Comprehensive Genomic Profiles of Melanoma in Veterans Compared to Reference Databases

Article Type
Changed
Wed, 08/13/2025 - 10:02
Display Headline

Comprehensive Genomic Profiles of Melanoma in Veterans Compared to Reference Databases

The veteran population, with its unique and diverse types of exposure and military service experiences, faces distinct health factors compared with the general population. These factors can be categorized into exposures during military service and those occurring postservice. While the latter phase incorporates psychological issues that may arise while transitioning to civilian life, the service period is associated with major physical, chemical, and psychological exposures that can impact veterans’ health. Carcinogenesis related to military exposures is concerning, and different types of malignancies have been associated with military exposures.1 The 2022 introduction of the Cancer Moonshot initiative served as a breeding ground for multiple projects aimed at investigation of exposure-related carcinogenesis, prompting increased attention and efforts to linking specific exposures to specific malignancies.2

Melanoma is the deadliest skin cancer, accounting for 1.3% of all cancer deaths.3 Although it may only account for 1% to 5% of skin cancer diagnoses, its incidence in the United States’ population has been increasing.4,5 There were 97,610 estimated new cases of melanoma in 2023, according to the National Cancer Institute.6

The incidence of melanoma may be higher in the military population compared with the general population.7 Melanoma is the fourth-most common cancer diagnosed in veterans.8

Several demographic characteristics of the US military population are associated with higher melanoma incidence and poorer prognosis, including male sex, older age, and White race. Apart from sun exposure—a known risk factor for melanoma development—other factors, such as service branch, seem to contribute to risk, with the highest melanoma rates noted in the Air Force.9 According to a study by Chang et al, veterans have a higher risk of stage III (18%) or stage IV (13%) melanoma at initial diagnosis.8

Molecular testing of metastatic melanoma is currently the standard of care for guiding the use of US Food and Drug Administration-approved targeted therapies such as BRAF, MEK, and KIT inhibitors. This comparative analysis details the melanoma comprehensive genomic profiles observed at a large US Department of Veterans Affairs (VA) medical center (VAMC) and those reported in reference databases.

Methods

A query to select all metastatic melanomas sent for comprehensive genomic profiling from the Kansas City VAMC (KCVAMC), identified 35 cases from 2019 through 2023 as the study population. The health records of these patients were reviewed to collect demographic information, military service history, melanoma history, other medical, social, and family histories. The comprehensive genomic profiling reports were reviewed to collect the reported pathogenic variants, microsatellite instability (MSI) status, and tumor mutational burden (TMB) for each case.

The Catalogue of Somatic Mutations in Cancer (COSMIC) was used to identify the most commonly mutated genes in melanomas from The Cancer Genome Atlas for the general population.4,5 The literature was consulted to determine the MSI status and TMB in melanomas from The Cancer Genome Atlas for separate reference populations.6,7 The frequency of MSI-high (MSI-H) status, TMB ≥ 10 mutations/megabase (mut/Mb), and mutations in each of the 20 most commonly mutated genes was determined and compared between melanomas from The Cancer Genome Atlas and KCVAMC cases. Corresponding P  values were calculated to identify significant differences. Values were calculated for the entire sample as well as a subgroup with Agent Orange (AO) exposure. The study was approved by the KCVAMC Institutional Review Board.

Results

The mean (SD) age of study participants was 72.9 (9.4) years (range, 39-90 years). The mean (SD) duration of military service was 1654 (1421) days (about 4 years, 6 months, and 10 days). Of the 35 patients included, 22 (63%) served during the Vietnam era (November 1, 1965, to April 30, 1975) and 2 (6%) served during the Persian Gulf War era (August 2, 1990, to February 28, 1991). Seventeen veterans (49%) served in the Army, 9 in the Navy (26%), 5 in the Air Force (14%), and 4 in the Marine Corps (11%). Definitive AO exposure was noted in 13 patients (37%) (Table 1).

0825FED-AVAHO-Mel-T1

Of the 35 patients, 24 (69%) had metastatic disease and the primary site of melanoma was unknown in 14 patients (40%). One patient (Patient 32) had an intraocular melanoma. The primary site was the trunk for 11 patients (31%), the face/head for 7 patients (20%) and extremities for 3 patients (9%). Eight patients (23%) were pT3 stage (thickness > 2 mm but < 4 mm), 7 patients (20%) were pT4 stage (thickness > 4 mm), and 5 patients (14%) were pT1 (thickness ≥ 1 mm). One patient had a primary lesion at pT2 stage, and 1 had a Tis stage lesion. Three patients (9%) had a family history of melanoma in a first-degree relative.

The list of genes mutated in melanoma cells in the study population is provided in the eAppendix.10,11 Twenty-seven patients (77%) had mutations in TERT promoter, 15 (43%) in CDKN2A/B, 13 (37%) in BRAF, 11 (31%) in NF1, 9 (26%) in TP53, and 8 (23%) in NRAS (Table 2). The majority of mutations in TERT promoter were c.- 146C>T (18 of 27 patients [67%]), whereas c.-124C>T was the second-most common (8 of 27 patients [30%]). The 2 observed mutations in the 13 patients with BRAF mutations were V600E and V600K, with almost equal distribution (54% and 46%, respectively). The mean (SD) TMB was 33.2 (39) mut/Mb (range, 1-203 mut/Mb). Ten patients (29%) had a TMB < 10 mut/Mb, whereas 24 (69%) had a TMB > 10 mut/Mb. The TMB could not be determined in 1 case. The frequency of TMB-high tumors in the study population compared with frequency in the reference population is shown in Table 3.12 Only 3 patients (0.64%) in the reference population had MSI-H tumors, and the microsatellite status could not be determined in those tumors (Table 4).13 Table 5 outlines statistically significant findings.

0825FED-AVAHO-Mel-T20825FED-AVAHO-Mel-T30825FED-AVAHO-Mel-T40825FED-AVAHO-Mel-T5
Agent Orange Subgroup

AO was a tactical herbicide used by the US military, named for the orange band around the storage barrels. Possible mutagenic properties of AO have been attributed to its byproduct, dioxin. Among the most common cancers known to be associated with AO exposure are bladder and prostate carcinoma and hematopoietic neoplasms. The association between genetic alterations and AO exposure was studied in veterans with prostate cancer.14 However, to our knowledge, insufficient information is available to determine whether an association exists between exposure to herbicides used in Vietnam or the contaminant dioxin and melanoma. Because a significant proportion of this study population had a well-documented history of AO exposure (37.1%), we were able to analyze them as a subgroup and to separately compare their mutation frequency with the general population.

Results were notable for different distributions of the most frequently mutated genes in the AO subgroup compared with the whole study population. As such, TERT promoter remained the most frequently mutated gene (92%), followed by CDKN2A/B (46%); however, frequency of mutations in NF1 (46%) outnumbered those of BRAF (31%), the fourth-most common mutation. Moreover, when compared with the general melanoma population, a significantly higher frequency of mutations in the NF1 gene was observed in the AO subgroup—not the entire study population.

Discussion

Given that veterans constitute a distinct population, there is reasonable interest in investigating characteristic health issues related to military service. Skin cancer—melanoma in particular—has been researched recently in a veteran population. The differences in demographics, tumor characteristics, and melanoma- specific survival in veterans compared with the general population have already been assessed. According to Chang et al, compared with the general population, veterans are more likely to present with metastatic disease and have lower 5-year survival rates.8

Melanoma is one of the most highly mutated malignancies.15 Fortunately, the most common mutation in melanoma, BRAF V600E, is now considered therapeutically targetable. However, there are still many mutations that are less often discussed and not well understood. Regardless of therapeutic implications, all mutations observed in melanoma are worth investigating because a tumor’s genomic profile also can provide prognostic and etiologic information. Developing comprehensive descriptions of melanoma mutational profiles in specific populations is critical to advancing etiologic understanding and informing prevention strategies.

Our results demonstrate the high prevalence of TERT promoter mutations with characteristic ultraviolet signature (C>T) in the study population. This aligns with general evidence that TERT promoter mutations are common in cutaneous melanomas: 77% of this study sample and up to 86% of all mutations are TERT promoter mutations, according to Davis et al.15TERT promoter mutations are positively associated with the initiation, invasion, and metastasis of melanoma. In certain subtypes, there is evidence that the presence of TERT promoter mutations is significantly associated with risk for extranodal metastasis and death.16 The second-most common mutated gene in the veteran study population was CDKN2A/B (43%), and the third-most mutated gene was BRAF (37%).

In chronically sun-exposed skin NF1, NRAS, and occasionally BRAF V600K mutations tend to predominate. BRAF V600E mutations, on the other hand, are rare in these melanomas.15 In our study population, the most prevalent melanoma site was the trunk (31%), which is considered a location with an intermittent pattern of sun exposure.17

This study population also had a higher frequency of CDKN2A/B mutations. High frequencies of CDKN2A/B mutations have been reported in familial melanomas, but only 1 patient with CDKN2A/B mutations had a known family history of melanoma.15 Tumors in the study population showed significantly lower frequency of mutations in ROS1, GRIN2A, KDR, KMT2C (MLL3), KMT2D (MLL2), LRP1B, PTPRT, PTCH1, FAT4, and PREX2 (P < .05).

In this study the subgroup of veterans with AO exposure differed from the whole study population. As such, CDKN2A/B mutations were observed with the same frequency as NF1 mutations (46% each); however, BRAF mutations constituted only 31% of the mutations. In addition, the frequency of NF1 mutations was significantly higher in the AO subgroup compared with the general population, but not in the whole study population.

Our sample also differed from the reference population by showing a significantly higher frequency of TMB-high (ie, ≥ 10 mut/Mb) tumors (71% vs 49%; P = .01).12 Interestingly, no significant difference in the frequency of TMB-high tumors was observed between the AO subgroup and the reference population (69% vs 49%; P = .16). There also was no statistically significant difference between the frequency of MSI-H tumors in our study population and the reference population (P = .64).13

One patient in the study population had uveal melanoma. Mutations encountered in this patient’s tumor differed from the general mutational profile of tumors. None of the 21 mutations depicted in Table 2 were present in this sample.10,11 On the other hand, those mutations frequently observed in intraocular melanomas, BAP1 and GNA11, were present in this patient.18 Additionally, this particular melanoma possessed mutations in genes RICTOR, RAD21, and PIK3R1.

Limitations

This study population consisted exclusively of male patients, introducing sex as a potential confounder in analyzing differences between the study population and the general population. As noted in a 2020 systematic review, there were no sex-based differences in the frequency of mutations in BRAF, NRAS, and KIT genes.19

Regarding NF1 mutations, only NF1-mutated acral and mucosal melanomas were more frequently observed in female patients, whereas nonacral NF1-mutated melanomas were more frequently observed in male patients.20 However, there is currently no clear evidence of whether the mutational landscapes of cutaneous melanoma differ by sex.21 Among the 11 cases with NF1-mutatation, site of origin was known in 6, 5 of which originated at nonacral sites. Although the AO subgroup also consisted entirely of male patients, this does not explain the observed increased frequency of NF1 mutations relative to the general population. No such difference was observed between the whole study population, which also consisted exclusively of male patients, and the general population. The similar frequencies of nonacral location in the whole study population (3 acral, 18 nonacral, 14 unknown site of origin) and AO subgroup (1 acral, 7 nonacral, 5 unknown site of origin) preclude location as an explanation.

The Cancer Genome Atlas Network proposed a framework for genomic classification of melanoma into 4 subtypes based on the pattern of the most prevalent significantly mutated genes: mutant BRAF, mutant RAS, mutant NF1, and triple–wild-type. According to that study, BRAF mutations were indeed associated with younger age, in contrast to the NF1-mutant genomic subtype, which was more prevalent in older individuals with higher TMB.22 This emphasizes the need to interpret the potential association of AO exposure and NF1 mutation in melanoma with caution, although additional studies are required to observe the difference between the veteran population and age-matched general population.

On the other hand, Yu et al reported no significant differences of TMB values between patients aged < 60 and ≥ 60 years with melanoma.23 In short, the observed differences we report in our limited study warrant additional investigation with larger sample sizes, sex-matched controlling, and age-matched controlling. The study was limited by its small sample size and the single location.

Conclusion

The genomic profile of melanomas in the veteran population appears to be similar to that of the general population with a few possible differences. Melanomas in the veteran study population showed a higher frequency of CDKN2A/B mutations; lower frequency of ROS1, GRIN2A, KDR, KMT2C (MLL3), KMT2D (MLL2), LRP1B, PTPRT, PTCH1, FAT4, and PREX2 mutations; and higher TMB. In addition, melanomas in the AO subgroup showed higher frequencies of NF1 mutations. The significance of such findings remains to be determined by further investigation.

References
  1. Bytnar JA, McGlynn KA, et al. Cancer incidence in the US military: An updated analysis. Cancer. 2024;130(1):96-106. doi:10.1002/cncr.34978
  2. Singer DS. A new phase of the Cancer Moonshot to end cancer as we know it. Nat Med. 2022;28(7):1345-1347. doi:10.1038/s41591-022-01881-5
  3. Koczkodaj P, Sulkowska U, Didkowska J, et al. Melanoma mortality trends in 28 European countries: a retrospective analysis for the years 1960-2020. Cancers (Basel). 2023;15(5):1514. Published 2023 Feb 28. doi:10.3390/cancers15051514
  4. Okobi OE, Abreo E, Sams NP, et al. Trends in melanoma incidence, prevalence, stage at diagnosis, and survival: an analysis of the United States Cancer Statistics (USCS) database. Cureus. 2024;16(10):e70697. doi:10.7759/cureus.70697
  5. Bartling SJ, Rivard SC, Meyerle JH. Melanoma in an active duty marine. Mil Med. 2017;182:e2034-e2039. doi:10.7205/MILMED-D-17-00127
  6. American Cancer Society. Cancer facts & figures 2023. American Cancer Society; 2023. Accessed June 20, 2025. https://www.cancer.org/content/dam/cancer-org/research/cancer-facts-and-statistics/annual-cancer-facts-and-figures/2023/2023-cancer-facts-and-figures.pdf
  7. Rezaei SJ, Kim J, Onyeka S, et al. Skin cancer and other dermatologic conditions among US veterans. JAMA Dermatol. 2024;160(10):1107-1111. doi:10.1001/jamadermatol.2024.3043
  8. Chang MS, La J, Trepanowski N, et al. Increased relative proportions of advanced melanoma among veterans: a comparative analysis with the Surveillance, Epidemiology, and End Results registry. J Am Acad Dermatol. 2022;87:72-79. doi:10.1016/j.jaad.2022.02.063
  9. Riemenschneider K, Liu J, Powers JG. Skin cancer in the military: a systematic review of melanoma and nonmelanoma skin cancer incidence, prevention, and screening among active duty and veteran personnel. J Am Acad Dermatol. 2018;78:1185-1192. doi:10.1016/j.jaad.2017.11.062
  10. Huang FW, Hodis E, Xu MJ, et al. Highly recurrent TERT promoter mutations in human melanoma. Science. 2013;339:957-959. doi:10.1126/science.1229259
  11. Tate JG, Bamford S, Jubb HC, et al. COSMIC: the Catalogue of Somatic Mutations in Cancer. Nucleic Acids Res. 2019;47:D941-D947. doi:10.1093/nar/gky1015
  12. Li M, Gao X, Wang X. Identification of tumor mutation burden-associated molecular and clinical features in cancer by analyzing multi-omics data. Front Immunol. 2023;14:1090838. doi:10.3389/fimmu.2023.1090838
  13. Bonneville R, Krook MA, Kautto EA, et al. Landscape of microsatellite instability across 39 cancer types. JCO Precis Oncol. 2017;2017:PO.17.00073. doi:10.1200/PO.17.00073
  14. Lui AJ, Pagadala MS, Zhong AY, et al. Agent Orange exposure and prostate cancer risk in the Million Veteran Program. medRxiv [Preprint]. 2023:2023.06.14.23291413. doi:10.1101/2023.06.14.23291413
  15. Davis EJ, Johnson DB, Sosman JA, et al. Melanoma: what do all the mutations mean? Cancer. 2018;124:3490-3499. doi:10.1002/cncr.31345
  16. Guo Y, Chen Y, Zhang L, et al. TERT promoter mutations and telomerase in melanoma. J Oncol. 2022;2022:6300329. doi:10.1155/2022/6300329
  17. Whiteman DC, Stickley M, Watt P, et al. Anatomic site, sun exposure, and risk of cutaneous melanoma. J Clin Oncol. 2006;24:3172-3177. doi:10.1200/JCO.2006.06.1325
  18. Decatur CL, Ong E, Garg N, et al. Driver mutations in uveal melanoma: associations with gene expression profile and patient outcomes. JAMA Ophthalmol. 2016;134:728-733. doi:10.1001/jamaophthalmol.2016.0903
  19. Gutiérrez-Castañeda LD, Nova JA, Tovar-Parra JD. Frequency of mutations in BRAF, NRAS, and KIT in different populations and histological subtypes of melanoma: a systemic review. Melanoma Res. 2020;30:62- 70. doi:10.1097/CMR.0000000000000628
  20. Thielmann CM, Chorti E, Matull J, et al. NF1-mutated melanomas reveal distinct clinical characteristics depending on tumour origin and respond favourably to immune checkpoint inhibitors. Eur J Cancer. 2021;159:113-124. doi:10.1016/j.ejca.2021.09.035
  21. D’Ecclesiis O, Caini S, Martinoli C, et al. Gender-dependent specificities in cutaneous melanoma predisposition, risk factors, somatic mutations, prognostic and predictive factors: a systematic review. Int J Environ Res Public Health. 2021;18:7945. doi:10.3390/ijerph18157945
  22. Cancer Genome Atlas Network. Genomic classification of cutaneous melanoma. Cell. 2015;161:1681-1696. doi:10.1016/j.cell.2015.05.044
  23. Yu Z, Wang J, Feng L, et al. Association of tumor mutational burden with age in solid tumors. J Clin Oncol. 2020;38:e13590-e13590. doi:10.1200/JCO.2020.38.15_suppl.e13590
Article PDF
Author and Disclosure Information

Daniel Mettman, MDa; Margaryta Stoieva, MDb; Maryam Abdo, MBChBb

Author affiliations
aKansas City Veterans Affairs Medical Center, Missouri
bUniversity of Kansas Medical Center, Kansas City

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

Correspondence: Margaryta Stoieva ([email protected])

Fed Pract. 2025;42(suppl 3). Published online August 15. doi:10.12788/fp.0607

Issue
Federal Practitioner - 42(suppl 3)
Publications
Topics
Page Number
S18-S24
Sections
Author and Disclosure Information

Daniel Mettman, MDa; Margaryta Stoieva, MDb; Maryam Abdo, MBChBb

Author affiliations
aKansas City Veterans Affairs Medical Center, Missouri
bUniversity of Kansas Medical Center, Kansas City

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

Correspondence: Margaryta Stoieva ([email protected])

Fed Pract. 2025;42(suppl 3). Published online August 15. doi:10.12788/fp.0607

Author and Disclosure Information

Daniel Mettman, MDa; Margaryta Stoieva, MDb; Maryam Abdo, MBChBb

Author affiliations
aKansas City Veterans Affairs Medical Center, Missouri
bUniversity of Kansas Medical Center, Kansas City

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

Correspondence: Margaryta Stoieva ([email protected])

Fed Pract. 2025;42(suppl 3). Published online August 15. doi:10.12788/fp.0607

Article PDF
Article PDF

The veteran population, with its unique and diverse types of exposure and military service experiences, faces distinct health factors compared with the general population. These factors can be categorized into exposures during military service and those occurring postservice. While the latter phase incorporates psychological issues that may arise while transitioning to civilian life, the service period is associated with major physical, chemical, and psychological exposures that can impact veterans’ health. Carcinogenesis related to military exposures is concerning, and different types of malignancies have been associated with military exposures.1 The 2022 introduction of the Cancer Moonshot initiative served as a breeding ground for multiple projects aimed at investigation of exposure-related carcinogenesis, prompting increased attention and efforts to linking specific exposures to specific malignancies.2

Melanoma is the deadliest skin cancer, accounting for 1.3% of all cancer deaths.3 Although it may only account for 1% to 5% of skin cancer diagnoses, its incidence in the United States’ population has been increasing.4,5 There were 97,610 estimated new cases of melanoma in 2023, according to the National Cancer Institute.6

The incidence of melanoma may be higher in the military population compared with the general population.7 Melanoma is the fourth-most common cancer diagnosed in veterans.8

Several demographic characteristics of the US military population are associated with higher melanoma incidence and poorer prognosis, including male sex, older age, and White race. Apart from sun exposure—a known risk factor for melanoma development—other factors, such as service branch, seem to contribute to risk, with the highest melanoma rates noted in the Air Force.9 According to a study by Chang et al, veterans have a higher risk of stage III (18%) or stage IV (13%) melanoma at initial diagnosis.8

Molecular testing of metastatic melanoma is currently the standard of care for guiding the use of US Food and Drug Administration-approved targeted therapies such as BRAF, MEK, and KIT inhibitors. This comparative analysis details the melanoma comprehensive genomic profiles observed at a large US Department of Veterans Affairs (VA) medical center (VAMC) and those reported in reference databases.

Methods

A query to select all metastatic melanomas sent for comprehensive genomic profiling from the Kansas City VAMC (KCVAMC), identified 35 cases from 2019 through 2023 as the study population. The health records of these patients were reviewed to collect demographic information, military service history, melanoma history, other medical, social, and family histories. The comprehensive genomic profiling reports were reviewed to collect the reported pathogenic variants, microsatellite instability (MSI) status, and tumor mutational burden (TMB) for each case.

The Catalogue of Somatic Mutations in Cancer (COSMIC) was used to identify the most commonly mutated genes in melanomas from The Cancer Genome Atlas for the general population.4,5 The literature was consulted to determine the MSI status and TMB in melanomas from The Cancer Genome Atlas for separate reference populations.6,7 The frequency of MSI-high (MSI-H) status, TMB ≥ 10 mutations/megabase (mut/Mb), and mutations in each of the 20 most commonly mutated genes was determined and compared between melanomas from The Cancer Genome Atlas and KCVAMC cases. Corresponding P  values were calculated to identify significant differences. Values were calculated for the entire sample as well as a subgroup with Agent Orange (AO) exposure. The study was approved by the KCVAMC Institutional Review Board.

Results

The mean (SD) age of study participants was 72.9 (9.4) years (range, 39-90 years). The mean (SD) duration of military service was 1654 (1421) days (about 4 years, 6 months, and 10 days). Of the 35 patients included, 22 (63%) served during the Vietnam era (November 1, 1965, to April 30, 1975) and 2 (6%) served during the Persian Gulf War era (August 2, 1990, to February 28, 1991). Seventeen veterans (49%) served in the Army, 9 in the Navy (26%), 5 in the Air Force (14%), and 4 in the Marine Corps (11%). Definitive AO exposure was noted in 13 patients (37%) (Table 1).

0825FED-AVAHO-Mel-T1

Of the 35 patients, 24 (69%) had metastatic disease and the primary site of melanoma was unknown in 14 patients (40%). One patient (Patient 32) had an intraocular melanoma. The primary site was the trunk for 11 patients (31%), the face/head for 7 patients (20%) and extremities for 3 patients (9%). Eight patients (23%) were pT3 stage (thickness > 2 mm but < 4 mm), 7 patients (20%) were pT4 stage (thickness > 4 mm), and 5 patients (14%) were pT1 (thickness ≥ 1 mm). One patient had a primary lesion at pT2 stage, and 1 had a Tis stage lesion. Three patients (9%) had a family history of melanoma in a first-degree relative.

The list of genes mutated in melanoma cells in the study population is provided in the eAppendix.10,11 Twenty-seven patients (77%) had mutations in TERT promoter, 15 (43%) in CDKN2A/B, 13 (37%) in BRAF, 11 (31%) in NF1, 9 (26%) in TP53, and 8 (23%) in NRAS (Table 2). The majority of mutations in TERT promoter were c.- 146C>T (18 of 27 patients [67%]), whereas c.-124C>T was the second-most common (8 of 27 patients [30%]). The 2 observed mutations in the 13 patients with BRAF mutations were V600E and V600K, with almost equal distribution (54% and 46%, respectively). The mean (SD) TMB was 33.2 (39) mut/Mb (range, 1-203 mut/Mb). Ten patients (29%) had a TMB < 10 mut/Mb, whereas 24 (69%) had a TMB > 10 mut/Mb. The TMB could not be determined in 1 case. The frequency of TMB-high tumors in the study population compared with frequency in the reference population is shown in Table 3.12 Only 3 patients (0.64%) in the reference population had MSI-H tumors, and the microsatellite status could not be determined in those tumors (Table 4).13 Table 5 outlines statistically significant findings.

0825FED-AVAHO-Mel-T20825FED-AVAHO-Mel-T30825FED-AVAHO-Mel-T40825FED-AVAHO-Mel-T5
Agent Orange Subgroup

AO was a tactical herbicide used by the US military, named for the orange band around the storage barrels. Possible mutagenic properties of AO have been attributed to its byproduct, dioxin. Among the most common cancers known to be associated with AO exposure are bladder and prostate carcinoma and hematopoietic neoplasms. The association between genetic alterations and AO exposure was studied in veterans with prostate cancer.14 However, to our knowledge, insufficient information is available to determine whether an association exists between exposure to herbicides used in Vietnam or the contaminant dioxin and melanoma. Because a significant proportion of this study population had a well-documented history of AO exposure (37.1%), we were able to analyze them as a subgroup and to separately compare their mutation frequency with the general population.

Results were notable for different distributions of the most frequently mutated genes in the AO subgroup compared with the whole study population. As such, TERT promoter remained the most frequently mutated gene (92%), followed by CDKN2A/B (46%); however, frequency of mutations in NF1 (46%) outnumbered those of BRAF (31%), the fourth-most common mutation. Moreover, when compared with the general melanoma population, a significantly higher frequency of mutations in the NF1 gene was observed in the AO subgroup—not the entire study population.

Discussion

Given that veterans constitute a distinct population, there is reasonable interest in investigating characteristic health issues related to military service. Skin cancer—melanoma in particular—has been researched recently in a veteran population. The differences in demographics, tumor characteristics, and melanoma- specific survival in veterans compared with the general population have already been assessed. According to Chang et al, compared with the general population, veterans are more likely to present with metastatic disease and have lower 5-year survival rates.8

Melanoma is one of the most highly mutated malignancies.15 Fortunately, the most common mutation in melanoma, BRAF V600E, is now considered therapeutically targetable. However, there are still many mutations that are less often discussed and not well understood. Regardless of therapeutic implications, all mutations observed in melanoma are worth investigating because a tumor’s genomic profile also can provide prognostic and etiologic information. Developing comprehensive descriptions of melanoma mutational profiles in specific populations is critical to advancing etiologic understanding and informing prevention strategies.

Our results demonstrate the high prevalence of TERT promoter mutations with characteristic ultraviolet signature (C>T) in the study population. This aligns with general evidence that TERT promoter mutations are common in cutaneous melanomas: 77% of this study sample and up to 86% of all mutations are TERT promoter mutations, according to Davis et al.15TERT promoter mutations are positively associated with the initiation, invasion, and metastasis of melanoma. In certain subtypes, there is evidence that the presence of TERT promoter mutations is significantly associated with risk for extranodal metastasis and death.16 The second-most common mutated gene in the veteran study population was CDKN2A/B (43%), and the third-most mutated gene was BRAF (37%).

In chronically sun-exposed skin NF1, NRAS, and occasionally BRAF V600K mutations tend to predominate. BRAF V600E mutations, on the other hand, are rare in these melanomas.15 In our study population, the most prevalent melanoma site was the trunk (31%), which is considered a location with an intermittent pattern of sun exposure.17

This study population also had a higher frequency of CDKN2A/B mutations. High frequencies of CDKN2A/B mutations have been reported in familial melanomas, but only 1 patient with CDKN2A/B mutations had a known family history of melanoma.15 Tumors in the study population showed significantly lower frequency of mutations in ROS1, GRIN2A, KDR, KMT2C (MLL3), KMT2D (MLL2), LRP1B, PTPRT, PTCH1, FAT4, and PREX2 (P < .05).

In this study the subgroup of veterans with AO exposure differed from the whole study population. As such, CDKN2A/B mutations were observed with the same frequency as NF1 mutations (46% each); however, BRAF mutations constituted only 31% of the mutations. In addition, the frequency of NF1 mutations was significantly higher in the AO subgroup compared with the general population, but not in the whole study population.

Our sample also differed from the reference population by showing a significantly higher frequency of TMB-high (ie, ≥ 10 mut/Mb) tumors (71% vs 49%; P = .01).12 Interestingly, no significant difference in the frequency of TMB-high tumors was observed between the AO subgroup and the reference population (69% vs 49%; P = .16). There also was no statistically significant difference between the frequency of MSI-H tumors in our study population and the reference population (P = .64).13

One patient in the study population had uveal melanoma. Mutations encountered in this patient’s tumor differed from the general mutational profile of tumors. None of the 21 mutations depicted in Table 2 were present in this sample.10,11 On the other hand, those mutations frequently observed in intraocular melanomas, BAP1 and GNA11, were present in this patient.18 Additionally, this particular melanoma possessed mutations in genes RICTOR, RAD21, and PIK3R1.

