User login
2023 Update on genetics in fetal growth
Whole exome sequencing’s role in diagnosing genetic causes of FGR with and without associated anomalies
Mone F, Mellis R, Gabriel H, et al. Should we offer prenatal exome sequencing for intrauterine growth restriction or short long bones? A systematic review and meta-analysis. Am J Obstet Gynecol. Published online October 7, 2022. doi:10.1016/j.ajog.2022.09.045
Multiple factors can play a role in FGR, including inherent maternal, placental, or fetal factors; the environment; and/or nutrition. However, prenatal diagnosis is an important consideration when exploring the underlying etiology for a growth-restricted fetus, especially in severe or early-onset cases. Many genetic conditions do not result in structural anomalies but can disrupt overall growth. Additionally, phenotyping in the prenatal period is limited and can miss more subtle physical differences that could point to a genetic cause.
When compared with karyotype, chromosomal microarray (CMA) has been shown to increase the diagnostic yield in cases of isolated early FGR by 5%,1,2 and the incidence of chromosomal abnormalities has been reported to be as high as 19% in this population. Let’s explore the data on exome sequencing for prenatal diagnosis in cases of isolated FGR.
Meta-analysis details
In this meta-analysis, the authors reviewed 19 cohort studies or case series that investigated the yield of prenatal sequencing in fetuses with intrauterine growth restriction (IUGR) or short long bones, both in association with and without additional anomalies. All cases had nondiagnostic cytogenetic results. Fetal DNA in most cases was obtained through amniocentesis. Variants classified as likely pathogenic and pathogenic were considered diagnostic. The authors then calculated the incremental yield of prenatal sequencing over cytogenetic studies as a pooled value, comparing the following groups:
- isolated FGR
- growth restriction with associated anomalies
- isolated short long bones
- short long bones with additional skeletal features.
Study outcomes
The total number of cases were as follows: isolated IUGR (n = 71), IUGR associated with additional anomalies (n = 45), isolated short long bones (n = 84), and short long bones associated with additional skeletal findings (n = 252). Causative pathogenic or likely pathogenic variants were identified in 224 (50%) cases. Apparent incremental yields with prenatal sequencing were as follows for the 4 groups (as illustrated in the FIGURE):
- 4% in isolated IUGR (95% confidence interval [CI], -5%–12%)
- 30% in IUGR with additional anomalies (95% CI, 13%–47%)
- 48% in isolated short long bones (95% CI, 26%–70%)
- 68% in short long bones with additional skeletal changes (95% CI, 58%–77%).
Overall, the authors concluded that prenatal sequencing does not improve prenatal diagnosis in cases of isolated IUGR. The majority of these cases were thought to be related to placental insufficiency.
Strengths and limitations
The main limitation of this study with regard to our discussion is the small study populationof isolated growth restriction. The authors indicate that the number of cases of isolated IUGR were too small to draw firm conclusions. Another limitation was the heterogeneity of the isolated FGR population, which was not limited to severe or early-onset cases. However, the authors did demonstrate that growth restriction in association with fetal anomalies has very high genetic yield rates with prenatal sequencing.
Not surprisingly, there is a high yield of diagnosing genetic conditions in pregnancies complicated by isolated or nonisolated short long bones or in cases of growth restriction with multisystem abnormalities. Based on the results of this study, the authors advise against sending for exome sequencing in cases of isolated growth restriction with coexisting evidence of placental insufficiency.
Continue to: Can whole exome sequencing diagnose genetic causes in cases of severe isolated FGR?...
Can whole exome sequencing diagnose genetic causes in cases of severe isolated FGR?
Zhou H, Fu F, Wang Y, et al. Genetic causes of isolated and severe fetal growth restriction in normal chromosomal microarray analysis. Int J Gynaecol Obstet. Published online December 10, 2022. doi:10.1002/ijgo.14620
Severe FGR is diagnosed based on an estimated fetal weight (EFW) or abdominal circumference (AC) below the third percentile. As we discussed in the above study by Mone and colleagues, it does not appear that prenatal sequencing significantly improves the diagnostic yield in all isolated FGR cases. However, this has not been previously explored in isolated severe FGR or cases of early-onset FGR (<32 weeks’ gestation). We know that several monogenic conditions are associated with severe and early-onset isolated fetal growth impairment, including but not limited to Cornelia de Lange syndrome, Smith-Lemli-Opitz syndrome, and Meier-Gorlin syndrome. Often, these syndromes can present in the prenatal period without other phenotypic findings. Therefore, this study explored the possibility that prenatal sequencing plays an important role for severe cases of FGR with nondiagnostic CMA and/or karyotype.
Retrospective study details
Zhou and colleagues retrospectively analyzed 51 cases of severe (EFW or AC below the third percentile) isolated FGR with negative CMA who underwent trio whole exome sequencing, which includes submitting fetal cells as well as both parental samples for testing. Patients with abnormal toxoplasmosis, rubella, cytomegalovirus, and herpes simplex virus (TORCH) tests; structural anomalies; and multiple gestation were excluded from the analysis. As in the study by Mone et al, variants classified as likely pathogenic and pathogenic were categorized as diagnostic.
Results
Eight of 51 cases (15.7%) with severe isolated FGR had diagnostic findings on trio whole exome sequencing as shown in the TABLE. Another 8 cases (15.7%) were found to have variants of unknown significance, of which 2 were later determined to be novel pathogenic variants. Genetic conditions uncovered in this cohort include Cornelia de Lange syndrome, pyruvate dehydrogenase deficiency, Dent disease, trichohepaticenteric syndrome, achondroplasia, osteogenesis imperfecta, Pendred syndrome, and both autosomal dominant type 3A and autosomal recessive type 1A deafness. All 10 cases with diagnostic whole exome sequencing or identified novel pathogenic variants were affected by early-onset FGR (<32 weeks’ gestation). Of these 10 cases, 7 patients underwent pregnancy termination.
To summarize, a total of 10 cases (19.6%) of severe isolated early-onset FGR with negative cytogenetic studies were subsequently diagnosed with an underlying genetic condition using prenatal trio whole exome sequencing.
Strengths and limitations
This study is retrospective and has a small sample size (n = 51) that was mostly limited to early-onset isolated severe FGR. However, the diagnostic yield (19.6%) of whole exome sequencing after negative CMA testing was noteworthy and shows that monogenic conditions are an important consideration in the evaluation of severe early-onset FGR, even in the absence of structural abnormalities.
As indications for exome sequencing during pregnancy continue to evolve, severe isolated FGR is emerging as a high-yield condition in which a subset of patients may benefit from the described testing strategy. We learned from our look at the prior study (Mone et al) that unselected isolated growth restriction with evident placental insufficiency may not benefit from exome sequencing, but this study differs in its selection of early-onset, severe cases—defined by diagnosis before 32 weeks’ gestation and an EFW or AC below the third percentile. Almost 20% of cases who met the aforementioned criteria received a genetic diagnosis from exome sequencing. We should remember to offer genetic counseling and diagnostic testing to our patients with severe growth restriction, even in the absence of additional structural anomalies.
Could epigenetic mechanisms of placental dysregulation explain low birthweight and future cardiometabolic disease?
Tekola-Ayele F, Zeng X, Chatterjee S, et al. Placental multi-omics integration identifies candidate functional genes for birthweight. Nat Commun. 2022;13:2384.
FGR has been linked to greater mortality in childhood and increased risk for cardiometabolic disease in adulthood. While genomewide associations studies (GWAS) have defined areas of interest linking genetic variants to low birthweight, their relationship to epigenetic changes in the placenta as well as biologic and functional mechanisms are not yet well understood.
Multiomics used to identify candidate functional genes for birthweight
This study analyzed the methylation and gene expression patterns of 291 placental samples, integrating findings into pathways of previously defined GWAS variants. Patient samples were obtained from participants in the Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD) Fetal Growth Studies–Singleton cohort. The cohort is ethnically diverse, with 97 Hispanic, 74 White, 71 Black, and 49 Asian participants. Of 286 single nucleotide polymorphisms (SNPs) found to be associated with birthweight, 273 were analyzed as part of the authors’ data set. These were found to have 7,901 unique protein-coding mRNAs (expression quantitative trait loci [eQTL]) and more than 100,000 nearby (within 1 Mb) CpG islands thought to be involved in changes in DNA methylation (methylation quantitative trait loci [mQTL]). Each functionally connected GWAS-eQTL-mQTL association is referred to as a triplet.
The next arm of the study investigatedthe connections and pathways within each triplet. Three possible scenarios were explored for birthweight GWAS SNPs using a causal interference test (CIT):
- the SNP alters placental DNA methylation, which then influences gene expression
- the SNP first alters placental DNA expression, which then influences methylation
- the SNP influences placental DNA expression and methylation independently, with no notable crossover between their pathways.
Triplets were investigated using the Mendelian randomization (MR) Steiger directionality test to validate the directionality of the pathways found by CIT. Lastly, the possibility of linkage disequilibrium was also studied using the moloc test.
Results
Using CIT, a causal relationship was predicted in 88 of 197 triplets, in which 84 (95.5%) indicated DNA methylation influences gene expression, and 4 (4.5%) indicated gene expression influences DNA methylation. The authors also used the MR Steiger test to investigate triplets to identify possible causal pathways. Using the MR Steiger test, only 3 of 45 (7%) triplets were found to have independent gene expression and methylation pathways. Thirty-eight of 45 (84%) triplets indicated that gene expression influences DNA methylation, and 7 (15%)triplets demonstrated that DNA methylation influences gene expression. Consistent predictions between CIT and the MR Steiger test revealed 3 triplets in which DNA methylation influences gene expression for the following genes: WNT3A, CTDNEP1, and RANBP2. Additionally, a strong colocalization signal was found among birthweight, DNA methylation, and gene expression for the following genes: PLEKHA1, FES, PRMT7, and CTDNEP1. Gene set enrichment analysis was performed as well and found that low birthweight is associated in substantial upregulation of genes associated with oxidative stress, immune response, adipogenesis, myogenesis, and the production of pancreatic ß cells.
Study strengths and limitations
The study is one of the first to identify regulatory targets for placental DNA methylation and gene expression in previously identified GWAS loci associated with low birthweight. For example, DNA methylation was found to influence gene expression of WNT3A, CTDNEP1, and RANBP2, which have previously been shown in animal studies to impact the vascularization and development of the placenta, embryogenesis, and fetal growth. The study also identified 4 genes (PLEKHA1, FES, PRMT7, and CTDNEP1) thought to have direct regulatory influence on placental DNA methylation and gene expression.
A limitation of the study is that it could not distinguish between whether the epigenetic changes we outlined have a maternal or fetal origin. Another limitation is that tissue used by the authors for analysis was a small placental biopsy, which does not accurately reflect the genetic heterogeneity of the placenta. Finally, this study does not establish causality between the identified epigenetic pathways and low birthweight. ●
We know that the placenta is critical to in utero development. This study begins to explore the genetic changes and programming in the placenta that may have profound effects on health and well-being both early and later in life.
- Li LS, Li DZ. A genetic approach to the etiologic investigation of isolated intrauterine growth restriction. Am J Obstet Gynecol. 2021;225:695-696. doi: 10.1016/j.ajog.2021 .07.021.
- Borrell A, Grande M, Pauta M, et al. Chromosomal microarray analysis in fetuses with growth restriction and normal karyotype: a systematic review and meta-analysis. Fetal Diagn Ther. 2018;44:1-9. doi: 10.1159/000479506.
Whole exome sequencing’s role in diagnosing genetic causes of FGR with and without associated anomalies
Mone F, Mellis R, Gabriel H, et al. Should we offer prenatal exome sequencing for intrauterine growth restriction or short long bones? A systematic review and meta-analysis. Am J Obstet Gynecol. Published online October 7, 2022. doi:10.1016/j.ajog.2022.09.045
Multiple factors can play a role in FGR, including inherent maternal, placental, or fetal factors; the environment; and/or nutrition. However, prenatal diagnosis is an important consideration when exploring the underlying etiology for a growth-restricted fetus, especially in severe or early-onset cases. Many genetic conditions do not result in structural anomalies but can disrupt overall growth. Additionally, phenotyping in the prenatal period is limited and can miss more subtle physical differences that could point to a genetic cause.
