risk factors, such as poor diet, is a highly relevant and possibly easily leveraged prevention for those at high risk. However, there is limited evidence for any particular dietary patterns or dietary features that are most important over time.7
Several dietary components have been shown to be associated with CRC risk,10 either as potentially chemopreventive (fiber, fruits and vegetables,11 dairy,12 supplemental vitamin D,13 calcium,14 and multivitamins15) or carcinogenic (red meat16 and alcohol17). Previous studies of veterans have similarly shown that higher intake of fiber and vitamin D reduced risk, and red meat is associated with an increased risk for finding CRC precursors during colonoscopy.18 However, these dietary categories are often analyzed in isolation. Studying healthy dietary patterns in aggregate may be more clinically relevant and easier to implement for prevention of CRC and its precursors.19-21 Healthy dietary patterns, such as the US Dietary Guidelines for Americans represented by the Healthy Eating Index (HEI), the Mediterranean diet (MD), and the Dietary Approaches to Stop Hypertension (DASH) diet, have been associated with lower risk for chronic disease.22-24 Despite the extant literature, no known studies have compared these dietary patterns for associations with risk of CRC precursor or CRC development among US veterans undergoing long-term screening and follow-up after a baseline colonoscopy.
The objective of this study was to test for associations between baseline scores of healthy dietary patterns and the most severe colonoscopy findings (MSCFs) over ≥ 10 years following a baseline screening colonoscopy in veterans.
Methods
Participants in the Cooperative Studies Program (CSP) #380 cohort study included 3121 asymptomatic veterans aged 50 to 75 years at baseline who had consented to initial screening colonoscopy between 1994 and 1997, with subsequent follow-up and surveillance.25 Prior to their colonoscopy, all participants completed a baseline study survey that included questions about cancer risk factors including family history of CRC, diet, physical activity, and medication use.
Included in this cross-sectional analysis were data from a sample of veteran participants of the CSP #380 cohort with 1 baseline colonoscopy, follow-up surveillance through 2009, a cancer risk factor survey collected at baseline, and complete demographic and clinical indicator data. Excluded from the analysis were 67 participants with insufficient responses to the dietary food frequency questionnaire (FFQ) and 31 participants with missing body mass index (BMI), 3023 veterans.
Measures
MSCF. The outcome of interest in this study was the MSCF recorded across all participant colonoscopies during the study period. MSCF was categorized as either (1) no neoplasia; (2) < 2 nonadvanced adenomas, including small adenomas (diameter < 10 mm) with tubular histology; or (3) advanced neoplasia (AN), which is characterized by adenomas > 10 mm in diameter, with villous histology, with high-grade dysplasia, or CRC.
Dietary patterns. Dietary pattern scores representing dietary quality and calculated based on recommendations of the US Dietary Guidelines for Americans using the HEI, MD, and DASH diets were independent variables.26-28 These 3 dietary patterns were chosen for their hypothesized relationship with CRC risk, but each weighs food categories differently (Appendix 1).22-24,29 Dietary pattern scores were calculated using the CSP #380 self-reported responses to 129 baseline survey questions adapted from a well-established and previously validated semiquantitative FFQ.30 The form was administered by mail twice to a sample of 127 participants at baseline and at 1 year. During this interval, men completed 1-week diet records twice, spaced about 6 months apart. Mean values for intake of most nutrients assessed by the 2 methods were similar. Intraclass correlation coefficients for the baseline and 1-year FFQ-assessed nutrient intakes that ranged from 0.47 for vitamin E (without supplements) to 0.80 for vitamin C (with supplements). Correlation coefficients between the energy-adjusted nutrient intakes were measured by diet records and the 1-year FFQ, which asked about diet during the year encompassing the diet records. Higher raw and percent scores indicated better alignment with recommendations from each respective dietary pattern. Percent scores were calculated as a standardizing method and used in analyses for ease of comparing the dietary patterns. Scoring can be found in Appendix 2.


Demographic characteristics and clinical indicators. Demographic characteristics included age categories, sex, and race/ethnicity. Clinical indicators included BMI, the number of comorbid conditions used to calculate the Charlson Comorbidity Index, family history of CRC in first-degree relatives, number of follow-up colonoscopies across the study period, and food-based vitamin D intake.31 These variables were chosen for their applicability found in previous CSP #380 cohort studies.18,32,33 Self-reported race and ethnicity were collapsed due to small numbers in some groups. The authors acknowledge these are distinct concepts and the variable has limited utility other than for controlling for systemic racism in the model.
Statistical Analyses
Descriptive statistics were used to describe distributional assumptions for all variables, including demographics, clinical indicators, colonoscopy results, and dietary patterns. Pairwise correlations between the total dietary pattern scores and food category scores were calculated with Pearson correlation (r).
Multinomial logistic regression models were created using SAS procedure LOGISTIC with the outcome of the categorical MSCF (no neoplasia, nonadvanced adenoma, or AN).34 A model was created for each independent predictor variable of interest (ie, the HEI, MD, or DASH percentage-standardized dietary pattern score and each food category comprising each dietary pattern score). All models were adjusted for age, sex, race/ethnicity, BMI, number of comorbidities, family history of CRC, number of follow-up colonoscopies, and estimated daily food-derived vitamin D intake. The demographic and clinical indicators were included in the models as they are known to be associated with CRC risk.18 The number of colonoscopies was included to control for surveillance intensity presuming risk for AN is reduced as polyps are removed. Because colonoscopy findings from an initial screening have unique clinical implications compared with follow- up and surveillance, MSCF was observed in 2 ways in sensitivity analyses: (1) baseline and (2) aggregate follow-up and surveillance only, excluding baseline findings.
Adjusted odds ratios (aORs) and 95% CIs for each of the MSCF outcomes with a reference finding of no neoplasia for the models are presented. We chose not to adjust for multiple comparisons across the different dietary patterns given the correlation between dietary pattern total and category scores but did adjust for multiple comparisons for dietary categories within each dietary pattern. Tests for statistical significance used α= .05 for the dietary pattern total scores and P values for the dietary category scores for each dietary pattern controlled for false discovery rate using the MULTTEST SAS procedure.35 All data manipulations and analyses were performed using SAS version 9.4.
Results
The study included 3023 patients. All were aged 50 to 75 years, 2923 (96.7%) were male and 2532 (83.8%) were non-Hispanic White (Table 1). Most participants were overweight or obese (n = 2535 [83.8%]), 2024 (67.0%) had ≤ 2 comorbidities, and 2602 (86.1%) had no family history of CRC. The MSCF for 1628 patients (53.9%) was no neoplasia, 966 patients (32.0%) was nonadvanced adenoma, and 429 participants (14.2%) had AN.

Mean percent scores were 58.5% for HEI, 38.2% for MD, and 63.1% for the DASH diet, with higher percentages indicating greater alignment with the recommendations for each diet (Table 2). All 3 dietary patterns scores standardized to percentages were strongly and significantly correlated in pairwise comparisons: HEI:MD, r = 0.62 (P < .001); HEI:DASH, r = 0.60 (P < .001); and MD:DASH, r = 0.72 (P < .001). Likewise, food category scores were significantly correlated across dietary patterns. For example, whole grain and fiber values from each dietary score were strongly correlated in pairwise comparisons: HEI Whole Grain:MD Grain, r = 0.64 (P < .001); HEI Whole Grain:DASH Fiber, r = 0.71 (P < .001); and MD Grain:DASH Fiber, r = 0.70 (P < .001).

Associations between individual participants' dietary pattern scores and the outcome of their pooled MSCF from baseline screening and ≥ 10 years of surveillance are presented in Table 3. For each single-point increases in dietary pattern scores (reflecting better dietary quality), aORs for nonadvanced adenoma vs no neoplasia were slightly lower but not statistically significantly: HEI, aOR, 1.00 (95% CI, 0.99-1.01); MD, aOR, 0.98 (95% CI, 0.94-1.02); and DASH, aOR, 0.99 (95% CI, 0.99-1.00). aORs for AN vs no neoplasia were slightly lower for each dietary pattern assessed, and only the MD and DASH scores were significantly different from 1.00: HEI, aOR, 1.00 (95% CI, 0.99-1.01); MD, aOR, 0.95 (95% CI, 0.90-1.00); and DASH, aOR, 0.99 (95% CI, 0.98-1.00).