Limitations

This study population consisted exclusively of male patients, introducing sex as a potential confounder in analyzing differences between the study population and the general population. As noted in a 2020 systematic review, there were no sex-based differences in the frequency of mutations in BRAF, NRAS, and KIT genes.19

Regarding NF1 mutations, only NF1-mutated acral and mucosal melanomas were more frequently observed in female patients, whereas nonacral NF1-mutated melanomas were more frequently observed in male patients.20 However, there is currently no clear evidence of whether the mutational landscapes of cutaneous melanoma differ by sex.21 Among the 11 cases with NF1-mutatation, site of origin was known in 6, 5 of which originated at nonacral sites. Although the AO subgroup also consisted entirely of male patients, this does not explain the observed increased frequency of NF1 mutations relative to the general population. No such difference was observed between the whole study population, which also consisted exclusively of male patients, and the general population. The similar frequencies of nonacral location in the whole study population (3 acral, 18 nonacral, 14 unknown site of origin) and AO subgroup (1 acral, 7 nonacral, 5 unknown site of origin) preclude location as an explanation.

The Cancer Genome Atlas Network proposed a framework for genomic classification of melanoma into 4 subtypes based on the pattern of the most prevalent significantly mutated genes: mutant BRAF, mutant RAS, mutant NF1, and triple–wild-type. According to that study, BRAF mutations were indeed associated with younger age, in contrast to the NF1-mutant genomic subtype, which was more prevalent in older individuals with higher TMB.22 This emphasizes the need to interpret the potential association of AO exposure and NF1 mutation in melanoma with caution, although additional studies are required to observe the difference between the veteran population and age-matched general population.

On the other hand, Yu et al reported no significant differences of TMB values between patients aged < 60 and ≥ 60 years with melanoma.23 In short, the observed differences we report in our limited study warrant additional investigation with larger sample sizes, sex-matched controlling, and age-matched controlling. The study was limited by its small sample size and the single location.

Conclusion

The genomic profile of melanomas in the veteran population appears to be similar to that of the general population with a few possible differences. Melanomas in the veteran study population showed a higher frequency of CDKN2A/B mutations; lower frequency of ROS1, GRIN2A, KDR, KMT2C (MLL3), KMT2D (MLL2), LRP1B, PTPRT, PTCH1, FAT4, and PREX2 mutations; and higher TMB. In addition, melanomas in the AO subgroup showed higher frequencies of NF1 mutations. The significance of such findings remains to be determined by further investigation.

The veteran population, with its unique and diverse types of exposure and military service experiences, faces distinct health factors compared with the general population. These factors can be categorized into exposures during military service and those occurring postservice. While the latter phase incorporates psychological issues that may arise while transitioning to civilian life, the service period is associated with major physical, chemical, and psychological exposures that can impact veterans’ health. Carcinogenesis related to military exposures is concerning, and different types of malignancies have been associated with military exposures.1 The 2022 introduction of the Cancer Moonshot initiative served as a breeding ground for multiple projects aimed at investigation of exposure-related carcinogenesis, prompting increased attention and efforts to linking specific exposures to specific malignancies.2

Melanoma is the deadliest skin cancer, accounting for 1.3% of all cancer deaths.3 Although it may only account for 1% to 5% of skin cancer diagnoses, its incidence in the United States’ population has been increasing.4,5 There were 97,610 estimated new cases of melanoma in 2023, according to the National Cancer Institute.6

The incidence of melanoma may be higher in the military population compared with the general population.7 Melanoma is the fourth-most common cancer diagnosed in veterans.8

Several demographic characteristics of the US military population are associated with higher melanoma incidence and poorer prognosis, including male sex, older age, and White race. Apart from sun exposure—a known risk factor for melanoma development—other factors, such as service branch, seem to contribute to risk, with the highest melanoma rates noted in the Air Force.9 According to a study by Chang et al, veterans have a higher risk of stage III (18%) or stage IV (13%) melanoma at initial diagnosis.8

Molecular testing of metastatic melanoma is currently the standard of care for guiding the use of US Food and Drug Administration-approved targeted therapies such as BRAF, MEK, and KIT inhibitors. This comparative analysis details the melanoma comprehensive genomic profiles observed at a large US Department of Veterans Affairs (VA) medical center (VAMC) and those reported in reference databases.

Methods

A query to select all metastatic melanomas sent for comprehensive genomic profiling from the Kansas City VAMC (KCVAMC), identified 35 cases from 2019 through 2023 as the study population. The health records of these patients were reviewed to collect demographic information, military service history, melanoma history, other medical, social, and family histories. The comprehensive genomic profiling reports were reviewed to collect the reported pathogenic variants, microsatellite instability (MSI) status, and tumor mutational burden (TMB) for each case.

The Catalogue of Somatic Mutations in Cancer (COSMIC) was used to identify the most commonly mutated genes in melanomas from The Cancer Genome Atlas for the general population.4,5 The literature was consulted to determine the MSI status and TMB in melanomas from The Cancer Genome Atlas for separate reference populations.6,7 The frequency of MSI-high (MSI-H) status, TMB ≥ 10 mutations/megabase (mut/Mb), and mutations in each of the 20 most commonly mutated genes was determined and compared between melanomas from The Cancer Genome Atlas and KCVAMC cases. Corresponding P  values were calculated to identify significant differences. Values were calculated for the entire sample as well as a subgroup with Agent Orange (AO) exposure. The study was approved by the KCVAMC Institutional Review Board.

Results

The mean (SD) age of study participants was 72.9 (9.4) years (range, 39-90 years). The mean (SD) duration of military service was 1654 (1421) days (about 4 years, 6 months, and 10 days). Of the 35 patients included, 22 (63%) served during the Vietnam era (November 1, 1965, to April 30, 1975) and 2 (6%) served during the Persian Gulf War era (August 2, 1990, to February 28, 1991). Seventeen veterans (49%) served in the Army, 9 in the Navy (26%), 5 in the Air Force (14%), and 4 in the Marine Corps (11%). Definitive AO exposure was noted in 13 patients (37%) (Table 1).

0825FED-AVAHO-Mel-T1

Of the 35 patients, 24 (69%) had metastatic disease and the primary site of melanoma was unknown in 14 patients (40%). One patient (Patient 32) had an intraocular melanoma. The primary site was the trunk for 11 patients (31%), the face/head for 7 patients (20%) and extremities for 3 patients (9%). Eight patients (23%) were pT3 stage (thickness > 2 mm but < 4 mm), 7 patients (20%) were pT4 stage (thickness > 4 mm), and 5 patients (14%) were pT1 (thickness ≥ 1 mm). One patient had a primary lesion at pT2 stage, and 1 had a Tis stage lesion. Three patients (9%) had a family history of melanoma in a first-degree relative.

The list of genes mutated in melanoma cells in the study population is provided in the eAppendix.10,11 Twenty-seven patients (77%) had mutations in TERT promoter, 15 (43%) in CDKN2A/B, 13 (37%) in BRAF, 11 (31%) in NF1, 9 (26%) in TP53, and 8 (23%) in NRAS (Table 2). The majority of mutations in TERT promoter were c.- 146C>T (18 of 27 patients [67%]), whereas c.-124C>T was the second-most common (8 of 27 patients [30%]). The 2 observed mutations in the 13 patients with BRAF mutations were V600E and V600K, with almost equal distribution (54% and 46%, respectively). The mean (SD) TMB was 33.2 (39) mut/Mb (range, 1-203 mut/Mb). Ten patients (29%) had a TMB < 10 mut/Mb, whereas 24 (69%) had a TMB > 10 mut/Mb. The TMB could not be determined in 1 case. The frequency of TMB-high tumors in the study population compared with frequency in the reference population is shown in Table 3.12 Only 3 patients (0.64%) in the reference population had MSI-H tumors, and the microsatellite status could not be determined in those tumors (Table 4).13 Table 5 outlines statistically significant findings.

0825FED-AVAHO-Mel-T20825FED-AVAHO-Mel-T30825FED-AVAHO-Mel-T40825FED-AVAHO-Mel-T5
Agent Orange Subgroup

AO was a tactical herbicide used by the US military, named for the orange band around the storage barrels. Possible mutagenic properties of AO have been attributed to its byproduct, dioxin. Among the most common cancers known to be associated with AO exposure are bladder and prostate carcinoma and hematopoietic neoplasms. The association between genetic alterations and AO exposure was studied in veterans with prostate cancer.14 However, to our knowledge, insufficient information is available to determine whether an association exists between exposure to herbicides used in Vietnam or the contaminant dioxin and melanoma. Because a significant proportion of this study population had a well-documented history of AO exposure (37.1%), we were able to analyze them as a subgroup and to separately compare their mutation frequency with the general population.

Results were notable for different distributions of the most frequently mutated genes in the AO subgroup compared with the whole study population. As such, TERT promoter remained the most frequently mutated gene (92%), followed by CDKN2A/B (46%); however, frequency of mutations in NF1 (46%) outnumbered those of BRAF (31%), the fourth-most common mutation. Moreover, when compared with the general melanoma population, a significantly higher frequency of mutations in the NF1 gene was observed in the AO subgroup—not the entire study population.

Discussion

Given that veterans constitute a distinct population, there is reasonable interest in investigating characteristic health issues related to military service. Skin cancer—melanoma in particular—has been researched recently in a veteran population. The differences in demographics, tumor characteristics, and melanoma- specific survival in veterans compared with the general population have already been assessed. According to Chang et al, compared with the general population, veterans are more likely to present with metastatic disease and have lower 5-year survival rates.8

Melanoma is one of the most highly mutated malignancies.15 Fortunately, the most common mutation in melanoma, BRAF V600E, is now considered therapeutically targetable. However, there are still many mutations that are less often discussed and not well understood. Regardless of therapeutic implications, all mutations observed in melanoma are worth investigating because a tumor’s genomic profile also can provide prognostic and etiologic information. Developing comprehensive descriptions of melanoma mutational profiles in specific populations is critical to advancing etiologic understanding and informing prevention strategies.

Our results demonstrate the high prevalence of TERT promoter mutations with characteristic ultraviolet signature (C>T) in the study population. This aligns with general evidence that TERT promoter mutations are common in cutaneous melanomas: 77% of this study sample and up to 86% of all mutations are TERT promoter mutations, according to Davis et al.15TERT promoter mutations are positively associated with the initiation, invasion, and metastasis of melanoma. In certain subtypes, there is evidence that the presence of TERT promoter mutations is significantly associated with risk for extranodal metastasis and death.16 The second-most common mutated gene in the veteran study population was CDKN2A/B (43%), and the third-most mutated gene was BRAF (37%).

In chronically sun-exposed skin NF1, NRAS, and occasionally BRAF V600K mutations tend to predominate. BRAF V600E mutations, on the other hand, are rare in these melanomas.15 In our study population, the most prevalent melanoma site was the trunk (31%), which is considered a location with an intermittent pattern of sun exposure.17

This study population also had a higher frequency of CDKN2A/B mutations. High frequencies of CDKN2A/B mutations have been reported in familial melanomas, but only 1 patient with CDKN2A/B mutations had a known family history of melanoma.15 Tumors in the study population showed significantly lower frequency of mutations in ROS1, GRIN2A, KDR, KMT2C (MLL3), KMT2D (MLL2), LRP1B, PTPRT, PTCH1, FAT4, and PREX2 (P < .05).

In this study the subgroup of veterans with AO exposure differed from the whole study population. As such, CDKN2A/B mutations were observed with the same frequency as NF1 mutations (46% each); however, BRAF mutations constituted only 31% of the mutations. In addition, the frequency of NF1 mutations was significantly higher in the AO subgroup compared with the general population, but not in the whole study population.

Our sample also differed from the reference population by showing a significantly higher frequency of TMB-high (ie, ≥ 10 mut/Mb) tumors (71% vs 49%; P = .01).12 Interestingly, no significant difference in the frequency of TMB-high tumors was observed between the AO subgroup and the reference population (69% vs 49%; P = .16). There also was no statistically significant difference between the frequency of MSI-H tumors in our study population and the reference population (P = .64).13

One patient in the study population had uveal melanoma. Mutations encountered in this patient’s tumor differed from the general mutational profile of tumors. None of the 21 mutations depicted in Table 2 were present in this sample.10,11 On the other hand, those mutations frequently observed in intraocular melanomas, BAP1 and GNA11, were present in this patient.18 Additionally, this particular melanoma possessed mutations in genes RICTOR, RAD21, and PIK3R1.

Limitations

This study population consisted exclusively of male patients, introducing sex as a potential confounder in analyzing differences between the study population and the general population. As noted in a 2020 systematic review, there were no sex-based differences in the frequency of mutations in BRAF, NRAS, and KIT genes.19

Regarding NF1 mutations, only NF1-mutated acral and mucosal melanomas were more frequently observed in female patients, whereas nonacral NF1-mutated melanomas were more frequently observed in male patients.20 However, there is currently no clear evidence of whether the mutational landscapes of cutaneous melanoma differ by sex.21 Among the 11 cases with NF1-mutatation, site of origin was known in 6, 5 of which originated at nonacral sites. Although the AO subgroup also consisted entirely of male patients, this does not explain the observed increased frequency of NF1 mutations relative to the general population. No such difference was observed between the whole study population, which also consisted exclusively of male patients, and the general population. The similar frequencies of nonacral location in the whole study population (3 acral, 18 nonacral, 14 unknown site of origin) and AO subgroup (1 acral, 7 nonacral, 5 unknown site of origin) preclude location as an explanation.

The Cancer Genome Atlas Network proposed a framework for genomic classification of melanoma into 4 subtypes based on the pattern of the most prevalent significantly mutated genes: mutant BRAF, mutant RAS, mutant NF1, and triple–wild-type. According to that study, BRAF mutations were indeed associated with younger age, in contrast to the NF1-mutant genomic subtype, which was more prevalent in older individuals with higher TMB.22 This emphasizes the need to interpret the potential association of AO exposure and NF1 mutation in melanoma with caution, although additional studies are required to observe the difference between the veteran population and age-matched general population.

On the other hand, Yu et al reported no significant differences of TMB values between patients aged < 60 and ≥ 60 years with melanoma.23 In short, the observed differences we report in our limited study warrant additional investigation with larger sample sizes, sex-matched controlling, and age-matched controlling. The study was limited by its small sample size and the single location.

Conclusion

The genomic profile of melanomas in the veteran population appears to be similar to that of the general population with a few possible differences. Melanomas in the veteran study population showed a higher frequency of CDKN2A/B mutations; lower frequency of ROS1, GRIN2A, KDR, KMT2C (MLL3), KMT2D (MLL2), LRP1B, PTPRT, PTCH1, FAT4, and PREX2 mutations; and higher TMB. In addition, melanomas in the AO subgroup showed higher frequencies of NF1 mutations. The significance of such findings remains to be determined by further investigation.

References
  1. Bytnar JA, McGlynn KA, et al. Cancer incidence in the US military: An updated analysis. Cancer. 2024;130(1):96-106. doi:10.1002/cncr.34978
  2. Singer DS. A new phase of the Cancer Moonshot to end cancer as we know it. Nat Med. 2022;28(7):1345-1347. doi:10.1038/s41591-022-01881-5
  3. Koczkodaj P, Sulkowska U, Didkowska J, et al. Melanoma mortality trends in 28 European countries: a retrospective analysis for the years 1960-2020. Cancers (Basel). 2023;15(5):1514. Published 2023 Feb 28. doi:10.3390/cancers15051514
  4. Okobi OE, Abreo E, Sams NP, et al. Trends in melanoma incidence, prevalence, stage at diagnosis, and survival: an analysis of the United States Cancer Statistics (USCS) database. Cureus. 2024;16(10):e70697. doi:10.7759/cureus.70697
  5. Bartling SJ, Rivard SC, Meyerle JH. Melanoma in an active duty marine. Mil Med. 2017;182:e2034-e2039. doi:10.7205/MILMED-D-17-00127
  6. American Cancer Society. Cancer facts & figures 2023. American Cancer Society; 2023. Accessed June 20, 2025. https://www.cancer.org/content/dam/cancer-org/research/cancer-facts-and-statistics/annual-cancer-facts-and-figures/2023/2023-cancer-facts-and-figures.pdf
  7. Rezaei SJ, Kim J, Onyeka S, et al. Skin cancer and other dermatologic conditions among US veterans. JAMA Dermatol. 2024;160(10):1107-1111. doi:10.1001/jamadermatol.2024.3043
  8. Chang MS, La J, Trepanowski N, et al. Increased relative proportions of advanced melanoma among veterans: a comparative analysis with the Surveillance, Epidemiology, and End Results registry. J Am Acad Dermatol. 2022;87:72-79. doi:10.1016/j.jaad.2022.02.063
  9. Riemenschneider K, Liu J, Powers JG. Skin cancer in the military: a systematic review of melanoma and nonmelanoma skin cancer incidence, prevention, and screening among active duty and veteran personnel. J Am Acad Dermatol. 2018;78:1185-1192. doi:10.1016/j.jaad.2017.11.062
  10. Huang FW, Hodis E, Xu MJ, et al. Highly recurrent TERT promoter mutations in human melanoma. Science. 2013;339:957-959. doi:10.1126/science.1229259
  11. Tate JG, Bamford S, Jubb HC, et al. COSMIC: the Catalogue of Somatic Mutations in Cancer. Nucleic Acids Res. 2019;47:D941-D947. doi:10.1093/nar/gky1015
  12. Li M, Gao X, Wang X. Identification of tumor mutation burden-associated molecular and clinical features in cancer by analyzing multi-omics data. Front Immunol. 2023;14:1090838. doi:10.3389/fimmu.2023.1090838
  13. Bonneville R, Krook MA, Kautto EA, et al. Landscape of microsatellite instability across 39 cancer types. JCO Precis Oncol. 2017;2017:PO.17.00073. doi:10.1200/PO.17.00073
  14. Lui AJ, Pagadala MS, Zhong AY, et al. Agent Orange exposure and prostate cancer risk in the Million Veteran Program. medRxiv [Preprint]. 2023:2023.06.14.23291413. doi:10.1101/2023.06.14.23291413
  15. Davis EJ, Johnson DB, Sosman JA, et al. Melanoma: what do all the mutations mean? Cancer. 2018;124:3490-3499. doi:10.1002/cncr.31345
  16. Guo Y, Chen Y, Zhang L, et al. TERT promoter mutations and telomerase in melanoma. J Oncol. 2022;2022:6300329. doi:10.1155/2022/6300329
  17. Whiteman DC, Stickley M, Watt P, et al. Anatomic site, sun exposure, and risk of cutaneous melanoma. J Clin Oncol. 2006;24:3172-3177. doi:10.1200/JCO.2006.06.1325
  18. Decatur CL, Ong E, Garg N, et al. Driver mutations in uveal melanoma: associations with gene expression profile and patient outcomes. JAMA Ophthalmol. 2016;134:728-733. doi:10.1001/jamaophthalmol.2016.0903
  19. Gutiérrez-Castañeda LD, Nova JA, Tovar-Parra JD. Frequency of mutations in BRAF, NRAS, and KIT in different populations and histological subtypes of melanoma: a systemic review. Melanoma Res. 2020;30:62- 70. doi:10.1097/CMR.0000000000000628
  20. Thielmann CM, Chorti E, Matull J, et al. NF1-mutated melanomas reveal distinct clinical characteristics depending on tumour origin and respond favourably to immune checkpoint inhibitors. Eur J Cancer. 2021;159:113-124. doi:10.1016/j.ejca.2021.09.035
  21. D’Ecclesiis O, Caini S, Martinoli C, et al. Gender-dependent specificities in cutaneous melanoma predisposition, risk factors, somatic mutations, prognostic and predictive factors: a systematic review. Int J Environ Res Public Health. 2021;18:7945. doi:10.3390/ijerph18157945
  22. Cancer Genome Atlas Network. Genomic classification of cutaneous melanoma. Cell. 2015;161:1681-1696. doi:10.1016/j.cell.2015.05.044
  23. Yu Z, Wang J, Feng L, et al. Association of tumor mutational burden with age in solid tumors. J Clin Oncol. 2020;38:e13590-e13590. doi:10.1200/JCO.2020.38.15_suppl.e13590
References
  1. Bytnar JA, McGlynn KA, et al. Cancer incidence in the US military: An updated analysis. Cancer. 2024;130(1):96-106. doi:10.1002/cncr.34978
  2. Singer DS. A new phase of the Cancer Moonshot to end cancer as we know it. Nat Med. 2022;28(7):1345-1347. doi:10.1038/s41591-022-01881-5
  3. Koczkodaj P, Sulkowska U, Didkowska J, et al. Melanoma mortality trends in 28 European countries: a retrospective analysis for the years 1960-2020. Cancers (Basel). 2023;15(5):1514. Published 2023 Feb 28. doi:10.3390/cancers15051514
  4. Okobi OE, Abreo E, Sams NP, et al. Trends in melanoma incidence, prevalence, stage at diagnosis, and survival: an analysis of the United States Cancer Statistics (USCS) database. Cureus. 2024;16(10):e70697. doi:10.7759/cureus.70697
  5. Bartling SJ, Rivard SC, Meyerle JH. Melanoma in an active duty marine. Mil Med. 2017;182:e2034-e2039. doi:10.7205/MILMED-D-17-00127
  6. American Cancer Society. Cancer facts & figures 2023. American Cancer Society; 2023. Accessed June 20, 2025. https://www.cancer.org/content/dam/cancer-org/research/cancer-facts-and-statistics/annual-cancer-facts-and-figures/2023/2023-cancer-facts-and-figures.pdf
  7. Rezaei SJ, Kim J, Onyeka S, et al. Skin cancer and other dermatologic conditions among US veterans. JAMA Dermatol. 2024;160(10):1107-1111. doi:10.1001/jamadermatol.2024.3043
  8. Chang MS, La J, Trepanowski N, et al. Increased relative proportions of advanced melanoma among veterans: a comparative analysis with the Surveillance, Epidemiology, and End Results registry. J Am Acad Dermatol. 2022;87:72-79. doi:10.1016/j.jaad.2022.02.063
  9. Riemenschneider K, Liu J, Powers JG. Skin cancer in the military: a systematic review of melanoma and nonmelanoma skin cancer incidence, prevention, and screening among active duty and veteran personnel. J Am Acad Dermatol. 2018;78:1185-1192. doi:10.1016/j.jaad.2017.11.062
  10. Huang FW, Hodis E, Xu MJ, et al. Highly recurrent TERT promoter mutations in human melanoma. Science. 2013;339:957-959. doi:10.1126/science.1229259
  11. Tate JG, Bamford S, Jubb HC, et al. COSMIC: the Catalogue of Somatic Mutations in Cancer. Nucleic Acids Res. 2019;47:D941-D947. doi:10.1093/nar/gky1015
  12. Li M, Gao X, Wang X. Identification of tumor mutation burden-associated molecular and clinical features in cancer by analyzing multi-omics data. Front Immunol. 2023;14:1090838. doi:10.3389/fimmu.2023.1090838
  13. Bonneville R, Krook MA, Kautto EA, et al. Landscape of microsatellite instability across 39 cancer types. JCO Precis Oncol. 2017;2017:PO.17.00073. doi:10.1200/PO.17.00073
  14. Lui AJ, Pagadala MS, Zhong AY, et al. Agent Orange exposure and prostate cancer risk in the Million Veteran Program. medRxiv [Preprint]. 2023:2023.06.14.23291413. doi:10.1101/2023.06.14.23291413
  15. Davis EJ, Johnson DB, Sosman JA, et al. Melanoma: what do all the mutations mean? Cancer. 2018;124:3490-3499. doi:10.1002/cncr.31345
  16. Guo Y, Chen Y, Zhang L, et al. TERT promoter mutations and telomerase in melanoma. J Oncol. 2022;2022:6300329. doi:10.1155/2022/6300329
  17. Whiteman DC, Stickley M, Watt P, et al. Anatomic site, sun exposure, and risk of cutaneous melanoma. J Clin Oncol. 2006;24:3172-3177. doi:10.1200/JCO.2006.06.1325
  18. Decatur CL, Ong E, Garg N, et al. Driver mutations in uveal melanoma: associations with gene expression profile and patient outcomes. JAMA Ophthalmol. 2016;134:728-733. doi:10.1001/jamaophthalmol.2016.0903
  19. Gutiérrez-Castañeda LD, Nova JA, Tovar-Parra JD. Frequency of mutations in BRAF, NRAS, and KIT in different populations and histological subtypes of melanoma: a systemic review. Melanoma Res. 2020;30:62- 70. doi:10.1097/CMR.0000000000000628
  20. Thielmann CM, Chorti E, Matull J, et al. NF1-mutated melanomas reveal distinct clinical characteristics depending on tumour origin and respond favourably to immune checkpoint inhibitors. Eur J Cancer. 2021;159:113-124. doi:10.1016/j.ejca.2021.09.035
  21. D’Ecclesiis O, Caini S, Martinoli C, et al. Gender-dependent specificities in cutaneous melanoma predisposition, risk factors, somatic mutations, prognostic and predictive factors: a systematic review. Int J Environ Res Public Health. 2021;18:7945. doi:10.3390/ijerph18157945
  22. Cancer Genome Atlas Network. Genomic classification of cutaneous melanoma. Cell. 2015;161:1681-1696. doi:10.1016/j.cell.2015.05.044
  23. Yu Z, Wang J, Feng L, et al. Association of tumor mutational burden with age in solid tumors. J Clin Oncol. 2020;38:e13590-e13590. doi:10.1200/JCO.2020.38.15_suppl.e13590
Issue
Federal Practitioner - 42(suppl 3)
Issue
Federal Practitioner - 42(suppl 3)
Page Number
S18-S24
Page Number
S18-S24
Publications
Publications
Topics
Article Type
Display Headline

Comprehensive Genomic Profiles of Melanoma in Veterans Compared to Reference Databases

Display Headline

Comprehensive Genomic Profiles of Melanoma in Veterans Compared to Reference Databases

Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Thu, 08/07/2025 - 15:21
Un-Gate On Date
Thu, 08/07/2025 - 15:21
Use ProPublica
CFC Schedule Remove Status
Thu, 08/07/2025 - 15:21
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
survey writer start date
Thu, 08/07/2025 - 15:21
Media Files
825FED_AVAHO_Mel_eApp.jpg

Colorectal Cancer Characteristics and Mortality From Propensity Score-Matched Cohorts of Urban and Rural Veterans

Article Type
Changed
Wed, 08/13/2025 - 15:45
Display Headline

Colorectal Cancer Characteristics and Mortality From Propensity Score-Matched Cohorts of Urban and Rural Veterans

Colorectal cancer (CRC) is the second-leading cause of cancer-related deaths in the United States, with an estimated 52,550 deaths in 2023.1 However, the disease burden varies among different segments of the population.2 While both CRC incidence and mortality have been decreasing due to screening and advances in treatment, there are disparities in incidence and mortality across the sociodemographic spectrum including race, ethnicity, education, and income.1-4 While CRC incidence is decreasing for older adults, it is increasing among those aged < 55 years.5 The incidence of CRC in adults aged 40 to 54 years has increased by 0.5% to 1.3% annually since the mid-1990s.6 The US Preventive Services Task Force now recommends starting CRC screening at age 45 years for asymptomatic adults with average risk.7

Disparities also exist across geographical boundaries and living environment. Rural Americans faces additional challenges in health and lifestyle that can affect CRC outcomes. Compared to their urban counterparts, rural residents are more likely to be older, have lower levels of education, higher levels of poverty, lack health insurance, and less access to health care practitioners (HCPs).8-10 Geographic proximity, defined as travel time or physical distance to a health facility, has been recognized as a predictor of inferior outcomes.11 These aspects of rural living may pose challenges for accessing care for CRC screening and treatment.11-13 National and local studies have shown disparities in CRC screening rates, incidence, and mortality between rural and urban populations.14-16

It is unclear whether rural/urban disparities persist under the Veterans Health Administration (VHA) health care delivery model. This study examined differences in baseline characteristics and mortality between rural and urban veterans newly diagnosed with CRC. We also focused on a subpopulation aged ≤ 45 years.

Methods

This study extracted national data from the US Department of Veterans Affairs (VA) Corporate Data Warehouse (CDW) hosted in the VA Informatics and Computing Infrastructure (VINCI) environment. VINCI is an initiative to improve access to VA data and facilitate the analysis of these data while ensuring veterans’ privacy and data security.17 CDW is the VHA business intelligence information repository, which extracts data from clinical and nonclinical sources following prescribed and validated protocols. Data extracted included demographics, diagnosis, and procedure codes for both inpatient and outpatient encounters, vital signs, and vital status. This study used data previously extracted from a national cohort of veterans that encompassed all patients who received a group of commonly prescribed medications, such as statins, proton pump inhibitors, histamine-2 blockers, acetaminophen-containing products, and hydrocortisone-containing skin applications. This cohort encompassed 8,648,754 veterans, from whom 2,460,727 had encounters during fiscal years (FY) 2016 to 2021 (study period). The cohort was used to ensure that subjects were VHA patients, allowing them to adequately capture their clinical profiles.

Patients were identified as rural or urban based on their residence address at the date of their first diagnosis of CRC. The Geospatial Service Support Center (GSSC) aggregates and updates veterans’ residence address records for all enrolled veterans from the National Change of Address database. The data contain 1 record per enrollee. GSSC Geocoded Enrollee File contains enrollee addresses and their rurality indicators, categorized as urban, rural, or highly rural.18 Rurality is defined by the Rural Urban Commuting Area (RUCA) categories developed by the Department of Agriculture and the Health Resources and Services Administration of the US Department of Health and Human Services.19 Urban areas had RUCA codes of 1.0 to 1.1, and highly rural areas had RUCA scores of 10.0. All other areas were classified as rural. Since the proportion of veterans from highly rural areas was small, we included residents from highly rural areas in the rural residents’ group.

Inclusion and Exclusion Criteria

All veterans newly diagnosed with CRC from FY 2016 to 2021 were included. We used the ninth and tenth clinical modification revisions of the International Classification of Diseases (ICD-9-CM and ICD-10-CM) to define CRC diagnosis (Supplemental materials).4,20 To ensure that patients were newly diagnosed with CRC, this study excluded patients with a previous ICD-9-CM code for CRC diagnosis since FY 2003.