When compared with karyotype, chromosomal microarray (CMA) has been shown to increase the diagnostic yield in cases of isolated early FGR by 5%,1,2 and the incidence of chromosomal abnormalities has been reported to be as high as 19% in this population. Let’s explore the data on exome sequencing for prenatal diagnosis in cases of isolated FGR.
Meta-analysis details
In this meta-analysis, the authors reviewed 19 cohort studies or case series that investigated the yield of prenatal sequencing in fetuses with intrauterine growth restriction (IUGR) or short long bones, both in association with and without additional anomalies. All cases had nondiagnostic cytogenetic results. Fetal DNA in most cases was obtained through amniocentesis. Variants classified as likely pathogenic and pathogenic were considered diagnostic. The authors then calculated the incremental yield of prenatal sequencing over cytogenetic studies as a pooled value, comparing the following groups:
- isolated FGR
- growth restriction with associated anomalies
- isolated short long bones
- short long bones with additional skeletal features.
Study outcomes
The total number of cases were as follows: isolated IUGR (n = 71), IUGR associated with additional anomalies (n = 45), isolated short long bones (n = 84), and short long bones associated with additional skeletal findings (n = 252). Causative pathogenic or likely pathogenic variants were identified in 224 (50%) cases. Apparent incremental yields with prenatal sequencing were as follows for the 4 groups (as illustrated in the FIGURE):
- 4% in isolated IUGR (95% confidence interval [CI], -5%–12%)
- 30% in IUGR with additional anomalies (95% CI, 13%–47%)
- 48% in isolated short long bones (95% CI, 26%–70%)
- 68% in short long bones with additional skeletal changes (95% CI, 58%–77%).
Overall, the authors concluded that prenatal sequencing does not improve prenatal diagnosis in cases of isolated IUGR. The majority of these cases were thought to be related to placental insufficiency.
Strengths and limitations
The main limitation of this study with regard to our discussion is the small study populationof isolated growth restriction. The authors indicate that the number of cases of isolated IUGR were too small to draw firm conclusions. Another limitation was the heterogeneity of the isolated FGR population, which was not limited to severe or early-onset cases. However, the authors did demonstrate that growth restriction in association with fetal anomalies has very high genetic yield rates with prenatal sequencing.
Not surprisingly, there is a high yield of diagnosing genetic conditions in pregnancies complicated by isolated or nonisolated short long bones or in cases of growth restriction with multisystem abnormalities. Based on the results of this study, the authors advise against sending for exome sequencing in cases of isolated growth restriction with coexisting evidence of placental insufficiency.
Continue to: Can whole exome sequencing diagnose genetic causes in cases of severe isolated FGR?...
Can whole exome sequencing diagnose genetic causes in cases of severe isolated FGR?
Zhou H, Fu F, Wang Y, et al. Genetic causes of isolated and severe fetal growth restriction in normal chromosomal microarray analysis. Int J Gynaecol Obstet. Published online December 10, 2022. doi:10.1002/ijgo.14620
Severe FGR is diagnosed based on an estimated fetal weight (EFW) or abdominal circumference (AC) below the third percentile. As we discussed in the above study by Mone and colleagues, it does not appear that prenatal sequencing significantly improves the diagnostic yield in all isolated FGR cases. However, this has not been previously explored in isolated severe FGR or cases of early-onset FGR (<32 weeks’ gestation). We know that several monogenic conditions are associated with severe and early-onset isolated fetal growth impairment, including but not limited to Cornelia de Lange syndrome, Smith-Lemli-Opitz syndrome, and Meier-Gorlin syndrome. Often, these syndromes can present in the prenatal period without other phenotypic findings. Therefore, this study explored the possibility that prenatal sequencing plays an important role for severe cases of FGR with nondiagnostic CMA and/or karyotype.
Retrospective study details
Zhou and colleagues retrospectively analyzed 51 cases of severe (EFW or AC below the third percentile) isolated FGR with negative CMA who underwent trio whole exome sequencing, which includes submitting fetal cells as well as both parental samples for testing. Patients with abnormal toxoplasmosis, rubella, cytomegalovirus, and herpes simplex virus (TORCH) tests; structural anomalies; and multiple gestation were excluded from the analysis. As in the study by Mone et al, variants classified as likely pathogenic and pathogenic were categorized as diagnostic.
Results
Eight of 51 cases (15.7%) with severe isolated FGR had diagnostic findings on trio whole exome sequencing as shown in the TABLE. Another 8 cases (15.7%) were found to have variants of unknown significance, of which 2 were later determined to be novel pathogenic variants. Genetic conditions uncovered in this cohort include Cornelia de Lange syndrome, pyruvate dehydrogenase deficiency, Dent disease, trichohepaticenteric syndrome, achondroplasia, osteogenesis imperfecta, Pendred syndrome, and both autosomal dominant type 3A and autosomal recessive type 1A deafness. All 10 cases with diagnostic whole exome sequencing or identified novel pathogenic variants were affected by early-onset FGR (<32 weeks’ gestation). Of these 10 cases, 7 patients underwent pregnancy termination.
To summarize, a total of 10 cases (19.6%) of severe isolated early-onset FGR with negative cytogenetic studies were subsequently diagnosed with an underlying genetic condition using prenatal trio whole exome sequencing.
Strengths and limitations
This study is retrospective and has a small sample size (n = 51) that was mostly limited to early-onset isolated severe FGR. However, the diagnostic yield (19.6%) of whole exome sequencing after negative CMA testing was noteworthy and shows that monogenic conditions are an important consideration in the evaluation of severe early-onset FGR, even in the absence of structural abnormalities.
As indications for exome sequencing during pregnancy continue to evolve, severe isolated FGR is emerging as a high-yield condition in which a subset of patients may benefit from the described testing strategy. We learned from our look at the prior study (Mone et al) that unselected isolated growth restriction with evident placental insufficiency may not benefit from exome sequencing, but this study differs in its selection of early-onset, severe cases—defined by diagnosis before 32 weeks’ gestation and an EFW or AC below the third percentile. Almost 20% of cases who met the aforementioned criteria received a genetic diagnosis from exome sequencing. We should remember to offer genetic counseling and diagnostic testing to our patients with severe growth restriction, even in the absence of additional structural anomalies.
Could epigenetic mechanisms of placental dysregulation explain low birthweight and future cardiometabolic disease?
Tekola-Ayele F, Zeng X, Chatterjee S, et al. Placental multi-omics integration identifies candidate functional genes for birthweight. Nat Commun. 2022;13:2384.
FGR has been linked to greater mortality in childhood and increased risk for cardiometabolic disease in adulthood. While genomewide associations studies (GWAS) have defined areas of interest linking genetic variants to low birthweight, their relationship to epigenetic changes in the placenta as well as biologic and functional mechanisms are not yet well understood.
Multiomics used to identify candidate functional genes for birthweight
This study analyzed the methylation and gene expression patterns of 291 placental samples, integrating findings into pathways of previously defined GWAS variants. Patient samples were obtained from participants in the Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD) Fetal Growth Studies–Singleton cohort. The cohort is ethnically diverse, with 97 Hispanic, 74 White, 71 Black, and 49 Asian participants. Of 286 single nucleotide polymorphisms (SNPs) found to be associated with birthweight, 273 were analyzed as part of the authors’ data set. These were found to have 7,901 unique protein-coding mRNAs (expression quantitative trait loci [eQTL]) and more than 100,000 nearby (within 1 Mb) CpG islands thought to be involved in changes in DNA methylation (methylation quantitative trait loci [mQTL]). Each functionally connected GWAS-eQTL-mQTL association is referred to as a triplet.
The next arm of the study investigatedthe connections and pathways within each triplet. Three possible scenarios were explored for birthweight GWAS SNPs using a causal interference test (CIT):
- the SNP alters placental DNA methylation, which then influences gene expression
- the SNP first alters placental DNA expression, which then influences methylation
- the SNP influences placental DNA expression and methylation independently, with no notable crossover between their pathways.
Triplets were investigated using the Mendelian randomization (MR) Steiger directionality test to validate the directionality of the pathways found by CIT. Lastly, the possibility of linkage disequilibrium was also studied using the moloc test.
Results
Using CIT, a causal relationship was predicted in 88 of 197 triplets, in which 84 (95.5%) indicated DNA methylation influences gene expression, and 4 (4.5%) indicated gene expression influences DNA methylation. The authors also used the MR Steiger test to investigate triplets to identify possible causal pathways. Using the MR Steiger test, only 3 of 45 (7%) triplets were found to have independent gene expression and methylation pathways. Thirty-eight of 45 (84%) triplets indicated that gene expression influences DNA methylation, and 7 (15%)triplets demonstrated that DNA methylation influences gene expression. Consistent predictions between CIT and the MR Steiger test revealed 3 triplets in which DNA methylation influences gene expression for the following genes: WNT3A, CTDNEP1, and RANBP2. Additionally, a strong colocalization signal was found among birthweight, DNA methylation, and gene expression for the following genes: PLEKHA1, FES, PRMT7, and CTDNEP1. Gene set enrichment analysis was performed as well and found that low birthweight is associated in substantial upregulation of genes associated with oxidative stress, immune response, adipogenesis, myogenesis, and the production of pancreatic ß cells.
Study strengths and limitations
The study is one of the first to identify regulatory targets for placental DNA methylation and gene expression in previously identified GWAS loci associated with low birthweight. For example, DNA methylation was found to influence gene expression of WNT3A, CTDNEP1, and RANBP2, which have previously been shown in animal studies to impact the vascularization and development of the placenta, embryogenesis, and fetal growth. The study also identified 4 genes (PLEKHA1, FES, PRMT7, and CTDNEP1) thought to have direct regulatory influence on placental DNA methylation and gene expression.
A limitation of the study is that it could not distinguish between whether the epigenetic changes we outlined have a maternal or fetal origin. Another limitation is that tissue used by the authors for analysis was a small placental biopsy, which does not accurately reflect the genetic heterogeneity of the placenta. Finally, this study does not establish causality between the identified epigenetic pathways and low birthweight. ●
We know that the placenta is critical to in utero development. This study begins to explore the genetic changes and programming in the placenta that may have profound effects on health and well-being both early and later in life.
Whole exome sequencing’s role in diagnosing genetic causes of FGR with and without associated anomalies
Mone F, Mellis R, Gabriel H, et al. Should we offer prenatal exome sequencing for intrauterine growth restriction or short long bones? A systematic review and meta-analysis. Am J Obstet Gynecol. Published online October 7, 2022. doi:10.1016/j.ajog.2022.09.045
Multiple factors can play a role in FGR, including inherent maternal, placental, or fetal factors; the environment; and/or nutrition. However, prenatal diagnosis is an important consideration when exploring the underlying etiology for a growth-restricted fetus, especially in severe or early-onset cases. Many genetic conditions do not result in structural anomalies but can disrupt overall growth. Additionally, phenotyping in the prenatal period is limited and can miss more subtle physical differences that could point to a genetic cause.
When compared with karyotype, chromosomal microarray (CMA) has been shown to increase the diagnostic yield in cases of isolated early FGR by 5%,1,2 and the incidence of chromosomal abnormalities has been reported to be as high as 19% in this population. Let’s explore the data on exome sequencing for prenatal diagnosis in cases of isolated FGR.
Meta-analysis details
In this meta-analysis, the authors reviewed 19 cohort studies or case series that investigated the yield of prenatal sequencing in fetuses with intrauterine growth restriction (IUGR) or short long bones, both in association with and without additional anomalies. All cases had nondiagnostic cytogenetic results. Fetal DNA in most cases was obtained through amniocentesis. Variants classified as likely pathogenic and pathogenic were considered diagnostic. The authors then calculated the incremental yield of prenatal sequencing over cytogenetic studies as a pooled value, comparing the following groups:
- isolated FGR
- growth restriction with associated anomalies
- isolated short long bones
- short long bones with additional skeletal features.