We observed lower odds for nonadvanced adenoma and AN among all these dietary patterns when there was greater alignment with the recommended intake of whole grains and fiber. In separate models conducted using food categories comprising the dietary patterns as independent variables and after correcting for multiple tests, higher scores for the HEI Refined Grain category were associated with higher odds for nonadvanced adenoma (aOR, 1.03 [95% CI, 1.01-1.05]; P = .01) and AN (aOR, 1.05 [95% CI, 1.02-1.08]; P < .001). Higher scores for the HEI Whole Grain category were associated with lower odds for nonadvanced adenoma (aOR, 0.97 [95% CI, 0.95-0.99]; P = .01) and AN (aOR, 0.96 [95% CI, 0.93-0.99]; P = .01). Higher scores for the MD Grain category were significantly associated with lower odds for nonadvanced adenoma (aOR, 0.44 [95% CI, 0.26-0.75]; P = .002) and AN (aOR, 0.29 [95% CI, 0.14-0.62]; P = .001). The DASH Grains category also was significantly associated with lower odds for AN (aOR, 0.86 [95% CI, 0.78-0.95]; P = .002).
Discussion
In this study of 3023 veterans undergoing first-time screening colonoscopy and ≥ 10 years of surveillance, we found that healthy dietary patterns, as assessed by the MD and DASH diet, were significantly associated with lower risk of AN. Additionally, we identified lower odds for AN and nonadvanced adenoma compared with no neoplasia for higher grain scores for all the dietary patterns studied. Other food categories that comprise the dietary pattern scores had mixed associations with the MSCF outcomes. Several other studies have examined associations between dietary patterns and risk for CRC but to our knowledge, no studies have explored these associations among US veterans.
These results also indicate study participants had better than average (based on a 50% threshold) dietary quality according to the HEI and DASH diet scoring methods we used, but poor dietary quality according to the MD scoring method. The mean HEI scores for the present study were higher than a US Department of Agriculture study by Dong et al that compared dietary quality between veterans and nonveterans using the HEI, for which veterans’ expected HEI score was 45.6 of 100.8 This could be explained by the fact that the participants needed to be healthy to be eligible and those with healthier behaviors overall may have self-selected into the study due to motivation for screening during a time when screening was not yet commonplace. 36 Similarly, participants of the present study had higher adherence to the DASH diet (63.1%) than adolescents with diabetes in a study by Günther et al. Conversely, firefighters who were coached to use a Mediterranean-style dietary pattern and dietary had higher adherence to MD than did participants in this study.27
A closer examination of specific food category component scores that comprise the 3 distinct dietary patterns revealed mixed results from the multinomial modeling, which may have to do with the guideline thresholds used to calculate the dietary scores. When analyzed separately in the logistic regression models for their associations with nonadvanced adenomas and AN compared with no neoplasia, higher MD and DASH fruit scores (but not HEI fruit scores) were found to be significant. Other studies have had mixed findings when attempting to test for associations of fruit intake with adenoma recurrence.10,37
This study had some unexpected findings. Vegetable intake was not associated with nonadvanced adenomas or AN risk. Studies of food categories have consistently found vegetable (specifically cruciferous ones) intake to be linked with lower odds for cancers.38 Likewise, the red meat category, which was only a unique food category in the MD score, was not associated with nonadvanced adenomas or AN. Despite consistent literature suggesting higher intake of red meat and processed meats increases CRC risk, in 2019 the Nutritional Recommendations Consortium indicated that the evidence was weak.39,40 This study showed higher DASH diet scores for low-fat dairy, which were maximized when participants reported at least 50% of their dairy servings per day as being low-fat, had lower odds for AN. Yet, the MD scores for low-fat dairy had no association with either outcome; their calculation was based on total number of servings per week. This difference in findings suggests the fat intake ratio may be more relevant to CRC risk than intake quantity.
The literature is mixed regarding fatty acid intake and CRC risk, which may be relevant to both dairy and meat intake. One systematic review and meta-analysis found dietary fat and types of fatty acid intake had no association with CRC risk.41 However, a more recent meta-analysis that assessed both dietary intake and plasma levels of fatty acids did find some statistically significant differences for various types of fatty acids and CRC risk.42
The findings in the present study that grain intake is associated with lower odds for more severe colonoscopy findings among veterans are notable.43 Lieberman et al, using the CSP #380 data, found that cereal fiber intake was associated with a lower odds for AN compared with hyperplastic polyps (OR, 0.98 [95% CI, 0.96- 1.00]).18 Similarly, Hullings et al determined that older adults in the highest quintile of cereal fiber intake had significantly lower odds of CRC than those in lower odds for CRC when compared with lowest quintile (OR, 0.89 [95% CI, 0.83- 0.96]; P < .001).44 These findings support existing guidance that prioritizes whole grains as a key source of dietary fiber for CRC prevention.
A recent literature review on fiber, fat, and CRC risk suggested a consensus regarding one protective mechanism: dietary fiber from grains modulates the gut microbiota by promoting butyrate synthesis.45 Butyrate is a short-chain fatty acid that supports energy production in colonocytes and has tumor-suppressing properties.46 Our findings suggest there could be more to learn about the relationship between butyrate production and reduction of CRC risk through metabolomic studies that use measurements of plasma butyrate. These studies may examine associations between not just a singular food or food category, but rather food patterns that include fruits, vegetables, nuts and seeds, and whole grains known to promote butyrate production and plasma butyrate.47
Improved understanding of mechanisms and risk-modifying lifestyle factors such as dietary patterns may enhance prevention strategies. Identifying the collective chemopreventive characteristics of a specific dietary pattern (eg, MD) will be helpful to clinicians and health care staff to promote healthy eating to reduce cancer risk. More studies are needed to understand whether such promotion is more clinically applicable and effective for patients, as compared with eating more or less of specific foods (eg, more whole grains, less red meat). Furthermore, considering important environmental factors collectively beyond dietary patterns may offer a way to better tailor screening and implement a variety of lifestyle interventions. In the literature, this is often referred to as a teachable moment when patients’ attentions are captured and may position them to be more receptive to guidance.48
Limitations
This study has several important limitations and leaves opportunities for future studies that explore the role of dietary patterns and AN or CRC risk. First, the FFQ data used to calculate dietary pattern scores used in analysis were only captured at baseline, and there are nearly 3 decades across the study period. However, it is widely assumed that the diets of older adults, like those included in this study, remain stable over time which is appropriate given our sample population was aged 50 to 75 years when the baseline FFQ data were collected.49-51 Additionally, while the HEI is a well-documented, standard scoring method for dietary quality, there are multitudes of dietary pattern scoring approaches for MD and DASH.23,52,53 Finally, findings from this study using the sample of veterans may not be generalizable to a broader population. Future longitudinal studies that test for a clinically significant change threshold are warranted.
Conclusion
Results of this study suggest future research should further explore the effects of dietary patterns, particularly intake of specific food groups in combination, as opposed to individual nutrients or food items, on AN and CRC risk. Possible studies might explore these dietary patterns for their mechanistic role in altering the microbiome metabolism, which may influence CRC outcomes or include diet in a more comprehensive, holistic risk score that could be used to predict colonic neoplasia risk or in intervention studies that assess the effects of dietary changes on long-term CRC prevention. We suggest there are differences in people’s dietary intake patterns that might be important to consider when implementing tailored approaches to CRC risk mitigation.
- Zauber AG, Winawer SJ, O’Brien MJ, et al. Colonoscopic polypectomy and long-term prevention of colorectalcancer deaths. N Engl J Med. 2012;366(8):687-696. doi:10.1056/NEJMoa1100370
- Nishihara R, Wu K, Lochhead P, et al. Long-term colorectal-cancer incidence and mortality after lower endoscopy. N Engl J Med. 2013;369(12):1095-1105. doi:10.1056/NEJMoa1301969
- Bretthauer M, Løberg M, Wieszczy P, et al. Effect of colonoscopy screening on risks of colorectal cancer and related death. N Engl J Med. 2022;387(17):1547-1556. doi:10.1056/NEJMoa2208375
- Cottet V, Bonithon-Kopp C, Kronborg O, et al. Dietary patterns and the risk of colorectal adenoma recurrence in a European intervention trial. Eur J Cancer Prev. 2005;14(1):21.
- Miller PE, Lesko SM, Muscat JE, Lazarus P, Hartman TJ. Dietary patterns and colorectal adenoma and cancer risk: a review of the epidemiological evidence. Nutr Cancer. 2010;62(4):413-424. doi:10.1080/01635580903407114
- Godos J, Bella F, Torrisi A, Sciacca S, Galvano F, Grosso G. Dietary patterns and risk of colorectal adenoma: a systematic review and meta-analysis of observational studies. J Hum Nutr Diet Off J Br Diet Assoc. 2016;29(6):757-767. doi:10.1111/jhn.12395
- Haggar FA, Boushey RP. Colorectal cancer epidemiology: incidence, mortality, survival, and risk factors. Clin Colon Rectal Surg. 2009;22(4):191-197. doi:10.1055/s-0029-1242458
- Dong D, Stewart H, Carlson AC. An Examination of Veterans’ Diet Quality. U.S. Department of Agriculture, Economic Research Service; 2019:32.
- El-Halabi MM, Rex DK, Saito A, Eckert GJ, Kahi CJ. Defining adenoma detection rate benchmarks in average-risk male veterans. Gastrointest Endosc. 2019;89(1):137-143. doi:10.1016/j.gie.2018.08.021
- Alberts DS, Hess LM, eds. Fundamentals of Cancer Prevention. Springer International Publishing; 2019. doi:10.1007/978-3-030-15935-1
- Dahm CC, Keogh RH, Spencer EA, et al. Dietary fiber and colorectal cancer risk: a nested case-control study using food diaries. J Natl Cancer Inst. 2010;102(9):614-626. doi:10.1093/jnci/djq092
- Aune D, Lau R, Chan DSM, et al. Dairy products and colorectal cancer risk: a systematic review and metaanalysis of cohort studies. Ann Oncol. 2012;23(1):37-45. doi:10.1093/annonc/mdr269
- Lee JE, Li H, Chan AT, et al. Circulating levels of vitamin D and colon and rectal cancer: the Physicians’ Health Study and a meta-analysis of prospective studies. Cancer Prev Res Phila Pa. 2011;4(5):735-743. doi:10.1158/1940-6207.CAPR-10-0289
- Carroll C, Cooper K, Papaioannou D, Hind D, Pilgrim H, Tappenden P. Supplemental calcium in the chemoprevention of colorectal cancer: a systematic review and meta-analysis. Clin Ther. 2010;32(5):789-803. doi:10.1016/j.clinthera.2010.04.024
- Park Y, Spiegelman D, Hunter DJ, et al. Intakes of vitamins A, C, and E and use of multiple vitamin supplements and risk of colon cancer: a pooled analysis of prospective cohort studies. Cancer Causes Control CCC. 2010;21(11):1745- 1757. doi:10.1007/s10552-010-9549-y
- Alexander DD, Weed DL, Miller PE, Mohamed MA. Red meat and colorectal cancer: a quantitative update on the state of the epidemiologic science. J Am Coll Nutr. 2015;34(6):521-543. doi:10.1080/07315724.2014.992553
- Park SY, Wilkens LR, Setiawan VW, Monroe KR, Haiman CA, Le Marchand L. Alcohol intake and colorectal cancer risk in the multiethnic cohort study. Am J Epidemiol. 2019;188(1):67-76. doi:10.1093/aje/kwy208
- Lieberman DA. Risk Factors for advanced colonic neoplasia and hyperplastic polyps in asymptomatic individuals. JAMA. 2003;290(22):2959. doi:10.1001/jama.290.22.2959
- Archambault AN, Jeon J, Lin Y, et al. Risk stratification for early-onset colorectal cancer using a combination of genetic and environmental risk scores: an international multi-center study. J Natl Cancer Inst. 2022;114(4):528-539. doi:10.1093/jnci/djac003
- Carr PR, Weigl K, Edelmann D, et al. Estimation of absolute risk of colorectal cancer based on healthy lifestyle, genetic risk, and colonoscopy status in a populationbased study. Gastroenterology. 2020;159(1):129-138.e9. doi:10.1053/j.gastro.2020.03.016
- Sullivan BA, Qin X, Miller C, et al. Screening colonoscopy findings are associated with noncolorectal cancer mortality. Clin Transl Gastroenterol. 2022;13(4):e00479. doi:10.14309/ctg.0000000000000479
- Erben V, Carr PR, Holleczek B, Stegmaier C, Hoffmeister M, Brenner H. Dietary patterns and risk of advanced colorectal neoplasms: A large population based screening study in Germany. Prev Med. 2018;111:101-109. doi:10.1016/j.ypmed.2018.02.025
- Donovan MG, Selmin OI, Doetschman TC, Romagnolo DF. Mediterranean diet: prevention of colorectal cancer. Front Nutr. 2017;4:59. doi:10.3389/fnut.2017.00059
- Mohseni R, Mohseni F, Alizadeh S, Abbasi S. The Association of Dietary Approaches to Stop Hypertension (DASH) diet with the risk of colorectal cancer: a meta-analysis of observational studies.Nutr Cancer. 2020;72(5):778-790. doi:10.1080/01635581.2019.1651880
- Lieberman DA, Weiss DG, Bond JH, Ahnen DJ, Garewal H, Chejfec G. Use of colonoscopy to screen asymptomatic adults for colorectal cancer. Veterans Affairs Cooperative Study Group 380. N Engl J Med. 2000;343(3):162-168. doi:10.1056/NEJM200007203430301
- Developing the Healthy Eating Index (HEI) | EGRP/ DCCPS/NCI/NIH. Accessed July 22, 2025. https://epi.grants.cancer.gov/hei/developing.html#2015c
- Reeve E, Piccici F, Feairheller DL. Validation of a Mediterranean diet scoring system for intervention based research. J Nutr Med Diet Care. 2021;7(1):053. doi:10.23937/2572-3278/1510053
- Günther AL, Liese AD, Bell RA, et al. ASSOCIATION BETWEEN THE DIETARY APPROACHES TO HYPERTENSION (DASH) DIET AND HYPERTENSION IN YOUTH WITH DIABETES. Hypertens Dallas Tex 1979. 2009;53(1):6-12. doi:10.1161/HYPERTENSIONAHA.108.116665
- Buckland G, Agudo A, Luján L, et al. Adherence to a Mediterranean diet and risk of gastric adenocarcinoma within the European Prospective Investigation into Cancer and Nutrition (EPIC) cohort study. Am J Clin Nutr. 2010;91(2):381- 390. doi:10.3945/ajcn.2009.28209
- Rimm EB, Giovannucci EL, Stampfer MJ, Colditz GA, Litin LB, Willett WC. Reproducibility and validity of an expanded self-administered semiquantitative food frequency questionnaire among male health professionals. Am J Epidemiol. 1992;135(10):1114-1126. doi:10.1093/oxfordjournals.aje.a116211
- Charlson ME, Pompei P, Ales KL, MacKenzie CR. A new method of classifying prognostic comorbidity in longitudinal studies: development and validation. J Chronic Dis. 1987;40(5):373-383. doi:10.1016/0021-9681(87)90171-8
- Lieberman DA, Weiss DG, Harford WV, et al. Fiveyear colon surveillance after screening colonoscopy. Gastroenterology. 2007;133(4):1077-1085. doi:10.1053/j.gastro.2007.07.006
- Lieberman D, Sullivan BA, Hauser ER, et al. Baseline colonoscopy findings associated with 10-year outcomes in a screening cohort undergoing colonoscopy surveillance. Gastroenterology. 2020;158(4):862-874.e8. doi:10.1053/j.gastro.2019.07.052
- PROC LOGISTIC: PROC LOGISTIC Statement : SAS/STAT(R) 9.22 User’s Guide. Accessed July 22, 2025. https://support.sas.com/documentation/cdl/en/statug/63347/HTML/default/viewer.htm#statug_logistic_sect004.htm
- PROC MULTTEST: PROC MULTTEST Statement : SAS/ STAT(R) 9.22 User’s Guide. Accessed July 22, 2025. https://support.sas.com/documentation/cdl/en/statug/63347/HTML/default/viewer.htm#statug_multtest_sect005.htm
- Elston DM. Participation bias, self-selection bias, and response bias. J Am Acad Dermatol. Published online June 18, 2021. doi:10.1016/j.jaad.2021.06.025
- Sansbury LB, Wanke K, Albert PS, et al. The effect of strict adherence to a high-fiber, high-fruit and -vegetable, and low-fat eating pattern on adenoma recurrence. Am J Epidemiol. 2009;170(5):576-584. doi:10.1093/aje/kwp169
- Borgas P, Gonzalez G, Veselkov K, Mirnezami R. Phytochemically rich dietary components and the risk of colorectal cancer: A systematic review and meta-analysis of observational studies. World J Clin Oncol. 2021;12(6):482- 499. doi:10.5306/wjco.v12.i6.482
- Papadimitriou N, Markozannes G, Kanellopoulou A, et al. An umbrella review of the evidence associating diet and cancer risk at 11 anatomical sites. Nat Commun. 2021;12(1):4579. doi:10.1038/s41467-021-24861-8
- Johnston BC, Zeraatkar D, Han MA, et al. Unprocessed red meat and processed meat consumption: dietary guideline recommendations from the nutritional recommendations (NutriRECS) Consortium. Ann Intern Med. 2019;171(10):756-764. doi:10.7326/M19-1621
- Kim M, Park K. Dietary fat intake and risk of colorectal cancer: a systematic review and meta-analysis of prospective studies. Nutrients. 2018;10(12):1963. doi:10.3390/nu10121963
- Lu Y, Li D, Wang L, et al. Comprehensive investigation on associations between dietary intake and blood levels of fatty acids and colorectal cancer risk. Nutrients. 2023;15(3):730. doi:10.3390/nu15030730
- Gherasim A, Arhire LI, Ni.a O, Popa AD, Graur M, Mihalache L. The relationship between lifestyle components and dietary patterns. Proc Nutr Soc. 2020;79(3):311-323. doi:10.1017/S0029665120006898
- Hullings AG, Sinha R, Liao LM, Freedman ND, Graubard BI, Loftfield E. Whole grain and dietary fiber intake and risk of colorectal cancer in the NIH-AARP Diet and Health Study cohort. Am J Clin Nutr. 2020;112(3):603- 612. doi:10.1093/ajcn/nqaa161
- Ocvirk S, Wilson AS, Appolonia CN, Thomas TK, O’Keefe SJD. Fiber, fat, and colorectal cancer: new insight into modifiable dietary risk factors. Curr Gastroenterol Rep. 2019;21(11):62. doi:10.1007/s11894-019-0725-2
- O’Keefe SJD. Diet, microorganisms and their metabolites, and colon cancer. Nat Rev Gastroenterol Hepatol. 2016;13(12):691-706. doi:10.1038/nrgastro.2016.165
- The health benefits and side effects of Butyrate Cleveland Clinic. July 11, 2022. Accessed July 22, 2025. https://health.clevelandclinic.org/butyrate-benefits/
- Knudsen MD, Wang L, Wang K, et al. Changes in lifestyle factors after endoscopic screening: a prospective study in the United States. Clin Gastroenterol Hepatol Off ClinPract J Am Gastroenterol Assoc. 2022;20(6):e1240-e1249. doi:10.1016/j.cgh.2021.07.014
- Thorpe MG, Milte CM, Crawford D, McNaughton SA. Education and lifestyle predict change in dietary patterns and diet quality of adults 55 years and over. Nutr J. 2019;18(1):67. doi:10.1186/s12937-019-0495-6
- Chapman K, Ogden J. How do people change their diet?: an exploration into mechanisms of dietary change. J Health Psychol. 2009;14(8):1229-1242. doi:10.1177/1359105309342289
- Djoussé L, Petrone AB, Weir NL, et al. Repeated versus single measurement of plasma omega-3 fatty acids and risk of heart failure. Eur J Nutr. 2014;53(6):1403-1408. doi:10.1007/s00394-013-0642-3
- Bach-Faig A, Berry EM, Lairon D, et al. Mediterranean diet pyramid today. Science and cultural updates. Public Health Nutr. 2011;14(12A):2274-2284. doi:10.1017/S1368980011002515
- Miller PE, Cross AJ, Subar AF, et al. Comparison of 4 established DASH diet indexes: examining associations of index scores and colorectal cancer123. Am J Clin Nutr. 2013;98(3):794-803. doi:10.3945/ajcn.113.063602
- Krebs-Smith SM, Pannucci TE, Subar AF, et al. Update of the Healthy Eating Index: HEI-2015. J Acad Nutr Diet. 2018;118(9):1591-1602. doi:10.1016/j.jand.2018.05.021
- P.R. Pehrsson, Cutrufelli RL, Gebhardt SE, et al. USDA Database for the Added Sugars Content of Selected Foods. USDA; 2005. www.ars.usda.gov/nutrientdata
- Zauber AG, Winawer SJ, O’Brien MJ, et al. Colonoscopic polypectomy and long-term prevention of colorectalcancer deaths. N Engl J Med. 2012;366(8):687-696. doi:10.1056/NEJMoa1100370
- Nishihara R, Wu K, Lochhead P, et al. Long-term colorectal-cancer incidence and mortality after lower endoscopy. N Engl J Med. 2013;369(12):1095-1105. doi:10.1056/NEJMoa1301969
- Bretthauer M, Løberg M, Wieszczy P, et al. Effect of colonoscopy screening on risks of colorectal cancer and related death. N Engl J Med. 2022;387(17):1547-1556. doi:10.1056/NEJMoa2208375
- Cottet V, Bonithon-Kopp C, Kronborg O, et al. Dietary patterns and the risk of colorectal adenoma recurrence in a European intervention trial. Eur J Cancer Prev. 2005;14(1):21.
- Miller PE, Lesko SM, Muscat JE, Lazarus P, Hartman TJ. Dietary patterns and colorectal adenoma and cancer risk: a review of the epidemiological evidence. Nutr Cancer. 2010;62(4):413-424. doi:10.1080/01635580903407114
- Godos J, Bella F, Torrisi A, Sciacca S, Galvano F, Grosso G. Dietary patterns and risk of colorectal adenoma: a systematic review and meta-analysis of observational studies. J Hum Nutr Diet Off J Br Diet Assoc. 2016;29(6):757-767. doi:10.1111/jhn.12395
- Haggar FA, Boushey RP. Colorectal cancer epidemiology: incidence, mortality, survival, and risk factors. Clin Colon Rectal Surg. 2009;22(4):191-197. doi:10.1055/s-0029-1242458
- Dong D, Stewart H, Carlson AC. An Examination of Veterans’ Diet Quality. U.S. Department of Agriculture, Economic Research Service; 2019:32.
- El-Halabi MM, Rex DK, Saito A, Eckert GJ, Kahi CJ. Defining adenoma detection rate benchmarks in average-risk male veterans. Gastrointest Endosc. 2019;89(1):137-143. doi:10.1016/j.gie.2018.08.021
- Alberts DS, Hess LM, eds. Fundamentals of Cancer Prevention. Springer International Publishing; 2019. doi:10.1007/978-3-030-15935-1
- Dahm CC, Keogh RH, Spencer EA, et al. Dietary fiber and colorectal cancer risk: a nested case-control study using food diaries. J Natl Cancer Inst. 2010;102(9):614-626. doi:10.1093/jnci/djq092
- Aune D, Lau R, Chan DSM, et al. Dairy products and colorectal cancer risk: a systematic review and metaanalysis of cohort studies. Ann Oncol. 2012;23(1):37-45. doi:10.1093/annonc/mdr269
- Lee JE, Li H, Chan AT, et al. Circulating levels of vitamin D and colon and rectal cancer: the Physicians’ Health Study and a meta-analysis of prospective studies. Cancer Prev Res Phila Pa. 2011;4(5):735-743. doi:10.1158/1940-6207.CAPR-10-0289
- Carroll C, Cooper K, Papaioannou D, Hind D, Pilgrim H, Tappenden P. Supplemental calcium in the chemoprevention of colorectal cancer: a systematic review and meta-analysis. Clin Ther. 2010;32(5):789-803. doi:10.1016/j.clinthera.2010.04.024
- Park Y, Spiegelman D, Hunter DJ, et al. Intakes of vitamins A, C, and E and use of multiple vitamin supplements and risk of colon cancer: a pooled analysis of prospective cohort studies. Cancer Causes Control CCC. 2010;21(11):1745- 1757. doi:10.1007/s10552-010-9549-y
- Alexander DD, Weed DL, Miller PE, Mohamed MA. Red meat and colorectal cancer: a quantitative update on the state of the epidemiologic science. J Am Coll Nutr. 2015;34(6):521-543. doi:10.1080/07315724.2014.992553
- Park SY, Wilkens LR, Setiawan VW, Monroe KR, Haiman CA, Le Marchand L. Alcohol intake and colorectal cancer risk in the multiethnic cohort study. Am J Epidemiol. 2019;188(1):67-76. doi:10.1093/aje/kwy208
- Lieberman DA. Risk Factors for advanced colonic neoplasia and hyperplastic polyps in asymptomatic individuals. JAMA. 2003;290(22):2959. doi:10.1001/jama.290.22.2959
- Archambault AN, Jeon J, Lin Y, et al. Risk stratification for early-onset colorectal cancer using a combination of genetic and environmental risk scores: an international multi-center study. J Natl Cancer Inst. 2022;114(4):528-539. doi:10.1093/jnci/djac003
- Carr PR, Weigl K, Edelmann D, et al. Estimation of absolute risk of colorectal cancer based on healthy lifestyle, genetic risk, and colonoscopy status in a populationbased study. Gastroenterology. 2020;159(1):129-138.e9. doi:10.1053/j.gastro.2020.03.016
- Sullivan BA, Qin X, Miller C, et al. Screening colonoscopy findings are associated with noncolorectal cancer mortality. Clin Transl Gastroenterol. 2022;13(4):e00479. doi:10.14309/ctg.0000000000000479
- Erben V, Carr PR, Holleczek B, Stegmaier C, Hoffmeister M, Brenner H. Dietary patterns and risk of advanced colorectal neoplasms: A large population based screening study in Germany. Prev Med. 2018;111:101-109. doi:10.1016/j.ypmed.2018.02.025
- Donovan MG, Selmin OI, Doetschman TC, Romagnolo DF. Mediterranean diet: prevention of colorectal cancer. Front Nutr. 2017;4:59. doi:10.3389/fnut.2017.00059
- Mohseni R, Mohseni F, Alizadeh S, Abbasi S. The Association of Dietary Approaches to Stop Hypertension (DASH) diet with the risk of colorectal cancer: a meta-analysis of observational studies.Nutr Cancer. 2020;72(5):778-790. doi:10.1080/01635581.2019.1651880
- Lieberman DA, Weiss DG, Bond JH, Ahnen DJ, Garewal H, Chejfec G. Use of colonoscopy to screen asymptomatic adults for colorectal cancer. Veterans Affairs Cooperative Study Group 380. N Engl J Med. 2000;343(3):162-168. doi:10.1056/NEJM200007203430301
- Developing the Healthy Eating Index (HEI) | EGRP/ DCCPS/NCI/NIH. Accessed July 22, 2025. https://epi.grants.cancer.gov/hei/developing.html#2015c
- Reeve E, Piccici F, Feairheller DL. Validation of a Mediterranean diet scoring system for intervention based research. J Nutr Med Diet Care. 2021;7(1):053. doi:10.23937/2572-3278/1510053
- Günther AL, Liese AD, Bell RA, et al. ASSOCIATION BETWEEN THE DIETARY APPROACHES TO HYPERTENSION (DASH) DIET AND HYPERTENSION IN YOUTH WITH DIABETES. Hypertens Dallas Tex 1979. 2009;53(1):6-12. doi:10.1161/HYPERTENSIONAHA.108.116665
- Buckland G, Agudo A, Luján L, et al. Adherence to a Mediterranean diet and risk of gastric adenocarcinoma within the European Prospective Investigation into Cancer and Nutrition (EPIC) cohort study. Am J Clin Nutr. 2010;91(2):381- 390. doi:10.3945/ajcn.2009.28209
- Rimm EB, Giovannucci EL, Stampfer MJ, Colditz GA, Litin LB, Willett WC. Reproducibility and validity of an expanded self-administered semiquantitative food frequency questionnaire among male health professionals. Am J Epidemiol. 1992;135(10):1114-1126. doi:10.1093/oxfordjournals.aje.a116211
- Charlson ME, Pompei P, Ales KL, MacKenzie CR. A new method of classifying prognostic comorbidity in longitudinal studies: development and validation. J Chronic Dis. 1987;40(5):373-383. doi:10.1016/0021-9681(87)90171-8
- Lieberman DA, Weiss DG, Harford WV, et al. Fiveyear colon surveillance after screening colonoscopy. Gastroenterology. 2007;133(4):1077-1085. doi:10.1053/j.gastro.2007.07.006
- Lieberman D, Sullivan BA, Hauser ER, et al. Baseline colonoscopy findings associated with 10-year outcomes in a screening cohort undergoing colonoscopy surveillance. Gastroenterology. 2020;158(4):862-874.e8. doi:10.1053/j.gastro.2019.07.052
- PROC LOGISTIC: PROC LOGISTIC Statement : SAS/STAT(R) 9.22 User’s Guide. Accessed July 22, 2025. https://support.sas.com/documentation/cdl/en/statug/63347/HTML/default/viewer.htm#statug_logistic_sect004.htm
- PROC MULTTEST: PROC MULTTEST Statement : SAS/ STAT(R) 9.22 User’s Guide. Accessed July 22, 2025. https://support.sas.com/documentation/cdl/en/statug/63347/HTML/default/viewer.htm#statug_multtest_sect005.htm
- Elston DM. Participation bias, self-selection bias, and response bias. J Am Acad Dermatol. Published online June 18, 2021. doi:10.1016/j.jaad.2021.06.025
- Sansbury LB, Wanke K, Albert PS, et al. The effect of strict adherence to a high-fiber, high-fruit and -vegetable, and low-fat eating pattern on adenoma recurrence. Am J Epidemiol. 2009;170(5):576-584. doi:10.1093/aje/kwp169
- Borgas P, Gonzalez G, Veselkov K, Mirnezami R. Phytochemically rich dietary components and the risk of colorectal cancer: A systematic review and meta-analysis of observational studies. World J Clin Oncol. 2021;12(6):482- 499. doi:10.5306/wjco.v12.i6.482
- Papadimitriou N, Markozannes G, Kanellopoulou A, et al. An umbrella review of the evidence associating diet and cancer risk at 11 anatomical sites. Nat Commun. 2021;12(1):4579. doi:10.1038/s41467-021-24861-8
- Johnston BC, Zeraatkar D, Han MA, et al. Unprocessed red meat and processed meat consumption: dietary guideline recommendations from the nutritional recommendations (NutriRECS) Consortium. Ann Intern Med. 2019;171(10):756-764. doi:10.7326/M19-1621
- Kim M, Park K. Dietary fat intake and risk of colorectal cancer: a systematic review and meta-analysis of prospective studies. Nutrients. 2018;10(12):1963. doi:10.3390/nu10121963
- Lu Y, Li D, Wang L, et al. Comprehensive investigation on associations between dietary intake and blood levels of fatty acids and colorectal cancer risk. Nutrients. 2023;15(3):730. doi:10.3390/nu15030730
- Gherasim A, Arhire LI, Ni.a O, Popa AD, Graur M, Mihalache L. The relationship between lifestyle components and dietary patterns. Proc Nutr Soc. 2020;79(3):311-323. doi:10.1017/S0029665120006898
- Hullings AG, Sinha R, Liao LM, Freedman ND, Graubard BI, Loftfield E. Whole grain and dietary fiber intake and risk of colorectal cancer in the NIH-AARP Diet and Health Study cohort. Am J Clin Nutr. 2020;112(3):603- 612. doi:10.1093/ajcn/nqaa161
- Ocvirk S, Wilson AS, Appolonia CN, Thomas TK, O’Keefe SJD. Fiber, fat, and colorectal cancer: new insight into modifiable dietary risk factors. Curr Gastroenterol Rep. 2019;21(11):62. doi:10.1007/s11894-019-0725-2
- O’Keefe SJD. Diet, microorganisms and their metabolites, and colon cancer. Nat Rev Gastroenterol Hepatol. 2016;13(12):691-706. doi:10.1038/nrgastro.2016.165
- The health benefits and side effects of Butyrate Cleveland Clinic. July 11, 2022. Accessed July 22, 2025. https://health.clevelandclinic.org/butyrate-benefits/
- Knudsen MD, Wang L, Wang K, et al. Changes in lifestyle factors after endoscopic screening: a prospective study in the United States. Clin Gastroenterol Hepatol Off ClinPract J Am Gastroenterol Assoc. 2022;20(6):e1240-e1249. doi:10.1016/j.cgh.2021.07.014
- Thorpe MG, Milte CM, Crawford D, McNaughton SA. Education and lifestyle predict change in dietary patterns and diet quality of adults 55 years and over. Nutr J. 2019;18(1):67. doi:10.1186/s12937-019-0495-6
- Chapman K, Ogden J. How do people change their diet?: an exploration into mechanisms of dietary change. J Health Psychol. 2009;14(8):1229-1242. doi:10.1177/1359105309342289
- Djoussé L, Petrone AB, Weir NL, et al. Repeated versus single measurement of plasma omega-3 fatty acids and risk of heart failure. Eur J Nutr. 2014;53(6):1403-1408. doi:10.1007/s00394-013-0642-3
- Bach-Faig A, Berry EM, Lairon D, et al. Mediterranean diet pyramid today. Science and cultural updates. Public Health Nutr. 2011;14(12A):2274-2284. doi:10.1017/S1368980011002515
- Miller PE, Cross AJ, Subar AF, et al. Comparison of 4 established DASH diet indexes: examining associations of index scores and colorectal cancer123. Am J Clin Nutr. 2013;98(3):794-803. doi:10.3945/ajcn.113.063602
- Krebs-Smith SM, Pannucci TE, Subar AF, et al. Update of the Healthy Eating Index: HEI-2015. J Acad Nutr Diet. 2018;118(9):1591-1602. doi:10.1016/j.jand.2018.05.021
- P.R. Pehrsson, Cutrufelli RL, Gebhardt SE, et al. USDA Database for the Added Sugars Content of Selected Foods. USDA; 2005. www.ars.usda.gov/nutrientdata
Associations Between Prescreening Dietary Patterns and Longitudinal Colonoscopy Outcomes in Veterans
Associations Between Prescreening Dietary Patterns and Longitudinal Colonoscopy Outcomes in Veterans
Clinical Outcomes of Stevens-Johnson Syndrome and Toxic Epidermal Necrolysis Based on Hospital Admission Type
Clinical Outcomes of Stevens-Johnson Syndrome and Toxic Epidermal Necrolysis Based on Hospital Admission Type
Stevens-Johnson syndrome (SJS) and toxic epidermal necrolysis (TEN) are rare, life-threatening conditions that involve widespread necrosis of the skin and mucous membranes.1 Guidelines for SJS and TEN recommend management in hospitals with access to inpatient dermatology to provide immediate interventions that are necessary for achieving optimal patient outcomes.2 A delay in admission of 5 days or more after onset of symptoms has been associated with increases in overall mortality, bacteremia, intensive care unit (ICU) admission, and length of stay.3 Patients who are not directly admitted to specialized facilities and require transfer from other hospitals may experience delays in receiving critical interventions, further increasing the risk for mortality and complications. In this study, we analyzed the clinical outcomes of patients with SJS/TEN in relation to their admission pathway.
Methods
A single-center retrospective chart review was performed at Atrium Health Wake Forest Baptist Medical Center (AHWFBMC) in Winston-Salem, North Carolina. Participants were identified using i2b2, an informatics tool compliant with the Health Insurance Portability and Accountability Act for integrating biology and the bedside. Inclusion criteria were having a diagnosis of SJS (International Classification of Diseases, Tenth Revision, code L51.1; International Classification of Diseases, Ninth Revision, code 695.13), TEN (International Classification of Diseases, Tenth Revision, code L51.2; International Classification of Diseases, Ninth Revision, code 695.15) or Lyell syndrome from January 2012 to December 2024. Patients with erythema multiforme or bullous drug eruption were excluded, as these conditions initially were misdiagnosed as SJS or TEN. Patients with only a reported history of prior SJS or TEN also were excluded.
The following clinical outcomes were assessed: demographics, comorbidities, age at disease onset, outside hospital transfer status, complications during admission, inpatient length of stay in days, age of mortality (if applicable), culprit medications, interventions received, Severity-of-Illness Score for Toxic Epidermal Necrolysis (SCORTEN) upon admission, site of admission (eg, floor bed, ICU, medical ICU, burn unit), and length of disease process prior to hospital admission. Patients then were categorized as either direct or transfer admissions based on the initial point of care and admission process. Direct admissions included patients who presented to the AHWFBMC emergency department and were subsequently admitted. Transfer patients included patients who initially presented to an outside hospital and were transferred to AHWFBMC. Data regarding the wait time for Physician Access Line requests and the time elapsed from the initial transfer call to arrival at the tertiary hospital also were collected—this is a method that outside hospitals can use to contact physicians at the tertiary hospital for a possible transfer. Statistical analysis was performed using unpaired t tests and X2 tests as necessary using GraphPad By Dotmatics Prism.
Results
A total of 112 patients were included in the analysis; of these, 71 had a diagnosis with biopsy confirmation of SJS, SJS/TEN overlap, or TEN (Table 1). Forty-one patients were excluded due to having a diagnosis of erythema multiforme or bullous drug eruption or a reported history of prior SJS or TEN without hospitalization. All biopsies were performed at AHWFBMC. Of the 71 confirmed patients with SJS/TEN, 54 (76%) were female with a mean age of 44 years. The majority of patients identified as Black (35 [49%]) or White (27 [38%]), along with Asian (7 [10%]) and other (2 [3%]). The most common comorbidity was cardiovascular disease in 42 (59%) patients, followed by type 2 diabetes in 36 (51%) patients. Among these 71 patients with SJS/TEN, 29 (41%) were directly admitted to the tertiary hospital, while 42 (59%) were transferred from outside hospitals.

Of the 71 confirmed patients with SJS/TEN, sulfonamides were identified as the most common inciting drug in 25 (41%) patients, followed by beta-lactam antibiotics in 16 (23%) patients (Table 2). This is consistent with previous literature of sulfamethoxazole with trimethoprim as the primary causative drug for SJS and TEN in the United States.1

Clinical Outcomes—Of the 71 patients, there were 23 (32%) cases of SJS, 29 (41%) cases of SJS/TEN overlap, and 19 (27%) cases of TEN (eTable). The initial and maximum affected body surface area (BSA) was higher in transfer admissions, with a mean maximum BSA of 38.55% in the transfer group compared to 19.14% in the direct admissions. The mean SCORTEN (range, 0-5) was 1.6 overall, with a higher mean score of 1.92 in the transfer group compared to 1.07 in the direct admissions.

Transfer patients had a longer mean stay at the tertiary hospital (13.71 d) compared to direct admissions (7.17 d). The mean time from symptom onset until tertiary hospital admission was 8.5 days; transfer and direct admission patients had similar mean time from symptom onset of 9.02 days and 7.86 days, respectively. Although the duration of cutaneous symptoms from onset until tertiary hospital admission was similar (P=.283) between direct admissions (7.86 d) and transfer patients (9.02 d), the transfer group presented with greater disease severity at the time of admission. Transfer patients had a higher mean maximum BSA involvement (38.55% vs 19.14% [P=.005]), elevated SCORTEN (1.92 vs 1.07 [P=.029]), and longer mean hospital stays (13.71 d vs 7.17 d [P<.0001]) compared to direct admissions.
Despite the absence of mortality in both groups, transfer patients showed a higher number of ICU admissions (19 vs 5 [P=.014]) and burn unit admissions (9 vs 2 [P=.096]), bacteremia (16 vs 4 [P=.025]), acute kidney injury (13 vs 10 [P=.755]), acute respiratory failure (12 vs 5 [P=.272]), and transaminitis (8 vs 3 [P=.319]).
Outside Hospital Treatments—All outside hospitals provided supportive care with intravenous fluids and acetaminophen; however, further care provided at outside hospitals varied (Table 3), with transfer patients most frequently being treated with diphenhydramine (69% [29/42]), antimicrobial medications (57% [24/42]), steroids (40%), and epinephrine (10% [4/42]). Some patients may have received more than one of these treatments. Based on outside hospital treatments, the primary care teams’ main clinical concerns were allergic reactions and infection, as 33 (79%) patients received diphenhydramine (29 [89%]) or epinephrine (4 [12%]) and 24 (52%) received antimicrobial medications. Of the 42 transfer patients, 24 (57%) received or continued these medications before transfer; the medications were promptly discontinued upon tertiary hospital admission.

Once the outside hospitals contacted the tertiary hospital for a referral, the mean length of time between the transfer request and Physician Access Line call was 17.13 minutes (Table 4). Following the transfer request, the mean length of time for arrival at the tertiary hospital was 6.22 hours. The mean length of stay at the outside hospital prior to the patient being transferred was 3.84 days.