Comorbidities were identified using diagnosis and procedure codes from inpatient and outpatient encounters, which were used to calculate the Charlson Comorbidity Index (CCI) at the time of CRC diagnosis using the weighted method described by Schneeweiss et al.21 We defined CRC high-risk conditions and CRC screening tests, including flexible sigmoidoscopy and stool tests, as described in previous studies (Supplemental materials).20

The main outcome was total mortality. The date of death was extracted from the VHA Death Ascertainment File, which contains mortality data from the Master Person Index file in CDW and the Social Security Administration Death Master File. We used the date of death from any cause, as cause of death was not available.

A propensity score (PS) was created to match rural (including highly rural) and urban residents at a ratio of 1:1. Using a standard procedure described in prior publications, multivariable logistic regression used all baseline characteristics to estimate the PS and perform nearest-number matching without replacement.22,23 A caliper of 0.01 maximized the matched cohort size and achieved balance (Supplemental materials). We then examined the balance of baseline characteristics between PS-matched groups.

Analyses

Cox proportional hazards regression analysis estimated the hazard ratio (HR) of death in rural residents compared to urban residents in the PS-matched cohort. The outcome event was the date of death during the study’s follow-up period (defined as period from first CRC diagnosis to death or study end), with censoring at the study’s end date (September 30, 2021). The proportional hazards assumption was assessed by inspecting the Kaplan-Meier curves. Multiple analyses examined the HR of total mortality in the PS-matched cohort, stratified by sex, race, and ethnicity. We also examined the HR of total mortality stratified by duration of follow-up.

Another PS-matching analysis among veterans aged ≤ 45 years was performed using the same techniques described earlier in this article. We performed a Cox proportional hazards regression analysis to compare mortality in PS-matched urban and rural veterans aged ≤ 45 years. The HR of death in all veterans aged ≤ 45 years (before PS-matching) was estimated using Cox proportional hazard regression analysis, adjusting for PS.

Dichotomous variables were compared using X2 tests and continuous variables were compared using t tests. Baseline characteristics with missing values were converted into categorical variables and the proportion of subjects with missing values was equalized between treatment groups after PS-matching. For subgroup analysis, we examined the HR of total mortality in each subgroup using separate Cox proportional hazards regression models similar to the primary analysis but adjusted for PS. Due to multiple comparisons in the subgroup analysis, the findings should be considered exploratory. Statistical tests were 2-tailed, and significance was defined as P < .05. Data management and statistical analyses were conducted from June 2022 to January 2023 using STATA, Version 17. The VA Orlando Healthcare System Institutional Review Board approved the study and waived requirements for informed consent because only deidentified data were used.

Results

After excluding 49 patients (Supplemental materials, available at doi:10.12788/fp.0560), we identified 30,219 veterans with newly diagnosed CRC between FY 2016 to 2021 (Table 1). Of these, 19,422 (64.3%) resided in urban areas and 10,797 (35.7%) resided in rural areas (Table 2). The mean (SD) duration from the first CRC diagnosis to death or study end was 832 (640) days, and the median (IQR) was 723 (246–1330) days. Overall, incident CRC diagnoses were numerically highest in FY 2016 and lowest in FY 2020 (Figure 1). Patients with CRC in rural areas vs urban areas were significantly older (mean, 71.2 years vs 70.8 years, respectively; P < .001), more likely to be male (96.7% vs 95.7%, respectively; P < .001), more likely to be White (83.6% vs 67.8%, respectively; P < .001) and more likely to be non-Hispanic (92.2% vs 87.5%, respectively; P < .001). In terms of general health, rural veterans with CRC were more likely to be overweight or obese (81.5% rural vs 78.5% urban; P < .001) but had fewer mean comorbidities as measured by CCI (5.66 rural vs 5.90 urban; P < .001). A higher proportion of rural veterans with CRC had received stool-based (fecal occult blood test or fecal immunochemical test) CRC screening tests (61.6% rural vs 57.2% urban; P < .001). Fewer rural patients presented with systemic symptoms or signs within 1 year of CRC diagnosis (54.4% rural vs 57.5% urban, P < .001). Among urban patients with CRC, 6959 (35.8%) deaths were observed, compared with 3766 (34.9%) among rural patients (P = .10).

0525FED-AVAHO-CRC_T10525FED-AVAHO-CRC_T20525FED-AVAHO-CRC_F1

There were 21,568 PS-matched veterans: 10,784 in each group. In the PS-matched cohort, baseline characteristics were similar between veterans in urban and rural communities, including age, sex, race/ethnicity, body mass index, and comorbidities. Among rural patients with CRC, 3763 deaths (34.9%) were observed compared with 3702 (34.3%) among urban veterans. There was no significant difference in the HR of mortality between rural and urban CRC residents (HR, 1.01; 95% CI, 0.97-1.06; P = .53) (Figure 2).

0525FED-AVAHO-CRC_F20525FED-AVAHO-CRC_F30525FED-AVAHO-CRC_F4

Among veterans aged ≤ 45 years, 551 were diagnosed with CRC (391 urban and 160 rural). We PS-matched 142 pairs of urban and rural veterans without residual differences in baseline characteristics (eAppendix 1). There was no significant difference in the HR of mortality between rural and urban veterans aged ≤ 45 years (HR, 0.97; 95% CI, 0.57-1.63; P = .90) (Figure 2). Similarly, no difference in mortality was observed adjusting for PS between all rural and urban veterans aged ≤ 45 years (HR, 1.03; 95% CI, 0.67-1.59; P = .88).

0525FED-AVAHO-CRC_eApp1

There was no difference in total mortality between rural and urban veterans in any subgroup except for American Indian or Alaska Native veterans (HR, 2.41; 95% CI, 1.29-4.50; P = .006) (eAppendix 2).

0525FED-AVAHO-CRC_eApp2

Discussion

This study examined characteristics of patients with CRC between urban and rural areas among veterans who were VHA patients. Similar to other studies, rural veterans with CRC were older, more likely to be White, and were obese, but exhibited fewer comorbidities (lower CCI and lower incidence of congestive heart failure, dementia, hemiplegia, kidney diseases, liver diseases and AIDS, but higher incidence of chronic obstructive lung disease).8,16 The incidence of CRC in this study population was lowest in FY 2020, which was reported by the Centers for Disease Control and Prevention and is attributed to COVID-19 pandemic disruption of health services.24 The overall mortality in this study was similar to rates reported in other studies from the VA Central Cancer Registry.4 In the PS-matched cohort, where baseline characteristics were similar between urban and rural patients with CRC, we found no disparities in CRC-specific mortality between veterans in rural and urban areas. Additionally, when analysis was restricted to veterans aged ≤ 45 years, the results remained consistent.

Subgroup analyses showed no significant difference in mortality between rural and urban areas by sex, race or ethnicity, except rural American Indian or Alaska Native veterans who had double the mortality of their urban counterparts (HR, 2.41; 95% CI, 1.29-4.50; P = .006). This finding is difficult to interpret due to the small number of events and the wide CI. While with a Bonferroni correction the adjusted P value was .08, which is not statistically significant, a previous study found that although CRC incidence was lower overall in American Indian or Alaska Native populations compared to non-Hispanic White populations, CRC incidence was higher among American Indian or Alaska Native individuals in some areas such as Alaska and the Northern Plains.25,26 Studies have noted that rural American Indian/Alaska Native populations experience greater poverty, less access to broadband internet, and limited access to care, contributing to poorer cancer outcomes and lower survival.27 Thus, the finding of disparity in mortality between rural and urban American Indian or Alaska Native veterans warrants further study.

Other studies have raised concerns that CRC disproportionately affects adults in rural areas with higher mortality rates.14-16 These disparities arise from sociodemographic factors and modifiable risk factors, including physical activity, dietary patterns, access to cancer screening, and gaps in quality treatment resources.16,28 These factors operate at multiple levels: from individual, local health system, to community and policy.2,27 For example, a South Carolina study (1996–2016) found that residents in rural areas were more likely to be diagnosed with advanced CRC, possibly indicating lower rates of CRC screening in rural areas. They also had higher likelihood of death from CRC.15 However, the study did not include any clinical parameters, such as comorbidities or obesity. A statewide, population-based study in Utah showed that rural men experienced a lower CRC survival in their unadjusted analysis.16 However, the study was small, with only 3948 urban and 712 rural residents. Additionally, there was no difference in total mortality in the whole cohort (HR, 0.96; 95% CI, 0.86-1.07) or in CRC-specific death (HR, 0.93; 95% CI, 0.81-1.08). A nationwide study also showed that CRC mortality rates were 8% higher in nonmetropolitan or rural areas than in the most urbanized areas containing large metropolitan counties.29 However, this study did not include descriptions of clinical confounders, such as comorbidities, making it difficult to ascertain whether the difference in CRC mortality was due to rurality or differences in baseline risk characteristics.

In this study, the lack of CRC-specific mortality disparities may be attributed to the structures and practices of VHA health care. Recent studies have noted that mortality of several chronic medical conditions treated at the VHA was lower than at non-VHA hospitals.30,31 One study that measured the quality of nonmetastatic CRC care based on National Comprehensive Cancer Network guidelines showed that > 72% of VHA patients received guideline-concordant care for each diagnostic and therapeutic measure, except for follow-up colonoscopy timing, which appear to be similar or superior to that of the private sector.30,32,33 Some of the VA initiative for CRC screening may bypass the urban-rurality divide such as the mailed fecal immunochemical test program for CRC. This program was implemented at the onset of the COVID-19 pandemic to avoid disruptions of medical care.34 Rural patients are more likely to undergo fecal immunochemical testing when compared to urban patients in this data. Beyond clinical care, the VHA uses processes to tackle social determinants of health such as housing, food security, and transportation, promoting equal access to health care, and promoting cultural competency among HCPs.35-37

The results suggest that solutions to CRC disparities between rural and urban areas need to consider known barriers to rural health care, including transportation, diminished rural health care workforce, and other social determinants of health.9,10,27,38 VHA makes considerable efforts to provide equitable care to all enrolled veterans, including specific programs for rural veterans, including ongoing outreach.39 This study demonstrated lack of disparity in CRC-specific mortality in veterans receiving VHA care, highlighting the importance of these efforts.

Strengths and Limitations

This study used the VHA cohort to compare patient characteristics and mortality between patients with CRC residing in rural and urban areas. The study provides nationwide perspectives on CRC across the geographical spectrum and used a longitudinal cohort with prolonged follow-up to account for comorbidities.

However, the study compared a cohort of rural and urban veterans enrolled in the VHA; hence, the results may not reflect CRC outcomes in veterans without access to VHA care. Rurality has been independently associated with decreased likelihood of meeting CRC screening guidelines among veterans and military service members.38 This study lacked sufficient information to compare CRC staging or treatment modalities among veterans. Although the data cannot identify CRC stage, the proportions of patients with metastatic CRC at diagnosis and CRC location were similar between groups. The study did not have information on their care outside of VHA setting.

This study could not ascertain whether disparities existed in CRC treatment modality since rural residence may result in referral to community-based CRC care, which did not appear in the data. To address these limitations, we used death from any cause as the primary outcome, since death is a hard outcome and is not subject to ascertainment bias. The relatively short follow-up time is another limitation, though subgroup analysis by follow-up did not show significant differences. Despite PS matching, residual unmeasured confounding may exist between urban and rural groups. The predominantly White, male VHA population with high CCI may limit the generalizability of the results.

Conclusions

Rural VHA enrollees had similar survival rates after CRC diagnosis compared to their urban counterparts in a PS-matched analysis. The VHA models of care—including mailed CRC screening tools, several socioeconomic determinants of health (housing, food security, and transportation), and promoting equal access to health care, as well as cultural competency among HCPs—HCPs—may help alleviate disparities across the rural-urban spectrum. The VHA should continue efforts to enroll veterans and provide comprehensive coordinated care in community partnerships.

References
  1. Siegel RL, Wagle NS, Cercek A, Smith RA, Jemal A. Colorectal cancer statistics, 2023. CA Cancer J Clin. 2023;73(3):233-254. doi:10.3322/caac.21772
  2. Carethers JM, Doubeni CA. Causes of socioeconomic disparities in colorectal cancer and intervention framework and strategies. Gastroenterology. 2020;158(2):354-367. doi:10.1053/j.gastro.2019.10.029
  3. Murphy G, Devesa SS, Cross AJ, Inskip PD, McGlynn KA, Cook MB. Sex disparities in colorectal cancer incidence by anatomic subsite, race and age. Int J Cancer. 2011;128(7):1668-75. doi:10.1002/ijc.25481
  4. Zullig LL, Smith VA, Jackson GL, et al. Colorectal cancer statistics from the Veterans Affairs central cancer registry. Clin Colorectal Cancer. 2016;15(4):e199-e204. doi:10.1016/j.clcc.2016.04.005
  5. Lin JS, Perdue LA, Henrikson NB, Bean SI, Blasi PR. Screening for Colorectal Cancer: An Evidence Update for the US Preventive Services Task Force. 2021. U.S. Preventive Services Task Force Evidence Syntheses, formerly Systematic Evidence Reviews:Chapter 1. Agency for Healthcare Research and Quality (US); 2021. Accessed February 18, 2025. https://www.ncbi.nlm.nih.gov/books/NBK570917/
  6. Siegel RL, Fedewa SA, Anderson WF, et al. Colorectal cancer incidence patterns in the United States, 1974-2013. J Natl Cancer Inst. 2017;109(8). doi:10.1093/jnci/djw322
  7. Davidson KW, Barry MJ, Mangione CM, et al. Screening for colorectal cancer: US Preventive Services Task Force recommendation statement. JAMA. 2021;325(19):1965-1977. doi:10.1001/jama.2021.6238
  8. Hines R, Markossian T, Johnson A, Dong F, Bayakly R. Geographic residency status and census tract socioeconomic status as determinants of colorectal cancer outcomes. Am J Public Health. 2014;104(3):e63-e71. doi:10.2105/AJPH.2013.301572
  9. Cauwels J. The many barriers to high-quality rural health care. 2022;(9):1-32. NEJM Catal Innov Care Deliv. Accessed April 24, 2025. https://catalyst.nejm.org/doi/pdf/10.1056/CAT.22.0254
  10. Gong G, Phillips SG, Hudson C, Curti D, Philips BU. Higher US rural mortality rates linked to socioeconomic status, physician shortages, and lack of health insurance. Health Aff (Millwood);38(12):2003-2010. doi:10.1377/hlthaff.2019.00722
  11. Aboagye JK, Kaiser HE, Hayanga AJ. Rural-urban differences in access to specialist providers of colorectal cancer care in the United States: a physician workforce issue. JAMA Surg. 2014;149(6):537-543. doi:10.1001/jamasurg.2013.5062
  12. Lyckholm LJ, Hackney MH, Smith TJ. Ethics of rural health care. Crit Rev Oncol Hematol. 2001;40(2):131-138. doi:10.1016/s1040-8428(01)00139-1
  13. Krieger N, Williams DR, Moss NE. Measuring social class in US public health research: concepts, methodologies, and guidelines. Annu Rev Public Health. 1997;18:341-378. doi:10.1146/annurev.publhealth.18.1.341
  14. Singh GK, Jemal A. Socioeconomic and racial/ethnic disparities in cancer mortality, incidence, and survival in the United States, 1950-2014: over six decades of changing patterns and widening inequalities. J Environ Public Health. 2017;2017:2819372. doi:10.1155/2017/2819372
  15. Adams SA, Zahnd WE, Ranganathan R, et al. Rural and racial disparities in colorectal cancer incidence and mortality in South Carolina, 1996 - 2016. J Rural Health. 2022;38(1):34-39. doi:10.1111/jrh.12580
  16. Rogers CR, Blackburn BE, Huntington M, et al. Rural- urban disparities in colorectal cancer survival and risk among men in Utah: a statewide population-based study. Cancer Causes Control. 2020;31(3):241-253. doi:10.1007/s10552-020-01268-2
  17. US Department of Veterans Affairs. VA Informatics and Computing Infrastructure (VINCI), VA HSR RES 13-457. https://vincicentral.vinci.med.va.gov [Source not verified]
  18. US Department of Veterans Affairs Information Resource Center. VIReC Research User Guide: PSSG Geocoded Enrollee Files, 2015 Edition. US Department of Veterans Affairs, Health Services Research & Development Service, Information Resource Center; May. 2016. [source not verified]
  19. Goldsmith HF, Puskin DS, Stiles DJ. Improving the operational definition of “rural areas” for federal programs. US Department of Health and Human Services; 1993. Accessed February 27, 2025. https://www.ruralhealthinfo.org/pdf/improving-the-operational-definition-of-rural-areas.pdf
  20. Adams MA, Kerr EA, Dominitz JA, et al. Development and validation of a new ICD-10-based screening colonoscopy overuse measure in a large integrated healthcare system: a retrospective observational study. BMJ Qual Saf. 2023;32(7):414-424. doi:10.1136/bmjqs-2021-014236
  21. Schneeweiss S, Wang PS, Avorn J, Glynn RJ. Improved comorbidity adjustment for predicting mortality in Medicare populations. Health Serv Res. 2003;38(4):1103-1120. doi:10.1111/1475-6773.00165
  22. Becker S, Ichino A. Estimation of average treatment effects based on propensity scores. The Stata Journal. 2002;2(4):358-377.
  23. Leuven E, Sianesi B. PSMATCH2: Stata module to perform full Mahalanobis and propensity score matching, common support graphing, and covariate imbalance testing. Statistical software components. Revised February 1, 2018. Accessed February 27, 2025. https://ideas.repec.org/c/boc/bocode/s432001.html.
  24. US Cancer Statistics Working Group. US cancer statistics data visualizations tool. Centers for Disease Control and Prevention. June 2024. Accessed February 27, 2025. https://www.cdc.gov/cancer/dataviz
  25. Cao J, Zhang S. Multiple Comparison Procedures. JAMA. 2014;312(5):543-544. doi:10.1001/jama.2014.9440
  26. Gopalani SV, Janitz AE, Martinez SA, et al. Trends in cancer incidence among American Indians and Alaska Natives and Non-Hispanic Whites in the United States, 1999-2015. Epidemiology. 2020;31(2):205-213. doi:10.1097/EDE.0000000000001140
  27. Zahnd WE, Murphy C, Knoll M, et al. The intersection of rural residence and minority race/ethnicity in cancer disparities in the United States. Int J Environ Res Public Health. 2021;18(4). doi:10.3390/ijerph18041384
  28. Blake KD, Moss JL, Gaysynsky A, Srinivasan S, Croyle RT. Making the case for investment in rural cancer control: an analysis of rural cancer incidence, mortality, and funding trends. Cancer Epidemiol Biomarkers Prev. 2017;26(7):992-997. doi:10.1158/1055-9965.EPI-17-0092
  29. Singh GK, Williams SD, Siahpush M, Mulhollen A. Socioeconomic, rural-urban, and racial inequalities in US cancer mortality: part i-all cancers and lung cancer and part iicolorectal, prostate, breast, and cervical cancers. J Cancer Epidemiol. 2011;2011:107497. doi:10.1155/2011/107497
  30. Jackson GL, Melton LD, Abbott DH, et al. Quality of nonmetastatic colorectal cancer care in the Department of Veterans Affairs. J Clin Oncol. 2010;28(19):3176-3181. doi:10.1200/JCO.2009.26.7948
  31. Yoon J, Phibbs CS, Ong MK, et al. Outcomes of veterans treated in Veterans Affairs hospitals vs non-Veterans Affairs hospitals. JAMA Netw Open. 2023;6(12):e2345898. doi:10.1001/jamanetworkopen.2023.45898
  32. Malin JL, Schneider EC, Epstein AM, Adams J, Emanuel EJ, Kahn KL. Results of the National Initiative for Cancer Care Quality: how can we improve the quality of cancer care in the United States? J Clin Oncol. 2006;24(4):626-634. doi:10.1200/JCO.2005.03.3365
  33. Levin B, Lieberman DA, McFarland B, et al. Screening and surveillance for the early detection of colorectal cancer and adenomatous polyps, 2008: a joint guideline from the American Cancer Society, the US Multi-Society Task Force on Colorectal Cancer, and the American College of Radiology. Gastroenterology. 2008;134(5):1570-1595. doi:10.1053/j.gastro.2008.02.002
  34. Deeds SA, Moore CB, Gunnink EJ, et al. Implementation of a mailed faecal immunochemical test programme for colorectal cancer screening among Veterans. BMJ Open Qual. 2022;11(4). doi:10.1136/bmjoq-2022-001927
  35. Yehia BR, Greenstone CL, Hosenfeld CB, Matthews KL, Zephyrin LC. The role of VA community care in addressing health and health care disparities. Med Care. 2017;55(Suppl 9 suppl 2):S4-S5. doi:10.1097/MLR.0000000000000768
  36. Wright BN, MacDermid Wadsworth S, Wellnitz A, Eicher- Miller HA. Reaching rural veterans: a new mechanism to connect rural, low-income US Veterans with resources and improve food security. J Public Health (Oxf). 2019;41(4):714-723. doi:10.1093/pubmed/fdy203
  37. Nelson RE, Byrne TH, Suo Y, et al. Association of temporary financial assistance with housing stability among US veterans in the supportive services for veteran families program. JAMA Netw Open. 2021;4(2):e2037047. doi:10.1001/jamanetworkopen.2020.37047
  38. McDaniel JT, Albright D, Lee HY, et al. Rural–urban disparities in colorectal cancer screening among military service members and Veterans. J Mil Veteran Fam Health. 2019;5(1):40-48. doi:10.3138/jmvfh.2018-0013
  39. US Department of Veterans Affairs, Office of Rural Health. The rural veteran outreach toolkit. Updated February 12, 2025. Accessed February 18, 2025. https://www.ruralhealth.va.gov/partners/toolkit.asp
Article PDF
Author and Disclosure Information

Minh Anh Le, MDa; Po-Hong Liu, MDb; Amar Mandalia, MDc; Sergio Romero, MDd; Ishak A. Mansi, MDa,c; Moheb Boktor, MDb

Author affiliations: 
aUniversity of Central Florida, Orlando 
bUniversity of Texas Southwestern Medical Center, Dallas 
cOrlando Veterans Affairs Medical Center, Florida 
dNorth Florida/South Georgia Veterans Health System, Gainesville

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

Correspondence: Ishak Mansi ([email protected])

Fed Pract. 2025;42(suppl 2). Published online May 8. doi:10.12788/fp.0560

Issue
Federal Practitioner - 42(5)s
Publications
Topics
Page Number
S22-S31
Sections
Author and Disclosure Information

Minh Anh Le, MDa; Po-Hong Liu, MDb; Amar Mandalia, MDc; Sergio Romero, MDd; Ishak A. Mansi, MDa,c; Moheb Boktor, MDb

Author affiliations: 
aUniversity of Central Florida, Orlando 
bUniversity of Texas Southwestern Medical Center, Dallas 
cOrlando Veterans Affairs Medical Center, Florida 
dNorth Florida/South Georgia Veterans Health System, Gainesville

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

Correspondence: Ishak Mansi ([email protected])

Fed Pract. 2025;42(suppl 2). Published online May 8. doi:10.12788/fp.0560

Author and Disclosure Information

Minh Anh Le, MDa; Po-Hong Liu, MDb; Amar Mandalia, MDc; Sergio Romero, MDd; Ishak A. Mansi, MDa,c; Moheb Boktor, MDb

Author affiliations: 
aUniversity of Central Florida, Orlando 
bUniversity of Texas Southwestern Medical Center, Dallas 
cOrlando Veterans Affairs Medical Center, Florida 
dNorth Florida/South Georgia Veterans Health System, Gainesville

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

Correspondence: Ishak Mansi ([email protected])

Fed Pract. 2025;42(suppl 2). Published online May 8. doi:10.12788/fp.0560

Article PDF
Article PDF

Colorectal cancer (CRC) is the second-leading cause of cancer-related deaths in the United States, with an estimated 52,550 deaths in 2023.1 However, the disease burden varies among different segments of the population.2 While both CRC incidence and mortality have been decreasing due to screening and advances in treatment, there are disparities in incidence and mortality across the sociodemographic spectrum including race, ethnicity, education, and income.1-4 While CRC incidence is decreasing for older adults, it is increasing among those aged < 55 years.5 The incidence of CRC in adults aged 40 to 54 years has increased by 0.5% to 1.3% annually since the mid-1990s.6 The US Preventive Services Task Force now recommends starting CRC screening at age 45 years for asymptomatic adults with average risk.7

Disparities also exist across geographical boundaries and living environment. Rural Americans faces additional challenges in health and lifestyle that can affect CRC outcomes. Compared to their urban counterparts, rural residents are more likely to be older, have lower levels of education, higher levels of poverty, lack health insurance, and less access to health care practitioners (HCPs).8-10 Geographic proximity, defined as travel time or physical distance to a health facility, has been recognized as a predictor of inferior outcomes.11 These aspects of rural living may pose challenges for accessing care for CRC screening and treatment.11-13 National and local studies have shown disparities in CRC screening rates, incidence, and mortality between rural and urban populations.14-16

It is unclear whether rural/urban disparities persist under the Veterans Health Administration (VHA) health care delivery model. This study examined differences in baseline characteristics and mortality between rural and urban veterans newly diagnosed with CRC. We also focused on a subpopulation aged ≤ 45 years.

Methods

This study extracted national data from the US Department of Veterans Affairs (VA) Corporate Data Warehouse (CDW) hosted in the VA Informatics and Computing Infrastructure (VINCI) environment. VINCI is an initiative to improve access to VA data and facilitate the analysis of these data while ensuring veterans’ privacy and data security.17 CDW is the VHA business intelligence information repository, which extracts data from clinical and nonclinical sources following prescribed and validated protocols. Data extracted included demographics, diagnosis, and procedure codes for both inpatient and outpatient encounters, vital signs, and vital status. This study used data previously extracted from a national cohort of veterans that encompassed all patients who received a group of commonly prescribed medications, such as statins, proton pump inhibitors, histamine-2 blockers, acetaminophen-containing products, and hydrocortisone-containing skin applications. This cohort encompassed 8,648,754 veterans, from whom 2,460,727 had encounters during fiscal years (FY) 2016 to 2021 (study period). The cohort was used to ensure that subjects were VHA patients, allowing them to adequately capture their clinical profiles.

Patients were identified as rural or urban based on their residence address at the date of their first diagnosis of CRC. The Geospatial Service Support Center (GSSC) aggregates and updates veterans’ residence address records for all enrolled veterans from the National Change of Address database. The data contain 1 record per enrollee. GSSC Geocoded Enrollee File contains enrollee addresses and their rurality indicators, categorized as urban, rural, or highly rural.18 Rurality is defined by the Rural Urban Commuting Area (RUCA) categories developed by the Department of Agriculture and the Health Resources and Services Administration of the US Department of Health and Human Services.19 Urban areas had RUCA codes of 1.0 to 1.1, and highly rural areas had RUCA scores of 10.0. All other areas were classified as rural. Since the proportion of veterans from highly rural areas was small, we included residents from highly rural areas in the rural residents’ group.

Inclusion and Exclusion Criteria

All veterans newly diagnosed with CRC from FY 2016 to 2021 were included. We used the ninth and tenth clinical modification revisions of the International Classification of Diseases (ICD-9-CM and ICD-10-CM) to define CRC diagnosis (Supplemental materials).4,20 To ensure that patients were newly diagnosed with CRC, this study excluded patients with a previous ICD-9-CM code for CRC diagnosis since FY 2003.

Comorbidities were identified using diagnosis and procedure codes from inpatient and outpatient encounters, which were used to calculate the Charlson Comorbidity Index (CCI) at the time of CRC diagnosis using the weighted method described by Schneeweiss et al.21 We defined CRC high-risk conditions and CRC screening tests, including flexible sigmoidoscopy and stool tests, as described in previous studies (Supplemental materials).20

The main outcome was total mortality. The date of death was extracted from the VHA Death Ascertainment File, which contains mortality data from the Master Person Index file in CDW and the Social Security Administration Death Master File. We used the date of death from any cause, as cause of death was not available.

A propensity score (PS) was created to match rural (including highly rural) and urban residents at a ratio of 1:1. Using a standard procedure described in prior publications, multivariable logistic regression used all baseline characteristics to estimate the PS and perform nearest-number matching without replacement.22,23 A caliper of 0.01 maximized the matched cohort size and achieved balance (Supplemental materials). We then examined the balance of baseline characteristics between PS-matched groups.

Analyses

Cox proportional hazards regression analysis estimated the hazard ratio (HR) of death in rural residents compared to urban residents in the PS-matched cohort. The outcome event was the date of death during the study’s follow-up period (defined as period from first CRC diagnosis to death or study end), with censoring at the study’s end date (September 30, 2021). The proportional hazards assumption was assessed by inspecting the Kaplan-Meier curves. Multiple analyses examined the HR of total mortality in the PS-matched cohort, stratified by sex, race, and ethnicity. We also examined the HR of total mortality stratified by duration of follow-up.

Another PS-matching analysis among veterans aged ≤ 45 years was performed using the same techniques described earlier in this article. We performed a Cox proportional hazards regression analysis to compare mortality in PS-matched urban and rural veterans aged ≤ 45 years. The HR of death in all veterans aged ≤ 45 years (before PS-matching) was estimated using Cox proportional hazard regression analysis, adjusting for PS.

Dichotomous variables were compared using X2 tests and continuous variables were compared using t tests. Baseline characteristics with missing values were converted into categorical variables and the proportion of subjects with missing values was equalized between treatment groups after PS-matching. For subgroup analysis, we examined the HR of total mortality in each subgroup using separate Cox proportional hazards regression models similar to the primary analysis but adjusted for PS. Due to multiple comparisons in the subgroup analysis, the findings should be considered exploratory. Statistical tests were 2-tailed, and significance was defined as P < .05. Data management and statistical analyses were conducted from June 2022 to January 2023 using STATA, Version 17. The VA Orlando Healthcare System Institutional Review Board approved the study and waived requirements for informed consent because only deidentified data were used.