Study outcomes
The total number of cases were as follows: isolated IUGR (n = 71), IUGR associated with additional anomalies (n = 45), isolated short long bones (n = 84), and short long bones associated with additional skeletal findings (n = 252). Causative pathogenic or likely pathogenic variants were identified in 224 (50%) cases. Apparent incremental yields with prenatal sequencing were as follows for the 4 groups (as illustrated in the FIGURE):
- 4% in isolated IUGR (95% confidence interval [CI], -5%–12%)
- 30% in IUGR with additional anomalies (95% CI, 13%–47%)
- 48% in isolated short long bones (95% CI, 26%–70%)
- 68% in short long bones with additional skeletal changes (95% CI, 58%–77%).
Overall, the authors concluded that prenatal sequencing does not improve prenatal diagnosis in cases of isolated IUGR. The majority of these cases were thought to be related to placental insufficiency.
Strengths and limitations
The main limitation of this study with regard to our discussion is the small study populationof isolated growth restriction. The authors indicate that the number of cases of isolated IUGR were too small to draw firm conclusions. Another limitation was the heterogeneity of the isolated FGR population, which was not limited to severe or early-onset cases. However, the authors did demonstrate that growth restriction in association with fetal anomalies has very high genetic yield rates with prenatal sequencing.
Not surprisingly, there is a high yield of diagnosing genetic conditions in pregnancies complicated by isolated or nonisolated short long bones or in cases of growth restriction with multisystem abnormalities. Based on the results of this study, the authors advise against sending for exome sequencing in cases of isolated growth restriction with coexisting evidence of placental insufficiency.
Continue to: Can whole exome sequencing diagnose genetic causes in cases of severe isolated FGR?...
Can whole exome sequencing diagnose genetic causes in cases of severe isolated FGR?
Zhou H, Fu F, Wang Y, et al. Genetic causes of isolated and severe fetal growth restriction in normal chromosomal microarray analysis. Int J Gynaecol Obstet. Published online December 10, 2022. doi:10.1002/ijgo.14620
Severe FGR is diagnosed based on an estimated fetal weight (EFW) or abdominal circumference (AC) below the third percentile. As we discussed in the above study by Mone and colleagues, it does not appear that prenatal sequencing significantly improves the diagnostic yield in all isolated FGR cases. However, this has not been previously explored in isolated severe FGR or cases of early-onset FGR (<32 weeks’ gestation). We know that several monogenic conditions are associated with severe and early-onset isolated fetal growth impairment, including but not limited to Cornelia de Lange syndrome, Smith-Lemli-Opitz syndrome, and Meier-Gorlin syndrome. Often, these syndromes can present in the prenatal period without other phenotypic findings. Therefore, this study explored the possibility that prenatal sequencing plays an important role for severe cases of FGR with nondiagnostic CMA and/or karyotype.
Retrospective study details
Zhou and colleagues retrospectively analyzed 51 cases of severe (EFW or AC below the third percentile) isolated FGR with negative CMA who underwent trio whole exome sequencing, which includes submitting fetal cells as well as both parental samples for testing. Patients with abnormal toxoplasmosis, rubella, cytomegalovirus, and herpes simplex virus (TORCH) tests; structural anomalies; and multiple gestation were excluded from the analysis. As in the study by Mone et al, variants classified as likely pathogenic and pathogenic were categorized as diagnostic.
Results
Eight of 51 cases (15.7%) with severe isolated FGR had diagnostic findings on trio whole exome sequencing as shown in the TABLE. Another 8 cases (15.7%) were found to have variants of unknown significance, of which 2 were later determined to be novel pathogenic variants. Genetic conditions uncovered in this cohort include Cornelia de Lange syndrome, pyruvate dehydrogenase deficiency, Dent disease, trichohepaticenteric syndrome, achondroplasia, osteogenesis imperfecta, Pendred syndrome, and both autosomal dominant type 3A and autosomal recessive type 1A deafness. All 10 cases with diagnostic whole exome sequencing or identified novel pathogenic variants were affected by early-onset FGR (<32 weeks’ gestation). Of these 10 cases, 7 patients underwent pregnancy termination.
To summarize, a total of 10 cases (19.6%) of severe isolated early-onset FGR with negative cytogenetic studies were subsequently diagnosed with an underlying genetic condition using prenatal trio whole exome sequencing.
Strengths and limitations
This study is retrospective and has a small sample size (n = 51) that was mostly limited to early-onset isolated severe FGR. However, the diagnostic yield (19.6%) of whole exome sequencing after negative CMA testing was noteworthy and shows that monogenic conditions are an important consideration in the evaluation of severe early-onset FGR, even in the absence of structural abnormalities.
As indications for exome sequencing during pregnancy continue to evolve, severe isolated FGR is emerging as a high-yield condition in which a subset of patients may benefit from the described testing strategy. We learned from our look at the prior study (Mone et al) that unselected isolated growth restriction with evident placental insufficiency may not benefit from exome sequencing, but this study differs in its selection of early-onset, severe cases—defined by diagnosis before 32 weeks’ gestation and an EFW or AC below the third percentile. Almost 20% of cases who met the aforementioned criteria received a genetic diagnosis from exome sequencing. We should remember to offer genetic counseling and diagnostic testing to our patients with severe growth restriction, even in the absence of additional structural anomalies.
Could epigenetic mechanisms of placental dysregulation explain low birthweight and future cardiometabolic disease?
Tekola-Ayele F, Zeng X, Chatterjee S, et al. Placental multi-omics integration identifies candidate functional genes for birthweight. Nat Commun. 2022;13:2384.
FGR has been linked to greater mortality in childhood and increased risk for cardiometabolic disease in adulthood. While genomewide associations studies (GWAS) have defined areas of interest linking genetic variants to low birthweight, their relationship to epigenetic changes in the placenta as well as biologic and functional mechanisms are not yet well understood.
Multiomics used to identify candidate functional genes for birthweight
This study analyzed the methylation and gene expression patterns of 291 placental samples, integrating findings into pathways of previously defined GWAS variants. Patient samples were obtained from participants in the Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD) Fetal Growth Studies–Singleton cohort. The cohort is ethnically diverse, with 97 Hispanic, 74 White, 71 Black, and 49 Asian participants. Of 286 single nucleotide polymorphisms (SNPs) found to be associated with birthweight, 273 were analyzed as part of the authors’ data set. These were found to have 7,901 unique protein-coding mRNAs (expression quantitative trait loci [eQTL]) and more than 100,000 nearby (within 1 Mb) CpG islands thought to be involved in changes in DNA methylation (methylation quantitative trait loci [mQTL]). Each functionally connected GWAS-eQTL-mQTL association is referred to as a triplet.
The next arm of the study investigatedthe connections and pathways within each triplet. Three possible scenarios were explored for birthweight GWAS SNPs using a causal interference test (CIT):
- the SNP alters placental DNA methylation, which then influences gene expression
- the SNP first alters placental DNA expression, which then influences methylation
- the SNP influences placental DNA expression and methylation independently, with no notable crossover between their pathways.
Triplets were investigated using the Mendelian randomization (MR) Steiger directionality test to validate the directionality of the pathways found by CIT. Lastly, the possibility of linkage disequilibrium was also studied using the moloc test.
Results
Using CIT, a causal relationship was predicted in 88 of 197 triplets, in which 84 (95.5%) indicated DNA methylation influences gene expression, and 4 (4.5%) indicated gene expression influences DNA methylation. The authors also used the MR Steiger test to investigate triplets to identify possible causal pathways. Using the MR Steiger test, only 3 of 45 (7%) triplets were found to have independent gene expression and methylation pathways. Thirty-eight of 45 (84%) triplets indicated that gene expression influences DNA methylation, and 7 (15%)triplets demonstrated that DNA methylation influences gene expression. Consistent predictions between CIT and the MR Steiger test revealed 3 triplets in which DNA methylation influences gene expression for the following genes: WNT3A, CTDNEP1, and RANBP2. Additionally, a strong colocalization signal was found among birthweight, DNA methylation, and gene expression for the following genes: PLEKHA1, FES, PRMT7, and CTDNEP1. Gene set enrichment analysis was performed as well and found that low birthweight is associated in substantial upregulation of genes associated with oxidative stress, immune response, adipogenesis, myogenesis, and the production of pancreatic ß cells.
Study strengths and limitations
The study is one of the first to identify regulatory targets for placental DNA methylation and gene expression in previously identified GWAS loci associated with low birthweight. For example, DNA methylation was found to influence gene expression of WNT3A, CTDNEP1, and RANBP2, which have previously been shown in animal studies to impact the vascularization and development of the placenta, embryogenesis, and fetal growth. The study also identified 4 genes (PLEKHA1, FES, PRMT7, and CTDNEP1) thought to have direct regulatory influence on placental DNA methylation and gene expression.
A limitation of the study is that it could not distinguish between whether the epigenetic changes we outlined have a maternal or fetal origin. Another limitation is that tissue used by the authors for analysis was a small placental biopsy, which does not accurately reflect the genetic heterogeneity of the placenta. Finally, this study does not establish causality between the identified epigenetic pathways and low birthweight. ●
We know that the placenta is critical to in utero development. This study begins to explore the genetic changes and programming in the placenta that may have profound effects on health and well-being both early and later in life.
- Li LS, Li DZ. A genetic approach to the etiologic investigation of isolated intrauterine growth restriction. Am J Obstet Gynecol. 2021;225:695-696. doi: 10.1016/j.ajog.2021 .07.021.
- Borrell A, Grande M, Pauta M, et al. Chromosomal microarray analysis in fetuses with growth restriction and normal karyotype: a systematic review and meta-analysis. Fetal Diagn Ther. 2018;44:1-9. doi: 10.1159/000479506.
- Li LS, Li DZ. A genetic approach to the etiologic investigation of isolated intrauterine growth restriction. Am J Obstet Gynecol. 2021;225:695-696. doi: 10.1016/j.ajog.2021 .07.021.
- Borrell A, Grande M, Pauta M, et al. Chromosomal microarray analysis in fetuses with growth restriction and normal karyotype: a systematic review and meta-analysis. Fetal Diagn Ther. 2018;44:1-9. doi: 10.1159/000479506.
Vulvar syringoma
To the Editor:
Syringomas are common benign tumors of the eccrine sweat glands that usually manifest clinically as multiple flesh-colored papules. They are most commonly seen on the face, neck, and chest of adolescent girls. Syringomas may appear at any site of the body but are rare in the vulva. We present a case of a 51-year-old woman who was referred to the Division of Gynecologic Oncology at the University of Alabama at Birmingham for further management of a tumor carrying a differential diagnosis of vulvar syringoma vs microcystic adnexal carcinoma (MAC).
A 51-year-old woman presented to dermatology (G.G.) and was referred to the Division of Gynecologic Oncology at the University of Alabama at Birmingham for further management of possible vulvar syringoma vs MAC. The patient previously had been evaluated at an outside community practice due to dyspareunia, vulvar discomfort, and vulvar irregularities of 1 month’s duration. At that time, a small biopsy was performed, and the histologic differential diagnosis included syringoma vs an adnexal carcinoma. Consequently, she was referred to gynecologic oncology for further management.
Pelvic examination revealed multilobular nodular areas overlying the clitoral hood that extended down to the labia majora. The nodular processes did not involve the clitoris, labia minora, or perineum. A mobile isolated lymph node measuring 2.0×1.0 cm in the right inguinal area also was noted. The patient’s clinical history was notable for right breast carcinoma treated with a right mastectomy with axillary lymph node dissection that showed metastatic disease. She also underwent adjuvant chemotherapy with paclitaxel and doxorubicin for breast carcinoma.