Comment
This retrospective study examined 71 patients with biopsy-confirmed SJS, SJS/TEN overlap, or TEN to evaluate differences in clinical outcomes between direct and transfer admissions. Transfer patients had a higher mean maximum affected BSA (38.55% vs 19.14% [P=.005]) and elevated SCORTEN (1.92 vs 1.07 [P=.029]); a higher number of transfer patients were admitted to the ICU (19 vs 5 [P=.014]) and burn unit (9 vs 2 [P=.096]), and this group also demonstrated longer hospitalization stays (13.71 vs 7.17 [P<.0001]). There were more complications among transfer patients, including bacteremia (16 vs 4 [P=.025]), which is consistent with findings from the existing literature.3
Once the decision for transfer of the patients included in our study was initiated and accepted, there was a prompt response and transfer of care; the mean length of time for Physician Access Line request was 17.13 minutes, and the mean transfer time to arrive at the tertiary hospital was 6.22 hours; however, patients spent an average of 3.84 days at outside hospitals, reflecting that transfer calls frequently were initiated due to urgent clinical decline of the patient rather than as an early intervention strategy. The management at outside hospitals often included the continuation of antimicrobial medications, which were discontinued upon transfer to AHWFBMC. Causative agents were either previously prescribed for a new medical condition or initiated for the management of suspected infections at outside hospitals. This may reflect the difficulty in correctly diagnosing SJS/TEN and initiating appropriate management at hospital facilities without an inpatient dermatologist.
The presence of inpatient dermatologists can improve the diagnostic accuracy and treatment of various conditions.4,5 Dermatology consultations added or changed 77% of treatment plans for 271 hospitalized patients.4 The impact of this intervention is reflected by the success of early dermatology consultations in reducing the length of hospitalization and use of inappropriate treatments in the care of skin diseases.6-8
Access to dermatologic care has been an identified need in inpatient hospitals that may limit the ability of hospitals to promptly treat serious conditions such as SJS/TEN.9 From an inpatient dermatology study from 2013 through 2019, 98.2% of 782 inpatient dermatologists reside in metropolitan areas, limiting the availability of care for rural patients; this study also found a decreasing number of facilities with inpatient dermatologists.10
The limitations of our study include a small sample size of 71 patients, which restricted the generalizability of our results. Our study also was based at a single tertiary center, which thereby limited the findings to this geographic area. It also was difficult to match patients by their demographic and comorbid conditions. The retrospective study design depended on the accuracy and completeness of medical records, which can introduce information bias. Future studies should compare the clinical outcomes of SJS/TEN based on burn unit and ICU admissions.
Conclusion
Prompt identification of SJS/TEN and rapid transfer to hospitals with inpatient dermatology are essential to optimize patient outcomes. Developing and validating SJS/TEN diagnosis and transfer protocols across multiple institutions may be helpful.
- Kridin K, Brüggen MC, Chua SL, et al. Assessment of treatment approaches and outcomes in Stevens-Johnson syndrome and toxic epidermal necrolysis: insights from a pan-European multicenter study. JAMA Dermatol. 2021;157:1182-1190. doi:10.1001/jamadermatol.2021.3154
- Seminario-Vidal L, Kroshinsky D, Malachowski SJ, et al. Society of Dermatology Hospitalists supportive care guidelines for the management of Stevens-Johnson syndrome/toxic epidermal necrolysis in adults. J Am Acad Dermatol. 2020;82:1553-1567. doi:10.1016 /j.jaad.2020.02.066
- Clark AE, Fook-Chong S, Choo K, et al. Delayed admission to a specialist referral center for Stevens-Johnson syndrome and toxic epidermal necrolysis is associated with increased mortality: a retrospective cohort study. JAAD Int. 2021;4:10-12. doi:10.1016/j.jdin.2021.03.008
- Davila M, Christenson LJ, Sontheimer RD. Epidemiology and outcomes of dermatology in-patient consultations in a Midwestern U.S. university hospital. Dermatol Online J. 2010;16:12.
- Hu L, Haynes H, Ferrazza D, et al. Impact of specialist consultations on inpatient admissions for dermatology-specific and related DRGs. J Gen Intern Med. 2013;28:1477-1482. doi:10.1007/s11606-013-2440-2
- Harr T, French LE. Toxic epidermal necrolysis and Stevens-Johnson syndrome. Orphanet J Rare Dis. 2010;5:39. doi:10.1186/1750-1172-5-39
- Li DG, Xia FD, Khosravi H, et al. Outcomes of early dermatology consultation for inpatients diagnosed with cellulitis. JAMA Dermatol. 2018;154:537-543. doi:10.1001/jamadermatol.2017.6197
- Milani-Nejad N, Zhang M, Kaffenberger BH. Association of dermatology consultations with patient care outcomes in hospitalized patients with inflammatory skin diseases. JAMA Dermatol. 2017;153:523-528. doi:10.1001/jamadermatol.2016.6130
- Messenger E, Kovarik CL, Lipoff JB. Access to inpatient dermatology care in Pennsylvania hospitals. Cutis. 2016;97:49-51.
- Hydol-Smith JA, Gallardo MA, Korman A, et al. The United States dermatology inpatient workforce between 2013 and 2019: a Medicare analysis reveals contraction of the workforce and vast access desertsa cross-sectional analysis. Arch Dermatol Res. 2024;316:103. doi:10.1007 /s00403-024-02845-0
Stevens-Johnson syndrome (SJS) and toxic epidermal necrolysis (TEN) are rare, life-threatening conditions that involve widespread necrosis of the skin and mucous membranes.1 Guidelines for SJS and TEN recommend management in hospitals with access to inpatient dermatology to provide immediate interventions that are necessary for achieving optimal patient outcomes.2 A delay in admission of 5 days or more after onset of symptoms has been associated with increases in overall mortality, bacteremia, intensive care unit (ICU) admission, and length of stay.3 Patients who are not directly admitted to specialized facilities and require transfer from other hospitals may experience delays in receiving critical interventions, further increasing the risk for mortality and complications. In this study, we analyzed the clinical outcomes of patients with SJS/TEN in relation to their admission pathway.
Methods
A single-center retrospective chart review was performed at Atrium Health Wake Forest Baptist Medical Center (AHWFBMC) in Winston-Salem, North Carolina. Participants were identified using i2b2, an informatics tool compliant with the Health Insurance Portability and Accountability Act for integrating biology and the bedside. Inclusion criteria were having a diagnosis of SJS (International Classification of Diseases, Tenth Revision, code L51.1; International Classification of Diseases, Ninth Revision, code 695.13), TEN (International Classification of Diseases, Tenth Revision, code L51.2; International Classification of Diseases, Ninth Revision, code 695.15) or Lyell syndrome from January 2012 to December 2024. Patients with erythema multiforme or bullous drug eruption were excluded, as these conditions initially were misdiagnosed as SJS or TEN. Patients with only a reported history of prior SJS or TEN also were excluded.
The following clinical outcomes were assessed: demographics, comorbidities, age at disease onset, outside hospital transfer status, complications during admission, inpatient length of stay in days, age of mortality (if applicable), culprit medications, interventions received, Severity-of-Illness Score for Toxic Epidermal Necrolysis (SCORTEN) upon admission, site of admission (eg, floor bed, ICU, medical ICU, burn unit), and length of disease process prior to hospital admission. Patients then were categorized as either direct or transfer admissions based on the initial point of care and admission process. Direct admissions included patients who presented to the AHWFBMC emergency department and were subsequently admitted. Transfer patients included patients who initially presented to an outside hospital and were transferred to AHWFBMC. Data regarding the wait time for Physician Access Line requests and the time elapsed from the initial transfer call to arrival at the tertiary hospital also were collected—this is a method that outside hospitals can use to contact physicians at the tertiary hospital for a possible transfer. Statistical analysis was performed using unpaired t tests and X2 tests as necessary using GraphPad By Dotmatics Prism.
Results
A total of 112 patients were included in the analysis; of these, 71 had a diagnosis with biopsy confirmation of SJS, SJS/TEN overlap, or TEN (Table 1). Forty-one patients were excluded due to having a diagnosis of erythema multiforme or bullous drug eruption or a reported history of prior SJS or TEN without hospitalization. All biopsies were performed at AHWFBMC. Of the 71 confirmed patients with SJS/TEN, 54 (76%) were female with a mean age of 44 years. The majority of patients identified as Black (35 [49%]) or White (27 [38%]), along with Asian (7 [10%]) and other (2 [3%]). The most common comorbidity was cardiovascular disease in 42 (59%) patients, followed by type 2 diabetes in 36 (51%) patients. Among these 71 patients with SJS/TEN, 29 (41%) were directly admitted to the tertiary hospital, while 42 (59%) were transferred from outside hospitals.

Of the 71 confirmed patients with SJS/TEN, sulfonamides were identified as the most common inciting drug in 25 (41%) patients, followed by beta-lactam antibiotics in 16 (23%) patients (Table 2). This is consistent with previous literature of sulfamethoxazole with trimethoprim as the primary causative drug for SJS and TEN in the United States.1

Clinical Outcomes—Of the 71 patients, there were 23 (32%) cases of SJS, 29 (41%) cases of SJS/TEN overlap, and 19 (27%) cases of TEN (eTable). The initial and maximum affected body surface area (BSA) was higher in transfer admissions, with a mean maximum BSA of 38.55% in the transfer group compared to 19.14% in the direct admissions. The mean SCORTEN (range, 0-5) was 1.6 overall, with a higher mean score of 1.92 in the transfer group compared to 1.07 in the direct admissions.

Transfer patients had a longer mean stay at the tertiary hospital (13.71 d) compared to direct admissions (7.17 d). The mean time from symptom onset until tertiary hospital admission was 8.5 days; transfer and direct admission patients had similar mean time from symptom onset of 9.02 days and 7.86 days, respectively. Although the duration of cutaneous symptoms from onset until tertiary hospital admission was similar (P=.283) between direct admissions (7.86 d) and transfer patients (9.02 d), the transfer group presented with greater disease severity at the time of admission. Transfer patients had a higher mean maximum BSA involvement (38.55% vs 19.14% [P=.005]), elevated SCORTEN (1.92 vs 1.07 [P=.029]), and longer mean hospital stays (13.71 d vs 7.17 d [P<.0001]) compared to direct admissions.
Despite the absence of mortality in both groups, transfer patients showed a higher number of ICU admissions (19 vs 5 [P=.014]) and burn unit admissions (9 vs 2 [P=.096]), bacteremia (16 vs 4 [P=.025]), acute kidney injury (13 vs 10 [P=.755]), acute respiratory failure (12 vs 5 [P=.272]), and transaminitis (8 vs 3 [P=.319]).
Outside Hospital Treatments—All outside hospitals provided supportive care with intravenous fluids and acetaminophen; however, further care provided at outside hospitals varied (Table 3), with transfer patients most frequently being treated with diphenhydramine (69% [29/42]), antimicrobial medications (57% [24/42]), steroids (40%), and epinephrine (10% [4/42]). Some patients may have received more than one of these treatments. Based on outside hospital treatments, the primary care teams’ main clinical concerns were allergic reactions and infection, as 33 (79%) patients received diphenhydramine (29 [89%]) or epinephrine (4 [12%]) and 24 (52%) received antimicrobial medications. Of the 42 transfer patients, 24 (57%) received or continued these medications before transfer; the medications were promptly discontinued upon tertiary hospital admission.

Once the outside hospitals contacted the tertiary hospital for a referral, the mean length of time between the transfer request and Physician Access Line call was 17.13 minutes (Table 4). Following the transfer request, the mean length of time for arrival at the tertiary hospital was 6.22 hours. The mean length of stay at the outside hospital prior to the patient being transferred was 3.84 days.

Comment
This retrospective study examined 71 patients with biopsy-confirmed SJS, SJS/TEN overlap, or TEN to evaluate differences in clinical outcomes between direct and transfer admissions. Transfer patients had a higher mean maximum affected BSA (38.55% vs 19.14% [P=.005]) and elevated SCORTEN (1.92 vs 1.07 [P=.029]); a higher number of transfer patients were admitted to the ICU (19 vs 5 [P=.014]) and burn unit (9 vs 2 [P=.096]), and this group also demonstrated longer hospitalization stays (13.71 vs 7.17 [P<.0001]). There were more complications among transfer patients, including bacteremia (16 vs 4 [P=.025]), which is consistent with findings from the existing literature.3
Once the decision for transfer of the patients included in our study was initiated and accepted, there was a prompt response and transfer of care; the mean length of time for Physician Access Line request was 17.13 minutes, and the mean transfer time to arrive at the tertiary hospital was 6.22 hours; however, patients spent an average of 3.84 days at outside hospitals, reflecting that transfer calls frequently were initiated due to urgent clinical decline of the patient rather than as an early intervention strategy. The management at outside hospitals often included the continuation of antimicrobial medications, which were discontinued upon transfer to AHWFBMC. Causative agents were either previously prescribed for a new medical condition or initiated for the management of suspected infections at outside hospitals. This may reflect the difficulty in correctly diagnosing SJS/TEN and initiating appropriate management at hospital facilities without an inpatient dermatologist.
The presence of inpatient dermatologists can improve the diagnostic accuracy and treatment of various conditions.4,5 Dermatology consultations added or changed 77% of treatment plans for 271 hospitalized patients.4 The impact of this intervention is reflected by the success of early dermatology consultations in reducing the length of hospitalization and use of inappropriate treatments in the care of skin diseases.6-8
Access to dermatologic care has been an identified need in inpatient hospitals that may limit the ability of hospitals to promptly treat serious conditions such as SJS/TEN.9 From an inpatient dermatology study from 2013 through 2019, 98.2% of 782 inpatient dermatologists reside in metropolitan areas, limiting the availability of care for rural patients; this study also found a decreasing number of facilities with inpatient dermatologists.10
The limitations of our study include a small sample size of 71 patients, which restricted the generalizability of our results. Our study also was based at a single tertiary center, which thereby limited the findings to this geographic area. It also was difficult to match patients by their demographic and comorbid conditions. The retrospective study design depended on the accuracy and completeness of medical records, which can introduce information bias. Future studies should compare the clinical outcomes of SJS/TEN based on burn unit and ICU admissions.
Conclusion
Prompt identification of SJS/TEN and rapid transfer to hospitals with inpatient dermatology are essential to optimize patient outcomes. Developing and validating SJS/TEN diagnosis and transfer protocols across multiple institutions may be helpful.
Stevens-Johnson syndrome (SJS) and toxic epidermal necrolysis (TEN) are rare, life-threatening conditions that involve widespread necrosis of the skin and mucous membranes.1 Guidelines for SJS and TEN recommend management in hospitals with access to inpatient dermatology to provide immediate interventions that are necessary for achieving optimal patient outcomes.2 A delay in admission of 5 days or more after onset of symptoms has been associated with increases in overall mortality, bacteremia, intensive care unit (ICU) admission, and length of stay.3 Patients who are not directly admitted to specialized facilities and require transfer from other hospitals may experience delays in receiving critical interventions, further increasing the risk for mortality and complications. In this study, we analyzed the clinical outcomes of patients with SJS/TEN in relation to their admission pathway.
Methods
A single-center retrospective chart review was performed at Atrium Health Wake Forest Baptist Medical Center (AHWFBMC) in Winston-Salem, North Carolina. Participants were identified using i2b2, an informatics tool compliant with the Health Insurance Portability and Accountability Act for integrating biology and the bedside. Inclusion criteria were having a diagnosis of SJS (International Classification of Diseases, Tenth Revision, code L51.1; International Classification of Diseases, Ninth Revision, code 695.13), TEN (International Classification of Diseases, Tenth Revision, code L51.2; International Classification of Diseases, Ninth Revision, code 695.15) or Lyell syndrome from January 2012 to December 2024. Patients with erythema multiforme or bullous drug eruption were excluded, as these conditions initially were misdiagnosed as SJS or TEN. Patients with only a reported history of prior SJS or TEN also were excluded.
The following clinical outcomes were assessed: demographics, comorbidities, age at disease onset, outside hospital transfer status, complications during admission, inpatient length of stay in days, age of mortality (if applicable), culprit medications, interventions received, Severity-of-Illness Score for Toxic Epidermal Necrolysis (SCORTEN) upon admission, site of admission (eg, floor bed, ICU, medical ICU, burn unit), and length of disease process prior to hospital admission. Patients then were categorized as either direct or transfer admissions based on the initial point of care and admission process. Direct admissions included patients who presented to the AHWFBMC emergency department and were subsequently admitted. Transfer patients included patients who initially presented to an outside hospital and were transferred to AHWFBMC. Data regarding the wait time for Physician Access Line requests and the time elapsed from the initial transfer call to arrival at the tertiary hospital also were collected—this is a method that outside hospitals can use to contact physicians at the tertiary hospital for a possible transfer. Statistical analysis was performed using unpaired t tests and X2 tests as necessary using GraphPad By Dotmatics Prism.
Results
A total of 112 patients were included in the analysis; of these, 71 had a diagnosis with biopsy confirmation of SJS, SJS/TEN overlap, or TEN (Table 1). Forty-one patients were excluded due to having a diagnosis of erythema multiforme or bullous drug eruption or a reported history of prior SJS or TEN without hospitalization. All biopsies were performed at AHWFBMC. Of the 71 confirmed patients with SJS/TEN, 54 (76%) were female with a mean age of 44 years. The majority of patients identified as Black (35 [49%]) or White (27 [38%]), along with Asian (7 [10%]) and other (2 [3%]). The most common comorbidity was cardiovascular disease in 42 (59%) patients, followed by type 2 diabetes in 36 (51%) patients. Among these 71 patients with SJS/TEN, 29 (41%) were directly admitted to the tertiary hospital, while 42 (59%) were transferred from outside hospitals.

Of the 71 confirmed patients with SJS/TEN, sulfonamides were identified as the most common inciting drug in 25 (41%) patients, followed by beta-lactam antibiotics in 16 (23%) patients (Table 2). This is consistent with previous literature of sulfamethoxazole with trimethoprim as the primary causative drug for SJS and TEN in the United States.1

Clinical Outcomes—Of the 71 patients, there were 23 (32%) cases of SJS, 29 (41%) cases of SJS/TEN overlap, and 19 (27%) cases of TEN (eTable). The initial and maximum affected body surface area (BSA) was higher in transfer admissions, with a mean maximum BSA of 38.55% in the transfer group compared to 19.14% in the direct admissions. The mean SCORTEN (range, 0-5) was 1.6 overall, with a higher mean score of 1.92 in the transfer group compared to 1.07 in the direct admissions.

Transfer patients had a longer mean stay at the tertiary hospital (13.71 d) compared to direct admissions (7.17 d). The mean time from symptom onset until tertiary hospital admission was 8.5 days; transfer and direct admission patients had similar mean time from symptom onset of 9.02 days and 7.86 days, respectively. Although the duration of cutaneous symptoms from onset until tertiary hospital admission was similar (P=.283) between direct admissions (7.86 d) and transfer patients (9.02 d), the transfer group presented with greater disease severity at the time of admission. Transfer patients had a higher mean maximum BSA involvement (38.55% vs 19.14% [P=.005]), elevated SCORTEN (1.92 vs 1.07 [P=.029]), and longer mean hospital stays (13.71 d vs 7.17 d [P<.0001]) compared to direct admissions.
Despite the absence of mortality in both groups, transfer patients showed a higher number of ICU admissions (19 vs 5 [P=.014]) and burn unit admissions (9 vs 2 [P=.096]), bacteremia (16 vs 4 [P=.025]), acute kidney injury (13 vs 10 [P=.755]), acute respiratory failure (12 vs 5 [P=.272]), and transaminitis (8 vs 3 [P=.319]).
Outside Hospital Treatments—All outside hospitals provided supportive care with intravenous fluids and acetaminophen; however, further care provided at outside hospitals varied (Table 3), with transfer patients most frequently being treated with diphenhydramine (69% [29/42]), antimicrobial medications (57% [24/42]), steroids (40%), and epinephrine (10% [4/42]). Some patients may have received more than one of these treatments. Based on outside hospital treatments, the primary care teams’ main clinical concerns were allergic reactions and infection, as 33 (79%) patients received diphenhydramine (29 [89%]) or epinephrine (4 [12%]) and 24 (52%) received antimicrobial medications. Of the 42 transfer patients, 24 (57%) received or continued these medications before transfer; the medications were promptly discontinued upon tertiary hospital admission.

Once the outside hospitals contacted the tertiary hospital for a referral, the mean length of time between the transfer request and Physician Access Line call was 17.13 minutes (Table 4). Following the transfer request, the mean length of time for arrival at the tertiary hospital was 6.22 hours. The mean length of stay at the outside hospital prior to the patient being transferred was 3.84 days.

Comment
This retrospective study examined 71 patients with biopsy-confirmed SJS, SJS/TEN overlap, or TEN to evaluate differences in clinical outcomes between direct and transfer admissions. Transfer patients had a higher mean maximum affected BSA (38.55% vs 19.14% [P=.005]) and elevated SCORTEN (1.92 vs 1.07 [P=.029]); a higher number of transfer patients were admitted to the ICU (19 vs 5 [P=.014]) and burn unit (9 vs 2 [P=.096]), and this group also demonstrated longer hospitalization stays (13.71 vs 7.17 [P<.0001]). There were more complications among transfer patients, including bacteremia (16 vs 4 [P=.025]), which is consistent with findings from the existing literature.3
Once the decision for transfer of the patients included in our study was initiated and accepted, there was a prompt response and transfer of care; the mean length of time for Physician Access Line request was 17.13 minutes, and the mean transfer time to arrive at the tertiary hospital was 6.22 hours; however, patients spent an average of 3.84 days at outside hospitals, reflecting that transfer calls frequently were initiated due to urgent clinical decline of the patient rather than as an early intervention strategy. The management at outside hospitals often included the continuation of antimicrobial medications, which were discontinued upon transfer to AHWFBMC. Causative agents were either previously prescribed for a new medical condition or initiated for the management of suspected infections at outside hospitals. This may reflect the difficulty in correctly diagnosing SJS/TEN and initiating appropriate management at hospital facilities without an inpatient dermatologist.
The presence of inpatient dermatologists can improve the diagnostic accuracy and treatment of various conditions.4,5 Dermatology consultations added or changed 77% of treatment plans for 271 hospitalized patients.4 The impact of this intervention is reflected by the success of early dermatology consultations in reducing the length of hospitalization and use of inappropriate treatments in the care of skin diseases.6-8
Access to dermatologic care has been an identified need in inpatient hospitals that may limit the ability of hospitals to promptly treat serious conditions such as SJS/TEN.9 From an inpatient dermatology study from 2013 through 2019, 98.2% of 782 inpatient dermatologists reside in metropolitan areas, limiting the availability of care for rural patients; this study also found a decreasing number of facilities with inpatient dermatologists.10
The limitations of our study include a small sample size of 71 patients, which restricted the generalizability of our results. Our study also was based at a single tertiary center, which thereby limited the findings to this geographic area. It also was difficult to match patients by their demographic and comorbid conditions. The retrospective study design depended on the accuracy and completeness of medical records, which can introduce information bias. Future studies should compare the clinical outcomes of SJS/TEN based on burn unit and ICU admissions.
Conclusion
Prompt identification of SJS/TEN and rapid transfer to hospitals with inpatient dermatology are essential to optimize patient outcomes. Developing and validating SJS/TEN diagnosis and transfer protocols across multiple institutions may be helpful.
- Kridin K, Brüggen MC, Chua SL, et al. Assessment of treatment approaches and outcomes in Stevens-Johnson syndrome and toxic epidermal necrolysis: insights from a pan-European multicenter study. JAMA Dermatol. 2021;157:1182-1190. doi:10.1001/jamadermatol.2021.3154
- Seminario-Vidal L, Kroshinsky D, Malachowski SJ, et al. Society of Dermatology Hospitalists supportive care guidelines for the management of Stevens-Johnson syndrome/toxic epidermal necrolysis in adults. J Am Acad Dermatol. 2020;82:1553-1567. doi:10.1016 /j.jaad.2020.02.066
- Clark AE, Fook-Chong S, Choo K, et al. Delayed admission to a specialist referral center for Stevens-Johnson syndrome and toxic epidermal necrolysis is associated with increased mortality: a retrospective cohort study. JAAD Int. 2021;4:10-12. doi:10.1016/j.jdin.2021.03.008
- Davila M, Christenson LJ, Sontheimer RD. Epidemiology and outcomes of dermatology in-patient consultations in a Midwestern U.S. university hospital. Dermatol Online J. 2010;16:12.
- Hu L, Haynes H, Ferrazza D, et al. Impact of specialist consultations on inpatient admissions for dermatology-specific and related DRGs. J Gen Intern Med. 2013;28:1477-1482. doi:10.1007/s11606-013-2440-2
- Harr T, French LE. Toxic epidermal necrolysis and Stevens-Johnson syndrome. Orphanet J Rare Dis. 2010;5:39. doi:10.1186/1750-1172-5-39
- Li DG, Xia FD, Khosravi H, et al. Outcomes of early dermatology consultation for inpatients diagnosed with cellulitis. JAMA Dermatol. 2018;154:537-543. doi:10.1001/jamadermatol.2017.6197
- Milani-Nejad N, Zhang M, Kaffenberger BH. Association of dermatology consultations with patient care outcomes in hospitalized patients with inflammatory skin diseases. JAMA Dermatol. 2017;153:523-528. doi:10.1001/jamadermatol.2016.6130
- Messenger E, Kovarik CL, Lipoff JB. Access to inpatient dermatology care in Pennsylvania hospitals. Cutis. 2016;97:49-51.
- Hydol-Smith JA, Gallardo MA, Korman A, et al. The United States dermatology inpatient workforce between 2013 and 2019: a Medicare analysis reveals contraction of the workforce and vast access desertsa cross-sectional analysis. Arch Dermatol Res. 2024;316:103. doi:10.1007 /s00403-024-02845-0
- Kridin K, Brüggen MC, Chua SL, et al. Assessment of treatment approaches and outcomes in Stevens-Johnson syndrome and toxic epidermal necrolysis: insights from a pan-European multicenter study. JAMA Dermatol. 2021;157:1182-1190. doi:10.1001/jamadermatol.2021.3154
- Seminario-Vidal L, Kroshinsky D, Malachowski SJ, et al. Society of Dermatology Hospitalists supportive care guidelines for the management of Stevens-Johnson syndrome/toxic epidermal necrolysis in adults. J Am Acad Dermatol. 2020;82:1553-1567. doi:10.1016 /j.jaad.2020.02.066
- Clark AE, Fook-Chong S, Choo K, et al. Delayed admission to a specialist referral center for Stevens-Johnson syndrome and toxic epidermal necrolysis is associated with increased mortality: a retrospective cohort study. JAAD Int. 2021;4:10-12. doi:10.1016/j.jdin.2021.03.008
- Davila M, Christenson LJ, Sontheimer RD. Epidemiology and outcomes of dermatology in-patient consultations in a Midwestern U.S. university hospital. Dermatol Online J. 2010;16:12.
- Hu L, Haynes H, Ferrazza D, et al. Impact of specialist consultations on inpatient admissions for dermatology-specific and related DRGs. J Gen Intern Med. 2013;28:1477-1482. doi:10.1007/s11606-013-2440-2
- Harr T, French LE. Toxic epidermal necrolysis and Stevens-Johnson syndrome. Orphanet J Rare Dis. 2010;5:39. doi:10.1186/1750-1172-5-39
- Li DG, Xia FD, Khosravi H, et al. Outcomes of early dermatology consultation for inpatients diagnosed with cellulitis. JAMA Dermatol. 2018;154:537-543. doi:10.1001/jamadermatol.2017.6197
- Milani-Nejad N, Zhang M, Kaffenberger BH. Association of dermatology consultations with patient care outcomes in hospitalized patients with inflammatory skin diseases. JAMA Dermatol. 2017;153:523-528. doi:10.1001/jamadermatol.2016.6130
- Messenger E, Kovarik CL, Lipoff JB. Access to inpatient dermatology care in Pennsylvania hospitals. Cutis. 2016;97:49-51.
- Hydol-Smith JA, Gallardo MA, Korman A, et al. The United States dermatology inpatient workforce between 2013 and 2019: a Medicare analysis reveals contraction of the workforce and vast access desertsa cross-sectional analysis. Arch Dermatol Res. 2024;316:103. doi:10.1007 /s00403-024-02845-0
Clinical Outcomes of Stevens-Johnson Syndrome and Toxic Epidermal Necrolysis Based on Hospital Admission Type
Clinical Outcomes of Stevens-Johnson Syndrome and Toxic Epidermal Necrolysis Based on Hospital Admission Type
PRACTICE POINTS
- Early identification and diagnosis of Stevens-Johnson syndrome and toxic epidermal necrolysis are essential to improving patient outcomes.
- Patients transferred from outside hospitals often present with more severe disease due to delays in diagnosis and initiation of appropriate treatment.
- Inpatient dermatology consultation plays a vital role in accurately diagnosing and managing life-threatening dermatologic conditions.
- Establishing timely interhospital transfer protocols may help expedite access to specialized treatment and improve patient outcomes.
Impact of Rapid Blood Culture Identification on Antibiotic De-escalation at a Veterans Affairs Medical Center
Impact of Rapid Blood Culture Identification on Antibiotic De-escalation at a Veterans Affairs Medical Center
About 530,000 to 628,000 episodes of bloodstream infections (BSI) occur annually in the US.1 Early identification and treatment of bacteremia are essential to improve patient outcomes because it allows for more timely targeted antibiotic therapy.2 Organism identification and susceptibility testing can take 2 to 5 days, prolonging the use of broad-spectrum empiric antibiotics and increasing the risk of adverse events.3,4 The Infectious Disease Society of America recommends the use of rapid diagnostic testing and antimicrobial stewardship programs (ASPs) to improve rates of antibiotic susceptibilities to targeted antibiotics and optimize resource utilization.3 Rapid blood culture identification (BCID) technologies reduce the duration of empiric antibiotics in patients with contaminated blood cultures, resulting in shorter hospital stays and saving money per each patient tested.4
In March 2023, Veteran Health Indiana (VHI) implemented the BioFire FilmArray Blood Culture Identification (BCID2), a BSI panel test that identifies select gram-negative bacteria, gram-positive bacteria, yeast, and antimicrobial resistance genes with an aggregate sensitivity of 99% and a specificity of 99.8%. The BCID2 presents clinically relevant information faster than traditional culture methods, allowing clinicians to make more efficient and educated antibiotic regimen decisions than with previous methods.5
It takes 24 to 48 hours from blood collection for culture incubation, positivity, and gram staining to occur at VHI. If the gram stain is positive, the blood culture is placed on the BioFire BCID2 in addition to traditional culture medium. BioFire BCID2 results are ready in 45 to 60 minutes. Results are uploaded into the electronic health record (EHR) ≤ 2 hours after they are obtained and the primary team is notified if the test is positive for certain critical results. Susceptibility testing of an identified organism typically requires an additional 24 to 48 hours for finalization. VHI Infectious Disease created an evidence-based antibiotic recommendation chart for certain medication(s) and alternate therapies based on the reported organism and its interpreted presence of resistance markers (eg, ceftriaxone for Escherichia coli when extended-spectrum beta lactamases are not detected vs meropenem if extended-spectrum beta lactamases marker are present). These charts optimize the antibiotic regimen while awaiting susceptibility finalizations.
Two previous studies describe the impact of rapid diagnostic testing technology at US Department of Veterans Affairs (VA) medical centers.6,7 In Texas, the ASP reviewed BCID panel results via clinical decision support software for about 1 hour per day.6 A Los Angeles study analyzed the impact of Biofire BCID with an interpretation guide centered on unnecessary vancomycin use and determined that shorter duration of the medication may have been the result of more frequent infectious disease consultation.7
This study assessed the time to optimal antibiotic de-escalation before and after the implementation of BioFire BCID2 with results reviewed by the ASP without active notification or assistance of any clinical decision support technology. The primary objective was to evaluate difference in time to optimal antibiotics from blood culture draw pre- vs postintervention. Secondary objectives included differences in time to organism identification, difference in time on broad-spectrum antibiotics, and difference in time to appropriate antibiotics.
Methods
This quasi-experimental retrospective chart review assessed the impact of BioFire BCID2 use on timely antibiotic de-escalation for patients who experienced a BSI at VHI between March 1, 2022, and October 1, 2023. Microbiology laboratory records identified eligible patients with positive blood cultures within the study time frame. Data were collected from the VHI EHR.
Patients were included if they had a positive bacterial blood culture and received ≥ 1 antibiotic indicated for bacteremia while receiving inpatient care. Patients were excluded if they died prior to blood culture results, transferred out of VHI, left against medical advice, or had untreated contaminants in blood culture results (ie, never received antibiotics aimed at the contaminated culture).
Patient lists were generated for before and after implementation of BioFire BCID2 (pre- and postintervention) using the VHI EHR and microbiology laboratory record system. The pre- and postinterventions groups were different sizes. As a result, a random sampling of the preintervention group was selected and included patients from March 1, 2022, through March 26, 2023. The postintervention group was smaller due to time constraints between initiation of BioFire BCID2 for data collection and included all patients from March 27, 2023, through October 1, 2023.
Optimal antibiotics were defined as escalation from inappropriate therapy to broader agent(s), de-escalation from broad-spectrum therapy to targeted agent(s), discontinuation of therapy due to an organism being identified as a contaminant, or optimization of a regimen to the preferred antimicrobial agent based on evidence-based consensus guidelines. Broad-spectrum antibiotics included: piperacillin/tazobactam, cefepime, ceftazidime, ceftazidime-avibactam, cefiderocol, carbapenems, fluroquinolones, vancomycin, daptomycin, ceftaroline, linezolid, or aztreonam. Appropriate antibiotics were defined as those with activity toward the final identified organism(s).
Deidentified participant data were entered into Microsoft Excel and kept on a secure VA server to complete statistical analyses. Parametric continuous data, such as age, were analyzed using the t-test, while nonparametric continuous data, such as time to optimal antibiotics, were analyzed using the Mann-Whitney U test. Categorical data, like sex and race, were analyzed using either Fisher exact test for small sample sizes or X2 test for a larger sample size. Statistical significance levels was defined as P < .05.
Results
Using patient lists drawn from the EHR and the microbiology laboratory records, 110 electronic charts were randomly selected for review. Fifteen patients were excluded: 8 had untreated contaminants, 4 died, and 3 were transferred out of VHI. Of the 95 patients included, 48 were in the preintervention group and 47 were in the postintervention group (Figure 1).