Results

After excluding 49 patients (Supplemental materials, available at doi:10.12788/fp.0560), we identified 30,219 veterans with newly diagnosed CRC between FY 2016 to 2021 (Table 1). Of these, 19,422 (64.3%) resided in urban areas and 10,797 (35.7%) resided in rural areas (Table 2). The mean (SD) duration from the first CRC diagnosis to death or study end was 832 (640) days, and the median (IQR) was 723 (246–1330) days. Overall, incident CRC diagnoses were numerically highest in FY 2016 and lowest in FY 2020 (Figure 1). Patients with CRC in rural areas vs urban areas were significantly older (mean, 71.2 years vs 70.8 years, respectively; P < .001), more likely to be male (96.7% vs 95.7%, respectively; P < .001), more likely to be White (83.6% vs 67.8%, respectively; P < .001) and more likely to be non-Hispanic (92.2% vs 87.5%, respectively; P < .001). In terms of general health, rural veterans with CRC were more likely to be overweight or obese (81.5% rural vs 78.5% urban; P < .001) but had fewer mean comorbidities as measured by CCI (5.66 rural vs 5.90 urban; P < .001). A higher proportion of rural veterans with CRC had received stool-based (fecal occult blood test or fecal immunochemical test) CRC screening tests (61.6% rural vs 57.2% urban; P < .001). Fewer rural patients presented with systemic symptoms or signs within 1 year of CRC diagnosis (54.4% rural vs 57.5% urban, P < .001). Among urban patients with CRC, 6959 (35.8%) deaths were observed, compared with 3766 (34.9%) among rural patients (P = .10).

0525FED-AVAHO-CRC_T10525FED-AVAHO-CRC_T20525FED-AVAHO-CRC_F1

There were 21,568 PS-matched veterans: 10,784 in each group. In the PS-matched cohort, baseline characteristics were similar between veterans in urban and rural communities, including age, sex, race/ethnicity, body mass index, and comorbidities. Among rural patients with CRC, 3763 deaths (34.9%) were observed compared with 3702 (34.3%) among urban veterans. There was no significant difference in the HR of mortality between rural and urban CRC residents (HR, 1.01; 95% CI, 0.97-1.06; P = .53) (Figure 2).

0525FED-AVAHO-CRC_F20525FED-AVAHO-CRC_F30525FED-AVAHO-CRC_F4

Among veterans aged ≤ 45 years, 551 were diagnosed with CRC (391 urban and 160 rural). We PS-matched 142 pairs of urban and rural veterans without residual differences in baseline characteristics (eAppendix 1). There was no significant difference in the HR of mortality between rural and urban veterans aged ≤ 45 years (HR, 0.97; 95% CI, 0.57-1.63; P = .90) (Figure 2). Similarly, no difference in mortality was observed adjusting for PS between all rural and urban veterans aged ≤ 45 years (HR, 1.03; 95% CI, 0.67-1.59; P = .88).

0525FED-AVAHO-CRC_eApp1

There was no difference in total mortality between rural and urban veterans in any subgroup except for American Indian or Alaska Native veterans (HR, 2.41; 95% CI, 1.29-4.50; P = .006) (eAppendix 2).

0525FED-AVAHO-CRC_eApp2

Discussion

This study examined characteristics of patients with CRC between urban and rural areas among veterans who were VHA patients. Similar to other studies, rural veterans with CRC were older, more likely to be White, and were obese, but exhibited fewer comorbidities (lower CCI and lower incidence of congestive heart failure, dementia, hemiplegia, kidney diseases, liver diseases and AIDS, but higher incidence of chronic obstructive lung disease).8,16 The incidence of CRC in this study population was lowest in FY 2020, which was reported by the Centers for Disease Control and Prevention and is attributed to COVID-19 pandemic disruption of health services.24 The overall mortality in this study was similar to rates reported in other studies from the VA Central Cancer Registry.4 In the PS-matched cohort, where baseline characteristics were similar between urban and rural patients with CRC, we found no disparities in CRC-specific mortality between veterans in rural and urban areas. Additionally, when analysis was restricted to veterans aged ≤ 45 years, the results remained consistent.

Subgroup analyses showed no significant difference in mortality between rural and urban areas by sex, race or ethnicity, except rural American Indian or Alaska Native veterans who had double the mortality of their urban counterparts (HR, 2.41; 95% CI, 1.29-4.50; P = .006). This finding is difficult to interpret due to the small number of events and the wide CI. While with a Bonferroni correction the adjusted P value was .08, which is not statistically significant, a previous study found that although CRC incidence was lower overall in American Indian or Alaska Native populations compared to non-Hispanic White populations, CRC incidence was higher among American Indian or Alaska Native individuals in some areas such as Alaska and the Northern Plains.25,26 Studies have noted that rural American Indian/Alaska Native populations experience greater poverty, less access to broadband internet, and limited access to care, contributing to poorer cancer outcomes and lower survival.27 Thus, the finding of disparity in mortality between rural and urban American Indian or Alaska Native veterans warrants further study.

Other studies have raised concerns that CRC disproportionately affects adults in rural areas with higher mortality rates.14-16 These disparities arise from sociodemographic factors and modifiable risk factors, including physical activity, dietary patterns, access to cancer screening, and gaps in quality treatment resources.16,28 These factors operate at multiple levels: from individual, local health system, to community and policy.2,27 For example, a South Carolina study (1996–2016) found that residents in rural areas were more likely to be diagnosed with advanced CRC, possibly indicating lower rates of CRC screening in rural areas. They also had higher likelihood of death from CRC.15 However, the study did not include any clinical parameters, such as comorbidities or obesity. A statewide, population-based study in Utah showed that rural men experienced a lower CRC survival in their unadjusted analysis.16 However, the study was small, with only 3948 urban and 712 rural residents. Additionally, there was no difference in total mortality in the whole cohort (HR, 0.96; 95% CI, 0.86-1.07) or in CRC-specific death (HR, 0.93; 95% CI, 0.81-1.08). A nationwide study also showed that CRC mortality rates were 8% higher in nonmetropolitan or rural areas than in the most urbanized areas containing large metropolitan counties.29 However, this study did not include descriptions of clinical confounders, such as comorbidities, making it difficult to ascertain whether the difference in CRC mortality was due to rurality or differences in baseline risk characteristics.

In this study, the lack of CRC-specific mortality disparities may be attributed to the structures and practices of VHA health care. Recent studies have noted that mortality of several chronic medical conditions treated at the VHA was lower than at non-VHA hospitals.30,31 One study that measured the quality of nonmetastatic CRC care based on National Comprehensive Cancer Network guidelines showed that > 72% of VHA patients received guideline-concordant care for each diagnostic and therapeutic measure, except for follow-up colonoscopy timing, which appear to be similar or superior to that of the private sector.30,32,33 Some of the VA initiative for CRC screening may bypass the urban-rurality divide such as the mailed fecal immunochemical test program for CRC. This program was implemented at the onset of the COVID-19 pandemic to avoid disruptions of medical care.34 Rural patients are more likely to undergo fecal immunochemical testing when compared to urban patients in this data. Beyond clinical care, the VHA uses processes to tackle social determinants of health such as housing, food security, and transportation, promoting equal access to health care, and promoting cultural competency among HCPs.35-37

The results suggest that solutions to CRC disparities between rural and urban areas need to consider known barriers to rural health care, including transportation, diminished rural health care workforce, and other social determinants of health.9,10,27,38 VHA makes considerable efforts to provide equitable care to all enrolled veterans, including specific programs for rural veterans, including ongoing outreach.39 This study demonstrated lack of disparity in CRC-specific mortality in veterans receiving VHA care, highlighting the importance of these efforts.

Strengths and Limitations

This study used the VHA cohort to compare patient characteristics and mortality between patients with CRC residing in rural and urban areas. The study provides nationwide perspectives on CRC across the geographical spectrum and used a longitudinal cohort with prolonged follow-up to account for comorbidities.

However, the study compared a cohort of rural and urban veterans enrolled in the VHA; hence, the results may not reflect CRC outcomes in veterans without access to VHA care. Rurality has been independently associated with decreased likelihood of meeting CRC screening guidelines among veterans and military service members.38 This study lacked sufficient information to compare CRC staging or treatment modalities among veterans. Although the data cannot identify CRC stage, the proportions of patients with metastatic CRC at diagnosis and CRC location were similar between groups. The study did not have information on their care outside of VHA setting.

This study could not ascertain whether disparities existed in CRC treatment modality since rural residence may result in referral to community-based CRC care, which did not appear in the data. To address these limitations, we used death from any cause as the primary outcome, since death is a hard outcome and is not subject to ascertainment bias. The relatively short follow-up time is another limitation, though subgroup analysis by follow-up did not show significant differences. Despite PS matching, residual unmeasured confounding may exist between urban and rural groups. The predominantly White, male VHA population with high CCI may limit the generalizability of the results.

Conclusions

Rural VHA enrollees had similar survival rates after CRC diagnosis compared to their urban counterparts in a PS-matched analysis. The VHA models of care—including mailed CRC screening tools, several socioeconomic determinants of health (housing, food security, and transportation), and promoting equal access to health care, as well as cultural competency among HCPs—HCPs—may help alleviate disparities across the rural-urban spectrum. The VHA should continue efforts to enroll veterans and provide comprehensive coordinated care in community partnerships.

Colorectal cancer (CRC) is the second-leading cause of cancer-related deaths in the United States, with an estimated 52,550 deaths in 2023.1 However, the disease burden varies among different segments of the population.2 While both CRC incidence and mortality have been decreasing due to screening and advances in treatment, there are disparities in incidence and mortality across the sociodemographic spectrum including race, ethnicity, education, and income.1-4 While CRC incidence is decreasing for older adults, it is increasing among those aged < 55 years.5 The incidence of CRC in adults aged 40 to 54 years has increased by 0.5% to 1.3% annually since the mid-1990s.6 The US Preventive Services Task Force now recommends starting CRC screening at age 45 years for asymptomatic adults with average risk.7

Disparities also exist across geographical boundaries and living environment. Rural Americans faces additional challenges in health and lifestyle that can affect CRC outcomes. Compared to their urban counterparts, rural residents are more likely to be older, have lower levels of education, higher levels of poverty, lack health insurance, and less access to health care practitioners (HCPs).8-10 Geographic proximity, defined as travel time or physical distance to a health facility, has been recognized as a predictor of inferior outcomes.11 These aspects of rural living may pose challenges for accessing care for CRC screening and treatment.11-13 National and local studies have shown disparities in CRC screening rates, incidence, and mortality between rural and urban populations.14-16

It is unclear whether rural/urban disparities persist under the Veterans Health Administration (VHA) health care delivery model. This study examined differences in baseline characteristics and mortality between rural and urban veterans newly diagnosed with CRC. We also focused on a subpopulation aged ≤ 45 years.

Methods

This study extracted national data from the US Department of Veterans Affairs (VA) Corporate Data Warehouse (CDW) hosted in the VA Informatics and Computing Infrastructure (VINCI) environment. VINCI is an initiative to improve access to VA data and facilitate the analysis of these data while ensuring veterans’ privacy and data security.17 CDW is the VHA business intelligence information repository, which extracts data from clinical and nonclinical sources following prescribed and validated protocols. Data extracted included demographics, diagnosis, and procedure codes for both inpatient and outpatient encounters, vital signs, and vital status. This study used data previously extracted from a national cohort of veterans that encompassed all patients who received a group of commonly prescribed medications, such as statins, proton pump inhibitors, histamine-2 blockers, acetaminophen-containing products, and hydrocortisone-containing skin applications. This cohort encompassed 8,648,754 veterans, from whom 2,460,727 had encounters during fiscal years (FY) 2016 to 2021 (study period). The cohort was used to ensure that subjects were VHA patients, allowing them to adequately capture their clinical profiles.

Patients were identified as rural or urban based on their residence address at the date of their first diagnosis of CRC. The Geospatial Service Support Center (GSSC) aggregates and updates veterans’ residence address records for all enrolled veterans from the National Change of Address database. The data contain 1 record per enrollee. GSSC Geocoded Enrollee File contains enrollee addresses and their rurality indicators, categorized as urban, rural, or highly rural.18 Rurality is defined by the Rural Urban Commuting Area (RUCA) categories developed by the Department of Agriculture and the Health Resources and Services Administration of the US Department of Health and Human Services.19 Urban areas had RUCA codes of 1.0 to 1.1, and highly rural areas had RUCA scores of 10.0. All other areas were classified as rural. Since the proportion of veterans from highly rural areas was small, we included residents from highly rural areas in the rural residents’ group.

Inclusion and Exclusion Criteria

All veterans newly diagnosed with CRC from FY 2016 to 2021 were included. We used the ninth and tenth clinical modification revisions of the International Classification of Diseases (ICD-9-CM and ICD-10-CM) to define CRC diagnosis (Supplemental materials).4,20 To ensure that patients were newly diagnosed with CRC, this study excluded patients with a previous ICD-9-CM code for CRC diagnosis since FY 2003.

Comorbidities were identified using diagnosis and procedure codes from inpatient and outpatient encounters, which were used to calculate the Charlson Comorbidity Index (CCI) at the time of CRC diagnosis using the weighted method described by Schneeweiss et al.21 We defined CRC high-risk conditions and CRC screening tests, including flexible sigmoidoscopy and stool tests, as described in previous studies (Supplemental materials).20

The main outcome was total mortality. The date of death was extracted from the VHA Death Ascertainment File, which contains mortality data from the Master Person Index file in CDW and the Social Security Administration Death Master File. We used the date of death from any cause, as cause of death was not available.

A propensity score (PS) was created to match rural (including highly rural) and urban residents at a ratio of 1:1. Using a standard procedure described in prior publications, multivariable logistic regression used all baseline characteristics to estimate the PS and perform nearest-number matching without replacement.22,23 A caliper of 0.01 maximized the matched cohort size and achieved balance (Supplemental materials). We then examined the balance of baseline characteristics between PS-matched groups.

Analyses

Cox proportional hazards regression analysis estimated the hazard ratio (HR) of death in rural residents compared to urban residents in the PS-matched cohort. The outcome event was the date of death during the study’s follow-up period (defined as period from first CRC diagnosis to death or study end), with censoring at the study’s end date (September 30, 2021). The proportional hazards assumption was assessed by inspecting the Kaplan-Meier curves. Multiple analyses examined the HR of total mortality in the PS-matched cohort, stratified by sex, race, and ethnicity. We also examined the HR of total mortality stratified by duration of follow-up.

Another PS-matching analysis among veterans aged ≤ 45 years was performed using the same techniques described earlier in this article. We performed a Cox proportional hazards regression analysis to compare mortality in PS-matched urban and rural veterans aged ≤ 45 years. The HR of death in all veterans aged ≤ 45 years (before PS-matching) was estimated using Cox proportional hazard regression analysis, adjusting for PS.

Dichotomous variables were compared using X2 tests and continuous variables were compared using t tests. Baseline characteristics with missing values were converted into categorical variables and the proportion of subjects with missing values was equalized between treatment groups after PS-matching. For subgroup analysis, we examined the HR of total mortality in each subgroup using separate Cox proportional hazards regression models similar to the primary analysis but adjusted for PS. Due to multiple comparisons in the subgroup analysis, the findings should be considered exploratory. Statistical tests were 2-tailed, and significance was defined as P < .05. Data management and statistical analyses were conducted from June 2022 to January 2023 using STATA, Version 17. The VA Orlando Healthcare System Institutional Review Board approved the study and waived requirements for informed consent because only deidentified data were used.

Results

After excluding 49 patients (Supplemental materials, available at doi:10.12788/fp.0560), we identified 30,219 veterans with newly diagnosed CRC between FY 2016 to 2021 (Table 1). Of these, 19,422 (64.3%) resided in urban areas and 10,797 (35.7%) resided in rural areas (Table 2). The mean (SD) duration from the first CRC diagnosis to death or study end was 832 (640) days, and the median (IQR) was 723 (246–1330) days. Overall, incident CRC diagnoses were numerically highest in FY 2016 and lowest in FY 2020 (Figure 1). Patients with CRC in rural areas vs urban areas were significantly older (mean, 71.2 years vs 70.8 years, respectively; P < .001), more likely to be male (96.7% vs 95.7%, respectively; P < .001), more likely to be White (83.6% vs 67.8%, respectively; P < .001) and more likely to be non-Hispanic (92.2% vs 87.5%, respectively; P < .001). In terms of general health, rural veterans with CRC were more likely to be overweight or obese (81.5% rural vs 78.5% urban; P < .001) but had fewer mean comorbidities as measured by CCI (5.66 rural vs 5.90 urban; P < .001). A higher proportion of rural veterans with CRC had received stool-based (fecal occult blood test or fecal immunochemical test) CRC screening tests (61.6% rural vs 57.2% urban; P < .001). Fewer rural patients presented with systemic symptoms or signs within 1 year of CRC diagnosis (54.4% rural vs 57.5% urban, P < .001). Among urban patients with CRC, 6959 (35.8%) deaths were observed, compared with 3766 (34.9%) among rural patients (P = .10).

0525FED-AVAHO-CRC_T10525FED-AVAHO-CRC_T20525FED-AVAHO-CRC_F1

There were 21,568 PS-matched veterans: 10,784 in each group. In the PS-matched cohort, baseline characteristics were similar between veterans in urban and rural communities, including age, sex, race/ethnicity, body mass index, and comorbidities. Among rural patients with CRC, 3763 deaths (34.9%) were observed compared with 3702 (34.3%) among urban veterans. There was no significant difference in the HR of mortality between rural and urban CRC residents (HR, 1.01; 95% CI, 0.97-1.06; P = .53) (Figure 2).

0525FED-AVAHO-CRC_F20525FED-AVAHO-CRC_F30525FED-AVAHO-CRC_F4

Among veterans aged ≤ 45 years, 551 were diagnosed with CRC (391 urban and 160 rural). We PS-matched 142 pairs of urban and rural veterans without residual differences in baseline characteristics (eAppendix 1). There was no significant difference in the HR of mortality between rural and urban veterans aged ≤ 45 years (HR, 0.97; 95% CI, 0.57-1.63; P = .90) (Figure 2). Similarly, no difference in mortality was observed adjusting for PS between all rural and urban veterans aged ≤ 45 years (HR, 1.03; 95% CI, 0.67-1.59; P = .88).

0525FED-AVAHO-CRC_eApp1

There was no difference in total mortality between rural and urban veterans in any subgroup except for American Indian or Alaska Native veterans (HR, 2.41; 95% CI, 1.29-4.50; P = .006) (eAppendix 2).

0525FED-AVAHO-CRC_eApp2

Discussion

This study examined characteristics of patients with CRC between urban and rural areas among veterans who were VHA patients. Similar to other studies, rural veterans with CRC were older, more likely to be White, and were obese, but exhibited fewer comorbidities (lower CCI and lower incidence of congestive heart failure, dementia, hemiplegia, kidney diseases, liver diseases and AIDS, but higher incidence of chronic obstructive lung disease).8,16 The incidence of CRC in this study population was lowest in FY 2020, which was reported by the Centers for Disease Control and Prevention and is attributed to COVID-19 pandemic disruption of health services.24 The overall mortality in this study was similar to rates reported in other studies from the VA Central Cancer Registry.4 In the PS-matched cohort, where baseline characteristics were similar between urban and rural patients with CRC, we found no disparities in CRC-specific mortality between veterans in rural and urban areas. Additionally, when analysis was restricted to veterans aged ≤ 45 years, the results remained consistent.

Subgroup analyses showed no significant difference in mortality between rural and urban areas by sex, race or ethnicity, except rural American Indian or Alaska Native veterans who had double the mortality of their urban counterparts (HR, 2.41; 95% CI, 1.29-4.50; P = .006). This finding is difficult to interpret due to the small number of events and the wide CI. While with a Bonferroni correction the adjusted P value was .08, which is not statistically significant, a previous study found that although CRC incidence was lower overall in American Indian or Alaska Native populations compared to non-Hispanic White populations, CRC incidence was higher among American Indian or Alaska Native individuals in some areas such as Alaska and the Northern Plains.25,26 Studies have noted that rural American Indian/Alaska Native populations experience greater poverty, less access to broadband internet, and limited access to care, contributing to poorer cancer outcomes and lower survival.27 Thus, the finding of disparity in mortality between rural and urban American Indian or Alaska Native veterans warrants further study.

Other studies have raised concerns that CRC disproportionately affects adults in rural areas with higher mortality rates.14-16 These disparities arise from sociodemographic factors and modifiable risk factors, including physical activity, dietary patterns, access to cancer screening, and gaps in quality treatment resources.16,28 These factors operate at multiple levels: from individual, local health system, to community and policy.2,27 For example, a South Carolina study (1996–2016) found that residents in rural areas were more likely to be diagnosed with advanced CRC, possibly indicating lower rates of CRC screening in rural areas. They also had higher likelihood of death from CRC.15 However, the study did not include any clinical parameters, such as comorbidities or obesity. A statewide, population-based study in Utah showed that rural men experienced a lower CRC survival in their unadjusted analysis.16 However, the study was small, with only 3948 urban and 712 rural residents. Additionally, there was no difference in total mortality in the whole cohort (HR, 0.96; 95% CI, 0.86-1.07) or in CRC-specific death (HR, 0.93; 95% CI, 0.81-1.08). A nationwide study also showed that CRC mortality rates were 8% higher in nonmetropolitan or rural areas than in the most urbanized areas containing large metropolitan counties.29 However, this study did not include descriptions of clinical confounders, such as comorbidities, making it difficult to ascertain whether the difference in CRC mortality was due to rurality or differences in baseline risk characteristics.

In this study, the lack of CRC-specific mortality disparities may be attributed to the structures and practices of VHA health care. Recent studies have noted that mortality of several chronic medical conditions treated at the VHA was lower than at non-VHA hospitals.30,31 One study that measured the quality of nonmetastatic CRC care based on National Comprehensive Cancer Network guidelines showed that > 72% of VHA patients received guideline-concordant care for each diagnostic and therapeutic measure, except for follow-up colonoscopy timing, which appear to be similar or superior to that of the private sector.30,32,33 Some of the VA initiative for CRC screening may bypass the urban-rurality divide such as the mailed fecal immunochemical test program for CRC. This program was implemented at the onset of the COVID-19 pandemic to avoid disruptions of medical care.34 Rural patients are more likely to undergo fecal immunochemical testing when compared to urban patients in this data. Beyond clinical care, the VHA uses processes to tackle social determinants of health such as housing, food security, and transportation, promoting equal access to health care, and promoting cultural competency among HCPs.35-37

The results suggest that solutions to CRC disparities between rural and urban areas need to consider known barriers to rural health care, including transportation, diminished rural health care workforce, and other social determinants of health.9,10,27,38 VHA makes considerable efforts to provide equitable care to all enrolled veterans, including specific programs for rural veterans, including ongoing outreach.39 This study demonstrated lack of disparity in CRC-specific mortality in veterans receiving VHA care, highlighting the importance of these efforts.

Strengths and Limitations

This study used the VHA cohort to compare patient characteristics and mortality between patients with CRC residing in rural and urban areas. The study provides nationwide perspectives on CRC across the geographical spectrum and used a longitudinal cohort with prolonged follow-up to account for comorbidities.

However, the study compared a cohort of rural and urban veterans enrolled in the VHA; hence, the results may not reflect CRC outcomes in veterans without access to VHA care. Rurality has been independently associated with decreased likelihood of meeting CRC screening guidelines among veterans and military service members.38 This study lacked sufficient information to compare CRC staging or treatment modalities among veterans. Although the data cannot identify CRC stage, the proportions of patients with metastatic CRC at diagnosis and CRC location were similar between groups. The study did not have information on their care outside of VHA setting.

This study could not ascertain whether disparities existed in CRC treatment modality since rural residence may result in referral to community-based CRC care, which did not appear in the data. To address these limitations, we used death from any cause as the primary outcome, since death is a hard outcome and is not subject to ascertainment bias. The relatively short follow-up time is another limitation, though subgroup analysis by follow-up did not show significant differences. Despite PS matching, residual unmeasured confounding may exist between urban and rural groups. The predominantly White, male VHA population with high CCI may limit the generalizability of the results.

Conclusions

Rural VHA enrollees had similar survival rates after CRC diagnosis compared to their urban counterparts in a PS-matched analysis. The VHA models of care—including mailed CRC screening tools, several socioeconomic determinants of health (housing, food security, and transportation), and promoting equal access to health care, as well as cultural competency among HCPs—HCPs—may help alleviate disparities across the rural-urban spectrum. The VHA should continue efforts to enroll veterans and provide comprehensive coordinated care in community partnerships.