After discussing the diagnostic differential and treatment options, the patient elected to undergo a bilateral partial radical vulvectomy with reconstruction and resection of the right inguinal lymph node. Gross examination of the vulvectomy specimen showed multiple flesh-colored papules (FIGURE 1). Histologic examination revealed a neoplasm with sweat gland differentiation that was broad and poorly circumscribed but confined to the dermis (FIGURES 2A and 2B). The neoplasm was composed of epithelial cells that formed ductlike structures, lined by 2 layers of cuboidal epithelium within a fibrous stroma (FIGURE 2C). A toluidine blue special stain was performed and demonstrated an increased amount of mast cells in the tissue (FIGURE 3). Immunohistochemical stains for gross cystic disease fluid protein, estrogen receptor (ER), and progesterone receptor (PR) were negative in the tumor cells. The lack of cytologic atypia, perineural invasion, and deep infiltration into the subcutis favored a syringoma. One month later, the case was presented at the Tumor Board Conference at the University of Alabama at Birmingham where a final diagnosis of vulvar syringoma was agreed upon and discussed with the patient. At that time, no recurrence was evident and follow-up was recommended.
Syringomas are benign tumors of the sweat glands that are fairly common and appear to have a predilection for women. Although most of the literature classifies them as eccrine neoplasms, the term syringoma can be used to describe neoplasms of either apocrine or eccrine lineage.1 To rule out an apocrine lineage of the tumor in our patient, we performed immunohistochemistry for gross cystic disease fluid protein, a marker of apocrine differentiation. This stain highlighted normal apocrine glands that were not involved in the tumor proliferation.
Syringomas may occur at any site on the body but are prone to occur on the periorbital area, especially the eyelids.1 Some of the atypical locations for a syringoma include the anterior neck, chest, abdomen, genitals, axillae, groin, and buttocks.2 Vulvar syringomas were first reported by Carneiro3 in 1971 as usually affecting adolescent girls and middle-aged women. There have been approximately 40 reported cases affecting women aged 8 to 78 years.4,5 Vulvar syringomas classically appear as firm or soft, flesh-colored to transparent, papular lesions. The 2 other clinical variants are miliumlike, whitish, cystic papules as well as lichenoid papules.6 Pérez-Bustillo et al5 reported a case of the lichenoid papule variant on the labia majora of a 78-year-old woman who presented with intermittent vulvar pruritus of 4 years’ duration. Due to this patient’s 9-year history of urinary incontinence, the lesions had been misdiagnosed as irritant dermatitis and associated lichen simplex chronicus (LSC). This case is a reminder to consider vulvar syringoma in patients with LSC who respond poorly to oral antihistamines and topical steroids.5 Rarely, multiple clinical variants may coexist. In a case reported by Dereli et al,7 a 19-year-old woman presented with concurrent classical and miliumlike forms of vulvar syringoma.
Vulvar syringomas usually present as multiple lesions involving both sides of the labia majora; however, Blasdale and McLelland8 reported a single isolated syringoma of the vulva on the anterior right labia minora that measured 1.0×0.5 cm, leading the lesion to be described as a giant syringoma.
Vulvar syringomas usually are asymptomatic and noticed during routine gynecologic examination. Therefore, it is believed that they likely are underdiagnosed.5 When symptomatic, they commonly present with constant9 or intermittent5 pruritus, which may intensify during menstruation, pregnancy, and summertime.6,10-12 Gerdsen et al10 documented a 27-year-old woman who presented with a 2-year history of pruritic vulvar skin lesions that became exacerbated during menstruation, which raised the possibility of cyclical hormonal changes being responsible for periodic exacerbation of vulvar pruritus during menstruation. In addition, patients may experience an increase in size and number of the lesions during pregnancy. Bal et al11 reported a 24-year-old primigravida with vulvar papular lesions that intensified during pregnancy. She had experienced intermittent vulvar pruritus for 12 years but had no change in symptoms during menstruation.11 Few studies have attempted to evaluate the presence of ER and PR in the syringomas. A study of 9 nonvulvar syringomas by Wallace and Smoller13 showed ER positivity in 1 case and PR positivity in 8 cases, lending support to the hormonal theory; however, in another case series of 15 vulvar syringomas, Huang et al6 failed to show ER and PR expression by immunohistochemical staining. A case report published 3 years earlier documented the first case of PR positivity on a vulvar syringoma.14 Our patient also was negative for ER and PR, which suggested that hormonal status is important in some but not all syringomas.
Patients with vulvar syringomas also might have coexisting extragenital syringomas in the neck,4 eyelids,6,7,10 and periorbital area,6 and thorough examination of the body is essential. If an extragenital syringoma is diagnosed, a vulvar syringoma should be considered, especially when the patient presents with unexplained genital symptoms. Although no proven hereditary transmission pattern has been established, family history of syringomas has been established in several cases.15 In a case series reported by Huang et al,6 4 of 18 patients reported a family history of periorbital syringomas. In our case, the patient did not report a family history of syringomas.
The differential diagnosis of vulvar lesions with pruritus is broad and includes Fox-Fordyce disease, lichen planus, LSC, epidermal cysts, senile angiomas, dystrophic calcinosis, xanthomas, steatocytomas, soft fibromas, condyloma acuminatum, and candidiasis. Vulvar syringomas might have a nonspecific appearance, and histologic examination is essential to confirm the diagnosis and rule out any malignant process such as MAC, vulvar intraepithelial neoplasia, extramammary Paget disease, or other glandular neoplasms of the vulva.
Microcystic adnexal carcinoma was first reported in 1982 by Goldstein et al16 as a locally aggressive neoplasm that can be confused with benign adnexal neoplasms, particularly desmoplastic trichoepithelioma, trichoadenoma, and syringoma. Microcystic adnexal carcinomas present as slow-growing, flesh-colored papules that may resemble syringomas and appear in similar body sites. Histologic examination is essential to differentiate between these two entities. Syringomas are tumors confined to the dermis and are composed of multiple small ducts lined by 2 layers of cuboidal epithelium within a dense fibrous stroma. Unlike syringomas, MACs usually infiltrate diffusely into the dermis and subcutis and may extend into the underlying muscle. Although bland cytologic features predominate, perineural invasion frequently is present in MACs. A potential pitfall of misdiagnosis can be caused by a superficial biopsy that may reveal benign histologic appearance, particularly in the upper level of the tumor where it may be confused with a syringoma or a benign follicular neoplasm.17
The initial biopsy performed on our patient was possibly not deep enough to render an unequivocal diagnosis and therefore bilateral partial radical vulvectomy was considered. After surgery, histologic examination of the resection specimen revealed a poorly circumscribed tumor confined to the dermis. The tumor was broad and the lack of deep infiltration into the subcutis and perineural invasion favored a syringoma (FIGURES 2A and 2B). These findings were consistent with case reports that documented syringomas as being more wide than deep on microscopic examination, whereas the opposite pertained to MAC.18 Cases of plaque-type syringomas that initially were misdiagnosed as MACs also have been reported.19 Because misdiagnosis may affect the treatment plan and potentially result in unnecessary surgery, caution should be taken when differentiating between these two entities. When a definitive diagnosis cannot be rendered on a superficial biopsy, a recommendation should be made for a deeper biopsy sampling the subcutis.
For the majority of the patients with vulvar syringomas, treatment is seldom required due to their asymptomatic nature; however, patients who present with symptoms usually report pruritus of variable intensities and patterns. A standardized treatment does not exist for vulvar syringomas, and oral or topical treatment might be used as an initial approach. Commonly prescribed medications with variable results include topical corticosteroids, oral antihistamines, and topical retinoids. In a case reported by Iwao et al,20 vulvar syringomas were successfully treated with tranilast, which has anti-inflammatory and immunomodulatory effects. This medication could have a possible dual action—inhibiting the release of chemical mediators from the mast cells and inhibiting the release of IL-1β from the eccrine duct, which could suppress the proliferation of stromal connective tissue. Our case was stained with toluidine blue and showed an increased number of mast cells in the tissue (FIGURE 3).Patients who are unresponsive to tranilast or have extensive disease resulting in cosmetic disfigurement might benefit from more invasive treatment methods including a variety of lasers, cryotherapy, electrosurgery, and excision. Excisions should include the entire tumor to avoid recurrence. In a case reported by Garman and Metry,21 the lesions were surgically excised using small 2- to 3-mm punches; however, several weeks later the lesions recurred. Our patient presented with a 1-month evolution of dyspareunia, vulvar discomfort, and vulvar irregularities that were probably not treated with oral or topical medications before being referred for surgery.
We report a case of a vulvar syringoma that presented diagnostic challenges in the initial biopsy, which prevented the exclusion of an MAC. After partial radical vulvectomy, histologic examination was more definitive, showing lack of deep infiltration into the subcutis or perineural invasion that are commonly seen in MAC. This case is an example of a notable pitfall in the diagnosis of vulvar syringoma on a limited biopsy leading to overtreatment. Raising awareness of this entity is the only modality to prevent misdiagnosis. We encourage reporting of further cases of syringomas, particularly those with atypical locations or patterns that may cause diagnostic problems. ●
- Ensure adequate depth of biopsy to assist in the histologic diagnosis of syringoma vs microcystic adnexal carcinoma.
- Vulvar syringomas also may contribute to notable pruritus and ultimately be the underlying etiology for secondary skin changes leading to a lichen simplex chronicus–like phenotype
- Bolognia JL, Jorizzo JL, Rapini RP. Dermatology. 2nd ed. Spain: Mosby Elsevier; 2008.
- Weedon D. Skin Pathology. 3rd ed. China: Churchill Livingstone Elsevier; 2010.
- Carneiro SJ, Gardner HL, Knox JM. Syringoma of the vulva. Arch Dermatol. 1971;103:494-496.
- Trager JD, Silvers J, Reed JA, et al. Neck and vulvar papules in an 8-year-old girl. Arch Dermatol. 1999;135:203, 206.
- Pérez-Bustillo A, Ruiz-González I, Delgado S, et al. Vulvar syringoma: a rare cause of vulvar pruritus. Actas DermoSifiliográficas. 2008; 99:580-581.
- Huang YH, Chuang YH, Kuo TT, et al. Vulvar syringoma: a clinicopathologic and immunohistologic study of 18 patients and results of treatment. J Am Acad Dermatol. 2003;48:735-739.
- Dereli T, Turk BG, Kazandi AC. Syringomas of the vulva. Int J Gynaecol Obstet. 2007;99:65-66.
- Blasdale C, McLelland J. Solitary giant vulval syringoma. Br J Dermatol. 1999;141:374-375.
- Kavala M, Can B, Zindanci I, et al. Vulvar pruritus caused by syringoma of the vulva. Int J Dermatol. 2008;47:831-832.
- Gerdsen R, Wenzel J, Uerlich M, et al. Periodic genital pruritus caused by syringoma of the vulva. Acta Obstet Gynecol Scand. 2002;81:369-370.
- Bal N, Aslan E, Kayaselcuk F, et al. Vulvar syringoma aggravated by pregnancy. Pathol Oncol Res. 2003;9:196-197.
- Turan C, Ugur M, Kutluay L, et al. Vulvar syringoma exacerbated during pregnancy. Eur J Obstet Gynecol Reprod Biol. 1996;64:141-142.
- Wallace ML, Smoller BR. Progesterone receptor positivity supports hormonal control of syringomas. J Cutan Pathol. 1995; 22:442-445.
- Yorganci A, Kale A, Dunder I, et al. Vulvar syringoma showing progesterone receptor positivity. BJOG. 2000;107:292-294.
- Draznin M. Hereditary syringomas: a case report. Dermatol Online J. 2004;10:19.
- Goldstein DJ, Barr RJ, Santa Cruz DJ. Microcystic adnexal carcinoma: a distinct clinicopathologic entity. Cancer. 1982;50:566-572.
- Hamsch C, Hartschuh W. Microcystic adnexal carcinomaaggressive infiltrative tumor often with innocent clinical appearance. J Dtsch Dermatol Ges. 2010;8:275-278.
- Henner MS, Shapiro PE, Ritter JH, et al. Solitary syringoma. report of five cases and clinicopathologic comparison with microcystic adnexal carcinoma of the skin. Am J Dermatopathol. 1995;17:465-470.
- Suwattee P, McClelland MC, Huiras EE, et al. Plaque-type syringoma: two cases misdiagnosed as microcystic adnexal carcinoma. J Cutan Pathol. 2008;35:570-574.
- Iwao F, Onozuka T, Kawashima T. Vulval syringoma successfully treated with tranilast. Br J Dermatol. 2005;153:1228-1230.