Baseline characteristics were similar between the 2 groups (Table 1). Most patients were White males aged > 70 years in the EHR. The urinary tract was the most common source of infection, impacting 12 patients in each group (Figure 2). Escherichia coli, Klebsiella, Staphylococcus, and Streptococcus were the most common bloodstream isolates identified.


The median time to optimal antibiotics in the preintervention group was 58.5 hours vs 43.4 hours in the postintervention group (P = .11). The median time to organism identification was 37.8 hours in the preintervention group vs 16.9 hours in the postintervention group (P < .001). The median time on broad-spectrum antibiotics was 45.2 hours in the preintervention group vs 46.6 hours in the postintervention group (P = .99). The median time on appropriate antibiotics in the preintervention group was 2.3 hours vs 1.9 hours in the postintervention group (P = .79). Differences in other measured outcomes between the groups were not statistically significant (Table 2).

Although implementation of rapid diagnostic technology reduced the median time to optimal antibiotics, the results were not statistically significant. Shorter time to organism identification in the postintervention group compared to the preintervention group was the lone statistically significant metric (P < .001).
Discussion
A lack of statistical significance in the primary outcome may have been due to nonadherence to facility de-escalation protocols or a suboptimal BioFire BCID2 result notification system. Additionally, use of rapid BCID at VHI may improve over time as clinicians become more familiar with the technology. Gaps in clinical pharmacy coverage during the night shift may have also contributed to delays in antibiotic optimization, particularly if other clinicians are not equipped with the knowledge or training to appropriately deescalate antibiotics based on microorganisms identified. A 2017 study by Donner et al concluded that physician interpretation of BCID results is suboptimal and should be augmented with clinical decision support tools as new technology becomes available.8 Despite the statistically insignificant results of this study, it did highlight potential areas of improvement which can lead to improved patient care.
Previous research has evaluated the impact of rapid BCID technology on antibiotic treatment and clinical outcomes. Chiasson et al found that median time to optimal therapy was 73.8 hours in the pre-BCID arm compared to 34.7 hours in the post- BCID arm (P ≤ .001), emphasizing the importance of combining rapid BCID with clinical decision support tools and pharmacy input.6 Senok et al found that BCID2 implementation led to a significant decrease in median time to culture result, which informed optimal antibiotic therapy and decreased 30-day mortality in the intensive care setting.9 In contrast, the current study did not stratify patients according to medical ward or illness severity even though clinicians may be less likely to de-escalate antibiotic therapy in critically ill patients.
Bae et al reported findings consistent with the current study and concluded that BCID did not affect the clinical outcomes of overall BSIs; however, it contributed to early administration of effective antibiotics in cases of BSIs caused by multidrug-resistant organisms.10 Results of this study were not stratified according to multidrug-resistant organisms because the sample size was too small. The current study also included patients with polymicrobial infections, which may have impacted the results due to a less streamlined approach to antibiotic optimization.
Limitations
This single-center, retrospective study had a small sample size, short time frame, and lacked patient diversity, and therefore may not be generalizable to other health care systems. The sample size was limited by shorter date range and smaller patient list between BioFire BCID2 implementation and data collection, which was used to determine the number of charts selected in each group. Some patients received antibiotics prior to blood cultures being drawn, which may falsely decrease time to optimal/ appropriate antibiotics and falsely increase time on broad spectrum/any antibiotics due to early antibiotic administration. The total number of patients on broad-spectrum antibiotics differed from the total number of patients for other outcomes because several patients never received the defined broad spectrum antibiotics.
Conclusions
When combined with a pre-existing ASP without active notification, the implementation of BioFire BCID2 did not return statistically significant data showing a decrease in time to optimal antibiotics, time to appropriate antibiotics, or time on broad-spectrum antibiotics at VHI. To make this program more successful, pharmacist intervention and clinical decision support tools may be needed.
Additional research is required to determine the optimal integration of antimicrobial stewardship, rapid diagnostic technology, and pharmacy services for maximum benefit. Even though the primary outcome was not statistically significant, the results may be clinically significant from a stewardship perspective. Realigning microbiology workflows to mimic other research, which emphasizes the importance of funneling rapid BCID results through the ASP, may improve outcomes. Future studies may be warranted following the implementation of clinical decision support tools to assess their impact on stewardship practices and patient outcomes.
- Goto M, Al-Hasan MN. Overall burden of bloodstream infection and nosocomial bloodstream infection in North America and Europe. Clin Microbiol Infect. 2013;19(6):501- 509. doi:10.1111/1469-0691.12195
- Pardo J, Klinker KP, Borgert SJ, Butler BM, Giglio PG, Rand KH. Clinical and economic impact of antimicrobial stewardship interventions with the FilmArray blood culture identification panel. Diagn Microbiol Infect Dis. 2016;84(2):159-164. doi:10.1016/j.diagmicrobio.2015.10.023.
- Barlam TF, Cosgrove SE, Abbo LM, et al. Implementing an antibiotic stewardship program: guidelines by the Infectious Diseases Society of America and the Society for Healthcare Epidemiology of America. Clin Infect Dis. 2016;62(10):e51-e77. doi:10.1093/cid/ciw118
- BIOFIRE® Blood Culture Identification 2 (BCID2) Panel. Biomerierux. Updated 2025. Accessed May 10, 2025. https://www.biofiredx.com/products/the-filmarray-panels/filmarraybcid/
- Huang AM, Newton D, Kunapuli A, et al. Impact of rapid organism identification via matrix-assisted laser desorption/ionization time-of-flight combined with antimicrobial stewardship team intervention in adult patients with bacteremia and candidemia. Clin Infect Dis. 2013;57(9):1237-1245. doi:10.1093/cid/cit498
- Chiasson JM, Smith WJ, Jodlowski TZ, Kouma MA, Cutrell JB. Impact of a rapid blood culture diagnostic panel on time to optimal antimicrobial therapy at a veterans affairs medical center. J Pharm Pract. 2022;35(5):722-729. doi:10.1177/08971900211000686
- Wu S, Watson RL, Graber CJ. 2007. Impact of combining rapid diagnostics with an interpretation guide on vancomycin usage for contaminant blood cultures growing coagulase- negative staphylococci (CoNS). Open Forum Infect Dis. 2019;6(Suppl 2):S674. doi:10.1093/ofid/ofz360.1687
- Donner LM, Campbell WS, Lyden E, Van Schooneveld TC. Assessment of rapid-blood-culture-identification result interpretation and antibiotic prescribing practices. J Clin Microbiol. 2017;55(5):1496-1507. doi:10.1128/JCM.02395-16
- Senok A, Dabal LA, Alfaresi M, et al. Clinical impact of the BIOFIRE blood culture identification 2 panel in adult patients with bloodstream infection: a multicentre observational study in the United Arab Emirates. Diagnostics (Basel). 2023;13(14):2433. doi:10.3390/diagnostics13142433
- Bae JY, Bae J, So MK, Choi HJ, Lee M. The impact of the rapid blood culture identification panel on antibiotic treatment and clinical outcomes in bloodstream infections, particularly those associated with multidrug-resistant micro-organisms. Diagnostics (Basel). 2023;13(23):3504. doi:10.3390/diagnostics13233504
About 530,000 to 628,000 episodes of bloodstream infections (BSI) occur annually in the US.1 Early identification and treatment of bacteremia are essential to improve patient outcomes because it allows for more timely targeted antibiotic therapy.2 Organism identification and susceptibility testing can take 2 to 5 days, prolonging the use of broad-spectrum empiric antibiotics and increasing the risk of adverse events.3,4 The Infectious Disease Society of America recommends the use of rapid diagnostic testing and antimicrobial stewardship programs (ASPs) to improve rates of antibiotic susceptibilities to targeted antibiotics and optimize resource utilization.3 Rapid blood culture identification (BCID) technologies reduce the duration of empiric antibiotics in patients with contaminated blood cultures, resulting in shorter hospital stays and saving money per each patient tested.4
In March 2023, Veteran Health Indiana (VHI) implemented the BioFire FilmArray Blood Culture Identification (BCID2), a BSI panel test that identifies select gram-negative bacteria, gram-positive bacteria, yeast, and antimicrobial resistance genes with an aggregate sensitivity of 99% and a specificity of 99.8%. The BCID2 presents clinically relevant information faster than traditional culture methods, allowing clinicians to make more efficient and educated antibiotic regimen decisions than with previous methods.5
It takes 24 to 48 hours from blood collection for culture incubation, positivity, and gram staining to occur at VHI. If the gram stain is positive, the blood culture is placed on the BioFire BCID2 in addition to traditional culture medium. BioFire BCID2 results are ready in 45 to 60 minutes. Results are uploaded into the electronic health record (EHR) ≤ 2 hours after they are obtained and the primary team is notified if the test is positive for certain critical results. Susceptibility testing of an identified organism typically requires an additional 24 to 48 hours for finalization. VHI Infectious Disease created an evidence-based antibiotic recommendation chart for certain medication(s) and alternate therapies based on the reported organism and its interpreted presence of resistance markers (eg, ceftriaxone for Escherichia coli when extended-spectrum beta lactamases are not detected vs meropenem if extended-spectrum beta lactamases marker are present). These charts optimize the antibiotic regimen while awaiting susceptibility finalizations.
Two previous studies describe the impact of rapid diagnostic testing technology at US Department of Veterans Affairs (VA) medical centers.6,7 In Texas, the ASP reviewed BCID panel results via clinical decision support software for about 1 hour per day.6 A Los Angeles study analyzed the impact of Biofire BCID with an interpretation guide centered on unnecessary vancomycin use and determined that shorter duration of the medication may have been the result of more frequent infectious disease consultation.7
This study assessed the time to optimal antibiotic de-escalation before and after the implementation of BioFire BCID2 with results reviewed by the ASP without active notification or assistance of any clinical decision support technology. The primary objective was to evaluate difference in time to optimal antibiotics from blood culture draw pre- vs postintervention. Secondary objectives included differences in time to organism identification, difference in time on broad-spectrum antibiotics, and difference in time to appropriate antibiotics.
Methods
This quasi-experimental retrospective chart review assessed the impact of BioFire BCID2 use on timely antibiotic de-escalation for patients who experienced a BSI at VHI between March 1, 2022, and October 1, 2023. Microbiology laboratory records identified eligible patients with positive blood cultures within the study time frame. Data were collected from the VHI EHR.
Patients were included if they had a positive bacterial blood culture and received ≥ 1 antibiotic indicated for bacteremia while receiving inpatient care. Patients were excluded if they died prior to blood culture results, transferred out of VHI, left against medical advice, or had untreated contaminants in blood culture results (ie, never received antibiotics aimed at the contaminated culture).
Patient lists were generated for before and after implementation of BioFire BCID2 (pre- and postintervention) using the VHI EHR and microbiology laboratory record system. The pre- and postinterventions groups were different sizes. As a result, a random sampling of the preintervention group was selected and included patients from March 1, 2022, through March 26, 2023. The postintervention group was smaller due to time constraints between initiation of BioFire BCID2 for data collection and included all patients from March 27, 2023, through October 1, 2023.
Optimal antibiotics were defined as escalation from inappropriate therapy to broader agent(s), de-escalation from broad-spectrum therapy to targeted agent(s), discontinuation of therapy due to an organism being identified as a contaminant, or optimization of a regimen to the preferred antimicrobial agent based on evidence-based consensus guidelines. Broad-spectrum antibiotics included: piperacillin/tazobactam, cefepime, ceftazidime, ceftazidime-avibactam, cefiderocol, carbapenems, fluroquinolones, vancomycin, daptomycin, ceftaroline, linezolid, or aztreonam. Appropriate antibiotics were defined as those with activity toward the final identified organism(s).
Deidentified participant data were entered into Microsoft Excel and kept on a secure VA server to complete statistical analyses. Parametric continuous data, such as age, were analyzed using the t-test, while nonparametric continuous data, such as time to optimal antibiotics, were analyzed using the Mann-Whitney U test. Categorical data, like sex and race, were analyzed using either Fisher exact test for small sample sizes or X2 test for a larger sample size. Statistical significance levels was defined as P < .05.
Results
Using patient lists drawn from the EHR and the microbiology laboratory records, 110 electronic charts were randomly selected for review. Fifteen patients were excluded: 8 had untreated contaminants, 4 died, and 3 were transferred out of VHI. Of the 95 patients included, 48 were in the preintervention group and 47 were in the postintervention group (Figure 1).

Baseline characteristics were similar between the 2 groups (Table 1). Most patients were White males aged > 70 years in the EHR. The urinary tract was the most common source of infection, impacting 12 patients in each group (Figure 2). Escherichia coli, Klebsiella, Staphylococcus, and Streptococcus were the most common bloodstream isolates identified.


The median time to optimal antibiotics in the preintervention group was 58.5 hours vs 43.4 hours in the postintervention group (P = .11). The median time to organism identification was 37.8 hours in the preintervention group vs 16.9 hours in the postintervention group (P < .001). The median time on broad-spectrum antibiotics was 45.2 hours in the preintervention group vs 46.6 hours in the postintervention group (P = .99). The median time on appropriate antibiotics in the preintervention group was 2.3 hours vs 1.9 hours in the postintervention group (P = .79). Differences in other measured outcomes between the groups were not statistically significant (Table 2).

Although implementation of rapid diagnostic technology reduced the median time to optimal antibiotics, the results were not statistically significant. Shorter time to organism identification in the postintervention group compared to the preintervention group was the lone statistically significant metric (P < .001).
Discussion
A lack of statistical significance in the primary outcome may have been due to nonadherence to facility de-escalation protocols or a suboptimal BioFire BCID2 result notification system. Additionally, use of rapid BCID at VHI may improve over time as clinicians become more familiar with the technology. Gaps in clinical pharmacy coverage during the night shift may have also contributed to delays in antibiotic optimization, particularly if other clinicians are not equipped with the knowledge or training to appropriately deescalate antibiotics based on microorganisms identified. A 2017 study by Donner et al concluded that physician interpretation of BCID results is suboptimal and should be augmented with clinical decision support tools as new technology becomes available.8 Despite the statistically insignificant results of this study, it did highlight potential areas of improvement which can lead to improved patient care.
Previous research has evaluated the impact of rapid BCID technology on antibiotic treatment and clinical outcomes. Chiasson et al found that median time to optimal therapy was 73.8 hours in the pre-BCID arm compared to 34.7 hours in the post- BCID arm (P ≤ .001), emphasizing the importance of combining rapid BCID with clinical decision support tools and pharmacy input.6 Senok et al found that BCID2 implementation led to a significant decrease in median time to culture result, which informed optimal antibiotic therapy and decreased 30-day mortality in the intensive care setting.9 In contrast, the current study did not stratify patients according to medical ward or illness severity even though clinicians may be less likely to de-escalate antibiotic therapy in critically ill patients.
Bae et al reported findings consistent with the current study and concluded that BCID did not affect the clinical outcomes of overall BSIs; however, it contributed to early administration of effective antibiotics in cases of BSIs caused by multidrug-resistant organisms.10 Results of this study were not stratified according to multidrug-resistant organisms because the sample size was too small. The current study also included patients with polymicrobial infections, which may have impacted the results due to a less streamlined approach to antibiotic optimization.
Limitations
This single-center, retrospective study had a small sample size, short time frame, and lacked patient diversity, and therefore may not be generalizable to other health care systems. The sample size was limited by shorter date range and smaller patient list between BioFire BCID2 implementation and data collection, which was used to determine the number of charts selected in each group. Some patients received antibiotics prior to blood cultures being drawn, which may falsely decrease time to optimal/ appropriate antibiotics and falsely increase time on broad spectrum/any antibiotics due to early antibiotic administration. The total number of patients on broad-spectrum antibiotics differed from the total number of patients for other outcomes because several patients never received the defined broad spectrum antibiotics.
Conclusions
When combined with a pre-existing ASP without active notification, the implementation of BioFire BCID2 did not return statistically significant data showing a decrease in time to optimal antibiotics, time to appropriate antibiotics, or time on broad-spectrum antibiotics at VHI. To make this program more successful, pharmacist intervention and clinical decision support tools may be needed.
Additional research is required to determine the optimal integration of antimicrobial stewardship, rapid diagnostic technology, and pharmacy services for maximum benefit. Even though the primary outcome was not statistically significant, the results may be clinically significant from a stewardship perspective. Realigning microbiology workflows to mimic other research, which emphasizes the importance of funneling rapid BCID results through the ASP, may improve outcomes. Future studies may be warranted following the implementation of clinical decision support tools to assess their impact on stewardship practices and patient outcomes.
About 530,000 to 628,000 episodes of bloodstream infections (BSI) occur annually in the US.1 Early identification and treatment of bacteremia are essential to improve patient outcomes because it allows for more timely targeted antibiotic therapy.2 Organism identification and susceptibility testing can take 2 to 5 days, prolonging the use of broad-spectrum empiric antibiotics and increasing the risk of adverse events.3,4 The Infectious Disease Society of America recommends the use of rapid diagnostic testing and antimicrobial stewardship programs (ASPs) to improve rates of antibiotic susceptibilities to targeted antibiotics and optimize resource utilization.3 Rapid blood culture identification (BCID) technologies reduce the duration of empiric antibiotics in patients with contaminated blood cultures, resulting in shorter hospital stays and saving money per each patient tested.4
In March 2023, Veteran Health Indiana (VHI) implemented the BioFire FilmArray Blood Culture Identification (BCID2), a BSI panel test that identifies select gram-negative bacteria, gram-positive bacteria, yeast, and antimicrobial resistance genes with an aggregate sensitivity of 99% and a specificity of 99.8%. The BCID2 presents clinically relevant information faster than traditional culture methods, allowing clinicians to make more efficient and educated antibiotic regimen decisions than with previous methods.5
It takes 24 to 48 hours from blood collection for culture incubation, positivity, and gram staining to occur at VHI. If the gram stain is positive, the blood culture is placed on the BioFire BCID2 in addition to traditional culture medium. BioFire BCID2 results are ready in 45 to 60 minutes. Results are uploaded into the electronic health record (EHR) ≤ 2 hours after they are obtained and the primary team is notified if the test is positive for certain critical results. Susceptibility testing of an identified organism typically requires an additional 24 to 48 hours for finalization. VHI Infectious Disease created an evidence-based antibiotic recommendation chart for certain medication(s) and alternate therapies based on the reported organism and its interpreted presence of resistance markers (eg, ceftriaxone for Escherichia coli when extended-spectrum beta lactamases are not detected vs meropenem if extended-spectrum beta lactamases marker are present). These charts optimize the antibiotic regimen while awaiting susceptibility finalizations.
Two previous studies describe the impact of rapid diagnostic testing technology at US Department of Veterans Affairs (VA) medical centers.6,7 In Texas, the ASP reviewed BCID panel results via clinical decision support software for about 1 hour per day.6 A Los Angeles study analyzed the impact of Biofire BCID with an interpretation guide centered on unnecessary vancomycin use and determined that shorter duration of the medication may have been the result of more frequent infectious disease consultation.7
This study assessed the time to optimal antibiotic de-escalation before and after the implementation of BioFire BCID2 with results reviewed by the ASP without active notification or assistance of any clinical decision support technology. The primary objective was to evaluate difference in time to optimal antibiotics from blood culture draw pre- vs postintervention. Secondary objectives included differences in time to organism identification, difference in time on broad-spectrum antibiotics, and difference in time to appropriate antibiotics.
Methods
This quasi-experimental retrospective chart review assessed the impact of BioFire BCID2 use on timely antibiotic de-escalation for patients who experienced a BSI at VHI between March 1, 2022, and October 1, 2023. Microbiology laboratory records identified eligible patients with positive blood cultures within the study time frame. Data were collected from the VHI EHR.
Patients were included if they had a positive bacterial blood culture and received ≥ 1 antibiotic indicated for bacteremia while receiving inpatient care. Patients were excluded if they died prior to blood culture results, transferred out of VHI, left against medical advice, or had untreated contaminants in blood culture results (ie, never received antibiotics aimed at the contaminated culture).
Patient lists were generated for before and after implementation of BioFire BCID2 (pre- and postintervention) using the VHI EHR and microbiology laboratory record system. The pre- and postinterventions groups were different sizes. As a result, a random sampling of the preintervention group was selected and included patients from March 1, 2022, through March 26, 2023. The postintervention group was smaller due to time constraints between initiation of BioFire BCID2 for data collection and included all patients from March 27, 2023, through October 1, 2023.
Optimal antibiotics were defined as escalation from inappropriate therapy to broader agent(s), de-escalation from broad-spectrum therapy to targeted agent(s), discontinuation of therapy due to an organism being identified as a contaminant, or optimization of a regimen to the preferred antimicrobial agent based on evidence-based consensus guidelines. Broad-spectrum antibiotics included: piperacillin/tazobactam, cefepime, ceftazidime, ceftazidime-avibactam, cefiderocol, carbapenems, fluroquinolones, vancomycin, daptomycin, ceftaroline, linezolid, or aztreonam. Appropriate antibiotics were defined as those with activity toward the final identified organism(s).
Deidentified participant data were entered into Microsoft Excel and kept on a secure VA server to complete statistical analyses. Parametric continuous data, such as age, were analyzed using the t-test, while nonparametric continuous data, such as time to optimal antibiotics, were analyzed using the Mann-Whitney U test. Categorical data, like sex and race, were analyzed using either Fisher exact test for small sample sizes or X2 test for a larger sample size. Statistical significance levels was defined as P < .05.
Results
Using patient lists drawn from the EHR and the microbiology laboratory records, 110 electronic charts were randomly selected for review. Fifteen patients were excluded: 8 had untreated contaminants, 4 died, and 3 were transferred out of VHI. Of the 95 patients included, 48 were in the preintervention group and 47 were in the postintervention group (Figure 1).