References
  1. Siegel RL, Wagle NS, Cercek A, Smith RA, Jemal A. Colorectal cancer statistics, 2023. CA Cancer J Clin. 2023;73(3):233-254. doi:10.3322/caac.21772
  2. Carethers JM, Doubeni CA. Causes of socioeconomic disparities in colorectal cancer and intervention framework and strategies. Gastroenterology. 2020;158(2):354-367. doi:10.1053/j.gastro.2019.10.029
  3. Murphy G, Devesa SS, Cross AJ, Inskip PD, McGlynn KA, Cook MB. Sex disparities in colorectal cancer incidence by anatomic subsite, race and age. Int J Cancer. 2011;128(7):1668-75. doi:10.1002/ijc.25481
  4. Zullig LL, Smith VA, Jackson GL, et al. Colorectal cancer statistics from the Veterans Affairs central cancer registry. Clin Colorectal Cancer. 2016;15(4):e199-e204. doi:10.1016/j.clcc.2016.04.005
  5. Lin JS, Perdue LA, Henrikson NB, Bean SI, Blasi PR. Screening for Colorectal Cancer: An Evidence Update for the US Preventive Services Task Force. 2021. U.S. Preventive Services Task Force Evidence Syntheses, formerly Systematic Evidence Reviews:Chapter 1. Agency for Healthcare Research and Quality (US); 2021. Accessed February 18, 2025. https://www.ncbi.nlm.nih.gov/books/NBK570917/
  6. Siegel RL, Fedewa SA, Anderson WF, et al. Colorectal cancer incidence patterns in the United States, 1974-2013. J Natl Cancer Inst. 2017;109(8). doi:10.1093/jnci/djw322
  7. Davidson KW, Barry MJ, Mangione CM, et al. Screening for colorectal cancer: US Preventive Services Task Force recommendation statement. JAMA. 2021;325(19):1965-1977. doi:10.1001/jama.2021.6238
  8. Hines R, Markossian T, Johnson A, Dong F, Bayakly R. Geographic residency status and census tract socioeconomic status as determinants of colorectal cancer outcomes. Am J Public Health. 2014;104(3):e63-e71. doi:10.2105/AJPH.2013.301572
  9. Cauwels J. The many barriers to high-quality rural health care. 2022;(9):1-32. NEJM Catal Innov Care Deliv. Accessed April 24, 2025. https://catalyst.nejm.org/doi/pdf/10.1056/CAT.22.0254
  10. Gong G, Phillips SG, Hudson C, Curti D, Philips BU. Higher US rural mortality rates linked to socioeconomic status, physician shortages, and lack of health insurance. Health Aff (Millwood);38(12):2003-2010. doi:10.1377/hlthaff.2019.00722
  11. Aboagye JK, Kaiser HE, Hayanga AJ. Rural-urban differences in access to specialist providers of colorectal cancer care in the United States: a physician workforce issue. JAMA Surg. 2014;149(6):537-543. doi:10.1001/jamasurg.2013.5062
  12. Lyckholm LJ, Hackney MH, Smith TJ. Ethics of rural health care. Crit Rev Oncol Hematol. 2001;40(2):131-138. doi:10.1016/s1040-8428(01)00139-1
  13. Krieger N, Williams DR, Moss NE. Measuring social class in US public health research: concepts, methodologies, and guidelines. Annu Rev Public Health. 1997;18:341-378. doi:10.1146/annurev.publhealth.18.1.341
  14. Singh GK, Jemal A. Socioeconomic and racial/ethnic disparities in cancer mortality, incidence, and survival in the United States, 1950-2014: over six decades of changing patterns and widening inequalities. J Environ Public Health. 2017;2017:2819372. doi:10.1155/2017/2819372
  15. Adams SA, Zahnd WE, Ranganathan R, et al. Rural and racial disparities in colorectal cancer incidence and mortality in South Carolina, 1996 - 2016. J Rural Health. 2022;38(1):34-39. doi:10.1111/jrh.12580
  16. Rogers CR, Blackburn BE, Huntington M, et al. Rural- urban disparities in colorectal cancer survival and risk among men in Utah: a statewide population-based study. Cancer Causes Control. 2020;31(3):241-253. doi:10.1007/s10552-020-01268-2
  17. US Department of Veterans Affairs. VA Informatics and Computing Infrastructure (VINCI), VA HSR RES 13-457. https://vincicentral.vinci.med.va.gov [Source not verified]
  18. US Department of Veterans Affairs Information Resource Center. VIReC Research User Guide: PSSG Geocoded Enrollee Files, 2015 Edition. US Department of Veterans Affairs, Health Services Research & Development Service, Information Resource Center; May. 2016. [source not verified]
  19. Goldsmith HF, Puskin DS, Stiles DJ. Improving the operational definition of “rural areas” for federal programs. US Department of Health and Human Services; 1993. Accessed February 27, 2025. https://www.ruralhealthinfo.org/pdf/improving-the-operational-definition-of-rural-areas.pdf
  20. Adams MA, Kerr EA, Dominitz JA, et al. Development and validation of a new ICD-10-based screening colonoscopy overuse measure in a large integrated healthcare system: a retrospective observational study. BMJ Qual Saf. 2023;32(7):414-424. doi:10.1136/bmjqs-2021-014236
  21. Schneeweiss S, Wang PS, Avorn J, Glynn RJ. Improved comorbidity adjustment for predicting mortality in Medicare populations. Health Serv Res. 2003;38(4):1103-1120. doi:10.1111/1475-6773.00165
  22. Becker S, Ichino A. Estimation of average treatment effects based on propensity scores. The Stata Journal. 2002;2(4):358-377.
  23. Leuven E, Sianesi B. PSMATCH2: Stata module to perform full Mahalanobis and propensity score matching, common support graphing, and covariate imbalance testing. Statistical software components. Revised February 1, 2018. Accessed February 27, 2025. https://ideas.repec.org/c/boc/bocode/s432001.html.
  24. US Cancer Statistics Working Group. US cancer statistics data visualizations tool. Centers for Disease Control and Prevention. June 2024. Accessed February 27, 2025. https://www.cdc.gov/cancer/dataviz
  25. Cao J, Zhang S. Multiple Comparison Procedures. JAMA. 2014;312(5):543-544. doi:10.1001/jama.2014.9440
  26. Gopalani SV, Janitz AE, Martinez SA, et al. Trends in cancer incidence among American Indians and Alaska Natives and Non-Hispanic Whites in the United States, 1999-2015. Epidemiology. 2020;31(2):205-213. doi:10.1097/EDE.0000000000001140
  27. Zahnd WE, Murphy C, Knoll M, et al. The intersection of rural residence and minority race/ethnicity in cancer disparities in the United States. Int J Environ Res Public Health. 2021;18(4). doi:10.3390/ijerph18041384
  28. Blake KD, Moss JL, Gaysynsky A, Srinivasan S, Croyle RT. Making the case for investment in rural cancer control: an analysis of rural cancer incidence, mortality, and funding trends. Cancer Epidemiol Biomarkers Prev. 2017;26(7):992-997. doi:10.1158/1055-9965.EPI-17-0092
  29. Singh GK, Williams SD, Siahpush M, Mulhollen A. Socioeconomic, rural-urban, and racial inequalities in US cancer mortality: part i-all cancers and lung cancer and part iicolorectal, prostate, breast, and cervical cancers. J Cancer Epidemiol. 2011;2011:107497. doi:10.1155/2011/107497
  30. Jackson GL, Melton LD, Abbott DH, et al. Quality of nonmetastatic colorectal cancer care in the Department of Veterans Affairs. J Clin Oncol. 2010;28(19):3176-3181. doi:10.1200/JCO.2009.26.7948
  31. Yoon J, Phibbs CS, Ong MK, et al. Outcomes of veterans treated in Veterans Affairs hospitals vs non-Veterans Affairs hospitals. JAMA Netw Open. 2023;6(12):e2345898. doi:10.1001/jamanetworkopen.2023.45898
  32. Malin JL, Schneider EC, Epstein AM, Adams J, Emanuel EJ, Kahn KL. Results of the National Initiative for Cancer Care Quality: how can we improve the quality of cancer care in the United States? J Clin Oncol. 2006;24(4):626-634. doi:10.1200/JCO.2005.03.3365
  33. Levin B, Lieberman DA, McFarland B, et al. Screening and surveillance for the early detection of colorectal cancer and adenomatous polyps, 2008: a joint guideline from the American Cancer Society, the US Multi-Society Task Force on Colorectal Cancer, and the American College of Radiology. Gastroenterology. 2008;134(5):1570-1595. doi:10.1053/j.gastro.2008.02.002
  34. Deeds SA, Moore CB, Gunnink EJ, et al. Implementation of a mailed faecal immunochemical test programme for colorectal cancer screening among Veterans. BMJ Open Qual. 2022;11(4). doi:10.1136/bmjoq-2022-001927
  35. Yehia BR, Greenstone CL, Hosenfeld CB, Matthews KL, Zephyrin LC. The role of VA community care in addressing health and health care disparities. Med Care. 2017;55(Suppl 9 suppl 2):S4-S5. doi:10.1097/MLR.0000000000000768
  36. Wright BN, MacDermid Wadsworth S, Wellnitz A, Eicher- Miller HA. Reaching rural veterans: a new mechanism to connect rural, low-income US Veterans with resources and improve food security. J Public Health (Oxf). 2019;41(4):714-723. doi:10.1093/pubmed/fdy203
  37. Nelson RE, Byrne TH, Suo Y, et al. Association of temporary financial assistance with housing stability among US veterans in the supportive services for veteran families program. JAMA Netw Open. 2021;4(2):e2037047. doi:10.1001/jamanetworkopen.2020.37047
  38. McDaniel JT, Albright D, Lee HY, et al. Rural–urban disparities in colorectal cancer screening among military service members and Veterans. J Mil Veteran Fam Health. 2019;5(1):40-48. doi:10.3138/jmvfh.2018-0013
  39. US Department of Veterans Affairs, Office of Rural Health. The rural veteran outreach toolkit. Updated February 12, 2025. Accessed February 18, 2025. https://www.ruralhealth.va.gov/partners/toolkit.asp
References
  1. Siegel RL, Wagle NS, Cercek A, Smith RA, Jemal A. Colorectal cancer statistics, 2023. CA Cancer J Clin. 2023;73(3):233-254. doi:10.3322/caac.21772
  2. Carethers JM, Doubeni CA. Causes of socioeconomic disparities in colorectal cancer and intervention framework and strategies. Gastroenterology. 2020;158(2):354-367. doi:10.1053/j.gastro.2019.10.029
  3. Murphy G, Devesa SS, Cross AJ, Inskip PD, McGlynn KA, Cook MB. Sex disparities in colorectal cancer incidence by anatomic subsite, race and age. Int J Cancer. 2011;128(7):1668-75. doi:10.1002/ijc.25481
  4. Zullig LL, Smith VA, Jackson GL, et al. Colorectal cancer statistics from the Veterans Affairs central cancer registry. Clin Colorectal Cancer. 2016;15(4):e199-e204. doi:10.1016/j.clcc.2016.04.005
  5. Lin JS, Perdue LA, Henrikson NB, Bean SI, Blasi PR. Screening for Colorectal Cancer: An Evidence Update for the US Preventive Services Task Force. 2021. U.S. Preventive Services Task Force Evidence Syntheses, formerly Systematic Evidence Reviews:Chapter 1. Agency for Healthcare Research and Quality (US); 2021. Accessed February 18, 2025. https://www.ncbi.nlm.nih.gov/books/NBK570917/
  6. Siegel RL, Fedewa SA, Anderson WF, et al. Colorectal cancer incidence patterns in the United States, 1974-2013. J Natl Cancer Inst. 2017;109(8). doi:10.1093/jnci/djw322
  7. Davidson KW, Barry MJ, Mangione CM, et al. Screening for colorectal cancer: US Preventive Services Task Force recommendation statement. JAMA. 2021;325(19):1965-1977. doi:10.1001/jama.2021.6238
  8. Hines R, Markossian T, Johnson A, Dong F, Bayakly R. Geographic residency status and census tract socioeconomic status as determinants of colorectal cancer outcomes. Am J Public Health. 2014;104(3):e63-e71. doi:10.2105/AJPH.2013.301572
  9. Cauwels J. The many barriers to high-quality rural health care. 2022;(9):1-32. NEJM Catal Innov Care Deliv. Accessed April 24, 2025. https://catalyst.nejm.org/doi/pdf/10.1056/CAT.22.0254
  10. Gong G, Phillips SG, Hudson C, Curti D, Philips BU. Higher US rural mortality rates linked to socioeconomic status, physician shortages, and lack of health insurance. Health Aff (Millwood);38(12):2003-2010. doi:10.1377/hlthaff.2019.00722
  11. Aboagye JK, Kaiser HE, Hayanga AJ. Rural-urban differences in access to specialist providers of colorectal cancer care in the United States: a physician workforce issue. JAMA Surg. 2014;149(6):537-543. doi:10.1001/jamasurg.2013.5062
  12. Lyckholm LJ, Hackney MH, Smith TJ. Ethics of rural health care. Crit Rev Oncol Hematol. 2001;40(2):131-138. doi:10.1016/s1040-8428(01)00139-1
  13. Krieger N, Williams DR, Moss NE. Measuring social class in US public health research: concepts, methodologies, and guidelines. Annu Rev Public Health. 1997;18:341-378. doi:10.1146/annurev.publhealth.18.1.341
  14. Singh GK, Jemal A. Socioeconomic and racial/ethnic disparities in cancer mortality, incidence, and survival in the United States, 1950-2014: over six decades of changing patterns and widening inequalities. J Environ Public Health. 2017;2017:2819372. doi:10.1155/2017/2819372
  15. Adams SA, Zahnd WE, Ranganathan R, et al. Rural and racial disparities in colorectal cancer incidence and mortality in South Carolina, 1996 - 2016. J Rural Health. 2022;38(1):34-39. doi:10.1111/jrh.12580
  16. Rogers CR, Blackburn BE, Huntington M, et al. Rural- urban disparities in colorectal cancer survival and risk among men in Utah: a statewide population-based study. Cancer Causes Control. 2020;31(3):241-253. doi:10.1007/s10552-020-01268-2
  17. US Department of Veterans Affairs. VA Informatics and Computing Infrastructure (VINCI), VA HSR RES 13-457. https://vincicentral.vinci.med.va.gov [Source not verified]
  18. US Department of Veterans Affairs Information Resource Center. VIReC Research User Guide: PSSG Geocoded Enrollee Files, 2015 Edition. US Department of Veterans Affairs, Health Services Research & Development Service, Information Resource Center; May. 2016. [source not verified]
  19. Goldsmith HF, Puskin DS, Stiles DJ. Improving the operational definition of “rural areas” for federal programs. US Department of Health and Human Services; 1993. Accessed February 27, 2025. https://www.ruralhealthinfo.org/pdf/improving-the-operational-definition-of-rural-areas.pdf
  20. Adams MA, Kerr EA, Dominitz JA, et al. Development and validation of a new ICD-10-based screening colonoscopy overuse measure in a large integrated healthcare system: a retrospective observational study. BMJ Qual Saf. 2023;32(7):414-424. doi:10.1136/bmjqs-2021-014236
  21. Schneeweiss S, Wang PS, Avorn J, Glynn RJ. Improved comorbidity adjustment for predicting mortality in Medicare populations. Health Serv Res. 2003;38(4):1103-1120. doi:10.1111/1475-6773.00165
  22. Becker S, Ichino A. Estimation of average treatment effects based on propensity scores. The Stata Journal. 2002;2(4):358-377.
  23. Leuven E, Sianesi B. PSMATCH2: Stata module to perform full Mahalanobis and propensity score matching, common support graphing, and covariate imbalance testing. Statistical software components. Revised February 1, 2018. Accessed February 27, 2025. https://ideas.repec.org/c/boc/bocode/s432001.html.
  24. US Cancer Statistics Working Group. US cancer statistics data visualizations tool. Centers for Disease Control and Prevention. June 2024. Accessed February 27, 2025. https://www.cdc.gov/cancer/dataviz
  25. Cao J, Zhang S. Multiple Comparison Procedures. JAMA. 2014;312(5):543-544. doi:10.1001/jama.2014.9440
  26. Gopalani SV, Janitz AE, Martinez SA, et al. Trends in cancer incidence among American Indians and Alaska Natives and Non-Hispanic Whites in the United States, 1999-2015. Epidemiology. 2020;31(2):205-213. doi:10.1097/EDE.0000000000001140
  27. Zahnd WE, Murphy C, Knoll M, et al. The intersection of rural residence and minority race/ethnicity in cancer disparities in the United States. Int J Environ Res Public Health. 2021;18(4). doi:10.3390/ijerph18041384
  28. Blake KD, Moss JL, Gaysynsky A, Srinivasan S, Croyle RT. Making the case for investment in rural cancer control: an analysis of rural cancer incidence, mortality, and funding trends. Cancer Epidemiol Biomarkers Prev. 2017;26(7):992-997. doi:10.1158/1055-9965.EPI-17-0092
  29. Singh GK, Williams SD, Siahpush M, Mulhollen A. Socioeconomic, rural-urban, and racial inequalities in US cancer mortality: part i-all cancers and lung cancer and part iicolorectal, prostate, breast, and cervical cancers. J Cancer Epidemiol. 2011;2011:107497. doi:10.1155/2011/107497
  30. Jackson GL, Melton LD, Abbott DH, et al. Quality of nonmetastatic colorectal cancer care in the Department of Veterans Affairs. J Clin Oncol. 2010;28(19):3176-3181. doi:10.1200/JCO.2009.26.7948
  31. Yoon J, Phibbs CS, Ong MK, et al. Outcomes of veterans treated in Veterans Affairs hospitals vs non-Veterans Affairs hospitals. JAMA Netw Open. 2023;6(12):e2345898. doi:10.1001/jamanetworkopen.2023.45898
  32. Malin JL, Schneider EC, Epstein AM, Adams J, Emanuel EJ, Kahn KL. Results of the National Initiative for Cancer Care Quality: how can we improve the quality of cancer care in the United States? J Clin Oncol. 2006;24(4):626-634. doi:10.1200/JCO.2005.03.3365
  33. Levin B, Lieberman DA, McFarland B, et al. Screening and surveillance for the early detection of colorectal cancer and adenomatous polyps, 2008: a joint guideline from the American Cancer Society, the US Multi-Society Task Force on Colorectal Cancer, and the American College of Radiology. Gastroenterology. 2008;134(5):1570-1595. doi:10.1053/j.gastro.2008.02.002
  34. Deeds SA, Moore CB, Gunnink EJ, et al. Implementation of a mailed faecal immunochemical test programme for colorectal cancer screening among Veterans. BMJ Open Qual. 2022;11(4). doi:10.1136/bmjoq-2022-001927
  35. Yehia BR, Greenstone CL, Hosenfeld CB, Matthews KL, Zephyrin LC. The role of VA community care in addressing health and health care disparities. Med Care. 2017;55(Suppl 9 suppl 2):S4-S5. doi:10.1097/MLR.0000000000000768
  36. Wright BN, MacDermid Wadsworth S, Wellnitz A, Eicher- Miller HA. Reaching rural veterans: a new mechanism to connect rural, low-income US Veterans with resources and improve food security. J Public Health (Oxf). 2019;41(4):714-723. doi:10.1093/pubmed/fdy203
  37. Nelson RE, Byrne TH, Suo Y, et al. Association of temporary financial assistance with housing stability among US veterans in the supportive services for veteran families program. JAMA Netw Open. 2021;4(2):e2037047. doi:10.1001/jamanetworkopen.2020.37047
  38. McDaniel JT, Albright D, Lee HY, et al. Rural–urban disparities in colorectal cancer screening among military service members and Veterans. J Mil Veteran Fam Health. 2019;5(1):40-48. doi:10.3138/jmvfh.2018-0013
  39. US Department of Veterans Affairs, Office of Rural Health. The rural veteran outreach toolkit. Updated February 12, 2025. Accessed February 18, 2025. https://www.ruralhealth.va.gov/partners/toolkit.asp
Issue
Federal Practitioner - 42(5)s
Issue
Federal Practitioner - 42(5)s
Page Number
S22-S31
Page Number
S22-S31
Publications
Publications
Topics
Article Type
Display Headline

Colorectal Cancer Characteristics and Mortality From Propensity Score-Matched Cohorts of Urban and Rural Veterans

Display Headline

Colorectal Cancer Characteristics and Mortality From Propensity Score-Matched Cohorts of Urban and Rural Veterans

Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Tue, 05/06/2025 - 15:51
Un-Gate On Date
Tue, 05/06/2025 - 15:51
Use ProPublica
CFC Schedule Remove Status
Tue, 05/06/2025 - 15:51
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
survey writer start date
Tue, 05/06/2025 - 15:51
Media Files

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

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

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

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

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

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

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

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

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

Methods

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

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

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

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

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

Statistical Analysis

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

Results

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

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

Abbreviation: CGM, continuous glucose monitor.

Discussion

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

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

Strengths and Limitations

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

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

Conclusions

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

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

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

Correspondence: Kelsey Floerchinger ([email protected])

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

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

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

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

Issue
Federal Practitioner - 41(suppl 5)
Publications
Topics
Page Number
S1-S5
Sections
Author and Disclosure Information

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

Correspondence: Kelsey Floerchinger ([email protected])

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

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

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

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

Author and Disclosure Information

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

Correspondence: Kelsey Floerchinger ([email protected])

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

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

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

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

Article PDF
Article PDF

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

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

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

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

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

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

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

Methods

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

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

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

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

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

Statistical Analysis

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

Results

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

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

Abbreviation: CGM, continuous glucose monitor.

Discussion

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

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

Strengths and Limitations

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

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

Conclusions

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

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

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

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

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

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

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

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

Methods

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

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

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

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

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

Statistical Analysis

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

Results

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

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

Abbreviation: CGM, continuous glucose monitor.

Discussion

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

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

Strengths and Limitations

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

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

Conclusions

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

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

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

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

Background

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

Methods

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

Results

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

Conclusions

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

Issue
Federal Practitioner - 41(suppl 4)
Publications
Topics
Page Number
S43
Sections

Background

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

Methods

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

Results

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

Conclusions

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

Background

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

Methods

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

Results

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

Conclusions

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

Issue
Federal Practitioner - 41(suppl 4)
Issue
Federal Practitioner - 41(suppl 4)
Page Number
S43
Page Number
S43
Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Eyebrow Default
Research
Gate On Date
Thu, 09/12/2024 - 14:00
Un-Gate On Date
Thu, 09/12/2024 - 14:00
Use ProPublica
CFC Schedule Remove Status
Thu, 09/12/2024 - 14:00
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
survey writer start date
Wed, 09/18/2024 - 20:58

Epidemiologic and Clinical Evaluation of the Bidirectional Link Between Molluscum Contagiosum and Atopic Dermatitis in Children

Article Type
Changed
Fri, 09/05/2025 - 12:04
Display Headline

Epidemiologic and Clinical Evaluation of the Bidirectional Link Between Molluscum Contagiosum and Atopic Dermatitis in Children

Molluscum contagiosum (MC), which is caused by a DNA virus in the Poxviridae family, is a common viral skin infection that primarily affects children.1-4 The reported incidence and prevalence of MC exhibit notable geographic variation. Worldwide, annual incidence rates per 1000 individuals range from 3.1 to 25, and prevalence ranges from 0.27% to 34.6%.2-7

Molluscum contagiosum is diagnosed clinically and typically manifests as smooth, flesh-colored papules measuring 2 to 6 mm in diameter with central umbilication. It can manifest as a single lesion or multiple clustered lesions, or in a disseminated pattern. The primary mode of transmission is through contact with skin, lesions, or contaminated personal items, or via self-inoculation. The majority of cases are asymptomatic, but in some patients, MC may be associated with pruritus, tenderness, erythema, or irritation. When present, secondary bacterial infections can cause localized inflammation and pain.1,3,4 The pathogenesis hinges on MC virus replication within keratinocytes, disrupting cellular differentiation and keratinization. The virus persists in the host by influencing the immune response through various mechanisms, including interference with signaling pathways, apoptosis inhibition, and antigen presentation disruption.3,4

Molluscum contagiosum typically follows a self-limiting trajectory, resolving over several months to 2 years.3,4 The resolution timeframe is intricately linked to variables such as the patient’s immune profile, lesion burden, and treatment approach. For symptomatic lesions, a variety of treatment options have been described, including physical ablation (eg, cryotherapy, curettage) and topical agents such as potassium hydroxide, cantharidin, imiquimod, and salicylic acid.3,4,8,9

Atopic dermatitis (AD) is a common chronic relapsing inflammatory skin disorder. In the United States, its prevalence ranges from 15% to 30% in children and from 2% to 10% in adults, with ongoing evidence of a growing global incidence.10-14 While AD can emerge at any age, typical onset is during early childhood. The clinical manifestation of AD includes a spectrum of eczematous features, often accompanied by persistent itching. The pathogenesis is multifactorial, involving a complex interplay of genetic, immunologic, and environmental factors. Key contributors to this multifaceted process encompass a compromised epidermal barrier, alterations in the skin microbiome, and an immune dysregulation promoting a type 2 immune response. Epidermal barrier dysfunction can be attributed to various factors, including diminished ceramide production, altered lipid composition, the release of inflammatory mediators, and mechanical damage from the persistent itch-scratch cycle.10-13,15 These factors or their interplay may enhance the susceptibility of patients with AD to infections. 

Several studies conducted across various geographic regions examining the relationship between MC and AD have reported variable findings.2,6,7,16-21 Published studies have reported a prevalence of AD in children with MC ranging from 13.2% to 43%.2,6,7,16-21 Although some studies suggest a higher rate of atopy in patients with MC, not all research has confirmed this association.16,21 Dohil et al2 reported a greater number of MC lesions in children with AD than those without an atopic background. Silverberg20 reported that in 10% (5/50) of children with MC, the onset of AD was triggered, and in 22% (11/50) MC was associated with flares of pre-existing AD.

In this study, we aimed to assess MC infection rates in children with AD, analyze the epidemiologic aspects and severity differences between atopic children with and without MC infection, and compare data from atopic and nonatopic children with MC.

Methods

In this retrospective cohort study, we analyzed the medical records of pediatric patients diagnosed with MC, AD, or both conditions at an outpatient dermatology practice in Netanya, HaSharon, Israel, from September 2013 to August 2022. Data were collected from the electronic medical records and included patient demographics, the clinical presentation of MC and/or AD at diagnosis, and the duration of both conditions. Only patients with complete data and at least 6 months of follow-up were included. Key epidemiologic characteristics assessed included patient sex, age at the initial visit, and age at the onset of MC and/or AD. Diagnoses of MC and AD were established through clinical examinations conducted by dermatologists. The clinical evaluation of AD encompassed the assessment of body surface area involvement (categorized as <5%, 5%-10%, or >10%). Atopic dermatitis severity was classified as mild, moderate, or severe using the validated Investigator Global Assessment Scale for Atopic Dermatitis.22 Clinical evaluation of MC included assessment of the number of lesions (categorized as 4, 5-9, or 10), presence of inflammatory lesions, and resolution times for individual lesions (categorized as <1 week, several weeks, or unknown), as well as the overall resolution time for all lesions (categorized as <6 months, 6-12 months, 13-18 months, or >18 months). The temporal relationship between the appearance of MC and AD also was assessed.

Statistical Analysis—Numbers and percentages were used for categorical variables. Continuous variables were represented by mean and standard deviation. Categorical variables were compared using the χ2 test, and continuous variables between groups were compared using the Student t test. All statistical tests were 2-sided, with statistical significance defined as P.05. Statistical analysis was performed using SPSS software version 28 (IBM).

Results

Study Population—A total of 610 children were included in the study; 263 (43%) were female and 347 (57%) were male. The patients ranged in age from 4 months to 10 years, with a mean (SD) age of 4.87 (1.82) years. Five hundred fifty-six (91%) patients had AD, and 336 (55%) had MC. Within this cohort, 274 (45%) children had AD only, 54 (9%) had MC only, and 282 (46%) had both AD and MC. Regarding the temporal sequence, among the 282 children who had both AD and MC, AD preceded MC in 203 (72%) cases, both conditions were diagnosed concomitantly in 43 (15%) cases, and MC preceded AD in 36 (13%) cases. For cases in which the MC diagnosis followed the diagnosis of AD, the mean (SD) time between each diagnosis was 3.17 (1.5) years.

Comparison of Atopic and Nonatopic Children With MC—Although a higher proportion of males were diagnosed with MC (with or without concurrent AD), the differences in sex distribution between the 2 groups did not reach statistical significance. Among all children with MC, the majority (81.5% [274/336]) were aged 1 to 6 years at presentation. Patients with MC as their sole diagnosis had a similar mean age compared with those with concurrent AD. However, a detailed age subgroup analysis revealed a notable distinction: in the group with MC as the sole diagnosis, the majority (95% [51/54]) were younger than 7 years. In contrast, in the combined MC and AD group, MC manifested across a wider age range, with 21% (58/282) of patients being older than 7 years. In MC cases associated with AD, a notably higher lesion count and increased local inflammatory response were observed compared to those without AD. The time for complete resolution of all MC lesions was substantially prolonged in patients with comorbid AD. Specifically, 93% (50/54) of patients with MC without comorbid AD achieved full resolution within 1 year, whereas 52% (146/282) of patients with comorbid AD required more than 1 year for resolution (eTable 1). 

CT116003099-eTable1

Comparison of Atopic Children With and Without MC—Sex, age distribution, and disease duration showed no differences between atopic patients with and without MC. Atopic patients with MC exhibited greater body surface area involvement and higher validated Investigator Global Assessment Scale for Atopic Dermatitis scores compared to atopic patients without MC (eTable 2).

CT116003099-eTable2

Comment

This study examined the relationship between MC and AD in pediatric patients, revealing a notable correlation and yielding valuable epidemiologic and clinical insights. Consistent with previous research, our study demonstrated a high prevalence of AD in children with MC.2,6,7,16-21 Previous studies indicated AD rates of 13% to 43% in pediatric patients with MC, whereas our study found a higher prevalence (84%), signifying a substantial majority of patients with MC in our cohort had AD. This discrepancy arises from factors such as demographic, genetic, and environmental differences, along with differences in access to medical care, referral practices, and diagnostic approaches across health care systems.14

Our temporal analysis of MC and AD diagnoses offers important insights. In the majority (72% [203/282]) of cases, the diagnosis of AD preceded MC, supporting previous research suggesting that the underlying pathophysiology of AD heightens susceptibility to MC.15,17-20 Less frequently, MC was diagnosed before or concurrently with AD, indicating that MC may occasionally trigger or exacerbate milder or undiagnosed AD, as previously proposed.20

A notable finding in our study was the expanded age range for MC onset in patients with AD, encompassing older age groups compared to patients with MC as their sole diagnosis, possibly due to persistent immune dysregulation. To the best of our knowledge, this specific observation has not been systematically reported or documented in prior cohort studies. Visible skin lesions of MC may have a psychological impact on patients, influencing self-consciousness and causing embarrassment and emotional distress. This may be more pronounced in older children, who are more aware of their appearance and social perceptions.23-25 These considerations should play a role in the management of MC. 

Our study revealed that children with AD and MC displayed higher lesion counts, increased local inflammatory responses, and a more protracted resolution period compared to nonatopic children. In more than 50% of children with AD, MC took more than 1 year for resolution, whereas the majority of those without AD achieved resolution within 1 year. These findings may be attributed to AD-related immune dysregulation, influencing the natural course of MC. Consequently, it suggests that while nonatopic children with MC usually are managed through observation, atopic patients may benefit from an intervention-oriented approach. 

Comparing atopic patients with and without MC showed a heightened occurrence of severe and extensive AD among those with concurrent MC. Several factors could contribute to this observation. On one hand, there could be a direct association between the extent and severity of AD, leading to an elevated susceptibility to MC. Conversely, MC might exacerbate immunologic dysregulation and intensify skin inflammation in atopic individuals.20 Additionally, itching related to both disorders may exacerbate inflammation and compromise the epidermal barrier, facilitating the spread of MC. This interplay suggests that each condition exacerbates the other in a self-reinforcing cycle. The importance of patient and caregiver education is underscored by recognizing these interactions. To manage both conditions effectively, health care providers should counsel patients and caregivers on maintaining proper skin care practices such as gentle cleansing with mild, fragrance-free products, regular moisturization, and avoidance of irritants, encourage them to avoid scratching, and recommend adopting an active treatment approach.

Our study had notable strengths. Firstly, a substantial sample size enhanced the statistical reliability of our findings. Additionally, valuable insights into the epidemiology and clinical aspects of AD and MC were obtained by utilizing real-world data from an outpatient dermatology practice. In our study, clinical evaluations covered body surface area involvement and disease severity for AD while also assessing lesion counts and the presence of inflammatory lesions for MC. This comprehensive approach facilitated a thorough analysis of both conditions. The extended data collection period not only allowed for observation of their clinical course and duration, but also enabled a detailed assessment of their interplay.

Our study also had several limitations. Primarily, its retrospective design relied on the accuracy and comprehensiveness of medical records, which may have introduced bias. The exclusion of some patients due to incomplete data further increased the potential for selection bias. Additionally, this study was conducted in a single outpatient dermatology practice in Israel, resulting in a study population composed predominantly of Jewish patients (94%), with a minority (6%) of Arab patients. Other ethnic groups, including Black, Asian, and Hispanic populations, were not represented. This reflects the country’s demographic composition rather than an intentional selection bias. However, the limited ethnic diversity reduces the generalizability of our findings. Differences in demographics, coding practices, health care utilization (eg, timeliness of seeking care, access to dermatology services), and treatment strategies also may impact the observed prevalence, clinical characteristics, and patient outcomes. Furthermore, while our study highlighted the potential advantages of a proactive treatment approach for atopic children with MC, it did not evaluate specific treatment protocols. Future research should aim to confirm the most efficacious therapeutic strategies for managing MC in atopic individuals and to include a more diverse population to better understand the applicability of findings across various ethnic groups.

Conclusion

Our study found a high prevalence of AD in children with MC and a strong bidirectional relationship between these conditions. Pediatric patients with AD display a broader age range for MC, greater lesion burden, increased local inflammatory responses, prolonged resolution times, and more extensive and severe AD.

Recognizing the interplay between MC and AD is crucial, highlighting the importance of health care providers educating patients and caregivers. Emphasizing skin hygiene, discouraging scratching, and implementing proactive treatment approaches can enhance the outcomes of both conditions. Further research into the underlying mechanisms of this association and effective therapeutic strategies for MC in atopic individuals is warranted.