- Garman M, Metry D. Vulvar syringomas in a 9-year-old child with review of the literature. Pediatr Dermatol. 2006;23:369372.
To the Editor:
Syringomas are common benign tumors of the eccrine sweat glands that usually manifest clinically as multiple flesh-colored papules. They are most commonly seen on the face, neck, and chest of adolescent girls. Syringomas may appear at any site of the body but are rare in the vulva. We present a case of a 51-year-old woman who was referred to the Division of Gynecologic Oncology at the University of Alabama at Birmingham for further management of a tumor carrying a differential diagnosis of vulvar syringoma vs microcystic adnexal carcinoma (MAC).
A 51-year-old woman presented to dermatology (G.G.) and was referred to the Division of Gynecologic Oncology at the University of Alabama at Birmingham for further management of possible vulvar syringoma vs MAC. The patient previously had been evaluated at an outside community practice due to dyspareunia, vulvar discomfort, and vulvar irregularities of 1 month’s duration. At that time, a small biopsy was performed, and the histologic differential diagnosis included syringoma vs an adnexal carcinoma. Consequently, she was referred to gynecologic oncology for further management.
Pelvic examination revealed multilobular nodular areas overlying the clitoral hood that extended down to the labia majora. The nodular processes did not involve the clitoris, labia minora, or perineum. A mobile isolated lymph node measuring 2.0×1.0 cm in the right inguinal area also was noted. The patient’s clinical history was notable for right breast carcinoma treated with a right mastectomy with axillary lymph node dissection that showed metastatic disease. She also underwent adjuvant chemotherapy with paclitaxel and doxorubicin for breast carcinoma.
After discussing the diagnostic differential and treatment options, the patient elected to undergo a bilateral partial radical vulvectomy with reconstruction and resection of the right inguinal lymph node. Gross examination of the vulvectomy specimen showed multiple flesh-colored papules (FIGURE 1). Histologic examination revealed a neoplasm with sweat gland differentiation that was broad and poorly circumscribed but confined to the dermis (FIGURES 2A and 2B). The neoplasm was composed of epithelial cells that formed ductlike structures, lined by 2 layers of cuboidal epithelium within a fibrous stroma (FIGURE 2C). A toluidine blue special stain was performed and demonstrated an increased amount of mast cells in the tissue (FIGURE 3). Immunohistochemical stains for gross cystic disease fluid protein, estrogen receptor (ER), and progesterone receptor (PR) were negative in the tumor cells. The lack of cytologic atypia, perineural invasion, and deep infiltration into the subcutis favored a syringoma. One month later, the case was presented at the Tumor Board Conference at the University of Alabama at Birmingham where a final diagnosis of vulvar syringoma was agreed upon and discussed with the patient. At that time, no recurrence was evident and follow-up was recommended.
Syringomas are benign tumors of the sweat glands that are fairly common and appear to have a predilection for women. Although most of the literature classifies them as eccrine neoplasms, the term syringoma can be used to describe neoplasms of either apocrine or eccrine lineage.1 To rule out an apocrine lineage of the tumor in our patient, we performed immunohistochemistry for gross cystic disease fluid protein, a marker of apocrine differentiation. This stain highlighted normal apocrine glands that were not involved in the tumor proliferation.
Syringomas may occur at any site on the body but are prone to occur on the periorbital area, especially the eyelids.1 Some of the atypical locations for a syringoma include the anterior neck, chest, abdomen, genitals, axillae, groin, and buttocks.2 Vulvar syringomas were first reported by Carneiro3 in 1971 as usually affecting adolescent girls and middle-aged women. There have been approximately 40 reported cases affecting women aged 8 to 78 years.4,5 Vulvar syringomas classically appear as firm or soft, flesh-colored to transparent, papular lesions. The 2 other clinical variants are miliumlike, whitish, cystic papules as well as lichenoid papules.6 Pérez-Bustillo et al5 reported a case of the lichenoid papule variant on the labia majora of a 78-year-old woman who presented with intermittent vulvar pruritus of 4 years’ duration. Due to this patient’s 9-year history of urinary incontinence, the lesions had been misdiagnosed as irritant dermatitis and associated lichen simplex chronicus (LSC). This case is a reminder to consider vulvar syringoma in patients with LSC who respond poorly to oral antihistamines and topical steroids.5 Rarely, multiple clinical variants may coexist. In a case reported by Dereli et al,7 a 19-year-old woman presented with concurrent classical and miliumlike forms of vulvar syringoma.
Vulvar syringomas usually present as multiple lesions involving both sides of the labia majora; however, Blasdale and McLelland8 reported a single isolated syringoma of the vulva on the anterior right labia minora that measured 1.0×0.5 cm, leading the lesion to be described as a giant syringoma.
Vulvar syringomas usually are asymptomatic and noticed during routine gynecologic examination. Therefore, it is believed that they likely are underdiagnosed.5 When symptomatic, they commonly present with constant9 or intermittent5 pruritus, which may intensify during menstruation, pregnancy, and summertime.6,10-12 Gerdsen et al10 documented a 27-year-old woman who presented with a 2-year history of pruritic vulvar skin lesions that became exacerbated during menstruation, which raised the possibility of cyclical hormonal changes being responsible for periodic exacerbation of vulvar pruritus during menstruation. In addition, patients may experience an increase in size and number of the lesions during pregnancy. Bal et al11 reported a 24-year-old primigravida with vulvar papular lesions that intensified during pregnancy. She had experienced intermittent vulvar pruritus for 12 years but had no change in symptoms during menstruation.11 Few studies have attempted to evaluate the presence of ER and PR in the syringomas. A study of 9 nonvulvar syringomas by Wallace and Smoller13 showed ER positivity in 1 case and PR positivity in 8 cases, lending support to the hormonal theory; however, in another case series of 15 vulvar syringomas, Huang et al6 failed to show ER and PR expression by immunohistochemical staining. A case report published 3 years earlier documented the first case of PR positivity on a vulvar syringoma.14 Our patient also was negative for ER and PR, which suggested that hormonal status is important in some but not all syringomas.
Patients with vulvar syringomas also might have coexisting extragenital syringomas in the neck,4 eyelids,6,7,10 and periorbital area,6 and thorough examination of the body is essential. If an extragenital syringoma is diagnosed, a vulvar syringoma should be considered, especially when the patient presents with unexplained genital symptoms. Although no proven hereditary transmission pattern has been established, family history of syringomas has been established in several cases.15 In a case series reported by Huang et al,6 4 of 18 patients reported a family history of periorbital syringomas. In our case, the patient did not report a family history of syringomas.
The differential diagnosis of vulvar lesions with pruritus is broad and includes Fox-Fordyce disease, lichen planus, LSC, epidermal cysts, senile angiomas, dystrophic calcinosis, xanthomas, steatocytomas, soft fibromas, condyloma acuminatum, and candidiasis. Vulvar syringomas might have a nonspecific appearance, and histologic examination is essential to confirm the diagnosis and rule out any malignant process such as MAC, vulvar intraepithelial neoplasia, extramammary Paget disease, or other glandular neoplasms of the vulva.
Microcystic adnexal carcinoma was first reported in 1982 by Goldstein et al16 as a locally aggressive neoplasm that can be confused with benign adnexal neoplasms, particularly desmoplastic trichoepithelioma, trichoadenoma, and syringoma. Microcystic adnexal carcinomas present as slow-growing, flesh-colored papules that may resemble syringomas and appear in similar body sites. Histologic examination is essential to differentiate between these two entities. Syringomas are tumors confined to the dermis and are composed of multiple small ducts lined by 2 layers of cuboidal epithelium within a dense fibrous stroma. Unlike syringomas, MACs usually infiltrate diffusely into the dermis and subcutis and may extend into the underlying muscle. Although bland cytologic features predominate, perineural invasion frequently is present in MACs. A potential pitfall of misdiagnosis can be caused by a superficial biopsy that may reveal benign histologic appearance, particularly in the upper level of the tumor where it may be confused with a syringoma or a benign follicular neoplasm.17
The initial biopsy performed on our patient was possibly not deep enough to render an unequivocal diagnosis and therefore bilateral partial radical vulvectomy was considered. After surgery, histologic examination of the resection specimen revealed a poorly circumscribed tumor confined to the dermis. The tumor was broad and the lack of deep infiltration into the subcutis and perineural invasion favored a syringoma (FIGURES 2A and 2B). These findings were consistent with case reports that documented syringomas as being more wide than deep on microscopic examination, whereas the opposite pertained to MAC.18 Cases of plaque-type syringomas that initially were misdiagnosed as MACs also have been reported.19 Because misdiagnosis may affect the treatment plan and potentially result in unnecessary surgery, caution should be taken when differentiating between these two entities. When a definitive diagnosis cannot be rendered on a superficial biopsy, a recommendation should be made for a deeper biopsy sampling the subcutis.
For the majority of the patients with vulvar syringomas, treatment is seldom required due to their asymptomatic nature; however, patients who present with symptoms usually report pruritus of variable intensities and patterns. A standardized treatment does not exist for vulvar syringomas, and oral or topical treatment might be used as an initial approach. Commonly prescribed medications with variable results include topical corticosteroids, oral antihistamines, and topical retinoids. In a case reported by Iwao et al,20 vulvar syringomas were successfully treated with tranilast, which has anti-inflammatory and immunomodulatory effects. This medication could have a possible dual action—inhibiting the release of chemical mediators from the mast cells and inhibiting the release of IL-1β from the eccrine duct, which could suppress the proliferation of stromal connective tissue. Our case was stained with toluidine blue and showed an increased number of mast cells in the tissue (FIGURE 3).Patients who are unresponsive to tranilast or have extensive disease resulting in cosmetic disfigurement might benefit from more invasive treatment methods including a variety of lasers, cryotherapy, electrosurgery, and excision. Excisions should include the entire tumor to avoid recurrence. In a case reported by Garman and Metry,21 the lesions were surgically excised using small 2- to 3-mm punches; however, several weeks later the lesions recurred. Our patient presented with a 1-month evolution of dyspareunia, vulvar discomfort, and vulvar irregularities that were probably not treated with oral or topical medications before being referred for surgery.
We report a case of a vulvar syringoma that presented diagnostic challenges in the initial biopsy, which prevented the exclusion of an MAC. After partial radical vulvectomy, histologic examination was more definitive, showing lack of deep infiltration into the subcutis or perineural invasion that are commonly seen in MAC. This case is an example of a notable pitfall in the diagnosis of vulvar syringoma on a limited biopsy leading to overtreatment. Raising awareness of this entity is the only modality to prevent misdiagnosis. We encourage reporting of further cases of syringomas, particularly those with atypical locations or patterns that may cause diagnostic problems. ●
- Ensure adequate depth of biopsy to assist in the histologic diagnosis of syringoma vs microcystic adnexal carcinoma.
- Vulvar syringomas also may contribute to notable pruritus and ultimately be the underlying etiology for secondary skin changes leading to a lichen simplex chronicus–like phenotype
To the Editor:
Syringomas are common benign tumors of the eccrine sweat glands that usually manifest clinically as multiple flesh-colored papules. They are most commonly seen on the face, neck, and chest of adolescent girls. Syringomas may appear at any site of the body but are rare in the vulva. We present a case of a 51-year-old woman who was referred to the Division of Gynecologic Oncology at the University of Alabama at Birmingham for further management of a tumor carrying a differential diagnosis of vulvar syringoma vs microcystic adnexal carcinoma (MAC).
A 51-year-old woman presented to dermatology (G.G.) and was referred to the Division of Gynecologic Oncology at the University of Alabama at Birmingham for further management of possible vulvar syringoma vs MAC. The patient previously had been evaluated at an outside community practice due to dyspareunia, vulvar discomfort, and vulvar irregularities of 1 month’s duration. At that time, a small biopsy was performed, and the histologic differential diagnosis included syringoma vs an adnexal carcinoma. Consequently, she was referred to gynecologic oncology for further management.