Baseline characteristics were similar between the 2 groups (Table 1). Most patients were White males aged > 70 years in the EHR. The urinary tract was the most common source of infection, impacting 12 patients in each group (Figure 2). Escherichia coli, Klebsiella, Staphylococcus, and Streptococcus were the most common bloodstream isolates identified.


The median time to optimal antibiotics in the preintervention group was 58.5 hours vs 43.4 hours in the postintervention group (P = .11). The median time to organism identification was 37.8 hours in the preintervention group vs 16.9 hours in the postintervention group (P < .001). The median time on broad-spectrum antibiotics was 45.2 hours in the preintervention group vs 46.6 hours in the postintervention group (P = .99). The median time on appropriate antibiotics in the preintervention group was 2.3 hours vs 1.9 hours in the postintervention group (P = .79). Differences in other measured outcomes between the groups were not statistically significant (Table 2).

Although implementation of rapid diagnostic technology reduced the median time to optimal antibiotics, the results were not statistically significant. Shorter time to organism identification in the postintervention group compared to the preintervention group was the lone statistically significant metric (P < .001).
Discussion
A lack of statistical significance in the primary outcome may have been due to nonadherence to facility de-escalation protocols or a suboptimal BioFire BCID2 result notification system. Additionally, use of rapid BCID at VHI may improve over time as clinicians become more familiar with the technology. Gaps in clinical pharmacy coverage during the night shift may have also contributed to delays in antibiotic optimization, particularly if other clinicians are not equipped with the knowledge or training to appropriately deescalate antibiotics based on microorganisms identified. A 2017 study by Donner et al concluded that physician interpretation of BCID results is suboptimal and should be augmented with clinical decision support tools as new technology becomes available.8 Despite the statistically insignificant results of this study, it did highlight potential areas of improvement which can lead to improved patient care.
Previous research has evaluated the impact of rapid BCID technology on antibiotic treatment and clinical outcomes. Chiasson et al found that median time to optimal therapy was 73.8 hours in the pre-BCID arm compared to 34.7 hours in the post- BCID arm (P ≤ .001), emphasizing the importance of combining rapid BCID with clinical decision support tools and pharmacy input.6 Senok et al found that BCID2 implementation led to a significant decrease in median time to culture result, which informed optimal antibiotic therapy and decreased 30-day mortality in the intensive care setting.9 In contrast, the current study did not stratify patients according to medical ward or illness severity even though clinicians may be less likely to de-escalate antibiotic therapy in critically ill patients.
Bae et al reported findings consistent with the current study and concluded that BCID did not affect the clinical outcomes of overall BSIs; however, it contributed to early administration of effective antibiotics in cases of BSIs caused by multidrug-resistant organisms.10 Results of this study were not stratified according to multidrug-resistant organisms because the sample size was too small. The current study also included patients with polymicrobial infections, which may have impacted the results due to a less streamlined approach to antibiotic optimization.
Limitations
This single-center, retrospective study had a small sample size, short time frame, and lacked patient diversity, and therefore may not be generalizable to other health care systems. The sample size was limited by shorter date range and smaller patient list between BioFire BCID2 implementation and data collection, which was used to determine the number of charts selected in each group. Some patients received antibiotics prior to blood cultures being drawn, which may falsely decrease time to optimal/ appropriate antibiotics and falsely increase time on broad spectrum/any antibiotics due to early antibiotic administration. The total number of patients on broad-spectrum antibiotics differed from the total number of patients for other outcomes because several patients never received the defined broad spectrum antibiotics.
Conclusions
When combined with a pre-existing ASP without active notification, the implementation of BioFire BCID2 did not return statistically significant data showing a decrease in time to optimal antibiotics, time to appropriate antibiotics, or time on broad-spectrum antibiotics at VHI. To make this program more successful, pharmacist intervention and clinical decision support tools may be needed.
Additional research is required to determine the optimal integration of antimicrobial stewardship, rapid diagnostic technology, and pharmacy services for maximum benefit. Even though the primary outcome was not statistically significant, the results may be clinically significant from a stewardship perspective. Realigning microbiology workflows to mimic other research, which emphasizes the importance of funneling rapid BCID results through the ASP, may improve outcomes. Future studies may be warranted following the implementation of clinical decision support tools to assess their impact on stewardship practices and patient outcomes.
- Goto M, Al-Hasan MN. Overall burden of bloodstream infection and nosocomial bloodstream infection in North America and Europe. Clin Microbiol Infect. 2013;19(6):501- 509. doi:10.1111/1469-0691.12195
- Pardo J, Klinker KP, Borgert SJ, Butler BM, Giglio PG, Rand KH. Clinical and economic impact of antimicrobial stewardship interventions with the FilmArray blood culture identification panel. Diagn Microbiol Infect Dis. 2016;84(2):159-164. doi:10.1016/j.diagmicrobio.2015.10.023.
- Barlam TF, Cosgrove SE, Abbo LM, et al. Implementing an antibiotic stewardship program: guidelines by the Infectious Diseases Society of America and the Society for Healthcare Epidemiology of America. Clin Infect Dis. 2016;62(10):e51-e77. doi:10.1093/cid/ciw118
- BIOFIRE® Blood Culture Identification 2 (BCID2) Panel. Biomerierux. Updated 2025. Accessed May 10, 2025. https://www.biofiredx.com/products/the-filmarray-panels/filmarraybcid/
- Huang AM, Newton D, Kunapuli A, et al. Impact of rapid organism identification via matrix-assisted laser desorption/ionization time-of-flight combined with antimicrobial stewardship team intervention in adult patients with bacteremia and candidemia. Clin Infect Dis. 2013;57(9):1237-1245. doi:10.1093/cid/cit498
- Chiasson JM, Smith WJ, Jodlowski TZ, Kouma MA, Cutrell JB. Impact of a rapid blood culture diagnostic panel on time to optimal antimicrobial therapy at a veterans affairs medical center. J Pharm Pract. 2022;35(5):722-729. doi:10.1177/08971900211000686
- Wu S, Watson RL, Graber CJ. 2007. Impact of combining rapid diagnostics with an interpretation guide on vancomycin usage for contaminant blood cultures growing coagulase- negative staphylococci (CoNS). Open Forum Infect Dis. 2019;6(Suppl 2):S674. doi:10.1093/ofid/ofz360.1687
- Donner LM, Campbell WS, Lyden E, Van Schooneveld TC. Assessment of rapid-blood-culture-identification result interpretation and antibiotic prescribing practices. J Clin Microbiol. 2017;55(5):1496-1507. doi:10.1128/JCM.02395-16
- Senok A, Dabal LA, Alfaresi M, et al. Clinical impact of the BIOFIRE blood culture identification 2 panel in adult patients with bloodstream infection: a multicentre observational study in the United Arab Emirates. Diagnostics (Basel). 2023;13(14):2433. doi:10.3390/diagnostics13142433
- Bae JY, Bae J, So MK, Choi HJ, Lee M. The impact of the rapid blood culture identification panel on antibiotic treatment and clinical outcomes in bloodstream infections, particularly those associated with multidrug-resistant micro-organisms. Diagnostics (Basel). 2023;13(23):3504. doi:10.3390/diagnostics13233504
- Goto M, Al-Hasan MN. Overall burden of bloodstream infection and nosocomial bloodstream infection in North America and Europe. Clin Microbiol Infect. 2013;19(6):501- 509. doi:10.1111/1469-0691.12195
- Pardo J, Klinker KP, Borgert SJ, Butler BM, Giglio PG, Rand KH. Clinical and economic impact of antimicrobial stewardship interventions with the FilmArray blood culture identification panel. Diagn Microbiol Infect Dis. 2016;84(2):159-164. doi:10.1016/j.diagmicrobio.2015.10.023.
- Barlam TF, Cosgrove SE, Abbo LM, et al. Implementing an antibiotic stewardship program: guidelines by the Infectious Diseases Society of America and the Society for Healthcare Epidemiology of America. Clin Infect Dis. 2016;62(10):e51-e77. doi:10.1093/cid/ciw118
- BIOFIRE® Blood Culture Identification 2 (BCID2) Panel. Biomerierux. Updated 2025. Accessed May 10, 2025. https://www.biofiredx.com/products/the-filmarray-panels/filmarraybcid/
- Huang AM, Newton D, Kunapuli A, et al. Impact of rapid organism identification via matrix-assisted laser desorption/ionization time-of-flight combined with antimicrobial stewardship team intervention in adult patients with bacteremia and candidemia. Clin Infect Dis. 2013;57(9):1237-1245. doi:10.1093/cid/cit498
- Chiasson JM, Smith WJ, Jodlowski TZ, Kouma MA, Cutrell JB. Impact of a rapid blood culture diagnostic panel on time to optimal antimicrobial therapy at a veterans affairs medical center. J Pharm Pract. 2022;35(5):722-729. doi:10.1177/08971900211000686
- Wu S, Watson RL, Graber CJ. 2007. Impact of combining rapid diagnostics with an interpretation guide on vancomycin usage for contaminant blood cultures growing coagulase- negative staphylococci (CoNS). Open Forum Infect Dis. 2019;6(Suppl 2):S674. doi:10.1093/ofid/ofz360.1687
- Donner LM, Campbell WS, Lyden E, Van Schooneveld TC. Assessment of rapid-blood-culture-identification result interpretation and antibiotic prescribing practices. J Clin Microbiol. 2017;55(5):1496-1507. doi:10.1128/JCM.02395-16
- Senok A, Dabal LA, Alfaresi M, et al. Clinical impact of the BIOFIRE blood culture identification 2 panel in adult patients with bloodstream infection: a multicentre observational study in the United Arab Emirates. Diagnostics (Basel). 2023;13(14):2433. doi:10.3390/diagnostics13142433
- Bae JY, Bae J, So MK, Choi HJ, Lee M. The impact of the rapid blood culture identification panel on antibiotic treatment and clinical outcomes in bloodstream infections, particularly those associated with multidrug-resistant micro-organisms. Diagnostics (Basel). 2023;13(23):3504. doi:10.3390/diagnostics13233504
Impact of Rapid Blood Culture Identification on Antibiotic De-escalation at a Veterans Affairs Medical Center
Impact of Rapid Blood Culture Identification on Antibiotic De-escalation at a Veterans Affairs Medical Center
Disparate Prednisone Starting Dosages for Systemic Corticosteroid-Naïve Veterans With Active Sarcoidosis
Disparate Prednisone Starting Dosages for Systemic Corticosteroid-Naïve Veterans With Active Sarcoidosis
Sarcoidosis is a multiorgan granulomatous disorder of unknown etiology that impacts many US veterans.1 At diagnosis, clinical manifestations vary and partially depend on the extent and severity of organ involvement, particularly of the lungs, heart, and eyes.2,3 Sarcoidosis may lead to progressive organ dysfunction, long-term disability, and death.1-3 Clinical practice guidelines recommend prednisone 20 to 40 mg daily or equivalent-prednisone dose followed by a slow tapering, as first-line pharmacotherapy for patients with active sarcoidosis who are naïve to systemic corticosteroids.2-4
Use of prolonged, high-dosage prednisone (> 40 mg daily) is discouraged due to a high risk of corticosteroid-related adverse events and associated health care costs.5,6 Research suggests that initial lower prednisone dosage (< 20 mg daily) may be effective in systemic corticosteroid-naïve patients with active sarcoidosis.3
Adherence to this regimen by specialists (eg, pulmonologists, dermatologists, ophthalmologists, rheumatologists, and cardiologists) has not been established. This study sought to determine the starting dosages for prednisone prescribed at the Jesse Brown Department of Veterans Affairs Medical Center (JBVAMC) to patients with active sarcoidosis who were systemic corticosteroid-naïve.
Methods
Patient data were reviewed from the Computerized Patient Record System (CPRS) for individuals diagnosed with sarcoidosis who were corticosteroid-naïve and prescribed initial prednisone dosages by health care practitioners (HCPs) from several specialties between 2014 and 2023 at JBVAMC. This 200-bed acute care facility serves about 62,000 veterans who live in Illinois or Indiana. JBVAMC is affiliated with the University of Illinois College of Medicine at Chicago, Northwestern University Feinberg School of Medicine, and the University of Chicago Pritzker School of Medicine; many JBVAMC HCPs hold academic appointments with these medical schools.
Patient demographics, prescriber specialty, and daily starting dosage were recorded. The decision to initiate prednisone therapy and its dosage were at the discretion of HCPs who diagnosed active sarcoidosis based on compatible clinical and ancillary test findings as documented in CPRS.2-4,6-10 Statistical analyses were conducted using a t test, and a threshold of P < .05 was considered statistically significant. This study was reviewed and determined to be exempt by the JBVAMC institutional review board.
Results
Sixty-eight patients who were systemic corticosteroid- naïve and had sarcoidosis were prescribed prednisone by HCPs at JBVAMC. Fifty-two were Black (76%), 62 were male (91%), and 53 were current or former smokers (78%). The mean (SD) age was 63 (11) years (Table 1). Forty patients (59%) had lung involvement, 6 had eye (9%), 6 had skin (9%), and 5 had musculoskeletal system (7%) involvement.

Pulmonologists predominantly prescribed initial dosage of 20 mg to 40 mg (median, 35 mg daily) (Figure). Other HCPs, including primary care, tended to prescribe prednisone < 20 mg (median, 17.5 mg; P < .05) (Table 2). The highest initial prednisone dosage was 80 mg daily, prescribed by a neurologist for a patient with neurosarcoidosis. Voortman et al recommend 20 to 40 mg prednisone daily for neurosarcoidosis.7 Both groups, pulmonologists and nonpulmonologists, had no significant differences in patient characteristics.


Discussion
Disparate prescription patterns of initial prednisone dosages were observed between pulmonologists and nonpulmonologists treating systemic corticosteroid-naïve patients with active sarcoidosis at JBVAMC. This study did not determine the underlying reasons for this phenomenon, nor its impact on patient outcomes.
Clinical practice guidelines have not been independently validated for each organ affected by sarcoidosis.2-4,6-10 Variations in clinical practice for other specialties may account for the variable prednisone starting dosage selection. For example, among 6 patients with active ocular sarcoidosis treated by ophthalmologists, 4 were prescribed an initial prednisone dosage of ≥ 10 mg daily. The American Academy of Ophthalmology recommends an initial short-term course of prednisone at 1 to 1.5 mg/kg daily, tapered down to the lowest effective dosage.10
Limitations
This study used a small, single-center predominantly older Black male patient cohort. The generalizability of these observations is unknown. A larger, multicenter prospective study is warranted to further evaluate these initial observations.
Conclusions
HCPs treating patients who are systemic corticosteroid-naïve with active sarcoidosis for whom prednisone is indicated should adhere to current clinical practice guidelines by prescribing prednisone in the 20 to 40 mg daily range.
- Seedahmed MI, Baugh AD, Albirair MT, et al. Epidemiology of sarcoidosis in U.S. veterans from 2003 to 2019. Ann Am Thorac Soc. 2023;20(6):797-806. doi:10.1513/AnnalsATS.202206-515OC
- Baughman RP, Valeyre D, Korsten P, et al. ERS clinical practice guidelines on treatment of sarcoidosis. Eur Respir J. 2021;58(6):2004079. doi:10.1183/13993003.04079-2020
- Rahaghi FF, Baughman RP, Saketkoo LA, et al. Delphi consensus recommendations for a treatment algorithm in pulmonary sarcoidosis. Eur Respir Rev. 2020;29(155):190146. doi:10.1183/16000617.0146-2019
- Kwon S, Judson MA. Clinical pharmacology in sarcoidosis: how to use and monitor sarcoidosis medications. J Clin Med. 2024;13(5):1250. doi:10.3390/jcm13051250
- Rice JB, White AG, Johnson M, et al. Quantitative characterization of the relationship between levels of extended corticosteroid use and related adverse events in a US population. Curr Med Res Opin. 2018;34(8):1519-1527. doi:10.1080/03007995.2018.1474090
- Rice JB, White AG, Johnson M, Wagh A, Qin Y, Bartels-Peculis L, et al. Healthcare resource use and cost associated with varying dosages of extended corticosteroid exposure in a US population. J Med Econ. 2018;21(9):846-852. doi:10.1080/13696998.2018.1474750
- Voortman M, Drent M, Baughman RP. Management of neurosarcoidosis: a clinical challenge. Curr Opin Neurol. 2019;32(3):475-483. doi:10.1097/WCO.0000000000000684
- Cheng RK, Kittleson MM, Beavers CJ, et al. Diagnosis and management of cardiac sarcoidosis: a scientific statement from the American Heart Association. Circulation. 2024;149(21):e1197-e1216. doi:10.1161/CIR.0000000000001240
- Cohen E, Lheure C, Ingen-Housz-Oro S, et al. Which firstline treatment for cutaneous sarcoidosis? A retrospective study of 120 patients. Eur J Dermatol. 2023;33(6):680-685. doi:10.1684/ejd.2023.4584
- American Academy of Ophthalmology. Ocular manifestations of sarcoidosis. EyeWiki. Accessed June 3, 2025. https://eyewiki.org/Ocular_Manifestations_of_Sarcoidosis
Sarcoidosis is a multiorgan granulomatous disorder of unknown etiology that impacts many US veterans.1 At diagnosis, clinical manifestations vary and partially depend on the extent and severity of organ involvement, particularly of the lungs, heart, and eyes.2,3 Sarcoidosis may lead to progressive organ dysfunction, long-term disability, and death.1-3 Clinical practice guidelines recommend prednisone 20 to 40 mg daily or equivalent-prednisone dose followed by a slow tapering, as first-line pharmacotherapy for patients with active sarcoidosis who are naïve to systemic corticosteroids.2-4
Use of prolonged, high-dosage prednisone (> 40 mg daily) is discouraged due to a high risk of corticosteroid-related adverse events and associated health care costs.5,6 Research suggests that initial lower prednisone dosage (< 20 mg daily) may be effective in systemic corticosteroid-naïve patients with active sarcoidosis.3
Adherence to this regimen by specialists (eg, pulmonologists, dermatologists, ophthalmologists, rheumatologists, and cardiologists) has not been established. This study sought to determine the starting dosages for prednisone prescribed at the Jesse Brown Department of Veterans Affairs Medical Center (JBVAMC) to patients with active sarcoidosis who were systemic corticosteroid-naïve.
Methods
Patient data were reviewed from the Computerized Patient Record System (CPRS) for individuals diagnosed with sarcoidosis who were corticosteroid-naïve and prescribed initial prednisone dosages by health care practitioners (HCPs) from several specialties between 2014 and 2023 at JBVAMC. This 200-bed acute care facility serves about 62,000 veterans who live in Illinois or Indiana. JBVAMC is affiliated with the University of Illinois College of Medicine at Chicago, Northwestern University Feinberg School of Medicine, and the University of Chicago Pritzker School of Medicine; many JBVAMC HCPs hold academic appointments with these medical schools.
Patient demographics, prescriber specialty, and daily starting dosage were recorded. The decision to initiate prednisone therapy and its dosage were at the discretion of HCPs who diagnosed active sarcoidosis based on compatible clinical and ancillary test findings as documented in CPRS.2-4,6-10 Statistical analyses were conducted using a t test, and a threshold of P < .05 was considered statistically significant. This study was reviewed and determined to be exempt by the JBVAMC institutional review board.
Results
Sixty-eight patients who were systemic corticosteroid- naïve and had sarcoidosis were prescribed prednisone by HCPs at JBVAMC. Fifty-two were Black (76%), 62 were male (91%), and 53 were current or former smokers (78%). The mean (SD) age was 63 (11) years (Table 1). Forty patients (59%) had lung involvement, 6 had eye (9%), 6 had skin (9%), and 5 had musculoskeletal system (7%) involvement.

Pulmonologists predominantly prescribed initial dosage of 20 mg to 40 mg (median, 35 mg daily) (Figure). Other HCPs, including primary care, tended to prescribe prednisone < 20 mg (median, 17.5 mg; P < .05) (Table 2). The highest initial prednisone dosage was 80 mg daily, prescribed by a neurologist for a patient with neurosarcoidosis. Voortman et al recommend 20 to 40 mg prednisone daily for neurosarcoidosis.7 Both groups, pulmonologists and nonpulmonologists, had no significant differences in patient characteristics.


Discussion
Disparate prescription patterns of initial prednisone dosages were observed between pulmonologists and nonpulmonologists treating systemic corticosteroid-naïve patients with active sarcoidosis at JBVAMC. This study did not determine the underlying reasons for this phenomenon, nor its impact on patient outcomes.
Clinical practice guidelines have not been independently validated for each organ affected by sarcoidosis.2-4,6-10 Variations in clinical practice for other specialties may account for the variable prednisone starting dosage selection. For example, among 6 patients with active ocular sarcoidosis treated by ophthalmologists, 4 were prescribed an initial prednisone dosage of ≥ 10 mg daily. The American Academy of Ophthalmology recommends an initial short-term course of prednisone at 1 to 1.5 mg/kg daily, tapered down to the lowest effective dosage.10
Limitations
This study used a small, single-center predominantly older Black male patient cohort. The generalizability of these observations is unknown. A larger, multicenter prospective study is warranted to further evaluate these initial observations.
Conclusions
HCPs treating patients who are systemic corticosteroid-naïve with active sarcoidosis for whom prednisone is indicated should adhere to current clinical practice guidelines by prescribing prednisone in the 20 to 40 mg daily range.
Sarcoidosis is a multiorgan granulomatous disorder of unknown etiology that impacts many US veterans.1 At diagnosis, clinical manifestations vary and partially depend on the extent and severity of organ involvement, particularly of the lungs, heart, and eyes.2,3 Sarcoidosis may lead to progressive organ dysfunction, long-term disability, and death.1-3 Clinical practice guidelines recommend prednisone 20 to 40 mg daily or equivalent-prednisone dose followed by a slow tapering, as first-line pharmacotherapy for patients with active sarcoidosis who are naïve to systemic corticosteroids.2-4
Use of prolonged, high-dosage prednisone (> 40 mg daily) is discouraged due to a high risk of corticosteroid-related adverse events and associated health care costs.5,6 Research suggests that initial lower prednisone dosage (< 20 mg daily) may be effective in systemic corticosteroid-naïve patients with active sarcoidosis.3
Adherence to this regimen by specialists (eg, pulmonologists, dermatologists, ophthalmologists, rheumatologists, and cardiologists) has not been established. This study sought to determine the starting dosages for prednisone prescribed at the Jesse Brown Department of Veterans Affairs Medical Center (JBVAMC) to patients with active sarcoidosis who were systemic corticosteroid-naïve.
Methods
Patient data were reviewed from the Computerized Patient Record System (CPRS) for individuals diagnosed with sarcoidosis who were corticosteroid-naïve and prescribed initial prednisone dosages by health care practitioners (HCPs) from several specialties between 2014 and 2023 at JBVAMC. This 200-bed acute care facility serves about 62,000 veterans who live in Illinois or Indiana. JBVAMC is affiliated with the University of Illinois College of Medicine at Chicago, Northwestern University Feinberg School of Medicine, and the University of Chicago Pritzker School of Medicine; many JBVAMC HCPs hold academic appointments with these medical schools.
Patient demographics, prescriber specialty, and daily starting dosage were recorded. The decision to initiate prednisone therapy and its dosage were at the discretion of HCPs who diagnosed active sarcoidosis based on compatible clinical and ancillary test findings as documented in CPRS.2-4,6-10 Statistical analyses were conducted using a t test, and a threshold of P < .05 was considered statistically significant. This study was reviewed and determined to be exempt by the JBVAMC institutional review board.
Results
Sixty-eight patients who were systemic corticosteroid- naïve and had sarcoidosis were prescribed prednisone by HCPs at JBVAMC. Fifty-two were Black (76%), 62 were male (91%), and 53 were current or former smokers (78%). The mean (SD) age was 63 (11) years (Table 1). Forty patients (59%) had lung involvement, 6 had eye (9%), 6 had skin (9%), and 5 had musculoskeletal system (7%) involvement.

Pulmonologists predominantly prescribed initial dosage of 20 mg to 40 mg (median, 35 mg daily) (Figure). Other HCPs, including primary care, tended to prescribe prednisone < 20 mg (median, 17.5 mg; P < .05) (Table 2). The highest initial prednisone dosage was 80 mg daily, prescribed by a neurologist for a patient with neurosarcoidosis. Voortman et al recommend 20 to 40 mg prednisone daily for neurosarcoidosis.7 Both groups, pulmonologists and nonpulmonologists, had no significant differences in patient characteristics.