Acknowledgments—The authors thank Zvi Segal, MD (Tel Hashomer, Israel) for his insightful contribution to the statistical analysis of the results. We would like to express our appreciation to the dedicated team of the dermatology practice in Netanya for the support throughout the performance of the study. Additionally, we thank all study participants and their parents for their participation and contribution to our research.

References
  1. Han H, Smythe C, Yousefian F, et al. Molluscum contagiosum virus evasion of immune surveillance: a review. J Drugs Dermatol. 2023;22182-189.
  2. Dohil MA, Lin P, Lee J, et al. The epidemiology of molluscum contagiosum in children. J Am Acad Dermatol. 2006;54:47-54.
  3. Silverberg NB. Pediatric molluscum: an update. Cutis. 2019;104:301-305;E1;E2.
  4. Forbat E, Al-Niaimi F, Ali FR. Molluscum contagiosum: review and update on management. Pediatr Dermatol. 2017;34:504-515.
  5. Olsen JR, Gallacher J, Piguet V, et al. Epidemiology of molluscum contagiosum in children: a systematic review. Fam Pract. 2014;31:130-136.
  6. Kakourou T, Zachariades A, Anastasiou T, et al. Molluscum contagiosum in Greek children: a case series. Int J Dermatol. 2005;44:221-223.
  7. Osio A, Deslandes E, Saada V, et al. Clinical characteristics of molluscum contagiosum in children in a private dermatology practice in the greater Paris area, France: a prospective study in 661 patients. Dermatology. 2011;222:314-320.
  8. Hebert AA, Bhatia N, Del Rosso JQ. Molluscum contagiosum: epidemiology, considerations, treatment options, and therapeutic gaps. J Clin Aesthet Dermatol. 2023;16(8 Suppl 1):S4-S11.
  9. Chao YC, Ko MJ, Tsai WC, et al. Comparative efficacy of treatments for molluscum contagiosum: a systematic review and network meta-analysis. J Dtsch Dermatol Ges. 2023;21:587-597.
  10. Garg N, Silverberg JI. Epidemiology of childhood atopic dermatitis. Clin Dermatol. 2015;33:281-288.
  11. Hale G, Davies E, Grindlay DJC, et al. What’s new in atopic eczema? an analysis of systematic reviews published in 2017. part 2: epidemiology, etiology, and risk factors. Clin Exp Dermatol. 2019;44:868-873.
  12. Tracy A, Bhatti S, Eichenfield LF. Update on pediatric atopic dermatitis. Cutis. 2020;106:143-146.
  13. Langan SM, Irvine AD, Weidinger S. Atopic dermatitis. Lancet. 2020;396:345-360.
  14. Silverberg JI. Public health burden and epidemiology of atopic dermatitis. Dermatol Clin. 2017;35:283-289.
  15. Manti S, Amorini M, Cuppari C, et al. Filaggrin mutations and molluscum contagiosum skin infection in patients with atopic dermatitis. Ann Allergy Asthma Immunol. 2017;119446-451.
  16. Seize M, Ianhez M, Cestari S. A study of the correlation between molluscum contagiosum and atopic dermatitis in children. An Bras Dermatol. 2011;86:663-668.
  17. Ren Z, Silverberg JI. Association of atopic dermatitis with bacterial, fungal, viral, and sexually transmitted skin infections. Dermatitis. 2020;31:157-164.
  18. Olsen JR, Piguet V, Gallacher J, et al. Molluscum contagiosum and associations with atopic eczema in children: a retrospective longitudinal study in primary care. Br J Gen Pract. 2016;66:E53-E58.
  19. Han JH, Yoon JW, Yook HJ, et al. Evaluation of atopic dermatitis and cutaneous infectious disorders using sequential pattern mining: a nationwide population-based cohort study. J Clin Med. 2022;11:3422.
  20. Silverberg NB. Molluscum contagiosum virus infection can trigger atopic dermatitis disease onset or flare. Cutis. 2018;102:191-194.
  21. Hayashida S, Furusho N, Uchi H, et al. Are lifetime prevalence of impetigo, molluscum and herpes infection really increased in children having atopic dermatitis? J Dermatol Sci. 2010;60:173-178.
  22. Simpson E, Bissonnette R, Eichenfield LF, et al. The Validated Investigator Global Assessment for Atopic Dermatitis (vIGA-AD): the development and reliability testing of a novel clinical outcome measurement instrument for the severity of atopic dermatitis. J Am Acad Dermatol. 2020;83:839-846.
  23. Olsen JR, Gallacher J, Finlay AY, et al. Time to resolution and effect on quality of life of molluscum contagiosum in children in the UK: a prospective community cohort study. Lancet Infect Dis. 2015;15:190-195.
  24. Ðurovic´ MR, Jankovic´ J, Spiric´ VT, et al. Does age influence the quality of life in children with atopic dermatitis? PLoS One. 2019;14:E0224618.
  25. Chernyshov PV. Stigmatization and self-perception in children with atopic dermatitis. Clin Cosmet Investig Dermatol. 2016;9:159-166.
Article PDF
Author and Disclosure Information

Drs. Anna Lyakhovitsky, Shemer, Galili, and Kassem are from the Department of Dermatology, Sheba Medical Center, Tel-HaShomer, Ramat-Gan, Israel. Drs. Anna Lyakhovitsky, Shemer, Galili and Kassem also are from and Dr. Magal is from the Gray School of Medical Sciences, Tel Aviv University, Israel. Drs. Hermush and Kaplan are from the Adelson School of Medicine, Ariel University, Israel. Dr. Hermush also is from the Laniado Medical Center, Natania, Israel. Dr. Daniel is from the Department of Dermatology, University of Mississippi Medical Center, Jackson, and the Department of Dermatology, University of Alabama at Birmingham. Dr. Keren Lyakhovitsky is from the Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel.

The authors have no relevant financial disclosures to report.

This study was conducted following the principles of the Declaration of Helsinki and was approved by the local ethical committee (0104-09-LND). The data supporting the findings are available from the corresponding author on request due to privacy/ethical restrictions.

Correspondence: Anna Lyakhovitsky, MD, Department of Dermatology, Sheba Medical Center, Tel-HaShomer, 52621 Ramat-Gan, Israel ([email protected]).

Cutis. 2025 September;116(3):99-102, E2-E3. doi:10.12788/cutis.1264

Issue
Cutis - 116(3)
Publications
Topics
Page Number
99-102
Sections
Author and Disclosure Information

Drs. Anna Lyakhovitsky, Shemer, Galili, and Kassem are from the Department of Dermatology, Sheba Medical Center, Tel-HaShomer, Ramat-Gan, Israel. Drs. Anna Lyakhovitsky, Shemer, Galili and Kassem also are from and Dr. Magal is from the Gray School of Medical Sciences, Tel Aviv University, Israel. Drs. Hermush and Kaplan are from the Adelson School of Medicine, Ariel University, Israel. Dr. Hermush also is from the Laniado Medical Center, Natania, Israel. Dr. Daniel is from the Department of Dermatology, University of Mississippi Medical Center, Jackson, and the Department of Dermatology, University of Alabama at Birmingham. Dr. Keren Lyakhovitsky is from the Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel.

The authors have no relevant financial disclosures to report.

This study was conducted following the principles of the Declaration of Helsinki and was approved by the local ethical committee (0104-09-LND). The data supporting the findings are available from the corresponding author on request due to privacy/ethical restrictions.

Correspondence: Anna Lyakhovitsky, MD, Department of Dermatology, Sheba Medical Center, Tel-HaShomer, 52621 Ramat-Gan, Israel ([email protected]).

Cutis. 2025 September;116(3):99-102, E2-E3. doi:10.12788/cutis.1264

Author and Disclosure Information

Drs. Anna Lyakhovitsky, Shemer, Galili, and Kassem are from the Department of Dermatology, Sheba Medical Center, Tel-HaShomer, Ramat-Gan, Israel. Drs. Anna Lyakhovitsky, Shemer, Galili and Kassem also are from and Dr. Magal is from the Gray School of Medical Sciences, Tel Aviv University, Israel. Drs. Hermush and Kaplan are from the Adelson School of Medicine, Ariel University, Israel. Dr. Hermush also is from the Laniado Medical Center, Natania, Israel. Dr. Daniel is from the Department of Dermatology, University of Mississippi Medical Center, Jackson, and the Department of Dermatology, University of Alabama at Birmingham. Dr. Keren Lyakhovitsky is from the Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel.

The authors have no relevant financial disclosures to report.

This study was conducted following the principles of the Declaration of Helsinki and was approved by the local ethical committee (0104-09-LND). The data supporting the findings are available from the corresponding author on request due to privacy/ethical restrictions.

Correspondence: Anna Lyakhovitsky, MD, Department of Dermatology, Sheba Medical Center, Tel-HaShomer, 52621 Ramat-Gan, Israel ([email protected]).

Cutis. 2025 September;116(3):99-102, E2-E3. doi:10.12788/cutis.1264

Article PDF
Article PDF

Molluscum contagiosum (MC), which is caused by a DNA virus in the Poxviridae family, is a common viral skin infection that primarily affects children.1-4 The reported incidence and prevalence of MC exhibit notable geographic variation. Worldwide, annual incidence rates per 1000 individuals range from 3.1 to 25, and prevalence ranges from 0.27% to 34.6%.2-7

Molluscum contagiosum is diagnosed clinically and typically manifests as smooth, flesh-colored papules measuring 2 to 6 mm in diameter with central umbilication. It can manifest as a single lesion or multiple clustered lesions, or in a disseminated pattern. The primary mode of transmission is through contact with skin, lesions, or contaminated personal items, or via self-inoculation. The majority of cases are asymptomatic, but in some patients, MC may be associated with pruritus, tenderness, erythema, or irritation. When present, secondary bacterial infections can cause localized inflammation and pain.1,3,4 The pathogenesis hinges on MC virus replication within keratinocytes, disrupting cellular differentiation and keratinization. The virus persists in the host by influencing the immune response through various mechanisms, including interference with signaling pathways, apoptosis inhibition, and antigen presentation disruption.3,4

Molluscum contagiosum typically follows a self-limiting trajectory, resolving over several months to 2 years.3,4 The resolution timeframe is intricately linked to variables such as the patient’s immune profile, lesion burden, and treatment approach. For symptomatic lesions, a variety of treatment options have been described, including physical ablation (eg, cryotherapy, curettage) and topical agents such as potassium hydroxide, cantharidin, imiquimod, and salicylic acid.3,4,8,9

Atopic dermatitis (AD) is a common chronic relapsing inflammatory skin disorder. In the United States, its prevalence ranges from 15% to 30% in children and from 2% to 10% in adults, with ongoing evidence of a growing global incidence.10-14 While AD can emerge at any age, typical onset is during early childhood. The clinical manifestation of AD includes a spectrum of eczematous features, often accompanied by persistent itching. The pathogenesis is multifactorial, involving a complex interplay of genetic, immunologic, and environmental factors. Key contributors to this multifaceted process encompass a compromised epidermal barrier, alterations in the skin microbiome, and an immune dysregulation promoting a type 2 immune response. Epidermal barrier dysfunction can be attributed to various factors, including diminished ceramide production, altered lipid composition, the release of inflammatory mediators, and mechanical damage from the persistent itch-scratch cycle.10-13,15 These factors or their interplay may enhance the susceptibility of patients with AD to infections. 

Several studies conducted across various geographic regions examining the relationship between MC and AD have reported variable findings.2,6,7,16-21 Published studies have reported a prevalence of AD in children with MC ranging from 13.2% to 43%.2,6,7,16-21 Although some studies suggest a higher rate of atopy in patients with MC, not all research has confirmed this association.16,21 Dohil et al2 reported a greater number of MC lesions in children with AD than those without an atopic background. Silverberg20 reported that in 10% (5/50) of children with MC, the onset of AD was triggered, and in 22% (11/50) MC was associated with flares of pre-existing AD.

In this study, we aimed to assess MC infection rates in children with AD, analyze the epidemiologic aspects and severity differences between atopic children with and without MC infection, and compare data from atopic and nonatopic children with MC.

Methods

In this retrospective cohort study, we analyzed the medical records of pediatric patients diagnosed with MC, AD, or both conditions at an outpatient dermatology practice in Netanya, HaSharon, Israel, from September 2013 to August 2022. Data were collected from the electronic medical records and included patient demographics, the clinical presentation of MC and/or AD at diagnosis, and the duration of both conditions. Only patients with complete data and at least 6 months of follow-up were included. Key epidemiologic characteristics assessed included patient sex, age at the initial visit, and age at the onset of MC and/or AD. Diagnoses of MC and AD were established through clinical examinations conducted by dermatologists. The clinical evaluation of AD encompassed the assessment of body surface area involvement (categorized as <5%, 5%-10%, or >10%). Atopic dermatitis severity was classified as mild, moderate, or severe using the validated Investigator Global Assessment Scale for Atopic Dermatitis.22 Clinical evaluation of MC included assessment of the number of lesions (categorized as 4, 5-9, or 10), presence of inflammatory lesions, and resolution times for individual lesions (categorized as <1 week, several weeks, or unknown), as well as the overall resolution time for all lesions (categorized as <6 months, 6-12 months, 13-18 months, or >18 months). The temporal relationship between the appearance of MC and AD also was assessed.

Statistical Analysis—Numbers and percentages were used for categorical variables. Continuous variables were represented by mean and standard deviation. Categorical variables were compared using the χ2 test, and continuous variables between groups were compared using the Student t test. All statistical tests were 2-sided, with statistical significance defined as P.05. Statistical analysis was performed using SPSS software version 28 (IBM).

Results

Study Population—A total of 610 children were included in the study; 263 (43%) were female and 347 (57%) were male. The patients ranged in age from 4 months to 10 years, with a mean (SD) age of 4.87 (1.82) years. Five hundred fifty-six (91%) patients had AD, and 336 (55%) had MC. Within this cohort, 274 (45%) children had AD only, 54 (9%) had MC only, and 282 (46%) had both AD and MC. Regarding the temporal sequence, among the 282 children who had both AD and MC, AD preceded MC in 203 (72%) cases, both conditions were diagnosed concomitantly in 43 (15%) cases, and MC preceded AD in 36 (13%) cases. For cases in which the MC diagnosis followed the diagnosis of AD, the mean (SD) time between each diagnosis was 3.17 (1.5) years.

Comparison of Atopic and Nonatopic Children With MC—Although a higher proportion of males were diagnosed with MC (with or without concurrent AD), the differences in sex distribution between the 2 groups did not reach statistical significance. Among all children with MC, the majority (81.5% [274/336]) were aged 1 to 6 years at presentation. Patients with MC as their sole diagnosis had a similar mean age compared with those with concurrent AD. However, a detailed age subgroup analysis revealed a notable distinction: in the group with MC as the sole diagnosis, the majority (95% [51/54]) were younger than 7 years. In contrast, in the combined MC and AD group, MC manifested across a wider age range, with 21% (58/282) of patients being older than 7 years. In MC cases associated with AD, a notably higher lesion count and increased local inflammatory response were observed compared to those without AD. The time for complete resolution of all MC lesions was substantially prolonged in patients with comorbid AD. Specifically, 93% (50/54) of patients with MC without comorbid AD achieved full resolution within 1 year, whereas 52% (146/282) of patients with comorbid AD required more than 1 year for resolution (eTable 1). 

CT116003099-eTable1

Comparison of Atopic Children With and Without MC—Sex, age distribution, and disease duration showed no differences between atopic patients with and without MC. Atopic patients with MC exhibited greater body surface area involvement and higher validated Investigator Global Assessment Scale for Atopic Dermatitis scores compared to atopic patients without MC (eTable 2).

CT116003099-eTable2

Comment

This study examined the relationship between MC and AD in pediatric patients, revealing a notable correlation and yielding valuable epidemiologic and clinical insights. Consistent with previous research, our study demonstrated a high prevalence of AD in children with MC.2,6,7,16-21 Previous studies indicated AD rates of 13% to 43% in pediatric patients with MC, whereas our study found a higher prevalence (84%), signifying a substantial majority of patients with MC in our cohort had AD. This discrepancy arises from factors such as demographic, genetic, and environmental differences, along with differences in access to medical care, referral practices, and diagnostic approaches across health care systems.14

Our temporal analysis of MC and AD diagnoses offers important insights. In the majority (72% [203/282]) of cases, the diagnosis of AD preceded MC, supporting previous research suggesting that the underlying pathophysiology of AD heightens susceptibility to MC.15,17-20 Less frequently, MC was diagnosed before or concurrently with AD, indicating that MC may occasionally trigger or exacerbate milder or undiagnosed AD, as previously proposed.20

A notable finding in our study was the expanded age range for MC onset in patients with AD, encompassing older age groups compared to patients with MC as their sole diagnosis, possibly due to persistent immune dysregulation. To the best of our knowledge, this specific observation has not been systematically reported or documented in prior cohort studies. Visible skin lesions of MC may have a psychological impact on patients, influencing self-consciousness and causing embarrassment and emotional distress. This may be more pronounced in older children, who are more aware of their appearance and social perceptions.23-25 These considerations should play a role in the management of MC. 

Our study revealed that children with AD and MC displayed higher lesion counts, increased local inflammatory responses, and a more protracted resolution period compared to nonatopic children. In more than 50% of children with AD, MC took more than 1 year for resolution, whereas the majority of those without AD achieved resolution within 1 year. These findings may be attributed to AD-related immune dysregulation, influencing the natural course of MC. Consequently, it suggests that while nonatopic children with MC usually are managed through observation, atopic patients may benefit from an intervention-oriented approach. 

Comparing atopic patients with and without MC showed a heightened occurrence of severe and extensive AD among those with concurrent MC. Several factors could contribute to this observation. On one hand, there could be a direct association between the extent and severity of AD, leading to an elevated susceptibility to MC. Conversely, MC might exacerbate immunologic dysregulation and intensify skin inflammation in atopic individuals.20 Additionally, itching related to both disorders may exacerbate inflammation and compromise the epidermal barrier, facilitating the spread of MC. This interplay suggests that each condition exacerbates the other in a self-reinforcing cycle. The importance of patient and caregiver education is underscored by recognizing these interactions. To manage both conditions effectively, health care providers should counsel patients and caregivers on maintaining proper skin care practices such as gentle cleansing with mild, fragrance-free products, regular moisturization, and avoidance of irritants, encourage them to avoid scratching, and recommend adopting an active treatment approach.

Our study had notable strengths. Firstly, a substantial sample size enhanced the statistical reliability of our findings. Additionally, valuable insights into the epidemiology and clinical aspects of AD and MC were obtained by utilizing real-world data from an outpatient dermatology practice. In our study, clinical evaluations covered body surface area involvement and disease severity for AD while also assessing lesion counts and the presence of inflammatory lesions for MC. This comprehensive approach facilitated a thorough analysis of both conditions. The extended data collection period not only allowed for observation of their clinical course and duration, but also enabled a detailed assessment of their interplay.

Our study also had several limitations. Primarily, its retrospective design relied on the accuracy and comprehensiveness of medical records, which may have introduced bias. The exclusion of some patients due to incomplete data further increased the potential for selection bias. Additionally, this study was conducted in a single outpatient dermatology practice in Israel, resulting in a study population composed predominantly of Jewish patients (94%), with a minority (6%) of Arab patients. Other ethnic groups, including Black, Asian, and Hispanic populations, were not represented. This reflects the country’s demographic composition rather than an intentional selection bias. However, the limited ethnic diversity reduces the generalizability of our findings. Differences in demographics, coding practices, health care utilization (eg, timeliness of seeking care, access to dermatology services), and treatment strategies also may impact the observed prevalence, clinical characteristics, and patient outcomes. Furthermore, while our study highlighted the potential advantages of a proactive treatment approach for atopic children with MC, it did not evaluate specific treatment protocols. Future research should aim to confirm the most efficacious therapeutic strategies for managing MC in atopic individuals and to include a more diverse population to better understand the applicability of findings across various ethnic groups.

Conclusion

Our study found a high prevalence of AD in children with MC and a strong bidirectional relationship between these conditions. Pediatric patients with AD display a broader age range for MC, greater lesion burden, increased local inflammatory responses, prolonged resolution times, and more extensive and severe AD.

Recognizing the interplay between MC and AD is crucial, highlighting the importance of health care providers educating patients and caregivers. Emphasizing skin hygiene, discouraging scratching, and implementing proactive treatment approaches can enhance the outcomes of both conditions. Further research into the underlying mechanisms of this association and effective therapeutic strategies for MC in atopic individuals is warranted.

Acknowledgments—The authors thank Zvi Segal, MD (Tel Hashomer, Israel) for his insightful contribution to the statistical analysis of the results. We would like to express our appreciation to the dedicated team of the dermatology practice in Netanya for the support throughout the performance of the study. Additionally, we thank all study participants and their parents for their participation and contribution to our research.

Molluscum contagiosum (MC), which is caused by a DNA virus in the Poxviridae family, is a common viral skin infection that primarily affects children.1-4 The reported incidence and prevalence of MC exhibit notable geographic variation. Worldwide, annual incidence rates per 1000 individuals range from 3.1 to 25, and prevalence ranges from 0.27% to 34.6%.2-7

Molluscum contagiosum is diagnosed clinically and typically manifests as smooth, flesh-colored papules measuring 2 to 6 mm in diameter with central umbilication. It can manifest as a single lesion or multiple clustered lesions, or in a disseminated pattern. The primary mode of transmission is through contact with skin, lesions, or contaminated personal items, or via self-inoculation. The majority of cases are asymptomatic, but in some patients, MC may be associated with pruritus, tenderness, erythema, or irritation. When present, secondary bacterial infections can cause localized inflammation and pain.1,3,4 The pathogenesis hinges on MC virus replication within keratinocytes, disrupting cellular differentiation and keratinization. The virus persists in the host by influencing the immune response through various mechanisms, including interference with signaling pathways, apoptosis inhibition, and antigen presentation disruption.3,4

Molluscum contagiosum typically follows a self-limiting trajectory, resolving over several months to 2 years.3,4 The resolution timeframe is intricately linked to variables such as the patient’s immune profile, lesion burden, and treatment approach. For symptomatic lesions, a variety of treatment options have been described, including physical ablation (eg, cryotherapy, curettage) and topical agents such as potassium hydroxide, cantharidin, imiquimod, and salicylic acid.3,4,8,9

Atopic dermatitis (AD) is a common chronic relapsing inflammatory skin disorder. In the United States, its prevalence ranges from 15% to 30% in children and from 2% to 10% in adults, with ongoing evidence of a growing global incidence.10-14 While AD can emerge at any age, typical onset is during early childhood. The clinical manifestation of AD includes a spectrum of eczematous features, often accompanied by persistent itching. The pathogenesis is multifactorial, involving a complex interplay of genetic, immunologic, and environmental factors. Key contributors to this multifaceted process encompass a compromised epidermal barrier, alterations in the skin microbiome, and an immune dysregulation promoting a type 2 immune response. Epidermal barrier dysfunction can be attributed to various factors, including diminished ceramide production, altered lipid composition, the release of inflammatory mediators, and mechanical damage from the persistent itch-scratch cycle.10-13,15 These factors or their interplay may enhance the susceptibility of patients with AD to infections. 

Several studies conducted across various geographic regions examining the relationship between MC and AD have reported variable findings.2,6,7,16-21 Published studies have reported a prevalence of AD in children with MC ranging from 13.2% to 43%.2,6,7,16-21 Although some studies suggest a higher rate of atopy in patients with MC, not all research has confirmed this association.16,21 Dohil et al2 reported a greater number of MC lesions in children with AD than those without an atopic background. Silverberg20 reported that in 10% (5/50) of children with MC, the onset of AD was triggered, and in 22% (11/50) MC was associated with flares of pre-existing AD.

In this study, we aimed to assess MC infection rates in children with AD, analyze the epidemiologic aspects and severity differences between atopic children with and without MC infection, and compare data from atopic and nonatopic children with MC.

Methods

In this retrospective cohort study, we analyzed the medical records of pediatric patients diagnosed with MC, AD, or both conditions at an outpatient dermatology practice in Netanya, HaSharon, Israel, from September 2013 to August 2022. Data were collected from the electronic medical records and included patient demographics, the clinical presentation of MC and/or AD at diagnosis, and the duration of both conditions. Only patients with complete data and at least 6 months of follow-up were included. Key epidemiologic characteristics assessed included patient sex, age at the initial visit, and age at the onset of MC and/or AD. Diagnoses of MC and AD were established through clinical examinations conducted by dermatologists. The clinical evaluation of AD encompassed the assessment of body surface area involvement (categorized as <5%, 5%-10%, or >10%). Atopic dermatitis severity was classified as mild, moderate, or severe using the validated Investigator Global Assessment Scale for Atopic Dermatitis.22 Clinical evaluation of MC included assessment of the number of lesions (categorized as 4, 5-9, or 10), presence of inflammatory lesions, and resolution times for individual lesions (categorized as <1 week, several weeks, or unknown), as well as the overall resolution time for all lesions (categorized as <6 months, 6-12 months, 13-18 months, or >18 months). The temporal relationship between the appearance of MC and AD also was assessed.

Statistical Analysis—Numbers and percentages were used for categorical variables. Continuous variables were represented by mean and standard deviation. Categorical variables were compared using the χ2 test, and continuous variables between groups were compared using the Student t test. All statistical tests were 2-sided, with statistical significance defined as P.05. Statistical analysis was performed using SPSS software version 28 (IBM).

Results

Study Population—A total of 610 children were included in the study; 263 (43%) were female and 347 (57%) were male. The patients ranged in age from 4 months to 10 years, with a mean (SD) age of 4.87 (1.82) years. Five hundred fifty-six (91%) patients had AD, and 336 (55%) had MC. Within this cohort, 274 (45%) children had AD only, 54 (9%) had MC only, and 282 (46%) had both AD and MC. Regarding the temporal sequence, among the 282 children who had both AD and MC, AD preceded MC in 203 (72%) cases, both conditions were diagnosed concomitantly in 43 (15%) cases, and MC preceded AD in 36 (13%) cases. For cases in which the MC diagnosis followed the diagnosis of AD, the mean (SD) time between each diagnosis was 3.17 (1.5) years.

Comparison of Atopic and Nonatopic Children With MC—Although a higher proportion of males were diagnosed with MC (with or without concurrent AD), the differences in sex distribution between the 2 groups did not reach statistical significance. Among all children with MC, the majority (81.5% [274/336]) were aged 1 to 6 years at presentation. Patients with MC as their sole diagnosis had a similar mean age compared with those with concurrent AD. However, a detailed age subgroup analysis revealed a notable distinction: in the group with MC as the sole diagnosis, the majority (95% [51/54]) were younger than 7 years. In contrast, in the combined MC and AD group, MC manifested across a wider age range, with 21% (58/282) of patients being older than 7 years. In MC cases associated with AD, a notably higher lesion count and increased local inflammatory response were observed compared to those without AD. The time for complete resolution of all MC lesions was substantially prolonged in patients with comorbid AD. Specifically, 93% (50/54) of patients with MC without comorbid AD achieved full resolution within 1 year, whereas 52% (146/282) of patients with comorbid AD required more than 1 year for resolution (eTable 1). 

CT116003099-eTable1

Comparison of Atopic Children With and Without MC—Sex, age distribution, and disease duration showed no differences between atopic patients with and without MC. Atopic patients with MC exhibited greater body surface area involvement and higher validated Investigator Global Assessment Scale for Atopic Dermatitis scores compared to atopic patients without MC (eTable 2).

CT116003099-eTable2

Comment

This study examined the relationship between MC and AD in pediatric patients, revealing a notable correlation and yielding valuable epidemiologic and clinical insights. Consistent with previous research, our study demonstrated a high prevalence of AD in children with MC.2,6,7,16-21 Previous studies indicated AD rates of 13% to 43% in pediatric patients with MC, whereas our study found a higher prevalence (84%), signifying a substantial majority of patients with MC in our cohort had AD. This discrepancy arises from factors such as demographic, genetic, and environmental differences, along with differences in access to medical care, referral practices, and diagnostic approaches across health care systems.14

Our temporal analysis of MC and AD diagnoses offers important insights. In the majority (72% [203/282]) of cases, the diagnosis of AD preceded MC, supporting previous research suggesting that the underlying pathophysiology of AD heightens susceptibility to MC.15,17-20 Less frequently, MC was diagnosed before or concurrently with AD, indicating that MC may occasionally trigger or exacerbate milder or undiagnosed AD, as previously proposed.20

A notable finding in our study was the expanded age range for MC onset in patients with AD, encompassing older age groups compared to patients with MC as their sole diagnosis, possibly due to persistent immune dysregulation. To the best of our knowledge, this specific observation has not been systematically reported or documented in prior cohort studies. Visible skin lesions of MC may have a psychological impact on patients, influencing self-consciousness and causing embarrassment and emotional distress. This may be more pronounced in older children, who are more aware of their appearance and social perceptions.23-25 These considerations should play a role in the management of MC. 

Our study revealed that children with AD and MC displayed higher lesion counts, increased local inflammatory responses, and a more protracted resolution period compared to nonatopic children. In more than 50% of children with AD, MC took more than 1 year for resolution, whereas the majority of those without AD achieved resolution within 1 year. These findings may be attributed to AD-related immune dysregulation, influencing the natural course of MC. Consequently, it suggests that while nonatopic children with MC usually are managed through observation, atopic patients may benefit from an intervention-oriented approach. 

Comparing atopic patients with and without MC showed a heightened occurrence of severe and extensive AD among those with concurrent MC. Several factors could contribute to this observation. On one hand, there could be a direct association between the extent and severity of AD, leading to an elevated susceptibility to MC. Conversely, MC might exacerbate immunologic dysregulation and intensify skin inflammation in atopic individuals.20 Additionally, itching related to both disorders may exacerbate inflammation and compromise the epidermal barrier, facilitating the spread of MC. This interplay suggests that each condition exacerbates the other in a self-reinforcing cycle. The importance of patient and caregiver education is underscored by recognizing these interactions. To manage both conditions effectively, health care providers should counsel patients and caregivers on maintaining proper skin care practices such as gentle cleansing with mild, fragrance-free products, regular moisturization, and avoidance of irritants, encourage them to avoid scratching, and recommend adopting an active treatment approach.