Pelvic examination revealed multilobular nodular areas overlying the clitoral hood that extended down to the labia majora. The nodular processes did not involve the clitoris, labia minora, or perineum. A mobile isolated lymph node measuring 2.0×1.0 cm in the right inguinal area also was noted. The patient’s clinical history was notable for right breast carcinoma treated with a right mastectomy with axillary lymph node dissection that showed metastatic disease. She also underwent adjuvant chemotherapy with paclitaxel and doxorubicin for breast carcinoma.
After discussing the diagnostic differential and treatment options, the patient elected to undergo a bilateral partial radical vulvectomy with reconstruction and resection of the right inguinal lymph node. Gross examination of the vulvectomy specimen showed multiple flesh-colored papules (FIGURE 1). Histologic examination revealed a neoplasm with sweat gland differentiation that was broad and poorly circumscribed but confined to the dermis (FIGURES 2A and 2B). The neoplasm was composed of epithelial cells that formed ductlike structures, lined by 2 layers of cuboidal epithelium within a fibrous stroma (FIGURE 2C). A toluidine blue special stain was performed and demonstrated an increased amount of mast cells in the tissue (FIGURE 3). Immunohistochemical stains for gross cystic disease fluid protein, estrogen receptor (ER), and progesterone receptor (PR) were negative in the tumor cells. The lack of cytologic atypia, perineural invasion, and deep infiltration into the subcutis favored a syringoma. One month later, the case was presented at the Tumor Board Conference at the University of Alabama at Birmingham where a final diagnosis of vulvar syringoma was agreed upon and discussed with the patient. At that time, no recurrence was evident and follow-up was recommended.
Syringomas are benign tumors of the sweat glands that are fairly common and appear to have a predilection for women. Although most of the literature classifies them as eccrine neoplasms, the term syringoma can be used to describe neoplasms of either apocrine or eccrine lineage.1 To rule out an apocrine lineage of the tumor in our patient, we performed immunohistochemistry for gross cystic disease fluid protein, a marker of apocrine differentiation. This stain highlighted normal apocrine glands that were not involved in the tumor proliferation.
Syringomas may occur at any site on the body but are prone to occur on the periorbital area, especially the eyelids.1 Some of the atypical locations for a syringoma include the anterior neck, chest, abdomen, genitals, axillae, groin, and buttocks.2 Vulvar syringomas were first reported by Carneiro3 in 1971 as usually affecting adolescent girls and middle-aged women. There have been approximately 40 reported cases affecting women aged 8 to 78 years.4,5 Vulvar syringomas classically appear as firm or soft, flesh-colored to transparent, papular lesions. The 2 other clinical variants are miliumlike, whitish, cystic papules as well as lichenoid papules.6 Pérez-Bustillo et al5 reported a case of the lichenoid papule variant on the labia majora of a 78-year-old woman who presented with intermittent vulvar pruritus of 4 years’ duration. Due to this patient’s 9-year history of urinary incontinence, the lesions had been misdiagnosed as irritant dermatitis and associated lichen simplex chronicus (LSC). This case is a reminder to consider vulvar syringoma in patients with LSC who respond poorly to oral antihistamines and topical steroids.5 Rarely, multiple clinical variants may coexist. In a case reported by Dereli et al,7 a 19-year-old woman presented with concurrent classical and miliumlike forms of vulvar syringoma.
Vulvar syringomas usually present as multiple lesions involving both sides of the labia majora; however, Blasdale and McLelland8 reported a single isolated syringoma of the vulva on the anterior right labia minora that measured 1.0×0.5 cm, leading the lesion to be described as a giant syringoma.
Vulvar syringomas usually are asymptomatic and noticed during routine gynecologic examination. Therefore, it is believed that they likely are underdiagnosed.5 When symptomatic, they commonly present with constant9 or intermittent5 pruritus, which may intensify during menstruation, pregnancy, and summertime.6,10-12 Gerdsen et al10 documented a 27-year-old woman who presented with a 2-year history of pruritic vulvar skin lesions that became exacerbated during menstruation, which raised the possibility of cyclical hormonal changes being responsible for periodic exacerbation of vulvar pruritus during menstruation. In addition, patients may experience an increase in size and number of the lesions during pregnancy. Bal et al11 reported a 24-year-old primigravida with vulvar papular lesions that intensified during pregnancy. She had experienced intermittent vulvar pruritus for 12 years but had no change in symptoms during menstruation.11 Few studies have attempted to evaluate the presence of ER and PR in the syringomas. A study of 9 nonvulvar syringomas by Wallace and Smoller13 showed ER positivity in 1 case and PR positivity in 8 cases, lending support to the hormonal theory; however, in another case series of 15 vulvar syringomas, Huang et al6 failed to show ER and PR expression by immunohistochemical staining. A case report published 3 years earlier documented the first case of PR positivity on a vulvar syringoma.14 Our patient also was negative for ER and PR, which suggested that hormonal status is important in some but not all syringomas.
Patients with vulvar syringomas also might have coexisting extragenital syringomas in the neck,4 eyelids,6,7,10 and periorbital area,6 and thorough examination of the body is essential. If an extragenital syringoma is diagnosed, a vulvar syringoma should be considered, especially when the patient presents with unexplained genital symptoms. Although no proven hereditary transmission pattern has been established, family history of syringomas has been established in several cases.15 In a case series reported by Huang et al,6 4 of 18 patients reported a family history of periorbital syringomas. In our case, the patient did not report a family history of syringomas.
The differential diagnosis of vulvar lesions with pruritus is broad and includes Fox-Fordyce disease, lichen planus, LSC, epidermal cysts, senile angiomas, dystrophic calcinosis, xanthomas, steatocytomas, soft fibromas, condyloma acuminatum, and candidiasis. Vulvar syringomas might have a nonspecific appearance, and histologic examination is essential to confirm the diagnosis and rule out any malignant process such as MAC, vulvar intraepithelial neoplasia, extramammary Paget disease, or other glandular neoplasms of the vulva.
Microcystic adnexal carcinoma was first reported in 1982 by Goldstein et al16 as a locally aggressive neoplasm that can be confused with benign adnexal neoplasms, particularly desmoplastic trichoepithelioma, trichoadenoma, and syringoma. Microcystic adnexal carcinomas present as slow-growing, flesh-colored papules that may resemble syringomas and appear in similar body sites. Histologic examination is essential to differentiate between these two entities. Syringomas are tumors confined to the dermis and are composed of multiple small ducts lined by 2 layers of cuboidal epithelium within a dense fibrous stroma. Unlike syringomas, MACs usually infiltrate diffusely into the dermis and subcutis and may extend into the underlying muscle. Although bland cytologic features predominate, perineural invasion frequently is present in MACs. A potential pitfall of misdiagnosis can be caused by a superficial biopsy that may reveal benign histologic appearance, particularly in the upper level of the tumor where it may be confused with a syringoma or a benign follicular neoplasm.17
The initial biopsy performed on our patient was possibly not deep enough to render an unequivocal diagnosis and therefore bilateral partial radical vulvectomy was considered. After surgery, histologic examination of the resection specimen revealed a poorly circumscribed tumor confined to the dermis. The tumor was broad and the lack of deep infiltration into the subcutis and perineural invasion favored a syringoma (FIGURES 2A and 2B). These findings were consistent with case reports that documented syringomas as being more wide than deep on microscopic examination, whereas the opposite pertained to MAC.18 Cases of plaque-type syringomas that initially were misdiagnosed as MACs also have been reported.19 Because misdiagnosis may affect the treatment plan and potentially result in unnecessary surgery, caution should be taken when differentiating between these two entities. When a definitive diagnosis cannot be rendered on a superficial biopsy, a recommendation should be made for a deeper biopsy sampling the subcutis.
For the majority of the patients with vulvar syringomas, treatment is seldom required due to their asymptomatic nature; however, patients who present with symptoms usually report pruritus of variable intensities and patterns. A standardized treatment does not exist for vulvar syringomas, and oral or topical treatment might be used as an initial approach. Commonly prescribed medications with variable results include topical corticosteroids, oral antihistamines, and topical retinoids. In a case reported by Iwao et al,20 vulvar syringomas were successfully treated with tranilast, which has anti-inflammatory and immunomodulatory effects. This medication could have a possible dual action—inhibiting the release of chemical mediators from the mast cells and inhibiting the release of IL-1β from the eccrine duct, which could suppress the proliferation of stromal connective tissue. Our case was stained with toluidine blue and showed an increased number of mast cells in the tissue (FIGURE 3).Patients who are unresponsive to tranilast or have extensive disease resulting in cosmetic disfigurement might benefit from more invasive treatment methods including a variety of lasers, cryotherapy, electrosurgery, and excision. Excisions should include the entire tumor to avoid recurrence. In a case reported by Garman and Metry,21 the lesions were surgically excised using small 2- to 3-mm punches; however, several weeks later the lesions recurred. Our patient presented with a 1-month evolution of dyspareunia, vulvar discomfort, and vulvar irregularities that were probably not treated with oral or topical medications before being referred for surgery.
We report a case of a vulvar syringoma that presented diagnostic challenges in the initial biopsy, which prevented the exclusion of an MAC. After partial radical vulvectomy, histologic examination was more definitive, showing lack of deep infiltration into the subcutis or perineural invasion that are commonly seen in MAC. This case is an example of a notable pitfall in the diagnosis of vulvar syringoma on a limited biopsy leading to overtreatment. Raising awareness of this entity is the only modality to prevent misdiagnosis. We encourage reporting of further cases of syringomas, particularly those with atypical locations or patterns that may cause diagnostic problems. ●
- Ensure adequate depth of biopsy to assist in the histologic diagnosis of syringoma vs microcystic adnexal carcinoma.
- Vulvar syringomas also may contribute to notable pruritus and ultimately be the underlying etiology for secondary skin changes leading to a lichen simplex chronicus–like phenotype
- Bolognia JL, Jorizzo JL, Rapini RP. Dermatology. 2nd ed. Spain: Mosby Elsevier; 2008.
- Weedon D. Skin Pathology. 3rd ed. China: Churchill Livingstone Elsevier; 2010.
- Carneiro SJ, Gardner HL, Knox JM. Syringoma of the vulva. Arch Dermatol. 1971;103:494-496.
- Trager JD, Silvers J, Reed JA, et al. Neck and vulvar papules in an 8-year-old girl. Arch Dermatol. 1999;135:203, 206.
- Pérez-Bustillo A, Ruiz-González I, Delgado S, et al. Vulvar syringoma: a rare cause of vulvar pruritus. Actas DermoSifiliográficas. 2008; 99:580-581.
- Huang YH, Chuang YH, Kuo TT, et al. Vulvar syringoma: a clinicopathologic and immunohistologic study of 18 patients and results of treatment. J Am Acad Dermatol. 2003;48:735-739.
- Dereli T, Turk BG, Kazandi AC. Syringomas of the vulva. Int J Gynaecol Obstet. 2007;99:65-66.
- Blasdale C, McLelland J. Solitary giant vulval syringoma. Br J Dermatol. 1999;141:374-375.
- Kavala M, Can B, Zindanci I, et al. Vulvar pruritus caused by syringoma of the vulva. Int J Dermatol. 2008;47:831-832.
- Gerdsen R, Wenzel J, Uerlich M, et al. Periodic genital pruritus caused by syringoma of the vulva. Acta Obstet Gynecol Scand. 2002;81:369-370.
- Bal N, Aslan E, Kayaselcuk F, et al. Vulvar syringoma aggravated by pregnancy. Pathol Oncol Res. 2003;9:196-197.
- Turan C, Ugur M, Kutluay L, et al. Vulvar syringoma exacerbated during pregnancy. Eur J Obstet Gynecol Reprod Biol. 1996;64:141-142.
- Wallace ML, Smoller BR. Progesterone receptor positivity supports hormonal control of syringomas. J Cutan Pathol. 1995; 22:442-445.
- Yorganci A, Kale A, Dunder I, et al. Vulvar syringoma showing progesterone receptor positivity. BJOG. 2000;107:292-294.
- Draznin M. Hereditary syringomas: a case report. Dermatol Online J. 2004;10:19.