Discussion
Disparate prescription patterns of initial prednisone dosages were observed between pulmonologists and nonpulmonologists treating systemic corticosteroid-naïve patients with active sarcoidosis at JBVAMC. This study did not determine the underlying reasons for this phenomenon, nor its impact on patient outcomes.
Clinical practice guidelines have not been independently validated for each organ affected by sarcoidosis.2-4,6-10 Variations in clinical practice for other specialties may account for the variable prednisone starting dosage selection. For example, among 6 patients with active ocular sarcoidosis treated by ophthalmologists, 4 were prescribed an initial prednisone dosage of ≥ 10 mg daily. The American Academy of Ophthalmology recommends an initial short-term course of prednisone at 1 to 1.5 mg/kg daily, tapered down to the lowest effective dosage.10
Limitations
This study used a small, single-center predominantly older Black male patient cohort. The generalizability of these observations is unknown. A larger, multicenter prospective study is warranted to further evaluate these initial observations.
Conclusions
HCPs treating patients who are systemic corticosteroid-naïve with active sarcoidosis for whom prednisone is indicated should adhere to current clinical practice guidelines by prescribing prednisone in the 20 to 40 mg daily range.
- Seedahmed MI, Baugh AD, Albirair MT, et al. Epidemiology of sarcoidosis in U.S. veterans from 2003 to 2019. Ann Am Thorac Soc. 2023;20(6):797-806. doi:10.1513/AnnalsATS.202206-515OC
- Baughman RP, Valeyre D, Korsten P, et al. ERS clinical practice guidelines on treatment of sarcoidosis. Eur Respir J. 2021;58(6):2004079. doi:10.1183/13993003.04079-2020
- Rahaghi FF, Baughman RP, Saketkoo LA, et al. Delphi consensus recommendations for a treatment algorithm in pulmonary sarcoidosis. Eur Respir Rev. 2020;29(155):190146. doi:10.1183/16000617.0146-2019
- Kwon S, Judson MA. Clinical pharmacology in sarcoidosis: how to use and monitor sarcoidosis medications. J Clin Med. 2024;13(5):1250. doi:10.3390/jcm13051250
- Rice JB, White AG, Johnson M, et al. Quantitative characterization of the relationship between levels of extended corticosteroid use and related adverse events in a US population. Curr Med Res Opin. 2018;34(8):1519-1527. doi:10.1080/03007995.2018.1474090
- Rice JB, White AG, Johnson M, Wagh A, Qin Y, Bartels-Peculis L, et al. Healthcare resource use and cost associated with varying dosages of extended corticosteroid exposure in a US population. J Med Econ. 2018;21(9):846-852. doi:10.1080/13696998.2018.1474750
- Voortman M, Drent M, Baughman RP. Management of neurosarcoidosis: a clinical challenge. Curr Opin Neurol. 2019;32(3):475-483. doi:10.1097/WCO.0000000000000684
- Cheng RK, Kittleson MM, Beavers CJ, et al. Diagnosis and management of cardiac sarcoidosis: a scientific statement from the American Heart Association. Circulation. 2024;149(21):e1197-e1216. doi:10.1161/CIR.0000000000001240
- Cohen E, Lheure C, Ingen-Housz-Oro S, et al. Which firstline treatment for cutaneous sarcoidosis? A retrospective study of 120 patients. Eur J Dermatol. 2023;33(6):680-685. doi:10.1684/ejd.2023.4584
- American Academy of Ophthalmology. Ocular manifestations of sarcoidosis. EyeWiki. Accessed June 3, 2025. https://eyewiki.org/Ocular_Manifestations_of_Sarcoidosis
- Seedahmed MI, Baugh AD, Albirair MT, et al. Epidemiology of sarcoidosis in U.S. veterans from 2003 to 2019. Ann Am Thorac Soc. 2023;20(6):797-806. doi:10.1513/AnnalsATS.202206-515OC
- Baughman RP, Valeyre D, Korsten P, et al. ERS clinical practice guidelines on treatment of sarcoidosis. Eur Respir J. 2021;58(6):2004079. doi:10.1183/13993003.04079-2020
- Rahaghi FF, Baughman RP, Saketkoo LA, et al. Delphi consensus recommendations for a treatment algorithm in pulmonary sarcoidosis. Eur Respir Rev. 2020;29(155):190146. doi:10.1183/16000617.0146-2019
- Kwon S, Judson MA. Clinical pharmacology in sarcoidosis: how to use and monitor sarcoidosis medications. J Clin Med. 2024;13(5):1250. doi:10.3390/jcm13051250
- Rice JB, White AG, Johnson M, et al. Quantitative characterization of the relationship between levels of extended corticosteroid use and related adverse events in a US population. Curr Med Res Opin. 2018;34(8):1519-1527. doi:10.1080/03007995.2018.1474090
- Rice JB, White AG, Johnson M, Wagh A, Qin Y, Bartels-Peculis L, et al. Healthcare resource use and cost associated with varying dosages of extended corticosteroid exposure in a US population. J Med Econ. 2018;21(9):846-852. doi:10.1080/13696998.2018.1474750
- Voortman M, Drent M, Baughman RP. Management of neurosarcoidosis: a clinical challenge. Curr Opin Neurol. 2019;32(3):475-483. doi:10.1097/WCO.0000000000000684
- Cheng RK, Kittleson MM, Beavers CJ, et al. Diagnosis and management of cardiac sarcoidosis: a scientific statement from the American Heart Association. Circulation. 2024;149(21):e1197-e1216. doi:10.1161/CIR.0000000000001240
- Cohen E, Lheure C, Ingen-Housz-Oro S, et al. Which firstline treatment for cutaneous sarcoidosis? A retrospective study of 120 patients. Eur J Dermatol. 2023;33(6):680-685. doi:10.1684/ejd.2023.4584
- American Academy of Ophthalmology. Ocular manifestations of sarcoidosis. EyeWiki. Accessed June 3, 2025. https://eyewiki.org/Ocular_Manifestations_of_Sarcoidosis
Disparate Prednisone Starting Dosages for Systemic Corticosteroid-Naïve Veterans With Active Sarcoidosis
Disparate Prednisone Starting Dosages for Systemic Corticosteroid-Naïve Veterans With Active Sarcoidosis
A Systemic Lupus Erythematosus Incidence Surveillance Report Among DoD Beneficiaries During the COVID-19 Pandemic
A Systemic Lupus Erythematosus Incidence Surveillance Report Among DoD Beneficiaries During the COVID-19 Pandemic
Systemic lupus erythematosus (SLE), or lupus, is a rare autoimmune disease estimated to occur in about 5.1 cases per 100,000 person-years in the United States in 2018.1 The disease predominantly affects females, with an incidence of 8.7 cases per 100,000 person-years vs 1.2 cases per 100,000 person-years in males, and is most common in patients aged 15 to 44 years.1,2
Lupus presents with a constellation of clinical signs and symptoms that evolve, along with hallmark laboratory findings indicative of immune dysregulation and polyclonal B-cell activation. Consequently, a wide array of autoantibodies may be produced, although the combination of epitope specificity can vary from patient to patient.3 Nevertheless, > 98% of individuals diagnosed with lupus produce antinuclear antibodies (ANA), making ANA positivity a near-universal serologic feature at the time of diagnosis.
The pathogenesis of lupus is complex. Research from the past 5 decades supports the role of certain viral infections—such as Epstein-Barr virus (EBV) and cytomegalovirus—as potential triggers.4 These viruses are thought to initiate disease through mechanisms including activation of interferon pathways, exposure of cryptic intracellular antigens, molecular mimicry, and epitope spreading. Subsequent clonal expansion and autoantibody production occur to varying degrees, influenced by viral load and host susceptibility factors.
During the COVID-19 pandemic, it became evident that SARS-CoV-2 exerts profound effects on immune regulation, influencing infection outcomes through mechanisms such as hyperactivation of innate immunity, especially in the lungs, leading to acute respiratory distress syndrome. Additionally, SARS-CoV-2 has been associated with polyclonal B-cell activation and the generation of autoantibodies. This association gained attention after Bastard et al identified anti–type I interferon antibodies in patients with severe COVID-19, predominantly among males with a genetic predisposition. These autoantibodies were shown to impair antiviral defenses and contribute to life-threatening pneumonia.5
Subsequent studies demonstrated the production of a wide spectrum of functional autoantibodies, including ANA, in patients with COVID-19.6,7 These findings were attributed to the acute expansion of autoreactive clones among naïve-derived immunoglobulin G1 antibody-secreting cells during the early stages of infection, with the degree of expansion correlating with disease severity.8,9 Although longitudinal data up to 15 months postinfection suggest this serologic abnormality resolves in more than two-thirds of patients, the number of individuals infected globally has raised serious public health concerns regarding the potential long-term sequelae, including the onset of lupus or other autoimmune diseases in COVID-19 survivors.6-9 A limited number of case reports describing the onset of lupus following SARS-CoV-2 infection support this hypothesis.10
This surveillance analysis investigates lupus incidence among patients within the Military Health System (MHS), encompassing all TRICARE beneficiaries, from January 2018 to December 2022. The objective of this analysis was to examine lupus incidence trends throughout the COVID-19 pandemic, stratified by sex, age, and active-duty status.
Methods
The MHS provides health care services to about 9.5 million US Department of Defense (DoD) beneficiaries. Outpatient health records and laboratory results for individuals receiving care at military treatment facilities (MTFs) between January 1, 2018, and December 31, 2022, were obtained from the Comprehensive Ambulatory/ Professional Encounter Record and MHS GENESIS. For beneficiaries receiving care in the private sector, data were sourced from the TRICARE Encounter Data—Non-Institutional database.
Laboratory test results, including ANA testing, were available only for individuals receiving care at MTFs. These laboratory data were extracted from the Composite Health Care System Chemistry database and MHS GENESIS laboratory systems for the same time frame. Inpatient data were not included in this analysis. Data from 2017 were used solely as a look-back (or washout) period to identify and exclude prevalent lupus cases diagnosed before 2018 and were not included in the final results.
Lupus cases were identified by the presence of a positive ANA test and appropriate International Classification of Diseases, 10th Revision, Clinical Modification (ICD-10-CM) codes. A positive ANA result was defined as either a qualitative result marked positive or a titer ≥ 1:80. The ICD-10-CM codes considered indicative of lupus included variations of M32, L93, or H01.12.
M32, L93, or H01.12. For cases with a positive ANA test, a lupus diagnosis required the presence of ≥ 2 lupus related ICD-10-CM codes. In the absence of ANA test results, a stricter criterion was applied: ≥ 4 lupus ICD-10-CM diagnosis codes recorded on separate days were required for inclusion.
Beneficiaries were excluded if they had a negative ANA result, only 1 lupus ICD- 10-CM diagnosis code, 1 positive ANA with only 1 corresponding ICD-10-CM code, or if their diagnosis occurred outside the defined study period. Patients and members of the public were not involved in the design, conduct, reporting, or dissemination of this study.
Results
Between January 1, 2017, and December 31, 2022, 99,946 TRICARE beneficiaries had some indication of lupus testing or diagnosis in their health records (Figure 1). Of these beneficiaries, 5335 had a positive ANA result and ≥ 2 ICD-10-CM lupus diagnosis codes. An additional 28,275 beneficiaries had ≥ 4 ICD-10-CM lupus diagnosis codes but no ANA test results. From these groups, the final sample included 10,760 beneficiaries who met the incident case definitions for SLE during the study period (2018 through 2022).

Most cases (85.1%, n = 9157) were diagnosed through TRICARE claims, while 1205 (11.2%) were diagnosed within the MHS. Another 398 (3.7%) had documentation of care both within and outside the MHS. Incident SLE cases declined by an average of 16% annually during the study period (Figure 2). This trend amounted to an overall reduction of 48.2%, from 2866 cases in 2018 to 1399 cases in 2022. This decline occurred despite total medical encounters among DoD beneficiaries remaining relatively stable during the pandemic years, with only a 3.5% change between 2018 and 2022.

The disease was more prevalent among female beneficiaries, with a female to- male ratio of 7:1 (Table 1). Among women, the number of new cases declined from 2519 in 2018 to 1223 in 2022, while the number of cases among men remained consistently < 350 annually. Similar trends were observed across other strata. Incident SLE cases were more common among nonactive-duty beneficiaries than active-duty service members, with a ratio of 18:1. New cases among active-duty members remained < 155 per year. Age-stratified data revealed that SLE was diagnosed predominantly in individuals aged ≥ 18 years, with a ratio of 37:1 compared with individuals aged < 18 years. Among children, the number of new cases remained < 75 per year throughout the study period.

A mean 56,850 ANA tests were conducted annually in centralized laboratories using standardized protocols (Table 2). The mean ANA positivity rate was 17.3%, which remained relatively stable from 2018 through 2022.

Discussion
This study examined the annual incidence of newly diagnosed SLE cases among all TRICARE beneficiaries from January 1, 2018, through December 31, 2022, covering both before and during the peak years of the COVID-19 pandemic. This analysis revealed a steady decline in SLE cases during this period. The reliability of these findings is reinforced by the comprehensiveness of the MHS, one of the largest US health care delivery systems, which maintains near-complete medical data capture for about 9.5 million DoD TRICARE beneficiaries across domestic and international settings.
SLE is a rare autoimmune disorder that presents a diagnostic challenge due to its wide range of nonspecific symptoms, many of which resemble other conditions. To reduce the likelihood of false-positive results and ensure diagnostic accuracy, this study adopted a stringent case definition. Incident cases were identified by the presence of ANA testing in conjunction with lupus-specific ICD-10-CM codes and required ≥ 4 lupus related diagnostic entries. This criterion was necessary due to the absence of ANA test results in data from private sector care settings. Our case definition aligns with established literature. For example, a Vanderbilt University chart review study demonstrated that combining ANA positivity with ≥ 4 lupus related ICD-10-CM codes achieves a positive predictive value of 100%, albeit with a sensitivity of 45%.11 Other studies similarly affirm the diagnostic validity of using recurrent ICD-10-CM codes to improve specificity in identifying lupus cases.12,13
The primary objective of this study was to examine the temporal trend in newly diagnosed lupus cases, rather than derive precise incidence rates. Although the TRICARE system includes about 9.5 million beneficiaries, this number represents a dynamic population with continual inflow and outflow. Accurate incidence rate calculation would require access to detailed denominator data, which were not readily available. In comparison with our findings, a study limited to active-duty service members reported fewer lupus cases. This discrepancy likely reflects differences in case definitions—specifically, the absence of laboratory data, the restricted range of diagnostic codes, and the requirement that diagnoses be rendered by specialists.14 Despite these differences, demographic patterns were consistent, with higher incidence observed in females and individuals aged ≥ 20 years.
A Centers for Disease Control and Prevention (CDC) study of lupus incidence in the general population also reported lower case counts.1 However, the CDC estimates were based on 5 state-level registries, which rely on clinician-reported cases and therefore may underestimate true disease burden. Moreover, the DoD beneficiary population differs markedly from the general population: it includes a large cohort of retirees, ensuring an older demographic; all members have comprehensive health care access; and active-duty personnel are subject to pre-enlistment medical screening. Taken together, these factors suggest this study may offer a more complete and systematically captured profile of lupus incidence.
We observed a marked decline of newly diagnosed SLE cases during the study period, which coincided with the widespread circulation of COVID-19. This decrease is unlikely to be attributable to reduced access to care during the pandemic. The MHS operates under a single-payer model, and the total number of patient encounters remained relatively stable throughout the pandemic.
To our knowledge, this is the only study to monitor lupus incidence in a large US population over the 5-year period encompassing before and during the COVID-19 pandemic. To date, only 4 large-scale surveillance studies have addressed similar questions. 14-17 Our findings are consistent with the most recent of these reports: an analysis limited to active-duty members of the US Armed Forces identified 1127 patients with newly diagnosed lupus between 2000 and 2022 and reported stable incidence trends throughout the pandemic.14 The other 3 studies adopted a different approach, comparing the emergence of autoimmune diseases, including lupus, between individuals with confirmed SARS-CoV-2 infection and those without. Each of these trials concluded that COVID-19 increases the risk of various autoimmune conditions, although the findings specific to lupus were inconsistent.15-17
Chang et al reported a significant increase in new lupus diagnoses (n = 2,926,016), with an adjusted hazard ratio (aHR) of 2.99 (95% CI, 2.68-3.34), spanning all ages and both sexes. The highest incidence was observed in individuals of Asian descent.15 Using German routine health care data from 2020, Tesch et al identified a heightened risk of autoimmune diseases, including lupus, among patients with a history of SARS-CoV-2 infection (n = 641,407; 9.4% children, 57.3% female, 6.4% hospitalized), compared with matched infection-naïve controls (n = 1,560,357).16 Both studies excluded vaccinated individuals.
These 2 studies diverged in their assessment of the relationship between COVID-19 severity and subsequent autoimmune risk. Chang et al found a higher incidence among nonhospitalized ambulatory patients, while Tesch et al reported that increased risk was associated with patients requiring intensive care unit admission.15,16
In contrast, based on a cohort of 4,197,188 individuals, Peng et al found no significant difference in lupus incidence among patients with SARS-CoV-2 infection (aHR, 1.05; 95% CI, 0.79-1.39).17 Notably, within the infected group, the incidence of SLE was significantly lower among vaccinated individuals compared with the unvaccinated group (aHR, 0.29; 95% CI, 0.18-0.47). Similar protective associations were observed for other antibody-mediated autoimmune disorders, including pemphigoid, Graves’ disease, and antiphospholipid antibody syndrome.
Limitations
Due to fundamental differences in study design, we were unable to directly reconcile our findings with those reported in the literature. This study lacked access to reliable documentation of COVID-19 infection status, primarily due to the widespread use of home testing among TRICARE beneficiaries. Additionally, the dataset did not include inpatient records and therefore did not permit evaluation of disease severity. Despite these constraints, it is plausible that the overall burden of COVID-19 infection within the study population was lower than that observed in the general US population.
As of December 2022, the DoD had reported about 750,000 confirmed COVID-19 cases among military personnel, civilian employees, dependents, and DoD contractors.18 Given that TRICARE beneficiaries represent about 2.8% of the total US population—and that > 90 million US individuals were infected between 2020 and 2022—the implied infection rate in our cohort appears to be about one-third of what might be expected.19 This discrepancy may be due to higher adherence to mitigation measures, such as social distancing and mask usage, among DoD-affiliated populations. COVID-19 vaccination was mandated for all active-duty service members, who constitute 5.4% of the study population. The remaining TRICARE beneficiaries also had access to guaranteed health care and vaccination coverage, likely contributing to high overall vaccination rates.
Because > 80% of the study population was composed of individuals from diverse civilian backgrounds, we expect the distribution of infection severity within the DoD beneficiary population to approximate that of the general US population.
Future Directions
The findings of this study offer circumstantial yet real-time evidence of the complexity underlying immune dysregulation at the intersection of host susceptibility and environmental exposures. The stability in ANA positivity rates during the study period mitigates concerns regarding undiagnosed subclinical lupus and may suggest that, overall, immune homeostasis was preserved among DoD beneficiaries.
It is noteworthy that during the COVID-19 pandemic, the incidence of several common infections—such as influenza and EBV—declined markedly, likely as a result of widespread social distancing and other public health interventions.20 Mitigation strategies implemented within the military may have conferred protection not only against COVID-19 but also against other community-acquired pathogens.
In light of these observations, we hypothesize that for COVID-19 to act as a trigger for SLE, a prolonged or repeated disruption of immune equilibrium may be required—potentially mediated by recurrent infections or sustained inflammatory states. The association between viral infections and autoimmunity is well established. Immune dysregulation leading to autoantibody production has been observed not only in the context of SARS-CoV-2 but also following infections with EBV, cytomegalovirus, enteroviruses, hepatitis B and C viruses, HIV, and parvovirus B19.21
This dysregulation is often transient, accompanied by compensatory immune regulatory responses. However, in individuals subjected to successive or overlapping infections, these regulatory mechanisms may become compromised or overwhelmed, due to emergent patterns of immune interference of varying severity. In such cases, a transient immune perturbation may progress into a bona fide autoimmune disease, contingent upon individual risk factors such as genetic predisposition, preexisting immune memory, and regenerative capacity.21
Therefore, we believe the significance of this study is 2-fold. First, lupus is known to develop gradually and may require 3 to 5 years to clinically manifest after the initial break in immunological tolerance.3 Continued public health surveillance represents a more pragmatic strategy than retrospective cohort construction, especially as histories of COVID-19 infection become increasingly complete and definitive. Our findings provide a valuable baseline reference point for future longitudinal studies.
The interpretation of surveillance outcomes—whether indicating an upward trend, a stable baseline, or a downward trend—offers distinct analytical value. Within this study population, we observed neither an upward trajectory that might suggest a direct causal link, nor a flat trend that would imply absence of association between COVID-19 and lupus pathogenesis. Instead, the observation of a downward trend invites consideration of nonlinear or protective influences. From this perspective, we recommend that future investigations adopt a holistic framework when assessing environmental contributions to immune dysregulation—particularly when evaluating the long-term immunopathological consequences of the COVID-19 pandemic on lupus and related autoimmune conditions.
Conclusions
This study identified a declining trend in incident lupus cases during the COVID-19 pandemic among the DoD beneficiary population. Further investigation is warranted to elucidate the underlying factors contributing to this decline. Conducting longitudinal epidemiologic studies and applying multivariable regression analyses will be essential to determine whether incidence rates revert to prepandemic baselines and how these trends may be influenced by evolving environmental factors within the general population.
Systemic lupus erythematosus (SLE), or lupus, is a rare autoimmune disease estimated to occur in about 5.1 cases per 100,000 person-years in the United States in 2018.1 The disease predominantly affects females, with an incidence of 8.7 cases per 100,000 person-years vs 1.2 cases per 100,000 person-years in males, and is most common in patients aged 15 to 44 years.1,2
Lupus presents with a constellation of clinical signs and symptoms that evolve, along with hallmark laboratory findings indicative of immune dysregulation and polyclonal B-cell activation. Consequently, a wide array of autoantibodies may be produced, although the combination of epitope specificity can vary from patient to patient.3 Nevertheless, > 98% of individuals diagnosed with lupus produce antinuclear antibodies (ANA), making ANA positivity a near-universal serologic feature at the time of diagnosis.
The pathogenesis of lupus is complex. Research from the past 5 decades supports the role of certain viral infections—such as Epstein-Barr virus (EBV) and cytomegalovirus—as potential triggers.4 These viruses are thought to initiate disease through mechanisms including activation of interferon pathways, exposure of cryptic intracellular antigens, molecular mimicry, and epitope spreading. Subsequent clonal expansion and autoantibody production occur to varying degrees, influenced by viral load and host susceptibility factors.
During the COVID-19 pandemic, it became evident that SARS-CoV-2 exerts profound effects on immune regulation, influencing infection outcomes through mechanisms such as hyperactivation of innate immunity, especially in the lungs, leading to acute respiratory distress syndrome. Additionally, SARS-CoV-2 has been associated with polyclonal B-cell activation and the generation of autoantibodies. This association gained attention after Bastard et al identified anti–type I interferon antibodies in patients with severe COVID-19, predominantly among males with a genetic predisposition. These autoantibodies were shown to impair antiviral defenses and contribute to life-threatening pneumonia.5
Subsequent studies demonstrated the production of a wide spectrum of functional autoantibodies, including ANA, in patients with COVID-19.6,7 These findings were attributed to the acute expansion of autoreactive clones among naïve-derived immunoglobulin G1 antibody-secreting cells during the early stages of infection, with the degree of expansion correlating with disease severity.8,9 Although longitudinal data up to 15 months postinfection suggest this serologic abnormality resolves in more than two-thirds of patients, the number of individuals infected globally has raised serious public health concerns regarding the potential long-term sequelae, including the onset of lupus or other autoimmune diseases in COVID-19 survivors.6-9 A limited number of case reports describing the onset of lupus following SARS-CoV-2 infection support this hypothesis.10
This surveillance analysis investigates lupus incidence among patients within the Military Health System (MHS), encompassing all TRICARE beneficiaries, from January 2018 to December 2022. The objective of this analysis was to examine lupus incidence trends throughout the COVID-19 pandemic, stratified by sex, age, and active-duty status.
Methods
The MHS provides health care services to about 9.5 million US Department of Defense (DoD) beneficiaries. Outpatient health records and laboratory results for individuals receiving care at military treatment facilities (MTFs) between January 1, 2018, and December 31, 2022, were obtained from the Comprehensive Ambulatory/ Professional Encounter Record and MHS GENESIS. For beneficiaries receiving care in the private sector, data were sourced from the TRICARE Encounter Data—Non-Institutional database.
Laboratory test results, including ANA testing, were available only for individuals receiving care at MTFs. These laboratory data were extracted from the Composite Health Care System Chemistry database and MHS GENESIS laboratory systems for the same time frame. Inpatient data were not included in this analysis. Data from 2017 were used solely as a look-back (or washout) period to identify and exclude prevalent lupus cases diagnosed before 2018 and were not included in the final results.
Lupus cases were identified by the presence of a positive ANA test and appropriate International Classification of Diseases, 10th Revision, Clinical Modification (ICD-10-CM) codes. A positive ANA result was defined as either a qualitative result marked positive or a titer ≥ 1:80. The ICD-10-CM codes considered indicative of lupus included variations of M32, L93, or H01.12.
M32, L93, or H01.12. For cases with a positive ANA test, a lupus diagnosis required the presence of ≥ 2 lupus related ICD-10-CM codes. In the absence of ANA test results, a stricter criterion was applied: ≥ 4 lupus ICD-10-CM diagnosis codes recorded on separate days were required for inclusion.
Beneficiaries were excluded if they had a negative ANA result, only 1 lupus ICD- 10-CM diagnosis code, 1 positive ANA with only 1 corresponding ICD-10-CM code, or if their diagnosis occurred outside the defined study period. Patients and members of the public were not involved in the design, conduct, reporting, or dissemination of this study.
Results
Between January 1, 2017, and December 31, 2022, 99,946 TRICARE beneficiaries had some indication of lupus testing or diagnosis in their health records (Figure 1). Of these beneficiaries, 5335 had a positive ANA result and ≥ 2 ICD-10-CM lupus diagnosis codes. An additional 28,275 beneficiaries had ≥ 4 ICD-10-CM lupus diagnosis codes but no ANA test results. From these groups, the final sample included 10,760 beneficiaries who met the incident case definitions for SLE during the study period (2018 through 2022).