Our study had notable strengths. Firstly, a substantial sample size enhanced the statistical reliability of our findings. Additionally, valuable insights into the epidemiology and clinical aspects of AD and MC were obtained by utilizing real-world data from an outpatient dermatology practice. In our study, clinical evaluations covered body surface area involvement and disease severity for AD while also assessing lesion counts and the presence of inflammatory lesions for MC. This comprehensive approach facilitated a thorough analysis of both conditions. The extended data collection period not only allowed for observation of their clinical course and duration, but also enabled a detailed assessment of their interplay.

Our study also had several limitations. Primarily, its retrospective design relied on the accuracy and comprehensiveness of medical records, which may have introduced bias. The exclusion of some patients due to incomplete data further increased the potential for selection bias. Additionally, this study was conducted in a single outpatient dermatology practice in Israel, resulting in a study population composed predominantly of Jewish patients (94%), with a minority (6%) of Arab patients. Other ethnic groups, including Black, Asian, and Hispanic populations, were not represented. This reflects the country’s demographic composition rather than an intentional selection bias. However, the limited ethnic diversity reduces the generalizability of our findings. Differences in demographics, coding practices, health care utilization (eg, timeliness of seeking care, access to dermatology services), and treatment strategies also may impact the observed prevalence, clinical characteristics, and patient outcomes. Furthermore, while our study highlighted the potential advantages of a proactive treatment approach for atopic children with MC, it did not evaluate specific treatment protocols. Future research should aim to confirm the most efficacious therapeutic strategies for managing MC in atopic individuals and to include a more diverse population to better understand the applicability of findings across various ethnic groups.

Conclusion

Our study found a high prevalence of AD in children with MC and a strong bidirectional relationship between these conditions. Pediatric patients with AD display a broader age range for MC, greater lesion burden, increased local inflammatory responses, prolonged resolution times, and more extensive and severe AD.

Recognizing the interplay between MC and AD is crucial, highlighting the importance of health care providers educating patients and caregivers. Emphasizing skin hygiene, discouraging scratching, and implementing proactive treatment approaches can enhance the outcomes of both conditions. Further research into the underlying mechanisms of this association and effective therapeutic strategies for MC in atopic individuals is warranted.

Acknowledgments—The authors thank Zvi Segal, MD (Tel Hashomer, Israel) for his insightful contribution to the statistical analysis of the results. We would like to express our appreciation to the dedicated team of the dermatology practice in Netanya for the support throughout the performance of the study. Additionally, we thank all study participants and their parents for their participation and contribution to our research.

References
  1. Han H, Smythe C, Yousefian F, et al. Molluscum contagiosum virus evasion of immune surveillance: a review. J Drugs Dermatol. 2023;22182-189.
  2. Dohil MA, Lin P, Lee J, et al. The epidemiology of molluscum contagiosum in children. J Am Acad Dermatol. 2006;54:47-54.
  3. Silverberg NB. Pediatric molluscum: an update. Cutis. 2019;104:301-305;E1;E2.
  4. Forbat E, Al-Niaimi F, Ali FR. Molluscum contagiosum: review and update on management. Pediatr Dermatol. 2017;34:504-515.
  5. Olsen JR, Gallacher J, Piguet V, et al. Epidemiology of molluscum contagiosum in children: a systematic review. Fam Pract. 2014;31:130-136.
  6. Kakourou T, Zachariades A, Anastasiou T, et al. Molluscum contagiosum in Greek children: a case series. Int J Dermatol. 2005;44:221-223.
  7. Osio A, Deslandes E, Saada V, et al. Clinical characteristics of molluscum contagiosum in children in a private dermatology practice in the greater Paris area, France: a prospective study in 661 patients. Dermatology. 2011;222:314-320.
  8. Hebert AA, Bhatia N, Del Rosso JQ. Molluscum contagiosum: epidemiology, considerations, treatment options, and therapeutic gaps. J Clin Aesthet Dermatol. 2023;16(8 Suppl 1):S4-S11.
  9. Chao YC, Ko MJ, Tsai WC, et al. Comparative efficacy of treatments for molluscum contagiosum: a systematic review and network meta-analysis. J Dtsch Dermatol Ges. 2023;21:587-597.
  10. Garg N, Silverberg JI. Epidemiology of childhood atopic dermatitis. Clin Dermatol. 2015;33:281-288.
  11. Hale G, Davies E, Grindlay DJC, et al. What’s new in atopic eczema? an analysis of systematic reviews published in 2017. part 2: epidemiology, etiology, and risk factors. Clin Exp Dermatol. 2019;44:868-873.
  12. Tracy A, Bhatti S, Eichenfield LF. Update on pediatric atopic dermatitis. Cutis. 2020;106:143-146.
  13. Langan SM, Irvine AD, Weidinger S. Atopic dermatitis. Lancet. 2020;396:345-360.
  14. Silverberg JI. Public health burden and epidemiology of atopic dermatitis. Dermatol Clin. 2017;35:283-289.
  15. Manti S, Amorini M, Cuppari C, et al. Filaggrin mutations and molluscum contagiosum skin infection in patients with atopic dermatitis. Ann Allergy Asthma Immunol. 2017;119446-451.
  16. Seize M, Ianhez M, Cestari S. A study of the correlation between molluscum contagiosum and atopic dermatitis in children. An Bras Dermatol. 2011;86:663-668.
  17. Ren Z, Silverberg JI. Association of atopic dermatitis with bacterial, fungal, viral, and sexually transmitted skin infections. Dermatitis. 2020;31:157-164.
  18. Olsen JR, Piguet V, Gallacher J, et al. Molluscum contagiosum and associations with atopic eczema in children: a retrospective longitudinal study in primary care. Br J Gen Pract. 2016;66:E53-E58.
  19. Han JH, Yoon JW, Yook HJ, et al. Evaluation of atopic dermatitis and cutaneous infectious disorders using sequential pattern mining: a nationwide population-based cohort study. J Clin Med. 2022;11:3422.
  20. Silverberg NB. Molluscum contagiosum virus infection can trigger atopic dermatitis disease onset or flare. Cutis. 2018;102:191-194.
  21. Hayashida S, Furusho N, Uchi H, et al. Are lifetime prevalence of impetigo, molluscum and herpes infection really increased in children having atopic dermatitis? J Dermatol Sci. 2010;60:173-178.
  22. Simpson E, Bissonnette R, Eichenfield LF, et al. The Validated Investigator Global Assessment for Atopic Dermatitis (vIGA-AD): the development and reliability testing of a novel clinical outcome measurement instrument for the severity of atopic dermatitis. J Am Acad Dermatol. 2020;83:839-846.
  23. Olsen JR, Gallacher J, Finlay AY, et al. Time to resolution and effect on quality of life of molluscum contagiosum in children in the UK: a prospective community cohort study. Lancet Infect Dis. 2015;15:190-195.
  24. Ðurovic´ MR, Jankovic´ J, Spiric´ VT, et al. Does age influence the quality of life in children with atopic dermatitis? PLoS One. 2019;14:E0224618.
  25. Chernyshov PV. Stigmatization and self-perception in children with atopic dermatitis. Clin Cosmet Investig Dermatol. 2016;9:159-166.
References
  1. Han H, Smythe C, Yousefian F, et al. Molluscum contagiosum virus evasion of immune surveillance: a review. J Drugs Dermatol. 2023;22182-189.
  2. Dohil MA, Lin P, Lee J, et al. The epidemiology of molluscum contagiosum in children. J Am Acad Dermatol. 2006;54:47-54.
  3. Silverberg NB. Pediatric molluscum: an update. Cutis. 2019;104:301-305;E1;E2.
  4. Forbat E, Al-Niaimi F, Ali FR. Molluscum contagiosum: review and update on management. Pediatr Dermatol. 2017;34:504-515.
  5. Olsen JR, Gallacher J, Piguet V, et al. Epidemiology of molluscum contagiosum in children: a systematic review. Fam Pract. 2014;31:130-136.
  6. Kakourou T, Zachariades A, Anastasiou T, et al. Molluscum contagiosum in Greek children: a case series. Int J Dermatol. 2005;44:221-223.
  7. Osio A, Deslandes E, Saada V, et al. Clinical characteristics of molluscum contagiosum in children in a private dermatology practice in the greater Paris area, France: a prospective study in 661 patients. Dermatology. 2011;222:314-320.
  8. Hebert AA, Bhatia N, Del Rosso JQ. Molluscum contagiosum: epidemiology, considerations, treatment options, and therapeutic gaps. J Clin Aesthet Dermatol. 2023;16(8 Suppl 1):S4-S11.
  9. Chao YC, Ko MJ, Tsai WC, et al. Comparative efficacy of treatments for molluscum contagiosum: a systematic review and network meta-analysis. J Dtsch Dermatol Ges. 2023;21:587-597.
  10. Garg N, Silverberg JI. Epidemiology of childhood atopic dermatitis. Clin Dermatol. 2015;33:281-288.
  11. Hale G, Davies E, Grindlay DJC, et al. What’s new in atopic eczema? an analysis of systematic reviews published in 2017. part 2: epidemiology, etiology, and risk factors. Clin Exp Dermatol. 2019;44:868-873.
  12. Tracy A, Bhatti S, Eichenfield LF. Update on pediatric atopic dermatitis. Cutis. 2020;106:143-146.
  13. Langan SM, Irvine AD, Weidinger S. Atopic dermatitis. Lancet. 2020;396:345-360.
  14. Silverberg JI. Public health burden and epidemiology of atopic dermatitis. Dermatol Clin. 2017;35:283-289.
  15. Manti S, Amorini M, Cuppari C, et al. Filaggrin mutations and molluscum contagiosum skin infection in patients with atopic dermatitis. Ann Allergy Asthma Immunol. 2017;119446-451.
  16. Seize M, Ianhez M, Cestari S. A study of the correlation between molluscum contagiosum and atopic dermatitis in children. An Bras Dermatol. 2011;86:663-668.
  17. Ren Z, Silverberg JI. Association of atopic dermatitis with bacterial, fungal, viral, and sexually transmitted skin infections. Dermatitis. 2020;31:157-164.
  18. Olsen JR, Piguet V, Gallacher J, et al. Molluscum contagiosum and associations with atopic eczema in children: a retrospective longitudinal study in primary care. Br J Gen Pract. 2016;66:E53-E58.
  19. Han JH, Yoon JW, Yook HJ, et al. Evaluation of atopic dermatitis and cutaneous infectious disorders using sequential pattern mining: a nationwide population-based cohort study. J Clin Med. 2022;11:3422.
  20. Silverberg NB. Molluscum contagiosum virus infection can trigger atopic dermatitis disease onset or flare. Cutis. 2018;102:191-194.
  21. Hayashida S, Furusho N, Uchi H, et al. Are lifetime prevalence of impetigo, molluscum and herpes infection really increased in children having atopic dermatitis? J Dermatol Sci. 2010;60:173-178.
  22. Simpson E, Bissonnette R, Eichenfield LF, et al. The Validated Investigator Global Assessment for Atopic Dermatitis (vIGA-AD): the development and reliability testing of a novel clinical outcome measurement instrument for the severity of atopic dermatitis. J Am Acad Dermatol. 2020;83:839-846.
  23. Olsen JR, Gallacher J, Finlay AY, et al. Time to resolution and effect on quality of life of molluscum contagiosum in children in the UK: a prospective community cohort study. Lancet Infect Dis. 2015;15:190-195.
  24. Ðurovic´ MR, Jankovic´ J, Spiric´ VT, et al. Does age influence the quality of life in children with atopic dermatitis? PLoS One. 2019;14:E0224618.
  25. Chernyshov PV. Stigmatization and self-perception in children with atopic dermatitis. Clin Cosmet Investig Dermatol. 2016;9:159-166.
Issue
Cutis - 116(3)
Issue
Cutis - 116(3)
Page Number
99-102
Page Number
99-102
Publications
Publications
Topics
Article Type
Display Headline

Epidemiologic and Clinical Evaluation of the Bidirectional Link Between Molluscum Contagiosum and Atopic Dermatitis in Children

Display Headline

Epidemiologic and Clinical Evaluation of the Bidirectional Link Between Molluscum Contagiosum and Atopic Dermatitis in Children

Sections
Inside the Article

Practice Points

  • There is a high prevalence of atopic dermatitis (AD) in children with molluscum contagiosum, with a strong bidirectional relationship between these conditions.
  • Children with AD display a broader age range for molluscum contagiosum, greater lesion burden, increased local inflammatory responses, prolonged resolution time, and more extensive and severe disease.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Fri, 09/05/2025 - 11:01
Un-Gate On Date
Fri, 09/05/2025 - 11:01
Use ProPublica
CFC Schedule Remove Status
Fri, 09/05/2025 - 11:01
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
survey writer start date
Fri, 09/05/2025 - 11:01

A Cross-Sectional Analysis of TikTok Skin Care Routines and the Associated Environmental Impact

Article Type
Changed
Fri, 09/05/2025 - 10:31
Display Headline

A Cross-Sectional Analysis of TikTok Skin Care Routines and the Associated Environmental Impact

To the Editor:

The popularity of the social media platform TikTok, which is known for its short-form videos, has surged in recent years. Viral videos demonstrating skin care routines reach millions of viewers,1 showcasing specific products, detailing beauty regimens, and setting fads that many users eagerly follow. These trends often influence consumer behavior—in 2023, viral videos using the tag #TikTokMadeMeBuy lead to a 14% growth in the sale of skin care products.2 However, they also encourage purchasing decisions that may escalate environmental waste through plastic packaging and single-use products. In this study, we analyzed videos on TikTok to assess the environmental impact of trending skin care routines. By examining the types of products promoted, their packaging, and the frequency with which they appear in viral content, we aimed to investigate how these trends, which may be imitated by users, impact the environment.

A search of TikTok videos using #skincareroutine was conducted on June 21, 2024. Sponsored content, non–English language videos, videos without demonstrated skin care routines, and videos showing makeup routines were excluded from our analysis. Data collected from each video included username, date posted, number of likes, total number of skin care products used, number of single-use skin care products used, average amount of product used, number of skin care applicators used, and number of single-use applicators used. Single-use items, defined as those intended for one-time use and subsequent disposal, were identified visually by packaging, manufacturer intent, and common consumer usage patterns. The amount of product used per application was graded on a scale of 1 to 3 (1=pea-sized amount or less; 2=single full pump/spray; 3=multiple pumps/sprays). Videos were categorized as personal (ie, skin care routine walk-throughs by the creator) or autonomous sensory meridian response (ASMR)(focused on product sounds and aesthetics).3 A Mann-Whitney U test was utilized to statistically compare the 2 groups. Statistical analysis was performed using Microsoft Excel (α=0.05). 

A total of 50 videos met the inclusion criteria and were included in the analysis. The average number of likes per video was 499,696.15, with skin care routines featuring an average of 6.4 unique products (Table). There was a weak positive correlation (r=0.1809) between the number of skin care products used and the number of likes. A total of 320 products were used across the videos, 23 of which were single-use (7.2%).On average, single-use skin care items were used 0.46 times per routine, comprising a mean 7.99% of total products per video. The average score for the amount of product used per application was 2.18. There was no difference in personal vs ASMR videos with regard to the total number of skin care products used or the average amount of product used per application (P>.05). Thirty-three (70.2%) of the 47 applicators used across all videos were single-use. An average of 0.94 applicators per routine were utilized, with a mean 68.83% being single-use applicators. Common single-use products were toner wipes and eye patches, and single-use applicators included cotton pads and plastic spatulas. 

CT116003107-Table

Our findings indicated a prevalence of multiple products and large amount of product used in trending skin care routines, suggesting a shift toward multistep skin care. This implies a high rate of product consumption that may accelerate the carbon footprint associated with skin care products,3 which could contribute to climate change and environmental degradation. Consumers also may feel compelled to purchase and discard numerous partially used products in order to keep up with the latest trends, exacerbating the environmental impact. Furthermore, the utilization of single-use products and applicators contributes to increased plastic waste, pollution, and resource depletion. Single-use items often are difficult to recycle due to their mixed materials and small size,4,5 and therefore they can accumulate in landfills and oceans. This impact can be mitigated by switching to reusable applicators, refillable packaging, and biodegradable materials. 

The substantial average number of likes per video indicates high engagement with skin care content among TikTok users. The continued popularity of complex multi­step skin care routines, despite a weak correlation between the number of skin care products used and the number of likes per video, likely stems from factors such as aesthetic appeal, ASMR effects, and creators’ established followings, which may drive user engagement to contribute to unsustainable consumption patterns. Factors such as presentation style, aesthetics, or creators’ pre-existing online following may have a major impact on how well a video performs on TikTok. The similarity between personal and ASMR videos, particularly in the number of products used and the amount applied, suggests that both formats employ common approaches to meet audience expectations and align with promotional trends, relying more on sensory and aesthetic strategies than substantive differences in skin care routines.

Our use of only one tag in our search as well as the subjective quantity scale limits the generalizability of these findings to broader TikTok skin care content.

Overall, our study underscores the role of brands and social media influencers in skin care education and promotion of sustainable practices. The extensive number of products used and generous application of each product in skin care routines demonstrated in TikTok videos may mislead viewers into believing that using more product improves outcomes, when often, less is more. We recommend that dermatologists counsel patients about informed skin care regimens that prioritize individual needs over social media fads.

References
  1. Pagani K, Lukac D, Martinez R, et al. Slugging: TikTokTM as a source of a viral “harmless” beauty trend. Clin Dermatol. 2022;40:810-812. doi:10.1016/j.clindermatol.2022.08.005
  2. Stern C. TikTok drives $31.7B in beauty sales: how viral trends are shaping the future of cosmetics. CosmeticsDesign. August 20, 2024. Accessed June 24, 2025. https://www.cosmeticsdesign.com/Article/2024/08/20/tiktok-drives-31.7b-in-beauty-sales-how-viral-trends-are-shaping-the-future-of-cosmetics/
  3. Fountain C. ASMR content saw huge growth on YouTube, but now creators are flocking to TikTok instead. Business Insider. July 4, 2022. Accessed June 24, 2025. https://www.businessinsider.com/asmr-tiktok-instead-of-youtube-growth-subscribers-2022-7
  4. Rathore S, Schuler B, Park J. Life cycle assessment of multiple dispensing systems used for cosmetic product packaging. Packaging Technol Sci. 2023;36:533-547. doi:10.1002/pts.2729
  5. Shaw S. How to actually recycle your empty beauty products. CNN Underscored. Updated April 17, 2024. Accessed June 24, 2025. https://www.cnn.com/cnn-underscored/beauty/how-to-recycle-beauty-products
Article PDF
Author and Disclosure Information

Aarushi K. Parikh is from Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey. Dr. Lipner is from the Department of Dermatology, Weill Cornell Medicine, New York, New York.

Aarushi K. Parikh has no relevant financial disclosures to report. Dr. Lipner has served as a consultant for BelleTorus Corporation and Moberg Pharma.

Correspondence: Shari R. Lipner, MD, PhD, 1305 York Avenue, New York, NY 10021 ([email protected]).

Cutis. 2025 September;116(3):107-108. doi:10.12788/cutis.1259

Issue
Cutis - 116(3)
Publications
Topics
Page Number
107-108
Sections
Author and Disclosure Information

Aarushi K. Parikh is from Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey. Dr. Lipner is from the Department of Dermatology, Weill Cornell Medicine, New York, New York.

Aarushi K. Parikh has no relevant financial disclosures to report. Dr. Lipner has served as a consultant for BelleTorus Corporation and Moberg Pharma.

Correspondence: Shari R. Lipner, MD, PhD, 1305 York Avenue, New York, NY 10021 ([email protected]).

Cutis. 2025 September;116(3):107-108. doi:10.12788/cutis.1259

Author and Disclosure Information

Aarushi K. Parikh is from Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey. Dr. Lipner is from the Department of Dermatology, Weill Cornell Medicine, New York, New York.

Aarushi K. Parikh has no relevant financial disclosures to report. Dr. Lipner has served as a consultant for BelleTorus Corporation and Moberg Pharma.

Correspondence: Shari R. Lipner, MD, PhD, 1305 York Avenue, New York, NY 10021 ([email protected]).

Cutis. 2025 September;116(3):107-108. doi:10.12788/cutis.1259

Article PDF
Article PDF

To the Editor:

The popularity of the social media platform TikTok, which is known for its short-form videos, has surged in recent years. Viral videos demonstrating skin care routines reach millions of viewers,1 showcasing specific products, detailing beauty regimens, and setting fads that many users eagerly follow. These trends often influence consumer behavior—in 2023, viral videos using the tag #TikTokMadeMeBuy lead to a 14% growth in the sale of skin care products.2 However, they also encourage purchasing decisions that may escalate environmental waste through plastic packaging and single-use products. In this study, we analyzed videos on TikTok to assess the environmental impact of trending skin care routines. By examining the types of products promoted, their packaging, and the frequency with which they appear in viral content, we aimed to investigate how these trends, which may be imitated by users, impact the environment.

A search of TikTok videos using #skincareroutine was conducted on June 21, 2024. Sponsored content, non–English language videos, videos without demonstrated skin care routines, and videos showing makeup routines were excluded from our analysis. Data collected from each video included username, date posted, number of likes, total number of skin care products used, number of single-use skin care products used, average amount of product used, number of skin care applicators used, and number of single-use applicators used. Single-use items, defined as those intended for one-time use and subsequent disposal, were identified visually by packaging, manufacturer intent, and common consumer usage patterns. The amount of product used per application was graded on a scale of 1 to 3 (1=pea-sized amount or less; 2=single full pump/spray; 3=multiple pumps/sprays). Videos were categorized as personal (ie, skin care routine walk-throughs by the creator) or autonomous sensory meridian response (ASMR)(focused on product sounds and aesthetics).3 A Mann-Whitney U test was utilized to statistically compare the 2 groups. Statistical analysis was performed using Microsoft Excel (α=0.05). 

A total of 50 videos met the inclusion criteria and were included in the analysis. The average number of likes per video was 499,696.15, with skin care routines featuring an average of 6.4 unique products (Table). There was a weak positive correlation (r=0.1809) between the number of skin care products used and the number of likes. A total of 320 products were used across the videos, 23 of which were single-use (7.2%).On average, single-use skin care items were used 0.46 times per routine, comprising a mean 7.99% of total products per video. The average score for the amount of product used per application was 2.18. There was no difference in personal vs ASMR videos with regard to the total number of skin care products used or the average amount of product used per application (P>.05). Thirty-three (70.2%) of the 47 applicators used across all videos were single-use. An average of 0.94 applicators per routine were utilized, with a mean 68.83% being single-use applicators. Common single-use products were toner wipes and eye patches, and single-use applicators included cotton pads and plastic spatulas. 

CT116003107-Table

Our findings indicated a prevalence of multiple products and large amount of product used in trending skin care routines, suggesting a shift toward multistep skin care. This implies a high rate of product consumption that may accelerate the carbon footprint associated with skin care products,3 which could contribute to climate change and environmental degradation. Consumers also may feel compelled to purchase and discard numerous partially used products in order to keep up with the latest trends, exacerbating the environmental impact. Furthermore, the utilization of single-use products and applicators contributes to increased plastic waste, pollution, and resource depletion. Single-use items often are difficult to recycle due to their mixed materials and small size,4,5 and therefore they can accumulate in landfills and oceans. This impact can be mitigated by switching to reusable applicators, refillable packaging, and biodegradable materials. 

The substantial average number of likes per video indicates high engagement with skin care content among TikTok users. The continued popularity of complex multi­step skin care routines, despite a weak correlation between the number of skin care products used and the number of likes per video, likely stems from factors such as aesthetic appeal, ASMR effects, and creators’ established followings, which may drive user engagement to contribute to unsustainable consumption patterns. Factors such as presentation style, aesthetics, or creators’ pre-existing online following may have a major impact on how well a video performs on TikTok. The similarity between personal and ASMR videos, particularly in the number of products used and the amount applied, suggests that both formats employ common approaches to meet audience expectations and align with promotional trends, relying more on sensory and aesthetic strategies than substantive differences in skin care routines.

Our use of only one tag in our search as well as the subjective quantity scale limits the generalizability of these findings to broader TikTok skin care content.

Overall, our study underscores the role of brands and social media influencers in skin care education and promotion of sustainable practices. The extensive number of products used and generous application of each product in skin care routines demonstrated in TikTok videos may mislead viewers into believing that using more product improves outcomes, when often, less is more. We recommend that dermatologists counsel patients about informed skin care regimens that prioritize individual needs over social media fads.

To the Editor:

The popularity of the social media platform TikTok, which is known for its short-form videos, has surged in recent years. Viral videos demonstrating skin care routines reach millions of viewers,1 showcasing specific products, detailing beauty regimens, and setting fads that many users eagerly follow. These trends often influence consumer behavior—in 2023, viral videos using the tag #TikTokMadeMeBuy lead to a 14% growth in the sale of skin care products.2 However, they also encourage purchasing decisions that may escalate environmental waste through plastic packaging and single-use products. In this study, we analyzed videos on TikTok to assess the environmental impact of trending skin care routines. By examining the types of products promoted, their packaging, and the frequency with which they appear in viral content, we aimed to investigate how these trends, which may be imitated by users, impact the environment.

A search of TikTok videos using #skincareroutine was conducted on June 21, 2024. Sponsored content, non–English language videos, videos without demonstrated skin care routines, and videos showing makeup routines were excluded from our analysis. Data collected from each video included username, date posted, number of likes, total number of skin care products used, number of single-use skin care products used, average amount of product used, number of skin care applicators used, and number of single-use applicators used. Single-use items, defined as those intended for one-time use and subsequent disposal, were identified visually by packaging, manufacturer intent, and common consumer usage patterns. The amount of product used per application was graded on a scale of 1 to 3 (1=pea-sized amount or less; 2=single full pump/spray; 3=multiple pumps/sprays). Videos were categorized as personal (ie, skin care routine walk-throughs by the creator) or autonomous sensory meridian response (ASMR)(focused on product sounds and aesthetics).3 A Mann-Whitney U test was utilized to statistically compare the 2 groups. Statistical analysis was performed using Microsoft Excel (α=0.05). 

A total of 50 videos met the inclusion criteria and were included in the analysis. The average number of likes per video was 499,696.15, with skin care routines featuring an average of 6.4 unique products (Table). There was a weak positive correlation (r=0.1809) between the number of skin care products used and the number of likes. A total of 320 products were used across the videos, 23 of which were single-use (7.2%).On average, single-use skin care items were used 0.46 times per routine, comprising a mean 7.99% of total products per video. The average score for the amount of product used per application was 2.18. There was no difference in personal vs ASMR videos with regard to the total number of skin care products used or the average amount of product used per application (P>.05). Thirty-three (70.2%) of the 47 applicators used across all videos were single-use. An average of 0.94 applicators per routine were utilized, with a mean 68.83% being single-use applicators. Common single-use products were toner wipes and eye patches, and single-use applicators included cotton pads and plastic spatulas. 

CT116003107-Table

Our findings indicated a prevalence of multiple products and large amount of product used in trending skin care routines, suggesting a shift toward multistep skin care. This implies a high rate of product consumption that may accelerate the carbon footprint associated with skin care products,3 which could contribute to climate change and environmental degradation. Consumers also may feel compelled to purchase and discard numerous partially used products in order to keep up with the latest trends, exacerbating the environmental impact. Furthermore, the utilization of single-use products and applicators contributes to increased plastic waste, pollution, and resource depletion. Single-use items often are difficult to recycle due to their mixed materials and small size,4,5 and therefore they can accumulate in landfills and oceans. This impact can be mitigated by switching to reusable applicators, refillable packaging, and biodegradable materials. 

The substantial average number of likes per video indicates high engagement with skin care content among TikTok users. The continued popularity of complex multi­step skin care routines, despite a weak correlation between the number of skin care products used and the number of likes per video, likely stems from factors such as aesthetic appeal, ASMR effects, and creators’ established followings, which may drive user engagement to contribute to unsustainable consumption patterns. Factors such as presentation style, aesthetics, or creators’ pre-existing online following may have a major impact on how well a video performs on TikTok. The similarity between personal and ASMR videos, particularly in the number of products used and the amount applied, suggests that both formats employ common approaches to meet audience expectations and align with promotional trends, relying more on sensory and aesthetic strategies than substantive differences in skin care routines.

Our use of only one tag in our search as well as the subjective quantity scale limits the generalizability of these findings to broader TikTok skin care content.

Overall, our study underscores the role of brands and social media influencers in skin care education and promotion of sustainable practices. The extensive number of products used and generous application of each product in skin care routines demonstrated in TikTok videos may mislead viewers into believing that using more product improves outcomes, when often, less is more. We recommend that dermatologists counsel patients about informed skin care regimens that prioritize individual needs over social media fads.