- Goldstein DJ, Barr RJ, Santa Cruz DJ. Microcystic adnexal carcinoma: a distinct clinicopathologic entity. Cancer. 1982;50:566-572.
- Hamsch C, Hartschuh W. Microcystic adnexal carcinomaaggressive infiltrative tumor often with innocent clinical appearance. J Dtsch Dermatol Ges. 2010;8:275-278.
- Henner MS, Shapiro PE, Ritter JH, et al. Solitary syringoma. report of five cases and clinicopathologic comparison with microcystic adnexal carcinoma of the skin. Am J Dermatopathol. 1995;17:465-470.
- Suwattee P, McClelland MC, Huiras EE, et al. Plaque-type syringoma: two cases misdiagnosed as microcystic adnexal carcinoma. J Cutan Pathol. 2008;35:570-574.
- Iwao F, Onozuka T, Kawashima T. Vulval syringoma successfully treated with tranilast. Br J Dermatol. 2005;153:1228-1230.
- Garman M, Metry D. Vulvar syringomas in a 9-year-old child with review of the literature. Pediatr Dermatol. 2006;23:369372.
- Bolognia JL, Jorizzo JL, Rapini RP. Dermatology. 2nd ed. Spain: Mosby Elsevier; 2008.
- Weedon D. Skin Pathology. 3rd ed. China: Churchill Livingstone Elsevier; 2010.
- Carneiro SJ, Gardner HL, Knox JM. Syringoma of the vulva. Arch Dermatol. 1971;103:494-496.
- Trager JD, Silvers J, Reed JA, et al. Neck and vulvar papules in an 8-year-old girl. Arch Dermatol. 1999;135:203, 206.
- Pérez-Bustillo A, Ruiz-González I, Delgado S, et al. Vulvar syringoma: a rare cause of vulvar pruritus. Actas DermoSifiliográficas. 2008; 99:580-581.
- Huang YH, Chuang YH, Kuo TT, et al. Vulvar syringoma: a clinicopathologic and immunohistologic study of 18 patients and results of treatment. J Am Acad Dermatol. 2003;48:735-739.
- Dereli T, Turk BG, Kazandi AC. Syringomas of the vulva. Int J Gynaecol Obstet. 2007;99:65-66.
- Blasdale C, McLelland J. Solitary giant vulval syringoma. Br J Dermatol. 1999;141:374-375.
- Kavala M, Can B, Zindanci I, et al. Vulvar pruritus caused by syringoma of the vulva. Int J Dermatol. 2008;47:831-832.
- Gerdsen R, Wenzel J, Uerlich M, et al. Periodic genital pruritus caused by syringoma of the vulva. Acta Obstet Gynecol Scand. 2002;81:369-370.
- Bal N, Aslan E, Kayaselcuk F, et al. Vulvar syringoma aggravated by pregnancy. Pathol Oncol Res. 2003;9:196-197.
- Turan C, Ugur M, Kutluay L, et al. Vulvar syringoma exacerbated during pregnancy. Eur J Obstet Gynecol Reprod Biol. 1996;64:141-142.
- Wallace ML, Smoller BR. Progesterone receptor positivity supports hormonal control of syringomas. J Cutan Pathol. 1995; 22:442-445.
- Yorganci A, Kale A, Dunder I, et al. Vulvar syringoma showing progesterone receptor positivity. BJOG. 2000;107:292-294.
- Draznin M. Hereditary syringomas: a case report. Dermatol Online J. 2004;10:19.
- Goldstein DJ, Barr RJ, Santa Cruz DJ. Microcystic adnexal carcinoma: a distinct clinicopathologic entity. Cancer. 1982;50:566-572.
- Hamsch C, Hartschuh W. Microcystic adnexal carcinomaaggressive infiltrative tumor often with innocent clinical appearance. J Dtsch Dermatol Ges. 2010;8:275-278.
- Henner MS, Shapiro PE, Ritter JH, et al. Solitary syringoma. report of five cases and clinicopathologic comparison with microcystic adnexal carcinoma of the skin. Am J Dermatopathol. 1995;17:465-470.
- Suwattee P, McClelland MC, Huiras EE, et al. Plaque-type syringoma: two cases misdiagnosed as microcystic adnexal carcinoma. J Cutan Pathol. 2008;35:570-574.
- Iwao F, Onozuka T, Kawashima T. Vulval syringoma successfully treated with tranilast. Br J Dermatol. 2005;153:1228-1230.
- Garman M, Metry D. Vulvar syringomas in a 9-year-old child with review of the literature. Pediatr Dermatol. 2006;23:369372.
Treatment of Angiosarcoma of the Head and Neck: A Systematic Review
Cutaneous angiosarcoma (cAS) is a rare malignancy arising from vascular or lymphatic tissue. It classically presents during the sixth or seventh decades of life as a raised purple papule or plaque on the head and neck areas.1 Primary cAS frequently mimics benign conditions, leading to delays in care. Such delays coupled with the aggressive nature of angiosarcomas leads to a poor prognosis. Five-year survival rates range from 11% to 50%, and more than half of patients die within 1 year of diagnosis.2-7
Currently, there is no consensus on the most effective treatments, as the rare nature of cAS has made the development of controlled clinical trials difficult. Wide local excision (WLE) is most frequently employed; however, the tumor’s infiltrative growth makes complete resection and negative surgical margins difficult to achieve.8 Recently, Mohs micrographic surgery (MMS) has been postulated as a treatment option. The tissue-sparing nature and intraoperative margin control of MMS may provide tumor eradication and cosmesis benefits reported with other cutaneous malignancies.9
Nearly all localized cASs are treated with surgical excision with or without adjuvant treatment modalities; however, it is unclear which of these modalities provide a survival benefit. We conducted a systematic review of the literature to compare treatment modalities for localized cAS of the head and neck regions and to compare treatments based on tumor stage.
METHODS
A literature search was performed to identify published studies indexed by MEDLINE, Cochrane Central Register of Controlled Trials (CENTRAL), Embase, and PubMed from January 1, 1977, to May 8, 2020, reporting on cAS and treatment modalities used. The search was conducted in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analysis guidelines.5 Data extracted included patient demographics, tumor characteristics (including T1 [≤5 cm] and T2 [>5 cm and ≤10 cm] based on the American Joint Committee on Cancer soft tissue sarcoma staging criteria), treatments used, follow-up time, overall survival (OS) rates, and complications.10,11
Studies were required to (1) include participants with head and neck cAS; (2) report original patient data following cAS treatment with surgical (WLE or MMS) and/or nonsurgical modalities (chemotherapy [CT], radiotherapy [RT], immunotherapy [IT]); (3) report outcome data related to OS rates following treatment; and (4) have articles published in English. Given the rare nature of cAS, there was no limitation on the number of participants needed.
The Newcastle-Ottawa scale for observational studies was used to assess the quality of studies.12 Higher scores indicate low risk of bias, while lower scores represent high risk of bias.
Continuous data were reported with means and SDs, while categorical variables were reported as percentages. Overall survival means and SDs were compared between treatment modalities using an independent sample t test with P<.05 considered statistically significant. Due to the heterogeneity of the data, a meta-analysis was not reported.
RESULTS
Literature Search and Risk of Bias Assessment
There were 283 manuscripts identified, 56 articles read in full, and 40 articles included in the review (Figure). Among the 16 studies not meeting inclusion criteria, 7 did not provide enough data to isolate head and neck cAS cases,1,13-18 6 did not report outcomes related to the current review,19-24 and 3 did not provide enough data to isolate different treatment outcomes.25-27 Among the included studies, 32 reported use of WLE: WLE alone (n=21)2,7,11,28-45; WLE with RT (n=24)2,3,11,28-31,33-36,38-41,43-51; WLE with CT (n=7)2,31,35,39,41,48,52; WLE with RT and CT (n=11)2,29,31,33-35,39,40,48,52,53; WLE with RT and IT (n=3)35,54,55; and WLE with RT, CT, and IT (n=1).53 Nine studies reported MMS: MMS alone (n=5)39,56-59; MMS with RT (n=3)32,50,60,61; and MMS with RT and CT (n=1).51
Risk of bias assessment identified low risk in 3 articles. High risk was identified in 5 case reports,57-61 and 1 study did not describe patient selection.43 Clayton et al56 showed intermediate risk, given the study controlled for 1 factor.
Patient Demographics
A total of 1295 patients were included. The pooled mean age of the patients was 67.5 years (range, 3–88 years), and 64.7% were male. There were 79 cases identified as T1 and 105 as T2. A total of 825 cases were treated using WLE with or without adjuvant therapy, while a total of 9 cases were treated using MMS with and without adjuvant therapies (Table). There were 461 cases treated without surgical excision: RT alone (n=261), CT alone (n=38), IT alone (n=35), RT with CT (n=81), RT with IT (n=34), and RT with CT and IT (n=12)(Table). The median follow-up period across all studies was 23.5 months (range, 1–228 months).
Comparison Between Surgical and Nonsurgical Modalities
Wide Local Excision—Wide local excision (n=825; 63.7%) alone or in combination with other therapies was the most frequently used treatment modality. The mean (SD) OS was longest for WLE with RT, CT, and IT (n=3; 39.3 [24.1]), followed by WLE with RT (n=447; 35.9 [34.3] months), WLE with CT (n=13; 32.4 [30.2] months), WLE alone (n=324; 29.6 [34.1] months), WLE with RT and IT (n=11; 23.5 [4.9] months), and WLE with RT and CT (n=27; 20.7 [13.1] months).
Nonsurgical Modalities—Nonsurgical methods were used less frequently than surgical methods (n=461; 35.6%). The mean (SD) OS time in descending order was as follows: RT with CT and IT (n=12; 34.9 [1.2] months), RT with CT (n=81; 30.4 [37.8] months), IT alone (n=35; 25.7 [no SD reported] months), RT with IT (n=34; 20.5 [8.6] months), CT alone (n=38; 20.1 [15.9] months), and RT alone (n=261; 12.8 [8.3] months).
When comparing mean (SD) OS outcomes between surgical and nonsurgical treatment modalities, only the addition of WLE to RT significantly increased OS when compared with RT alone (WLE, 35.9 [34.3] months; RT alone, 12.8 [8.3] months; P=.001). When WLE was added to CT or both RT and CT, there was no significant difference with OS when compared with CT alone (WLE with CT, 32.4 [30.2] months; CT alone, 20.1 [15.9] months; P=.065); or both RT and CT in combination (WLE with RT and CT, 20.7 [13.1] months; RT and CT, 30.4 [37.8] months; P=.204).
Comparison Between T1 and T2 cAS
T1 Angiosarcoma—There were 79 patients identified as having T1 tumors across 16 studies.2,31,32,34,39-41,46,48-50,53,58-60,62 The mean (SD) OS was longest for WLE with RT, CT, and IT (n=2; 56.0 [6.0] months), followed by WLE with CT (n=4; 54.5 [41.0] months); WLE with RT (n=30; 39.7 [41.2] months); WLE alone (n=22; 37.2 [37.3] months); WLE with both RT and CT (n=7; 25.5 [18.7] months); RT with IT (n=2; 20.0 [11.0] months); RT with CT (n=6; 15.7 [6.8] months); and RT alone (n=1; 13 [no SD]) months)(eTable).
T2 Angiosarcoma—There were 105 patients with T2 tumors in 15 studies.2,31,32,34,39-41,46,48-50,52,53,57,62 The mean (SD) OS for each treatment modality in descending order was as follows: RT with CT and IT (n=1; 36 [no SD reported] months); RT with CT (n=23; 34.3 [46.3] months); WLE with RT (n=21; 26.3 [23.8] months); WLE with CT (n=8; 21.5 [16.6] months); WLE alone (n=16; 19.8 [15.6] months); WLE with RT and CT (n=14; 19.2 [10.5] months); RT alone (n=17; 10.1 [5.5] months); CT alone (n=2; 6.7 [3.7] months); and WLE with RT, CT, and IT (n=1; 6.0 [no SD] months)(eTable).