Most cases (85.1%, n = 9157) were diagnosed through TRICARE claims, while 1205 (11.2%) were diagnosed within the MHS. Another 398 (3.7%) had documentation of care both within and outside the MHS. Incident SLE cases declined by an average of 16% annually during the study period (Figure 2). This trend amounted to an overall reduction of 48.2%, from 2866 cases in 2018 to 1399 cases in 2022. This decline occurred despite total medical encounters among DoD beneficiaries remaining relatively stable during the pandemic years, with only a 3.5% change between 2018 and 2022.

The disease was more prevalent among female beneficiaries, with a female to- male ratio of 7:1 (Table 1). Among women, the number of new cases declined from 2519 in 2018 to 1223 in 2022, while the number of cases among men remained consistently < 350 annually. Similar trends were observed across other strata. Incident SLE cases were more common among nonactive-duty beneficiaries than active-duty service members, with a ratio of 18:1. New cases among active-duty members remained < 155 per year. Age-stratified data revealed that SLE was diagnosed predominantly in individuals aged ≥ 18 years, with a ratio of 37:1 compared with individuals aged < 18 years. Among children, the number of new cases remained < 75 per year throughout the study period.

A mean 56,850 ANA tests were conducted annually in centralized laboratories using standardized protocols (Table 2). The mean ANA positivity rate was 17.3%, which remained relatively stable from 2018 through 2022.

Discussion
This study examined the annual incidence of newly diagnosed SLE cases among all TRICARE beneficiaries from January 1, 2018, through December 31, 2022, covering both before and during the peak years of the COVID-19 pandemic. This analysis revealed a steady decline in SLE cases during this period. The reliability of these findings is reinforced by the comprehensiveness of the MHS, one of the largest US health care delivery systems, which maintains near-complete medical data capture for about 9.5 million DoD TRICARE beneficiaries across domestic and international settings.
SLE is a rare autoimmune disorder that presents a diagnostic challenge due to its wide range of nonspecific symptoms, many of which resemble other conditions. To reduce the likelihood of false-positive results and ensure diagnostic accuracy, this study adopted a stringent case definition. Incident cases were identified by the presence of ANA testing in conjunction with lupus-specific ICD-10-CM codes and required ≥ 4 lupus related diagnostic entries. This criterion was necessary due to the absence of ANA test results in data from private sector care settings. Our case definition aligns with established literature. For example, a Vanderbilt University chart review study demonstrated that combining ANA positivity with ≥ 4 lupus related ICD-10-CM codes achieves a positive predictive value of 100%, albeit with a sensitivity of 45%.11 Other studies similarly affirm the diagnostic validity of using recurrent ICD-10-CM codes to improve specificity in identifying lupus cases.12,13
The primary objective of this study was to examine the temporal trend in newly diagnosed lupus cases, rather than derive precise incidence rates. Although the TRICARE system includes about 9.5 million beneficiaries, this number represents a dynamic population with continual inflow and outflow. Accurate incidence rate calculation would require access to detailed denominator data, which were not readily available. In comparison with our findings, a study limited to active-duty service members reported fewer lupus cases. This discrepancy likely reflects differences in case definitions—specifically, the absence of laboratory data, the restricted range of diagnostic codes, and the requirement that diagnoses be rendered by specialists.14 Despite these differences, demographic patterns were consistent, with higher incidence observed in females and individuals aged ≥ 20 years.
A Centers for Disease Control and Prevention (CDC) study of lupus incidence in the general population also reported lower case counts.1 However, the CDC estimates were based on 5 state-level registries, which rely on clinician-reported cases and therefore may underestimate true disease burden. Moreover, the DoD beneficiary population differs markedly from the general population: it includes a large cohort of retirees, ensuring an older demographic; all members have comprehensive health care access; and active-duty personnel are subject to pre-enlistment medical screening. Taken together, these factors suggest this study may offer a more complete and systematically captured profile of lupus incidence.
We observed a marked decline of newly diagnosed SLE cases during the study period, which coincided with the widespread circulation of COVID-19. This decrease is unlikely to be attributable to reduced access to care during the pandemic. The MHS operates under a single-payer model, and the total number of patient encounters remained relatively stable throughout the pandemic.
To our knowledge, this is the only study to monitor lupus incidence in a large US population over the 5-year period encompassing before and during the COVID-19 pandemic. To date, only 4 large-scale surveillance studies have addressed similar questions. 14-17 Our findings are consistent with the most recent of these reports: an analysis limited to active-duty members of the US Armed Forces identified 1127 patients with newly diagnosed lupus between 2000 and 2022 and reported stable incidence trends throughout the pandemic.14 The other 3 studies adopted a different approach, comparing the emergence of autoimmune diseases, including lupus, between individuals with confirmed SARS-CoV-2 infection and those without. Each of these trials concluded that COVID-19 increases the risk of various autoimmune conditions, although the findings specific to lupus were inconsistent.15-17
Chang et al reported a significant increase in new lupus diagnoses (n = 2,926,016), with an adjusted hazard ratio (aHR) of 2.99 (95% CI, 2.68-3.34), spanning all ages and both sexes. The highest incidence was observed in individuals of Asian descent.15 Using German routine health care data from 2020, Tesch et al identified a heightened risk of autoimmune diseases, including lupus, among patients with a history of SARS-CoV-2 infection (n = 641,407; 9.4% children, 57.3% female, 6.4% hospitalized), compared with matched infection-naïve controls (n = 1,560,357).16 Both studies excluded vaccinated individuals.
These 2 studies diverged in their assessment of the relationship between COVID-19 severity and subsequent autoimmune risk. Chang et al found a higher incidence among nonhospitalized ambulatory patients, while Tesch et al reported that increased risk was associated with patients requiring intensive care unit admission.15,16
In contrast, based on a cohort of 4,197,188 individuals, Peng et al found no significant difference in lupus incidence among patients with SARS-CoV-2 infection (aHR, 1.05; 95% CI, 0.79-1.39).17 Notably, within the infected group, the incidence of SLE was significantly lower among vaccinated individuals compared with the unvaccinated group (aHR, 0.29; 95% CI, 0.18-0.47). Similar protective associations were observed for other antibody-mediated autoimmune disorders, including pemphigoid, Graves’ disease, and antiphospholipid antibody syndrome.
Limitations
Due to fundamental differences in study design, we were unable to directly reconcile our findings with those reported in the literature. This study lacked access to reliable documentation of COVID-19 infection status, primarily due to the widespread use of home testing among TRICARE beneficiaries. Additionally, the dataset did not include inpatient records and therefore did not permit evaluation of disease severity. Despite these constraints, it is plausible that the overall burden of COVID-19 infection within the study population was lower than that observed in the general US population.
As of December 2022, the DoD had reported about 750,000 confirmed COVID-19 cases among military personnel, civilian employees, dependents, and DoD contractors.18 Given that TRICARE beneficiaries represent about 2.8% of the total US population—and that > 90 million US individuals were infected between 2020 and 2022—the implied infection rate in our cohort appears to be about one-third of what might be expected.19 This discrepancy may be due to higher adherence to mitigation measures, such as social distancing and mask usage, among DoD-affiliated populations. COVID-19 vaccination was mandated for all active-duty service members, who constitute 5.4% of the study population. The remaining TRICARE beneficiaries also had access to guaranteed health care and vaccination coverage, likely contributing to high overall vaccination rates.
Because > 80% of the study population was composed of individuals from diverse civilian backgrounds, we expect the distribution of infection severity within the DoD beneficiary population to approximate that of the general US population.
Future Directions
The findings of this study offer circumstantial yet real-time evidence of the complexity underlying immune dysregulation at the intersection of host susceptibility and environmental exposures. The stability in ANA positivity rates during the study period mitigates concerns regarding undiagnosed subclinical lupus and may suggest that, overall, immune homeostasis was preserved among DoD beneficiaries.
It is noteworthy that during the COVID-19 pandemic, the incidence of several common infections—such as influenza and EBV—declined markedly, likely as a result of widespread social distancing and other public health interventions.20 Mitigation strategies implemented within the military may have conferred protection not only against COVID-19 but also against other community-acquired pathogens.
In light of these observations, we hypothesize that for COVID-19 to act as a trigger for SLE, a prolonged or repeated disruption of immune equilibrium may be required—potentially mediated by recurrent infections or sustained inflammatory states. The association between viral infections and autoimmunity is well established. Immune dysregulation leading to autoantibody production has been observed not only in the context of SARS-CoV-2 but also following infections with EBV, cytomegalovirus, enteroviruses, hepatitis B and C viruses, HIV, and parvovirus B19.21
This dysregulation is often transient, accompanied by compensatory immune regulatory responses. However, in individuals subjected to successive or overlapping infections, these regulatory mechanisms may become compromised or overwhelmed, due to emergent patterns of immune interference of varying severity. In such cases, a transient immune perturbation may progress into a bona fide autoimmune disease, contingent upon individual risk factors such as genetic predisposition, preexisting immune memory, and regenerative capacity.21
Therefore, we believe the significance of this study is 2-fold. First, lupus is known to develop gradually and may require 3 to 5 years to clinically manifest after the initial break in immunological tolerance.3 Continued public health surveillance represents a more pragmatic strategy than retrospective cohort construction, especially as histories of COVID-19 infection become increasingly complete and definitive. Our findings provide a valuable baseline reference point for future longitudinal studies.
The interpretation of surveillance outcomes—whether indicating an upward trend, a stable baseline, or a downward trend—offers distinct analytical value. Within this study population, we observed neither an upward trajectory that might suggest a direct causal link, nor a flat trend that would imply absence of association between COVID-19 and lupus pathogenesis. Instead, the observation of a downward trend invites consideration of nonlinear or protective influences. From this perspective, we recommend that future investigations adopt a holistic framework when assessing environmental contributions to immune dysregulation—particularly when evaluating the long-term immunopathological consequences of the COVID-19 pandemic on lupus and related autoimmune conditions.
Conclusions
This study identified a declining trend in incident lupus cases during the COVID-19 pandemic among the DoD beneficiary population. Further investigation is warranted to elucidate the underlying factors contributing to this decline. Conducting longitudinal epidemiologic studies and applying multivariable regression analyses will be essential to determine whether incidence rates revert to prepandemic baselines and how these trends may be influenced by evolving environmental factors within the general population.
Systemic lupus erythematosus (SLE), or lupus, is a rare autoimmune disease estimated to occur in about 5.1 cases per 100,000 person-years in the United States in 2018.1 The disease predominantly affects females, with an incidence of 8.7 cases per 100,000 person-years vs 1.2 cases per 100,000 person-years in males, and is most common in patients aged 15 to 44 years.1,2
Lupus presents with a constellation of clinical signs and symptoms that evolve, along with hallmark laboratory findings indicative of immune dysregulation and polyclonal B-cell activation. Consequently, a wide array of autoantibodies may be produced, although the combination of epitope specificity can vary from patient to patient.3 Nevertheless, > 98% of individuals diagnosed with lupus produce antinuclear antibodies (ANA), making ANA positivity a near-universal serologic feature at the time of diagnosis.
The pathogenesis of lupus is complex. Research from the past 5 decades supports the role of certain viral infections—such as Epstein-Barr virus (EBV) and cytomegalovirus—as potential triggers.4 These viruses are thought to initiate disease through mechanisms including activation of interferon pathways, exposure of cryptic intracellular antigens, molecular mimicry, and epitope spreading. Subsequent clonal expansion and autoantibody production occur to varying degrees, influenced by viral load and host susceptibility factors.
During the COVID-19 pandemic, it became evident that SARS-CoV-2 exerts profound effects on immune regulation, influencing infection outcomes through mechanisms such as hyperactivation of innate immunity, especially in the lungs, leading to acute respiratory distress syndrome. Additionally, SARS-CoV-2 has been associated with polyclonal B-cell activation and the generation of autoantibodies. This association gained attention after Bastard et al identified anti–type I interferon antibodies in patients with severe COVID-19, predominantly among males with a genetic predisposition. These autoantibodies were shown to impair antiviral defenses and contribute to life-threatening pneumonia.5
Subsequent studies demonstrated the production of a wide spectrum of functional autoantibodies, including ANA, in patients with COVID-19.6,7 These findings were attributed to the acute expansion of autoreactive clones among naïve-derived immunoglobulin G1 antibody-secreting cells during the early stages of infection, with the degree of expansion correlating with disease severity.8,9 Although longitudinal data up to 15 months postinfection suggest this serologic abnormality resolves in more than two-thirds of patients, the number of individuals infected globally has raised serious public health concerns regarding the potential long-term sequelae, including the onset of lupus or other autoimmune diseases in COVID-19 survivors.6-9 A limited number of case reports describing the onset of lupus following SARS-CoV-2 infection support this hypothesis.10
This surveillance analysis investigates lupus incidence among patients within the Military Health System (MHS), encompassing all TRICARE beneficiaries, from January 2018 to December 2022. The objective of this analysis was to examine lupus incidence trends throughout the COVID-19 pandemic, stratified by sex, age, and active-duty status.
Methods
The MHS provides health care services to about 9.5 million US Department of Defense (DoD) beneficiaries. Outpatient health records and laboratory results for individuals receiving care at military treatment facilities (MTFs) between January 1, 2018, and December 31, 2022, were obtained from the Comprehensive Ambulatory/ Professional Encounter Record and MHS GENESIS. For beneficiaries receiving care in the private sector, data were sourced from the TRICARE Encounter Data—Non-Institutional database.
Laboratory test results, including ANA testing, were available only for individuals receiving care at MTFs. These laboratory data were extracted from the Composite Health Care System Chemistry database and MHS GENESIS laboratory systems for the same time frame. Inpatient data were not included in this analysis. Data from 2017 were used solely as a look-back (or washout) period to identify and exclude prevalent lupus cases diagnosed before 2018 and were not included in the final results.
Lupus cases were identified by the presence of a positive ANA test and appropriate International Classification of Diseases, 10th Revision, Clinical Modification (ICD-10-CM) codes. A positive ANA result was defined as either a qualitative result marked positive or a titer ≥ 1:80. The ICD-10-CM codes considered indicative of lupus included variations of M32, L93, or H01.12.
M32, L93, or H01.12. For cases with a positive ANA test, a lupus diagnosis required the presence of ≥ 2 lupus related ICD-10-CM codes. In the absence of ANA test results, a stricter criterion was applied: ≥ 4 lupus ICD-10-CM diagnosis codes recorded on separate days were required for inclusion.
Beneficiaries were excluded if they had a negative ANA result, only 1 lupus ICD- 10-CM diagnosis code, 1 positive ANA with only 1 corresponding ICD-10-CM code, or if their diagnosis occurred outside the defined study period. Patients and members of the public were not involved in the design, conduct, reporting, or dissemination of this study.
Results
Between January 1, 2017, and December 31, 2022, 99,946 TRICARE beneficiaries had some indication of lupus testing or diagnosis in their health records (Figure 1). Of these beneficiaries, 5335 had a positive ANA result and ≥ 2 ICD-10-CM lupus diagnosis codes. An additional 28,275 beneficiaries had ≥ 4 ICD-10-CM lupus diagnosis codes but no ANA test results. From these groups, the final sample included 10,760 beneficiaries who met the incident case definitions for SLE during the study period (2018 through 2022).

Most cases (85.1%, n = 9157) were diagnosed through TRICARE claims, while 1205 (11.2%) were diagnosed within the MHS. Another 398 (3.7%) had documentation of care both within and outside the MHS. Incident SLE cases declined by an average of 16% annually during the study period (Figure 2). This trend amounted to an overall reduction of 48.2%, from 2866 cases in 2018 to 1399 cases in 2022. This decline occurred despite total medical encounters among DoD beneficiaries remaining relatively stable during the pandemic years, with only a 3.5% change between 2018 and 2022.

The disease was more prevalent among female beneficiaries, with a female to- male ratio of 7:1 (Table 1). Among women, the number of new cases declined from 2519 in 2018 to 1223 in 2022, while the number of cases among men remained consistently < 350 annually. Similar trends were observed across other strata. Incident SLE cases were more common among nonactive-duty beneficiaries than active-duty service members, with a ratio of 18:1. New cases among active-duty members remained < 155 per year. Age-stratified data revealed that SLE was diagnosed predominantly in individuals aged ≥ 18 years, with a ratio of 37:1 compared with individuals aged < 18 years. Among children, the number of new cases remained < 75 per year throughout the study period.

A mean 56,850 ANA tests were conducted annually in centralized laboratories using standardized protocols (Table 2). The mean ANA positivity rate was 17.3%, which remained relatively stable from 2018 through 2022.

Discussion
This study examined the annual incidence of newly diagnosed SLE cases among all TRICARE beneficiaries from January 1, 2018, through December 31, 2022, covering both before and during the peak years of the COVID-19 pandemic. This analysis revealed a steady decline in SLE cases during this period. The reliability of these findings is reinforced by the comprehensiveness of the MHS, one of the largest US health care delivery systems, which maintains near-complete medical data capture for about 9.5 million DoD TRICARE beneficiaries across domestic and international settings.
SLE is a rare autoimmune disorder that presents a diagnostic challenge due to its wide range of nonspecific symptoms, many of which resemble other conditions. To reduce the likelihood of false-positive results and ensure diagnostic accuracy, this study adopted a stringent case definition. Incident cases were identified by the presence of ANA testing in conjunction with lupus-specific ICD-10-CM codes and required ≥ 4 lupus related diagnostic entries. This criterion was necessary due to the absence of ANA test results in data from private sector care settings. Our case definition aligns with established literature. For example, a Vanderbilt University chart review study demonstrated that combining ANA positivity with ≥ 4 lupus related ICD-10-CM codes achieves a positive predictive value of 100%, albeit with a sensitivity of 45%.11 Other studies similarly affirm the diagnostic validity of using recurrent ICD-10-CM codes to improve specificity in identifying lupus cases.12,13
The primary objective of this study was to examine the temporal trend in newly diagnosed lupus cases, rather than derive precise incidence rates. Although the TRICARE system includes about 9.5 million beneficiaries, this number represents a dynamic population with continual inflow and outflow. Accurate incidence rate calculation would require access to detailed denominator data, which were not readily available. In comparison with our findings, a study limited to active-duty service members reported fewer lupus cases. This discrepancy likely reflects differences in case definitions—specifically, the absence of laboratory data, the restricted range of diagnostic codes, and the requirement that diagnoses be rendered by specialists.14 Despite these differences, demographic patterns were consistent, with higher incidence observed in females and individuals aged ≥ 20 years.
A Centers for Disease Control and Prevention (CDC) study of lupus incidence in the general population also reported lower case counts.1 However, the CDC estimates were based on 5 state-level registries, which rely on clinician-reported cases and therefore may underestimate true disease burden. Moreover, the DoD beneficiary population differs markedly from the general population: it includes a large cohort of retirees, ensuring an older demographic; all members have comprehensive health care access; and active-duty personnel are subject to pre-enlistment medical screening. Taken together, these factors suggest this study may offer a more complete and systematically captured profile of lupus incidence.
We observed a marked decline of newly diagnosed SLE cases during the study period, which coincided with the widespread circulation of COVID-19. This decrease is unlikely to be attributable to reduced access to care during the pandemic. The MHS operates under a single-payer model, and the total number of patient encounters remained relatively stable throughout the pandemic.
To our knowledge, this is the only study to monitor lupus incidence in a large US population over the 5-year period encompassing before and during the COVID-19 pandemic. To date, only 4 large-scale surveillance studies have addressed similar questions. 14-17 Our findings are consistent with the most recent of these reports: an analysis limited to active-duty members of the US Armed Forces identified 1127 patients with newly diagnosed lupus between 2000 and 2022 and reported stable incidence trends throughout the pandemic.14 The other 3 studies adopted a different approach, comparing the emergence of autoimmune diseases, including lupus, between individuals with confirmed SARS-CoV-2 infection and those without. Each of these trials concluded that COVID-19 increases the risk of various autoimmune conditions, although the findings specific to lupus were inconsistent.15-17
Chang et al reported a significant increase in new lupus diagnoses (n = 2,926,016), with an adjusted hazard ratio (aHR) of 2.99 (95% CI, 2.68-3.34), spanning all ages and both sexes. The highest incidence was observed in individuals of Asian descent.15 Using German routine health care data from 2020, Tesch et al identified a heightened risk of autoimmune diseases, including lupus, among patients with a history of SARS-CoV-2 infection (n = 641,407; 9.4% children, 57.3% female, 6.4% hospitalized), compared with matched infection-naïve controls (n = 1,560,357).16 Both studies excluded vaccinated individuals.
These 2 studies diverged in their assessment of the relationship between COVID-19 severity and subsequent autoimmune risk. Chang et al found a higher incidence among nonhospitalized ambulatory patients, while Tesch et al reported that increased risk was associated with patients requiring intensive care unit admission.15,16
In contrast, based on a cohort of 4,197,188 individuals, Peng et al found no significant difference in lupus incidence among patients with SARS-CoV-2 infection (aHR, 1.05; 95% CI, 0.79-1.39).17 Notably, within the infected group, the incidence of SLE was significantly lower among vaccinated individuals compared with the unvaccinated group (aHR, 0.29; 95% CI, 0.18-0.47). Similar protective associations were observed for other antibody-mediated autoimmune disorders, including pemphigoid, Graves’ disease, and antiphospholipid antibody syndrome.
Limitations
Due to fundamental differences in study design, we were unable to directly reconcile our findings with those reported in the literature. This study lacked access to reliable documentation of COVID-19 infection status, primarily due to the widespread use of home testing among TRICARE beneficiaries. Additionally, the dataset did not include inpatient records and therefore did not permit evaluation of disease severity. Despite these constraints, it is plausible that the overall burden of COVID-19 infection within the study population was lower than that observed in the general US population.
As of December 2022, the DoD had reported about 750,000 confirmed COVID-19 cases among military personnel, civilian employees, dependents, and DoD contractors.18 Given that TRICARE beneficiaries represent about 2.8% of the total US population—and that > 90 million US individuals were infected between 2020 and 2022—the implied infection rate in our cohort appears to be about one-third of what might be expected.19 This discrepancy may be due to higher adherence to mitigation measures, such as social distancing and mask usage, among DoD-affiliated populations. COVID-19 vaccination was mandated for all active-duty service members, who constitute 5.4% of the study population. The remaining TRICARE beneficiaries also had access to guaranteed health care and vaccination coverage, likely contributing to high overall vaccination rates.
Because > 80% of the study population was composed of individuals from diverse civilian backgrounds, we expect the distribution of infection severity within the DoD beneficiary population to approximate that of the general US population.
Future Directions
The findings of this study offer circumstantial yet real-time evidence of the complexity underlying immune dysregulation at the intersection of host susceptibility and environmental exposures. The stability in ANA positivity rates during the study period mitigates concerns regarding undiagnosed subclinical lupus and may suggest that, overall, immune homeostasis was preserved among DoD beneficiaries.
It is noteworthy that during the COVID-19 pandemic, the incidence of several common infections—such as influenza and EBV—declined markedly, likely as a result of widespread social distancing and other public health interventions.20 Mitigation strategies implemented within the military may have conferred protection not only against COVID-19 but also against other community-acquired pathogens.
In light of these observations, we hypothesize that for COVID-19 to act as a trigger for SLE, a prolonged or repeated disruption of immune equilibrium may be required—potentially mediated by recurrent infections or sustained inflammatory states. The association between viral infections and autoimmunity is well established. Immune dysregulation leading to autoantibody production has been observed not only in the context of SARS-CoV-2 but also following infections with EBV, cytomegalovirus, enteroviruses, hepatitis B and C viruses, HIV, and parvovirus B19.21
This dysregulation is often transient, accompanied by compensatory immune regulatory responses. However, in individuals subjected to successive or overlapping infections, these regulatory mechanisms may become compromised or overwhelmed, due to emergent patterns of immune interference of varying severity. In such cases, a transient immune perturbation may progress into a bona fide autoimmune disease, contingent upon individual risk factors such as genetic predisposition, preexisting immune memory, and regenerative capacity.21
Therefore, we believe the significance of this study is 2-fold. First, lupus is known to develop gradually and may require 3 to 5 years to clinically manifest after the initial break in immunological tolerance.3 Continued public health surveillance represents a more pragmatic strategy than retrospective cohort construction, especially as histories of COVID-19 infection become increasingly complete and definitive. Our findings provide a valuable baseline reference point for future longitudinal studies.
The interpretation of surveillance outcomes—whether indicating an upward trend, a stable baseline, or a downward trend—offers distinct analytical value. Within this study population, we observed neither an upward trajectory that might suggest a direct causal link, nor a flat trend that would imply absence of association between COVID-19 and lupus pathogenesis. Instead, the observation of a downward trend invites consideration of nonlinear or protective influences. From this perspective, we recommend that future investigations adopt a holistic framework when assessing environmental contributions to immune dysregulation—particularly when evaluating the long-term immunopathological consequences of the COVID-19 pandemic on lupus and related autoimmune conditions.
Conclusions
This study identified a declining trend in incident lupus cases during the COVID-19 pandemic among the DoD beneficiary population. Further investigation is warranted to elucidate the underlying factors contributing to this decline. Conducting longitudinal epidemiologic studies and applying multivariable regression analyses will be essential to determine whether incidence rates revert to prepandemic baselines and how these trends may be influenced by evolving environmental factors within the general population.
A Systemic Lupus Erythematosus Incidence Surveillance Report Among DoD Beneficiaries During the COVID-19 Pandemic
A Systemic Lupus Erythematosus Incidence Surveillance Report Among DoD Beneficiaries During the COVID-19 Pandemic
- Izmirly PM, Ferucci ED, Somers EC, et al. Incidence rates of systemic lupus erythematosus in the USA: estimates from a meta-analysis of the Centers for Disease Control and Prevention national lupus registries. Lupus Sci Med. 2021;8(1):e000614. doi:10.1136/lupus-2021-000614
- Centers for Disease Control and Prevention. People with lupus. May 15, 2024. Accessed May 10, 2025. https:// www.cdc.gov/lupus/data-research/index.html
- Arbuckle MR, McClain MT, Rubertone MV, et al. Development of autoantibodies before the clinical onset of systemic lupus erythematosus. N Engl J Med. 2003;349(16):1526-1533. doi:10.1056/nejmoa021933
- Li ZX, Zeng S, Wu HX, Zhou Y. The risk of systemic lupus erythematosus associated with Epstein–Barr virus infection: a systematic review and meta-analysis. Clin Exp Med. 2019;19(1):23-36. doi:10.1007/s10238-018-0535-0
- Bastard P, Rosen LB, Zhang Q, et al. Autoantibodies against type I IFNs in patients with life-threatening COVID-19. Science. 2020;370(6515):eabd4585. doi:10.1126/science.abd4585
- Chang SE, Feng A, Meng W, et al. New-onset IgG autoantibodies in hospitalized patients with COVID-19. Nat Commun. 2021;12(1):5417. doi:10.1038/s41467-021-25509-3
- Lee SJ, Yoon T, Ha JW, et al. Prevalence, clinical significance, and persistence of autoantibodies in COVID-19. Virol J. 2023;20(1):236. doi:10.1186/s12985-023-02191-z
- Woodruff MC, Ramonell RP, Haddad NS, et al. Dysregulated naive B cells and de novo autoreactivity in severe COVID-19. Nature. 2022;611(7934):139-147. doi:10.1038/s41586-022-05273-0
- Taeschler P, Cervia C, Zurbuchen Y, et al. Autoantibodies in COVID-19 correlate with antiviral humoral responses and distinct immune signatures. Allergy. 2022;77(8):2415-2430. doi:10.1111/all.15302
- Gracia-Ramos AE, Martin-Nares E, Hernández-Molina G. New onset of autoimmune diseases following COVID-19 diagnosis. Cells. 2021;10(12):3592 doi:10.3390/cells10123592
- Barnado A, Carroll R, Denny JC, Crofford L. Using IC-10-CM codes to identify patients with systemic lupus erythematosus in the electronic health record [abstract]. Arthritis Rheumatol. 2018;70(suppl 9):abstract 1692. Accessed May 10, 2025. https://acrabstracts.org/abstract/using-icd-10-cm-codes-to-identify-patients-with-systemic-lupus-erythematosus-in-the-electronic-health-record
- Feldman C, Curtis JR, Oates JC, Yazdany J, Izmirly P. Validating claims-based algorithms for a systemic lupus erythematosus diagnosis in Medicare data for informed use of the Lupus Index: a tool for geospatial research. Lupus Sci Med. 2024;11(2):e001329. doi:10.1136/lupus-2024-001329
- Moe SR, Haukeland H, Brunborg C, et al. POS1472: Accuracy of disease-specific ICD-10 code for incident systemic lupus erythematosus; results from a population-based cohort study set in Norway [abstract]. Ann Rheum Dis. 2023;82(suppl 1):1090-1091. doi:10.1136/annrheumdis-2023-eular.1189
- Denagamage P, Mabila SL, McQuistan AA. Trends and disparities in systemic lupus erythematosus incidence among U.S. active component service members, 2000–2022. MSMR. 2023;30(12):2-5.
- Chang R, Yen-Ting Chen T, Wang SI, Hung YM, Chen HY, Wei CJ. Risk of autoimmune diseases in patients with COVID-19: a retrospective cohort study. EClinicalMedicine. 2023;56:101783. doi:10.1016/j.eclinm.2022.101783
- Tesch F, Ehm F, Vivirito A, et al. Incident autoimmune diseases in association with SARS-CoV-2 infection: a matched cohort study. Clin Rheumatol. 2023;42(10):2905- 2914. doi:10.1007/s10067-023-06670-0
- Peng K, Li X, Yang D, et al. Risk of autoimmune diseases following COVID-19 and the potential protective effect from vaccination: a population-based cohort study. EClinicalMedicine. 2023;63:102154. doi:10.1016/j.eclinm.2023.102154
- US Department of Defense. Coronavirus: DOD response. Updated December 20, 2022. Accessed May 10, 2025. https://www.defense.gov/Spotlights/Coronavirus-DOD-Response/
- Elflein J. Number of cumulative cases of COVID-19 in the United States from January 20, 2020 to November 11, 2022, by week. Statista. https://www.statista.com/statistics/1103185/cumulative-coronavirus-covid19-cases-number-us-by-day
- Ye Z, Chen L, Zhong H, Cao L, Fu P, Xu J. Epidemiology and clinical characteristics of Epstein-Barr virus infection among children in Shanghai, China, 2017- 2022. Front Cell Infect Microbiol. 2023;13:1139068. doi:10.3389/fcimb.2023.1139068
- Johnson D, Jiang W. Infectious diseases, autoantibodies, and autoimmunity. J Autoimmun. 2023;137:102962. doi:10.1016/j.jaut.2022.102962
Irritable Bowel Syndrome Risk in Acne Patients: Implications for Dermatologic Care
To the Editor:
Acne vulgaris and irritable bowel syndrome (IBS) are both associated with microbial dysbiosis and chronic inflammation.1-3 While the prevalence of IBS among patients with acne has been examined previously,4,5 there has been limited focus on the risk for new-onset IBS following acne diagnosis. Current evidence suggests isotretinoin may be associated with a lower risk for IBS compared to oral antibiotics6; however, evidence supporting this association is limited outside these cohorts, highlighting the need for further investigation. In this large-scale study, we sought to investigate the incidence of new-onset IBS among patients with acne compared with healthy controls as well as to evaluate whether oral acne treatments (ie, oral antibiotics or isotretinoin) are associated with new-onset IBS in this population.
A retrospective cohort study was conducted using data from the US Collaborative Network in TriNetX from October 2014 to October 2024. Patients were identified using International Classification of Diseases, Tenth Revision, Clinical Modification codes, Current Procedural Terminology codes, Anatomical Therapeutic Chemical Classification System codes, and RxNorm codes (Table 1). These codes were selected based on prior literature review, clinical relevance, and their ability to capture diagnoses of acne and IBS as well as relevant exclusion criteria. Patients were considered eligible if they were between the ages of 18 and 90 years. Individuals with a history of IBS, inflammatory bowel disease, infectious gastroenteritis, or celiac disease were excluded from our analysis.