References
  1. Pagani K, Lukac D, Martinez R, et al. Slugging: TikTokTM as a source of a viral “harmless” beauty trend. Clin Dermatol. 2022;40:810-812. doi:10.1016/j.clindermatol.2022.08.005
  2. Stern C. TikTok drives $31.7B in beauty sales: how viral trends are shaping the future of cosmetics. CosmeticsDesign. August 20, 2024. Accessed June 24, 2025. https://www.cosmeticsdesign.com/Article/2024/08/20/tiktok-drives-31.7b-in-beauty-sales-how-viral-trends-are-shaping-the-future-of-cosmetics/
  3. Fountain C. ASMR content saw huge growth on YouTube, but now creators are flocking to TikTok instead. Business Insider. July 4, 2022. Accessed June 24, 2025. https://www.businessinsider.com/asmr-tiktok-instead-of-youtube-growth-subscribers-2022-7
  4. Rathore S, Schuler B, Park J. Life cycle assessment of multiple dispensing systems used for cosmetic product packaging. Packaging Technol Sci. 2023;36:533-547. doi:10.1002/pts.2729
  5. Shaw S. How to actually recycle your empty beauty products. CNN Underscored. Updated April 17, 2024. Accessed June 24, 2025. https://www.cnn.com/cnn-underscored/beauty/how-to-recycle-beauty-products
References
  1. Pagani K, Lukac D, Martinez R, et al. Slugging: TikTokTM as a source of a viral “harmless” beauty trend. Clin Dermatol. 2022;40:810-812. doi:10.1016/j.clindermatol.2022.08.005
  2. Stern C. TikTok drives $31.7B in beauty sales: how viral trends are shaping the future of cosmetics. CosmeticsDesign. August 20, 2024. Accessed June 24, 2025. https://www.cosmeticsdesign.com/Article/2024/08/20/tiktok-drives-31.7b-in-beauty-sales-how-viral-trends-are-shaping-the-future-of-cosmetics/
  3. Fountain C. ASMR content saw huge growth on YouTube, but now creators are flocking to TikTok instead. Business Insider. July 4, 2022. Accessed June 24, 2025. https://www.businessinsider.com/asmr-tiktok-instead-of-youtube-growth-subscribers-2022-7
  4. Rathore S, Schuler B, Park J. Life cycle assessment of multiple dispensing systems used for cosmetic product packaging. Packaging Technol Sci. 2023;36:533-547. doi:10.1002/pts.2729
  5. Shaw S. How to actually recycle your empty beauty products. CNN Underscored. Updated April 17, 2024. Accessed June 24, 2025. https://www.cnn.com/cnn-underscored/beauty/how-to-recycle-beauty-products
Issue
Cutis - 116(3)
Issue
Cutis - 116(3)
Page Number
107-108
Page Number
107-108
Publications
Publications
Topics
Article Type
Display Headline

A Cross-Sectional Analysis of TikTok Skin Care Routines and the Associated Environmental Impact

Display Headline

A Cross-Sectional Analysis of TikTok Skin Care Routines and the Associated Environmental Impact

Sections
Inside the Article

PRACTICE POINTS

  • Social media platforms are increasingly influential in shaping consumer skin care habits, particularly among younger demographics.
  • Dermatologists should be aware of the aestheticdriven nature of online skin care trends when advising patients on product use.
  • Viral skin care routines often feature multiple products and applicators, potentially encouraging excessive product use and waste.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Thu, 09/04/2025 - 18:18
Un-Gate On Date
Thu, 09/04/2025 - 18:18
Use ProPublica
CFC Schedule Remove Status
Thu, 09/04/2025 - 18:18
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
survey writer start date
Thu, 09/04/2025 - 18:18

Demographic and Clinical Factors Associated With PD-L1 Testing of Veterans With Advanced Non-Small Cell Lung Cancer

Article Type
Changed
Thu, 09/04/2025 - 16:12

Background

Programmed death-ligand 1 (PD-L1) checkpoint inhibitors revolutionized the treatment of advanced non-small cell lung cancer (aNSCLC) by improving overall survival compared to chemotherapy. PD-L1 biomarker testing is paramount for informing treatment decisions in aNSCLC. Real-world data describing patterns of PD-L1 testing within the Veteran Health Administration (VHA) are limited. This retrospective study seeks to evaluate demographic and clinical factors associated with PD-L1 testing in VHA.

Methods

Veterans diagnosed with aNSCLC from 2019-2022 were identified using VHA’s Corporate Data Warehouse. Wilcoxon Rank Sum and Chi- Square tests measured association between receipt of PD-L1 testing and patient demographic and clinical characteristics at aNSCLC diagnosis. Logistic regression assessed predictors of PD-L1 testing, and subgroup analyses were performed for significant interactions.

Results

Our study included 4575 patients with aNSCLC; 57.0% received PD-L1 testing. The likelihood of PD-L1 testing increased among patients diagnosed with aNSCLC after 2019 vs during 2019 (OR≥1.118, p≤0.035) and in Black vs White patients (OR=1.227, p=0.011). However, the following had decreased likelihood of PD-L1 testing: patients with stage IIIB vs IV cancer (OR=0.683, p=0.004); non vs current/former smokers (OR=0.733, p=0.039); squamous (OR=0.863, p=0.030) or NOS (OR=0.695,p=0.013) vs. adenocarcinoma histology. Interactions were observed between patient residential region and residential rurality (p=0.003), and region and receipt of oncology community care consults (OCCC) (p=0.030). Patients in rural Midwest (OR=0.445,p=0.004) and rural South (OR=0.566, p=0.032) were less likely to receive PD-L1 testing than Metropolitan patients. Across patients with OCCC, Western US patients were more likely to receive PD-L1 testing (OR=1.554, p=0.001) than patients in other regions. However, within Midwestern patients, those without a OCCC were more likely to receive PD-L1 testing (OR=1.724, p< 0.001) than those with a OCCC. High comorbidity index (CCI≥3) is associated with an increased likelihood of PD-L1 testing in a univariable model (OR=1.286 vs. CCI=0,p=0.009), but not in the multivariable model (p=0.278).

Conclusions

We identified demographic and clinical factors, including regional differences in rurality and OCCC patterns, associated with PD-L1 testing. These factors can focus ongoing efforts to improve PD-L1 testing and efforts to be more in line with recommended care.

Issue
Federal Practitioner - 42(9)s
Publications
Topics
Page Number
S37-S38
Sections

Background

Programmed death-ligand 1 (PD-L1) checkpoint inhibitors revolutionized the treatment of advanced non-small cell lung cancer (aNSCLC) by improving overall survival compared to chemotherapy. PD-L1 biomarker testing is paramount for informing treatment decisions in aNSCLC. Real-world data describing patterns of PD-L1 testing within the Veteran Health Administration (VHA) are limited. This retrospective study seeks to evaluate demographic and clinical factors associated with PD-L1 testing in VHA.

Methods

Veterans diagnosed with aNSCLC from 2019-2022 were identified using VHA’s Corporate Data Warehouse. Wilcoxon Rank Sum and Chi- Square tests measured association between receipt of PD-L1 testing and patient demographic and clinical characteristics at aNSCLC diagnosis. Logistic regression assessed predictors of PD-L1 testing, and subgroup analyses were performed for significant interactions.

Results

Our study included 4575 patients with aNSCLC; 57.0% received PD-L1 testing. The likelihood of PD-L1 testing increased among patients diagnosed with aNSCLC after 2019 vs during 2019 (OR≥1.118, p≤0.035) and in Black vs White patients (OR=1.227, p=0.011). However, the following had decreased likelihood of PD-L1 testing: patients with stage IIIB vs IV cancer (OR=0.683, p=0.004); non vs current/former smokers (OR=0.733, p=0.039); squamous (OR=0.863, p=0.030) or NOS (OR=0.695,p=0.013) vs. adenocarcinoma histology. Interactions were observed between patient residential region and residential rurality (p=0.003), and region and receipt of oncology community care consults (OCCC) (p=0.030). Patients in rural Midwest (OR=0.445,p=0.004) and rural South (OR=0.566, p=0.032) were less likely to receive PD-L1 testing than Metropolitan patients. Across patients with OCCC, Western US patients were more likely to receive PD-L1 testing (OR=1.554, p=0.001) than patients in other regions. However, within Midwestern patients, those without a OCCC were more likely to receive PD-L1 testing (OR=1.724, p< 0.001) than those with a OCCC. High comorbidity index (CCI≥3) is associated with an increased likelihood of PD-L1 testing in a univariable model (OR=1.286 vs. CCI=0,p=0.009), but not in the multivariable model (p=0.278).

Conclusions

We identified demographic and clinical factors, including regional differences in rurality and OCCC patterns, associated with PD-L1 testing. These factors can focus ongoing efforts to improve PD-L1 testing and efforts to be more in line with recommended care.

Background

Programmed death-ligand 1 (PD-L1) checkpoint inhibitors revolutionized the treatment of advanced non-small cell lung cancer (aNSCLC) by improving overall survival compared to chemotherapy. PD-L1 biomarker testing is paramount for informing treatment decisions in aNSCLC. Real-world data describing patterns of PD-L1 testing within the Veteran Health Administration (VHA) are limited. This retrospective study seeks to evaluate demographic and clinical factors associated with PD-L1 testing in VHA.

Methods

Veterans diagnosed with aNSCLC from 2019-2022 were identified using VHA’s Corporate Data Warehouse. Wilcoxon Rank Sum and Chi- Square tests measured association between receipt of PD-L1 testing and patient demographic and clinical characteristics at aNSCLC diagnosis. Logistic regression assessed predictors of PD-L1 testing, and subgroup analyses were performed for significant interactions.

Results

Our study included 4575 patients with aNSCLC; 57.0% received PD-L1 testing. The likelihood of PD-L1 testing increased among patients diagnosed with aNSCLC after 2019 vs during 2019 (OR≥1.118, p≤0.035) and in Black vs White patients (OR=1.227, p=0.011). However, the following had decreased likelihood of PD-L1 testing: patients with stage IIIB vs IV cancer (OR=0.683, p=0.004); non vs current/former smokers (OR=0.733, p=0.039); squamous (OR=0.863, p=0.030) or NOS (OR=0.695,p=0.013) vs. adenocarcinoma histology. Interactions were observed between patient residential region and residential rurality (p=0.003), and region and receipt of oncology community care consults (OCCC) (p=0.030). Patients in rural Midwest (OR=0.445,p=0.004) and rural South (OR=0.566, p=0.032) were less likely to receive PD-L1 testing than Metropolitan patients. Across patients with OCCC, Western US patients were more likely to receive PD-L1 testing (OR=1.554, p=0.001) than patients in other regions. However, within Midwestern patients, those without a OCCC were more likely to receive PD-L1 testing (OR=1.724, p< 0.001) than those with a OCCC. High comorbidity index (CCI≥3) is associated with an increased likelihood of PD-L1 testing in a univariable model (OR=1.286 vs. CCI=0,p=0.009), but not in the multivariable model (p=0.278).

Conclusions

We identified demographic and clinical factors, including regional differences in rurality and OCCC patterns, associated with PD-L1 testing. These factors can focus ongoing efforts to improve PD-L1 testing and efforts to be more in line with recommended care.

Issue
Federal Practitioner - 42(9)s
Issue
Federal Practitioner - 42(9)s
Page Number
S37-S38
Page Number
S37-S38
Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Eyebrow Default
Research
Gate On Date
Thu, 09/04/2025 - 15:31
Un-Gate On Date
Thu, 09/04/2025 - 15:31
Use ProPublica
CFC Schedule Remove Status
Thu, 09/04/2025 - 15:31
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
survey writer start date
Thu, 09/04/2025 - 15:31

Survival Outcomes of Skin Adnexal Tumors: A National Cancer Database Analysis

Article Type
Changed
Thu, 09/04/2025 - 16:11

Purpose

Skin adnexal tumors (SAT) include a group of benign and malignant appendageal tumors that arise from hair follicles, sebaceous glands, or sweat glands. They typically appear as small, painless bumps or nodules on the skin, and are more common in men compared to women. The 5-year overall SAT survival rate ranges from 74-90%. To better understand the differences in survival outcomes based on subtypes of SAT, the National Cancer Database (NCDB) was analyzed.

Methods

A retrospective cohort study of 11,627 patients with histologically confirmed SAT between 2004 and 2021 was conducted across 1,500 Commission on Cancer facilities located in the US and Puerto Rico. Demographic factors such as sex, age, and race were analyzed using Pearson Chi-squared tests, and survival outcomes were analyzed by Kaplan- Meier survival analysis. P value < 0.05 was considered statistically significant.

Results

Most patients with SAT were male (57.3%). The average age at diagnosis was 65.9 (SD=14.4, range 0-90). Of the patient sample, 87.2% were White, 7.6% Black, 2.5% Asian, and 2.7% other. Several subtypes disproportionately affected Black individuals, including apocrine adenocarcinoma (15.7%) and hidradenocarcinoma (13.6%). The estimated 5-year survival of SAT was 74.9% with an overall survival of 135.8 months (SE=1.1). Sebaceous carcinoma (which accounts for 41.8% of all cases) had the lowest average survival time of 119.6 months (SE=1.8), while digital papillary adenocarcinoma had the highest survival at around 183.5 months (SE=4.6).

Conclusions

This study supports a higher frequency of SAT among men. While White patients were more likely to get SAT overall, including the most common sebaceous carcinoma, Black race were associated with higher frequency of rarer subtypes. The average age of diagnosis of SAT mimics other non-melanoma skin cancers, but has a lower overall survival rate. Future studies should consider other risk factors that may be impacting the differences in survival outcomes to guide treatment and address health disparities among the various subtypes.

Issue
Federal Practitioner - 42(9)s
Publications
Topics
Page Number
S37
Sections

Purpose

Skin adnexal tumors (SAT) include a group of benign and malignant appendageal tumors that arise from hair follicles, sebaceous glands, or sweat glands. They typically appear as small, painless bumps or nodules on the skin, and are more common in men compared to women. The 5-year overall SAT survival rate ranges from 74-90%. To better understand the differences in survival outcomes based on subtypes of SAT, the National Cancer Database (NCDB) was analyzed.

Methods

A retrospective cohort study of 11,627 patients with histologically confirmed SAT between 2004 and 2021 was conducted across 1,500 Commission on Cancer facilities located in the US and Puerto Rico. Demographic factors such as sex, age, and race were analyzed using Pearson Chi-squared tests, and survival outcomes were analyzed by Kaplan- Meier survival analysis. P value < 0.05 was considered statistically significant.

Results

Most patients with SAT were male (57.3%). The average age at diagnosis was 65.9 (SD=14.4, range 0-90). Of the patient sample, 87.2% were White, 7.6% Black, 2.5% Asian, and 2.7% other. Several subtypes disproportionately affected Black individuals, including apocrine adenocarcinoma (15.7%) and hidradenocarcinoma (13.6%). The estimated 5-year survival of SAT was 74.9% with an overall survival of 135.8 months (SE=1.1). Sebaceous carcinoma (which accounts for 41.8% of all cases) had the lowest average survival time of 119.6 months (SE=1.8), while digital papillary adenocarcinoma had the highest survival at around 183.5 months (SE=4.6).

Conclusions

This study supports a higher frequency of SAT among men. While White patients were more likely to get SAT overall, including the most common sebaceous carcinoma, Black race were associated with higher frequency of rarer subtypes. The average age of diagnosis of SAT mimics other non-melanoma skin cancers, but has a lower overall survival rate. Future studies should consider other risk factors that may be impacting the differences in survival outcomes to guide treatment and address health disparities among the various subtypes.

Purpose

Skin adnexal tumors (SAT) include a group of benign and malignant appendageal tumors that arise from hair follicles, sebaceous glands, or sweat glands. They typically appear as small, painless bumps or nodules on the skin, and are more common in men compared to women. The 5-year overall SAT survival rate ranges from 74-90%. To better understand the differences in survival outcomes based on subtypes of SAT, the National Cancer Database (NCDB) was analyzed.

Methods

A retrospective cohort study of 11,627 patients with histologically confirmed SAT between 2004 and 2021 was conducted across 1,500 Commission on Cancer facilities located in the US and Puerto Rico. Demographic factors such as sex, age, and race were analyzed using Pearson Chi-squared tests, and survival outcomes were analyzed by Kaplan- Meier survival analysis. P value < 0.05 was considered statistically significant.

Results

Most patients with SAT were male (57.3%). The average age at diagnosis was 65.9 (SD=14.4, range 0-90). Of the patient sample, 87.2% were White, 7.6% Black, 2.5% Asian, and 2.7% other. Several subtypes disproportionately affected Black individuals, including apocrine adenocarcinoma (15.7%) and hidradenocarcinoma (13.6%). The estimated 5-year survival of SAT was 74.9% with an overall survival of 135.8 months (SE=1.1). Sebaceous carcinoma (which accounts for 41.8% of all cases) had the lowest average survival time of 119.6 months (SE=1.8), while digital papillary adenocarcinoma had the highest survival at around 183.5 months (SE=4.6).

Conclusions

This study supports a higher frequency of SAT among men. While White patients were more likely to get SAT overall, including the most common sebaceous carcinoma, Black race were associated with higher frequency of rarer subtypes. The average age of diagnosis of SAT mimics other non-melanoma skin cancers, but has a lower overall survival rate. Future studies should consider other risk factors that may be impacting the differences in survival outcomes to guide treatment and address health disparities among the various subtypes.

Issue
Federal Practitioner - 42(9)s
Issue
Federal Practitioner - 42(9)s
Page Number
S37
Page Number
S37
Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Eyebrow Default
Research
Gate On Date
Thu, 09/04/2025 - 15:09
Un-Gate On Date
Thu, 09/04/2025 - 15:09
Use ProPublica
CFC Schedule Remove Status
Thu, 09/04/2025 - 15:09
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
survey writer start date
Thu, 09/04/2025 - 15:09

Timeliness of Specialty Palliative Care for Veterans With Cancer: An Analysis of Administrative Data

Article Type
Changed
Thu, 09/04/2025 - 16:09

Background

Studies show that early referral to Specialty Palliative Care (SPC) can improve patient- reported outcomes among Veterans with cancer; quality metrics include referral within 8 weeks of an advanced cancer diagnosis. In this study, we explored timeliness of specialty referrals and compared various factors.

Methods

We identified our cohort using Department of Veterans Affairs (VA) Corporate Data Warehouse (CDW). Eligibility criteria included active or history of cancer—using a peer-reviewed, in-house list of ICD-9 and ICD-10 codes—between 2013-2023. We stratified our cohort of Veterans using factors including cancer stage, rurality, and care assessment needs (CAN) scores. We performed survival analyses to look at time to SPC from initial diagnosis and peak CAN score. Predictors of utilization were evaluated using multinomial regression and Cox proportional hazards models through R.

Results

Using CDW’s oncology domain, we identified 475,775 Veterans. 28% received SPC. Most received it near the end of their life as evidenced by the mortality rates (79.5%) in the early period following SPC consultation. Median time to SPC was 515 days. There was a significant difference in utilization rates between urban and rural Veterans (Wilcoxon W-statistic = 2.31E+10, p < 0.001). Peak CAN scores ranged from 0 to 0.81, median peak of 0.057 and interquartile range of 0.1. Multinomial regression model indicated statistically significant associations of advanced cancer (Stages 3 and 4) with timing of SPC. Stage 4 cancer showed the strongest association with receipt of palliative care within 60 days of initial diagnosis (OR 4.8, 95% CI: 4.69-4.93, p < 0.001), suggesting higher stage disease increases the likelihood of palliative care referral and accelerates the timing of these referrals.

Conclusions

We found Veterans received SPC from a broad range of peak CAN scores (0 to 0.81), suggesting that absolute CAN scores may not be clinically actionable indicators but perhaps indicative of changes in condition warranting referral. Stage IV cancer at diagnosis was associated with early SPC. The significant differences in utilization rates between urban and rural patients highlight potential access barriers that should be addressed.

Issue
Federal Practitioner - 42(9)s
Publications
Topics
Page Number
S36-S37
Sections

Background

Studies show that early referral to Specialty Palliative Care (SPC) can improve patient- reported outcomes among Veterans with cancer; quality metrics include referral within 8 weeks of an advanced cancer diagnosis. In this study, we explored timeliness of specialty referrals and compared various factors.

Methods

We identified our cohort using Department of Veterans Affairs (VA) Corporate Data Warehouse (CDW). Eligibility criteria included active or history of cancer—using a peer-reviewed, in-house list of ICD-9 and ICD-10 codes—between 2013-2023. We stratified our cohort of Veterans using factors including cancer stage, rurality, and care assessment needs (CAN) scores. We performed survival analyses to look at time to SPC from initial diagnosis and peak CAN score. Predictors of utilization were evaluated using multinomial regression and Cox proportional hazards models through R.

Results

Using CDW’s oncology domain, we identified 475,775 Veterans. 28% received SPC. Most received it near the end of their life as evidenced by the mortality rates (79.5%) in the early period following SPC consultation. Median time to SPC was 515 days. There was a significant difference in utilization rates between urban and rural Veterans (Wilcoxon W-statistic = 2.31E+10, p < 0.001). Peak CAN scores ranged from 0 to 0.81, median peak of 0.057 and interquartile range of 0.1. Multinomial regression model indicated statistically significant associations of advanced cancer (Stages 3 and 4) with timing of SPC. Stage 4 cancer showed the strongest association with receipt of palliative care within 60 days of initial diagnosis (OR 4.8, 95% CI: 4.69-4.93, p < 0.001), suggesting higher stage disease increases the likelihood of palliative care referral and accelerates the timing of these referrals.

Conclusions

We found Veterans received SPC from a broad range of peak CAN scores (0 to 0.81), suggesting that absolute CAN scores may not be clinically actionable indicators but perhaps indicative of changes in condition warranting referral. Stage IV cancer at diagnosis was associated with early SPC. The significant differences in utilization rates between urban and rural patients highlight potential access barriers that should be addressed.

Background

Studies show that early referral to Specialty Palliative Care (SPC) can improve patient- reported outcomes among Veterans with cancer; quality metrics include referral within 8 weeks of an advanced cancer diagnosis. In this study, we explored timeliness of specialty referrals and compared various factors.

Methods

We identified our cohort using Department of Veterans Affairs (VA) Corporate Data Warehouse (CDW). Eligibility criteria included active or history of cancer—using a peer-reviewed, in-house list of ICD-9 and ICD-10 codes—between 2013-2023. We stratified our cohort of Veterans using factors including cancer stage, rurality, and care assessment needs (CAN) scores. We performed survival analyses to look at time to SPC from initial diagnosis and peak CAN score. Predictors of utilization were evaluated using multinomial regression and Cox proportional hazards models through R.

Results

Using CDW’s oncology domain, we identified 475,775 Veterans. 28% received SPC. Most received it near the end of their life as evidenced by the mortality rates (79.5%) in the early period following SPC consultation. Median time to SPC was 515 days. There was a significant difference in utilization rates between urban and rural Veterans (Wilcoxon W-statistic = 2.31E+10, p < 0.001). Peak CAN scores ranged from 0 to 0.81, median peak of 0.057 and interquartile range of 0.1. Multinomial regression model indicated statistically significant associations of advanced cancer (Stages 3 and 4) with timing of SPC. Stage 4 cancer showed the strongest association with receipt of palliative care within 60 days of initial diagnosis (OR 4.8, 95% CI: 4.69-4.93, p < 0.001), suggesting higher stage disease increases the likelihood of palliative care referral and accelerates the timing of these referrals.

Conclusions

We found Veterans received SPC from a broad range of peak CAN scores (0 to 0.81), suggesting that absolute CAN scores may not be clinically actionable indicators but perhaps indicative of changes in condition warranting referral. Stage IV cancer at diagnosis was associated with early SPC. The significant differences in utilization rates between urban and rural patients highlight potential access barriers that should be addressed.

Issue
Federal Practitioner - 42(9)s
Issue
Federal Practitioner - 42(9)s
Page Number
S36-S37
Page Number
S36-S37
Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Eyebrow Default
Research
Gate On Date
Thu, 09/04/2025 - 14:57
Un-Gate On Date
Thu, 09/04/2025 - 14:57
Use ProPublica
CFC Schedule Remove Status
Thu, 09/04/2025 - 14:57
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
survey writer start date
Thu, 09/04/2025 - 14:57

Uncovering Food Insecurity in Veterans with Cancer Distress

Article Type
Changed
Thu, 09/04/2025 - 16:08

Background

To close the food insecurity gap by providing food assistance and increasing opportunities for screening in Veterans receiving cancer treatment at a VA outpatient cancer clinic. Food Insecurity is associated with chronic disease such as cancer given insufficient access to nutritious foods leading to nutritional deficiencies and worsening health outcomes. The rates of food insecurity among Veterans revealed 28% of female veterans and 16% overall in male Veterans were faced with limited or uncertain access to adequate food.

Methods

A pivotal distress screening occurs at time of education consult or cycle 1 day 1 of antineoplastic therapy. A positive screening for any practical concern generates a discussion about food insecurity. A positive distress screen triggers an oncology social work referral to complete a systematic screening assessing circumstances and offering resources for needs (ACORN).

Results

Root cause analysis uncovered 24% of Veterans with cancer screened positive for food insecurity in the 9E oncology outpatient clinic. Post-implementation of robust screenings and conversation initiatives identified 36 unique Veterans who received 251 meals from July to December 2024.

Sustainability/Scalability

Prospective screening of Veterans at the time of a cancer diagnosis and ongoing screening during cancer treatment is the first step toward uncovering food insecurity and addressing this social determinate of health. A standard operating procedure following VA guidance and distress management guidelines should be updated as required. Oversight of the cancer leadership team annually evaluates the distress process, and the findings are reported to the cancer committee.

Conclusions

Uncovering food insecurity in Veterans at time of diagnosis and during cancer treatment is critical to optimize treatment outcomes. A systematic and robust screening standard operating procedure is key to implement. Veterans are a unique population with a spectrum of socioeconomic needs. Case management conferences or weekly huddles to discuss the Veteran’s needs will ensure food insecurity is addressed. Collection and analysis of screening data will highlight a program’s food insecurity need and supports community partnerships to available food resources and the opportunity to create a cancer outpatient clinic food hub for Veterans receiving cancer treatment.

Issue
Federal Practitioner - 42(9)s
Publications
Topics
Page Number
S36
Sections

Background

To close the food insecurity gap by providing food assistance and increasing opportunities for screening in Veterans receiving cancer treatment at a VA outpatient cancer clinic. Food Insecurity is associated with chronic disease such as cancer given insufficient access to nutritious foods leading to nutritional deficiencies and worsening health outcomes. The rates of food insecurity among Veterans revealed 28% of female veterans and 16% overall in male Veterans were faced with limited or uncertain access to adequate food.

Methods

A pivotal distress screening occurs at time of education consult or cycle 1 day 1 of antineoplastic therapy. A positive screening for any practical concern generates a discussion about food insecurity. A positive distress screen triggers an oncology social work referral to complete a systematic screening assessing circumstances and offering resources for needs (ACORN).

Results

Root cause analysis uncovered 24% of Veterans with cancer screened positive for food insecurity in the 9E oncology outpatient clinic. Post-implementation of robust screenings and conversation initiatives identified 36 unique Veterans who received 251 meals from July to December 2024.

Sustainability/Scalability

Prospective screening of Veterans at the time of a cancer diagnosis and ongoing screening during cancer treatment is the first step toward uncovering food insecurity and addressing this social determinate of health. A standard operating procedure following VA guidance and distress management guidelines should be updated as required. Oversight of the cancer leadership team annually evaluates the distress process, and the findings are reported to the cancer committee.

Conclusions

Uncovering food insecurity in Veterans at time of diagnosis and during cancer treatment is critical to optimize treatment outcomes. A systematic and robust screening standard operating procedure is key to implement. Veterans are a unique population with a spectrum of socioeconomic needs. Case management conferences or weekly huddles to discuss the Veteran’s needs will ensure food insecurity is addressed. Collection and analysis of screening data will highlight a program’s food insecurity need and supports community partnerships to available food resources and the opportunity to create a cancer outpatient clinic food hub for Veterans receiving cancer treatment.

Background

To close the food insecurity gap by providing food assistance and increasing opportunities for screening in Veterans receiving cancer treatment at a VA outpatient cancer clinic. Food Insecurity is associated with chronic disease such as cancer given insufficient access to nutritious foods leading to nutritional deficiencies and worsening health outcomes. The rates of food insecurity among Veterans revealed 28% of female veterans and 16% overall in male Veterans were faced with limited or uncertain access to adequate food.

Methods

A pivotal distress screening occurs at time of education consult or cycle 1 day 1 of antineoplastic therapy. A positive screening for any practical concern generates a discussion about food insecurity. A positive distress screen triggers an oncology social work referral to complete a systematic screening assessing circumstances and offering resources for needs (ACORN).

Results

Root cause analysis uncovered 24% of Veterans with cancer screened positive for food insecurity in the 9E oncology outpatient clinic. Post-implementation of robust screenings and conversation initiatives identified 36 unique Veterans who received 251 meals from July to December 2024.

Sustainability/Scalability

Prospective screening of Veterans at the time of a cancer diagnosis and ongoing screening during cancer treatment is the first step toward uncovering food insecurity and addressing this social determinate of health. A standard operating procedure following VA guidance and distress management guidelines should be updated as required. Oversight of the cancer leadership team annually evaluates the distress process, and the findings are reported to the cancer committee.

Conclusions

Uncovering food insecurity in Veterans at time of diagnosis and during cancer treatment is critical to optimize treatment outcomes. A systematic and robust screening standard operating procedure is key to implement. Veterans are a unique population with a spectrum of socioeconomic needs. Case management conferences or weekly huddles to discuss the Veteran’s needs will ensure food insecurity is addressed. Collection and analysis of screening data will highlight a program’s food insecurity need and supports community partnerships to available food resources and the opportunity to create a cancer outpatient clinic food hub for Veterans receiving cancer treatment.

Issue
Federal Practitioner - 42(9)s
Issue
Federal Practitioner - 42(9)s
Page Number
S36
Page Number
S36
Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Eyebrow Default
Quality Improvement
Gate On Date
Thu, 09/04/2025 - 13:51
Un-Gate On Date
Thu, 09/04/2025 - 13:51
Use ProPublica
CFC Schedule Remove Status
Thu, 09/04/2025 - 13:51
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
survey writer start date
Thu, 09/04/2025 - 13:51