To examine potential associations between acne and IBS, 2 primary cohorts were established: patients with acne who were managed without systemic medications and healthy controls (ie, patients with no history of acne) who had no exposure to systemic acne treatments (Figure). Further, to assess the relationship between oral acne treatments (macrolides, tetracyclines, isotretinoin) and IBS, additional cohorts were created for each therapy and were compared to a cohort of patients with acne who were managed without systemic medications. To control for potential concomitant treatments, patients who had received any systemic treatment other than the specific therapy for their treatment cohort were excluded from our analysis.

To account for potential confounders, all cohorts were 1:1 propensity score matched by demographics, tobacco and alcohol use, type 2 diabetes, obesity, anxiety, and depression (eTable). Each cohort was followed for 2 years after their index of event: the date of acne diagnosis for the acne cohort, the date of systemic treatment initiation for the treatment cohorts, and the date of the general adult encounter without abnormal findings for the control cohort. The primary outcome was the incidence of IBS, assessed by odds ratio (OR) and 95% CIs.
We identified 375,944 patients with acne managed without systemic treatment and 3,148,443 healthy controls who met study criteria. After the 1:1 propensity score match, each cohort included 49,690 patients (eTable). In the 2-year period after acne diagnosis, patients were more likely to develop IBS compared with controls (1421 vs 1285 [OR, 1.10; 95% CI, 1.02-1.19])(Table 2). Patients with acne who were treated with tetracyclines (n=208,971) were 30% more likely to develop IBS than those managed without systemic medications (1114 vs 856 [OR, 1.30; 95% CI, 1.19-1.42]). Within the tetracycline cohort, doxycycline-treated patients were 25% more likely to develop IBS compared with those treated with minocycline (213 vs 170 [OR, 1.25; 95% CI, 1.02-1.53]). Similarly, the use of macrolides (n=136,334) for acne treatment was significantly associated with an increased risk for IBS (1023 vs 595 [OR, 1.73; 95% CI, 1.57-1.92; P<.0001]) compared with controls. No statistically significant association was observed between isotretinoin and the incidence of IBS (Table 2).


In this large-scale cohort study, acne was associated with an increased likelihood of developing IBS within 2 years of an acne diagnosis compared with healthy controls. While a prior study also identified this association, it had a broader follow-up window ranging from 8 to 10 years.2 In contrast, our analysis specifically quantified the risk within the first 2 years of diagnosis. This distinction suggested potential for earlier IBS onset in patients with acne than has previously been recognized and may serve as an early clinical indicator for IBS risk in this population.
Our findings further suggested an association between oral tetracyclines and macrolides and an increased risk for IBS. This aligns with existing literature suggesting that oral antibiotic use can disrupt the gut microbiota and lead to potential gastrointestinal complications7 and reinforces the importance of careful antibiotic stewardship in dermatologic practice.
Although isotretinoin initially was surrounded by substantial controversy regarding its potential impact on gut health—particularly in inflammatory bowel disease8—our results do not support an increased risk for IBS among patients with acne who use isotretinoin. These findings challenge previous concerns and align with research suggesting that isotretinoin could be a safer alternative to antibiotic use for eligible patients who have a history of gastrointestinal disorders.6
This study highlights an important but underrecognized link between acne and IBS risk, emphasizing the need for early monitoring of gastrointestinal symptoms and careful antibiotic stewardship in dermatologic practice. Gastroenterology consultation may be advisable for patients with acne who have persistent gastrointestinal symptoms to facilitate a more integrated, patient-centered approach to care.
Limitations of this study include potential misclassification of International Classification of Diseases, Tenth Revision, Clinical Modification codes, selection bias, and residual confounding from unmeasured factors such as diet, lifestyle, disease severity, and treatment adherence due to the reliance on electronic health record data.
Our findings build upon prior evidence linking acne and IBS and offer important insights into the timing of this association following acne diagnosis. Future research should explore biological mechanisms underlying the gut-skin axis and evaluate targeted interventions to mitigate IBS risk in patients with acne.
Menees S, Chey W. The gut microbiome and irritable bowel syndrome. F1000Res. 2018;7:F1000 Faculty Rev-1029. doi:10.12688/f1000research.14592.1
Yu-Wen C, Chun-Ying W, Yi-Ju C. Gastrointestinal comorbidities in patients with acne vulgaris: a population-based retrospective study. JAAD Int. 2025;18:62-68. doi:10.1016/j.jdin.2024.08.022
Deng Y, Wang H, Zhou J, et al. Patients with acne vulgaris have a distinct gut microbiota in comparison with healthy controls. Acta Derm Venereol. 2018;98:783-790. doi:10.2340/00015555-2968
Demirbas¸ A, Elmas ÖF. The relationship between acne vulgaris and irritable bowel syndrome: a preliminary study. J Cosmet Dermatol. 2021;20:316-320. doi:10.1111/jocd.13481
Daye M, Cihan FG, Is¸ık B, et al. Evaluation of bowel habits in patients with acne vulgaris. Int J Clin Pract. 2021;75:e14903. doi:10.1111/ijcp.14903
Kridin K, Ludwig RJ. Isotretinoin and the risk of inflammatory bowel disease and irritable bowel syndrome: a large-scale global study. J Am Acad Dermatol. 2023;88:824-830. doi:10.1016/j.jaad.2022.12.015
Villarreal AA, Aberger FJ, Benrud R, et al. Use of broad-spectrum antibiotics and the development of irritable bowel syndrome. WMJ. 2012;111:17-20.
Yu C-L, Chou P-Y, Liang C-S, et al. Isotretinoin exposure and risk of inflammatory bowel disease: a systematic review with meta-analysis and trial sequential analysis. Am J Clin Dermatol. 2023;24:721-730. doi:10.1007/s40257-023-00765-9
To the Editor:
Acne vulgaris and irritable bowel syndrome (IBS) are both associated with microbial dysbiosis and chronic inflammation.1-3 While the prevalence of IBS among patients with acne has been examined previously,4,5 there has been limited focus on the risk for new-onset IBS following acne diagnosis. Current evidence suggests isotretinoin may be associated with a lower risk for IBS compared to oral antibiotics6; however, evidence supporting this association is limited outside these cohorts, highlighting the need for further investigation. In this large-scale study, we sought to investigate the incidence of new-onset IBS among patients with acne compared with healthy controls as well as to evaluate whether oral acne treatments (ie, oral antibiotics or isotretinoin) are associated with new-onset IBS in this population.
A retrospective cohort study was conducted using data from the US Collaborative Network in TriNetX from October 2014 to October 2024. Patients were identified using International Classification of Diseases, Tenth Revision, Clinical Modification codes, Current Procedural Terminology codes, Anatomical Therapeutic Chemical Classification System codes, and RxNorm codes (Table 1). These codes were selected based on prior literature review, clinical relevance, and their ability to capture diagnoses of acne and IBS as well as relevant exclusion criteria. Patients were considered eligible if they were between the ages of 18 and 90 years. Individuals with a history of IBS, inflammatory bowel disease, infectious gastroenteritis, or celiac disease were excluded from our analysis.

To examine potential associations between acne and IBS, 2 primary cohorts were established: patients with acne who were managed without systemic medications and healthy controls (ie, patients with no history of acne) who had no exposure to systemic acne treatments (Figure). Further, to assess the relationship between oral acne treatments (macrolides, tetracyclines, isotretinoin) and IBS, additional cohorts were created for each therapy and were compared to a cohort of patients with acne who were managed without systemic medications. To control for potential concomitant treatments, patients who had received any systemic treatment other than the specific therapy for their treatment cohort were excluded from our analysis.

To account for potential confounders, all cohorts were 1:1 propensity score matched by demographics, tobacco and alcohol use, type 2 diabetes, obesity, anxiety, and depression (eTable). Each cohort was followed for 2 years after their index of event: the date of acne diagnosis for the acne cohort, the date of systemic treatment initiation for the treatment cohorts, and the date of the general adult encounter without abnormal findings for the control cohort. The primary outcome was the incidence of IBS, assessed by odds ratio (OR) and 95% CIs.
We identified 375,944 patients with acne managed without systemic treatment and 3,148,443 healthy controls who met study criteria. After the 1:1 propensity score match, each cohort included 49,690 patients (eTable). In the 2-year period after acne diagnosis, patients were more likely to develop IBS compared with controls (1421 vs 1285 [OR, 1.10; 95% CI, 1.02-1.19])(Table 2). Patients with acne who were treated with tetracyclines (n=208,971) were 30% more likely to develop IBS than those managed without systemic medications (1114 vs 856 [OR, 1.30; 95% CI, 1.19-1.42]). Within the tetracycline cohort, doxycycline-treated patients were 25% more likely to develop IBS compared with those treated with minocycline (213 vs 170 [OR, 1.25; 95% CI, 1.02-1.53]). Similarly, the use of macrolides (n=136,334) for acne treatment was significantly associated with an increased risk for IBS (1023 vs 595 [OR, 1.73; 95% CI, 1.57-1.92; P<.0001]) compared with controls. No statistically significant association was observed between isotretinoin and the incidence of IBS (Table 2).


In this large-scale cohort study, acne was associated with an increased likelihood of developing IBS within 2 years of an acne diagnosis compared with healthy controls. While a prior study also identified this association, it had a broader follow-up window ranging from 8 to 10 years.2 In contrast, our analysis specifically quantified the risk within the first 2 years of diagnosis. This distinction suggested potential for earlier IBS onset in patients with acne than has previously been recognized and may serve as an early clinical indicator for IBS risk in this population.
Our findings further suggested an association between oral tetracyclines and macrolides and an increased risk for IBS. This aligns with existing literature suggesting that oral antibiotic use can disrupt the gut microbiota and lead to potential gastrointestinal complications7 and reinforces the importance of careful antibiotic stewardship in dermatologic practice.
Although isotretinoin initially was surrounded by substantial controversy regarding its potential impact on gut health—particularly in inflammatory bowel disease8—our results do not support an increased risk for IBS among patients with acne who use isotretinoin. These findings challenge previous concerns and align with research suggesting that isotretinoin could be a safer alternative to antibiotic use for eligible patients who have a history of gastrointestinal disorders.6
This study highlights an important but underrecognized link between acne and IBS risk, emphasizing the need for early monitoring of gastrointestinal symptoms and careful antibiotic stewardship in dermatologic practice. Gastroenterology consultation may be advisable for patients with acne who have persistent gastrointestinal symptoms to facilitate a more integrated, patient-centered approach to care.
Limitations of this study include potential misclassification of International Classification of Diseases, Tenth Revision, Clinical Modification codes, selection bias, and residual confounding from unmeasured factors such as diet, lifestyle, disease severity, and treatment adherence due to the reliance on electronic health record data.
Our findings build upon prior evidence linking acne and IBS and offer important insights into the timing of this association following acne diagnosis. Future research should explore biological mechanisms underlying the gut-skin axis and evaluate targeted interventions to mitigate IBS risk in patients with acne.
To the Editor:
Acne vulgaris and irritable bowel syndrome (IBS) are both associated with microbial dysbiosis and chronic inflammation.1-3 While the prevalence of IBS among patients with acne has been examined previously,4,5 there has been limited focus on the risk for new-onset IBS following acne diagnosis. Current evidence suggests isotretinoin may be associated with a lower risk for IBS compared to oral antibiotics6; however, evidence supporting this association is limited outside these cohorts, highlighting the need for further investigation. In this large-scale study, we sought to investigate the incidence of new-onset IBS among patients with acne compared with healthy controls as well as to evaluate whether oral acne treatments (ie, oral antibiotics or isotretinoin) are associated with new-onset IBS in this population.
A retrospective cohort study was conducted using data from the US Collaborative Network in TriNetX from October 2014 to October 2024. Patients were identified using International Classification of Diseases, Tenth Revision, Clinical Modification codes, Current Procedural Terminology codes, Anatomical Therapeutic Chemical Classification System codes, and RxNorm codes (Table 1). These codes were selected based on prior literature review, clinical relevance, and their ability to capture diagnoses of acne and IBS as well as relevant exclusion criteria. Patients were considered eligible if they were between the ages of 18 and 90 years. Individuals with a history of IBS, inflammatory bowel disease, infectious gastroenteritis, or celiac disease were excluded from our analysis.

To examine potential associations between acne and IBS, 2 primary cohorts were established: patients with acne who were managed without systemic medications and healthy controls (ie, patients with no history of acne) who had no exposure to systemic acne treatments (Figure). Further, to assess the relationship between oral acne treatments (macrolides, tetracyclines, isotretinoin) and IBS, additional cohorts were created for each therapy and were compared to a cohort of patients with acne who were managed without systemic medications. To control for potential concomitant treatments, patients who had received any systemic treatment other than the specific therapy for their treatment cohort were excluded from our analysis.

To account for potential confounders, all cohorts were 1:1 propensity score matched by demographics, tobacco and alcohol use, type 2 diabetes, obesity, anxiety, and depression (eTable). Each cohort was followed for 2 years after their index of event: the date of acne diagnosis for the acne cohort, the date of systemic treatment initiation for the treatment cohorts, and the date of the general adult encounter without abnormal findings for the control cohort. The primary outcome was the incidence of IBS, assessed by odds ratio (OR) and 95% CIs.
We identified 375,944 patients with acne managed without systemic treatment and 3,148,443 healthy controls who met study criteria. After the 1:1 propensity score match, each cohort included 49,690 patients (eTable). In the 2-year period after acne diagnosis, patients were more likely to develop IBS compared with controls (1421 vs 1285 [OR, 1.10; 95% CI, 1.02-1.19])(Table 2). Patients with acne who were treated with tetracyclines (n=208,971) were 30% more likely to develop IBS than those managed without systemic medications (1114 vs 856 [OR, 1.30; 95% CI, 1.19-1.42]). Within the tetracycline cohort, doxycycline-treated patients were 25% more likely to develop IBS compared with those treated with minocycline (213 vs 170 [OR, 1.25; 95% CI, 1.02-1.53]). Similarly, the use of macrolides (n=136,334) for acne treatment was significantly associated with an increased risk for IBS (1023 vs 595 [OR, 1.73; 95% CI, 1.57-1.92; P<.0001]) compared with controls. No statistically significant association was observed between isotretinoin and the incidence of IBS (Table 2).


In this large-scale cohort study, acne was associated with an increased likelihood of developing IBS within 2 years of an acne diagnosis compared with healthy controls. While a prior study also identified this association, it had a broader follow-up window ranging from 8 to 10 years.2 In contrast, our analysis specifically quantified the risk within the first 2 years of diagnosis. This distinction suggested potential for earlier IBS onset in patients with acne than has previously been recognized and may serve as an early clinical indicator for IBS risk in this population.
Our findings further suggested an association between oral tetracyclines and macrolides and an increased risk for IBS. This aligns with existing literature suggesting that oral antibiotic use can disrupt the gut microbiota and lead to potential gastrointestinal complications7 and reinforces the importance of careful antibiotic stewardship in dermatologic practice.
Although isotretinoin initially was surrounded by substantial controversy regarding its potential impact on gut health—particularly in inflammatory bowel disease8—our results do not support an increased risk for IBS among patients with acne who use isotretinoin. These findings challenge previous concerns and align with research suggesting that isotretinoin could be a safer alternative to antibiotic use for eligible patients who have a history of gastrointestinal disorders.6
This study highlights an important but underrecognized link between acne and IBS risk, emphasizing the need for early monitoring of gastrointestinal symptoms and careful antibiotic stewardship in dermatologic practice. Gastroenterology consultation may be advisable for patients with acne who have persistent gastrointestinal symptoms to facilitate a more integrated, patient-centered approach to care.
Limitations of this study include potential misclassification of International Classification of Diseases, Tenth Revision, Clinical Modification codes, selection bias, and residual confounding from unmeasured factors such as diet, lifestyle, disease severity, and treatment adherence due to the reliance on electronic health record data.
Our findings build upon prior evidence linking acne and IBS and offer important insights into the timing of this association following acne diagnosis. Future research should explore biological mechanisms underlying the gut-skin axis and evaluate targeted interventions to mitigate IBS risk in patients with acne.
Menees S, Chey W. The gut microbiome and irritable bowel syndrome. F1000Res. 2018;7:F1000 Faculty Rev-1029. doi:10.12688/f1000research.14592.1
Yu-Wen C, Chun-Ying W, Yi-Ju C. Gastrointestinal comorbidities in patients with acne vulgaris: a population-based retrospective study. JAAD Int. 2025;18:62-68. doi:10.1016/j.jdin.2024.08.022
Deng Y, Wang H, Zhou J, et al. Patients with acne vulgaris have a distinct gut microbiota in comparison with healthy controls. Acta Derm Venereol. 2018;98:783-790. doi:10.2340/00015555-2968
Demirbas¸ A, Elmas ÖF. The relationship between acne vulgaris and irritable bowel syndrome: a preliminary study. J Cosmet Dermatol. 2021;20:316-320. doi:10.1111/jocd.13481
Daye M, Cihan FG, Is¸ık B, et al. Evaluation of bowel habits in patients with acne vulgaris. Int J Clin Pract. 2021;75:e14903. doi:10.1111/ijcp.14903
Kridin K, Ludwig RJ. Isotretinoin and the risk of inflammatory bowel disease and irritable bowel syndrome: a large-scale global study. J Am Acad Dermatol. 2023;88:824-830. doi:10.1016/j.jaad.2022.12.015
Villarreal AA, Aberger FJ, Benrud R, et al. Use of broad-spectrum antibiotics and the development of irritable bowel syndrome. WMJ. 2012;111:17-20.
Yu C-L, Chou P-Y, Liang C-S, et al. Isotretinoin exposure and risk of inflammatory bowel disease: a systematic review with meta-analysis and trial sequential analysis. Am J Clin Dermatol. 2023;24:721-730. doi:10.1007/s40257-023-00765-9
Menees S, Chey W. The gut microbiome and irritable bowel syndrome. F1000Res. 2018;7:F1000 Faculty Rev-1029. doi:10.12688/f1000research.14592.1
Yu-Wen C, Chun-Ying W, Yi-Ju C. Gastrointestinal comorbidities in patients with acne vulgaris: a population-based retrospective study. JAAD Int. 2025;18:62-68. doi:10.1016/j.jdin.2024.08.022
Deng Y, Wang H, Zhou J, et al. Patients with acne vulgaris have a distinct gut microbiota in comparison with healthy controls. Acta Derm Venereol. 2018;98:783-790. doi:10.2340/00015555-2968
Demirbas¸ A, Elmas ÖF. The relationship between acne vulgaris and irritable bowel syndrome: a preliminary study. J Cosmet Dermatol. 2021;20:316-320. doi:10.1111/jocd.13481
Daye M, Cihan FG, Is¸ık B, et al. Evaluation of bowel habits in patients with acne vulgaris. Int J Clin Pract. 2021;75:e14903. doi:10.1111/ijcp.14903
Kridin K, Ludwig RJ. Isotretinoin and the risk of inflammatory bowel disease and irritable bowel syndrome: a large-scale global study. J Am Acad Dermatol. 2023;88:824-830. doi:10.1016/j.jaad.2022.12.015
Villarreal AA, Aberger FJ, Benrud R, et al. Use of broad-spectrum antibiotics and the development of irritable bowel syndrome. WMJ. 2012;111:17-20.
Yu C-L, Chou P-Y, Liang C-S, et al. Isotretinoin exposure and risk of inflammatory bowel disease: a systematic review with meta-analysis and trial sequential analysis. Am J Clin Dermatol. 2023;24:721-730. doi:10.1007/s40257-023-00765-9