Mohs Micrographic Surgery—The use of MMS was only identified in case reports or small observational studies for a total of 9 patients. Five cASs were treated with MMS alone for a mean (SD) OS of 37 (21.5) months, with 4 reporting cAS staging: 2 were T158,59 (mean [SD] OS, 37.0 [17.0] months) and 2 were T2 tumors39,57 (mean [SD] OS, 44.5 [26.5] months). Mohs micrographic surgery with RT was used for 3 tumors (mean [SD] OS, 34.0 [26.9] months); 2 were T150,60 (mean [SD] OS, 42.0 [30.0] months) and 1 unreported staging (eTable).56 Mohs micrographic surgery with both RT and CT was used in 1 patient (unreported staging; OS, 82 months).51
Complications
Complications were rare and mainly associated with CT and RT. Four studies reported radiation dermatitis with RT.53,55,62,63 Two studies reported peripheral neuropathy and myelotoxicity with CT.35,51 Only 1 study reported poor wound healing due to surgical complications.29
COMMENT
Cutaneous angiosarcomas are rare and have limited treatment guidelines. Surgical excision does appear to be an effective adjunct to nonsurgical treatments, particularly WLE combined with RT, CT, and IT. Although MMS ultimately may be useful for cAS, the limited number and substantial heterogeneity of reported cases precludes definitive conclusions at this time.
Achieving margin control during WLE is associated with higher OS when treating angiosarcoma,36,46 which is particularly true for T1 tumors where margin control is imperative, and many cases are treated with a combination of WLE and RT. Overall survival times are lower for T2 tumors, as these tumors are larger and most likely have spread; therefore, more aggressive combination treatments were more prevalent. In these cases, complete margin control may be difficult to achieve and may not be as critical to the outcome if another form of adjuvant therapy can be administered promptly.24,64
When surgery is contraindicated, RT with or without CT was the most commonly reported treatment modality. However, these treatments were notably less effective than when used in combination with surgical resection. The use of RT alone has a recurrence rate reported up to 100% in certain studies, suggesting the need to utilize RT in combination with other modalities.23,39 It is important to note that RT often is used as monotherapy in palliative treatment, which may indirectly skew survival rates.2
Limitations of the study include a lack of randomized controlled trials. Most reports were retrospective reviews or case series, and tumor staging was sparsely reported. Finally, although MMS may provide utility in the treatment of cAS, the sample size of 9 precluded definitive conclusions from being formed about its efficacy.
CONCLUSION
Cutaneous angiosarcoma is rare and has limited data comparing different treatment modalities. The paucity of data currently limits definitive recommendations; however, both surgical and nonsurgical modalities have demonstrated potential efficacy in the treatment of cAS and may benefit from additional research. Clinicians should consider a multidisciplinary approach for patients with a diagnosis of cAS to tailor treatments on a case-by-case basis.
- Rodríguez-Jiménez P, Jimenez YD, Reolid A, et al. State of the art of Mohs surgery for rare cutaneous tumors in the Spanish Registry of Mohs Surgery (REGESMOHS). Int J Dermatol. 2020;59:321-325.
- Alqumber NA, Choi JW, Kang MK. The management and prognosis of facial and scalp angiosarcoma: a retrospective analysis of 15 patients. Ann Plast Surg. 2019;83:55-62.
- Pawlik TM, Paulino AF, McGinn CJ, et al. Cutaneous angiosarcoma of the scalp: a multidisciplinary approach. Cancer. 2003;98:1716-1726.
- Deyrup AT, McKenney JK, Tighiouart M, et al. Sporadic cutaneous angiosarcomas: a proposal for risk stratification based on 69 cases. Am J Surg Pathol. 2008;32:72-77.
- Meis-Kindblom JM, Kindblom LG. Angiosarcoma of soft tissue: a study of 80 cases. Am J Surg Pathol. 1998;22:683-697.
- Harbour P, Song DH. The skin and subcutaneous tissue. In: Brunicardi FC, Andersen DK, Billiar TR, et al, eds. Schwartz’s Principles of Surgery. 11th ed. McGraw-Hill Education; 2019. Accessed April 24, 2023. https://accesssurgery.mhmedical.com/content.aspx?bookid=2576§ionid=216206374
- Oashi K, Namikawa K, Tsutsumida A, et al. Surgery with curative intent is associated with prolonged survival in patients with cutaneous angiosarcoma of the scalp and face—a retrospective study of 38 untreated cases in the Japanese population. Eur J Surg Oncol. 2018;44:823-829.
- Young RJ, Brown NJ, Reed MW, et al. Angiosarcoma. Lancet Oncol. 2010;11:983-991.
- Tolkachjov SN, Brodland DG, Coldiron BM, et al. Understanding Mohs micrographic surgery: a review and practical guide for the nondermatologist. Mayo Clin Proc. 2017;92:1261-1271.
- Amin M, Edge SB, Greene FL, et al, eds. AJCC Cancer Staging Manual. 8th ed. Springer; 2017.
- Holden CA, Spittle MF, Jones EW. Angiosarcoma of the face and scalp, prognosis and treatment. Cancer. 1987;59:1046-1057.
- Cook DA, Reed DA. Appraising the quality of medical education research methods: the Medical Education Research Study Quality Instrument and the Newcastle-Ottawa Scale-Education. Acad Med. 2015;90:1067-1076.
- Lee BL, Chen CF, Chen PC, et al. Investigation of prognostic features in primary cutaneous and soft tissue angiosarcoma after surgical resection: a retrospective study. Ann Plast Surg. 2017;78(3 suppl 2):S41-S46.
- Shen CJ, Parzuchowski AS, Kummerlowe MN, et al. Combined modality therapy improves overall survival for angiosarcoma. Acta Oncol. 2017;56:1235-1238.
- Breakey RW, Crowley TP, Anderson IB, et al. The surgical management of head and neck sarcoma: the Newcastle experience. J Plast Reconstr Aesthet Surg. 2017;70:78-84.
- Singla S, Papavasiliou P, Powers B, et al. Challenges in the treatment of angiosarcoma: a single institution experience. Am J Surg. 2014;208:254-259.
- Sasaki R, Soejima T, Kishi K, et al. Angiosarcoma treated with radiotherapy: impact of tumor type and size on outcome. Int J Radiat Oncol Biol Phys. 2002;52:1032-1040.
- Naka N, Ohsawa M, Tomita Y, et al. Angiosarcoma in Japan. A review of 99 cases. Cancer. 1995;75:989-996.
- DeMartelaere SL, Roberts D, Burgess MA, et al. Neoadjuvant chemotherapy-specific and overall treatment outcomes in patients with cutaneous angiosarcoma of the face with periorbital involvement. Head Neck. 2008;30:639-646.
- Ward JR, Feigenberg SJ, Mendenhall NP, et al. Radiation therapy for angiosarcoma. Head Neck. 2003;25:873-878.
- Letsa I, Benson C, Al-Muderis O, et al. Angiosarcoma of the face and scalp: effective systemic treatment in the older patient. J Geriatr Oncol. 2014;5:276-280.
- Buehler D, Rice SR, Moody JS, et al. Angiosarcoma outcomes and prognostic factors: a 25-year single institution experience. Am J Clin Oncol. 2014;37:473-479.
- Patel SH, Hayden RE, Hinni ML, et al. Angiosarcoma of the scalp and face: the Mayo Clinic experience. JAMA Otolaryngol Head Neck Surg. 2015;141:335-340.
- Guadagnolo BA, Zagars GK, Araujo D, et al. Outcomes after definitive treatment for cutaneous angiosarcoma of the face and scalp. Head Neck. 2011;33:661-667.
- Zhang Y, Yan Y, Zhu M, et al. Clinical outcomes in primary scalp angiosarcoma. Oncol Lett. 2019;18:5091-5096.
- Kamo R, Ishii M. Histological differentiation, histogenesis and prognosis of cutaneous angiosarcoma. Osaka City Med J. 2011;57:31-44.
- Ito T, Uchi H, Nakahara T, et al. Cutaneous angiosarcoma of the head and face: a single-center analysis of treatment outcomes in 43 patients in Japan. J Cancer Res Clin Oncol. 2016;142:1387-1394.
- Aust MR, Olsen KD, Lewis JE, et al. Angiosarcomas of the head and neck: clinical and pathologic characteristics. Ann Otol Rhinol Laryngol. 1997;106:943-951.
- Buschmann A, Lehnhardt M, Toman N, et al. Surgical treatment of angiosarcoma of the scalp: less is more. Ann Plast Surg. 2008;61:399-403.
- Cassidy RJ, Switchenko JM, Yushak ML, et al. The importance of surgery in scalp angiosarcomas. Surg Oncol. 2018;27:A3-A8.
- Choi JH, Ahn KC, Chang H, et al. Surgical treatment and prognosis of angiosarcoma of the scalp: a retrospective analysis of 14 patients in a single institution. Biomed Res Int. 2015;2015:321896.
- Chow TL, Kwan WW, Kwan CK. Treatment of cutaneous angiosarcoma of the scalp and face in Chinese patients: local experience at a regional hospital in Hong Kong. Hong Kong Med J. 2018;24:25-31.
- Donghi D, Kerl K, Dummer R, et al. Cutaneous angiosarcoma: own experience over 13 years. clinical features, disease course and immunohistochemical profile. J Eur Acad Dermatol Venereol. 2010;24:1230-1234.
- Ferrari A, Casanova M, Bisogno G, et al. Malignant vascular tumors in children and adolescents: a report from the Italian and German Soft Tissue Sarcoma Cooperative Group. Med Pediatr Oncol. 2002;39:109-114.
- Fujisawa Y, Nakamura Y, Kawachi Y, et al. Comparison between taxane-based chemotherapy with conventional surgery-based therapy for cutaneous angiosarcoma: a single-center experience. J Dermatolog Treat. 2014;25:419-423.
- Hodgkinson DJ, Soule EH, Woods JE. Cutaneous angiosarcoma of the head and neck. Cancer. 1979;44:1106-1113.
- Lim SY, Pyon JK, Mun GH, et al. Surgical treatment of angiosarcoma of the scalp with superficial parotidectomy. Ann Plast Surg. 2010;64:180-182.
- Maddox JC, Evans HL. Angiosarcoma of skin and soft tissue: a study of forty-four cases. Cancer. 1981;48:1907-1921.
- Mark RJ, Tran LM, Sercarz J, et al. Angiosarcoma of the head and neck. The UCLA experience 1955 through 1990. Arch Otolaryngol Head Neck Surg. 1993;119:973-978.
- Morgan MB, Swann M, Somach S, et al. Cutaneous angiosarcoma: a case series with prognostic correlation. J Am Acad Dermatol. 2004;50:867-874.
- Mullins B, Hackman T. Angiosarcoma of the head and neck. Int Arch Otorhinolaryngol. 2015;19:191-195.
- Ogawa K, Takahashi K, Asato Y, et al. Treatment and prognosis of angiosarcoma of the scalp and face: a retrospective analysis of 48 patients. Br J Radiol. 2012;85:E1127-E1133.
- Panje WR, Moran WJ, Bostwick DG, et al. Angiosarcoma of the head and neck: review of 11 cases. Laryngoscope. 1986;96:1381-1384.
- Perez MC, Padhya TA, Messina JL, et al. Cutaneous angiosarcoma: a single-institution experience. Ann Surg Oncol. 2013;20:3391-3397.
- Veness M, Cooper S. Treatment of cutaneous angiosarcomas of the head and neck. Australas Radiol. 1995;39:277-281.
- Barttelbort SW, Stahl R, Ariyan S. Cutaneous angiosarcoma of the face and scalp. Plast Reconstr Surg. 1989;84:55-59.
- Bernstein JM, Irish JC, Brown DH, et al. Survival outcomes for cutaneous angiosarcoma of the scalp versus face. Head Neck. 2017;39:1205-1211.
- Köhler HF, Neves RI, Brechtbühl ER, et al. Cutaneous angiosarcoma of the head and neck: report of 23 cases from a single institution. Otolaryngol Head Neck Surg. 2008;139:519-524.
- Morales PH, Lindberg RD, Barkley HT Jr. Soft tissue angiosarcomas. Int J Radiat Oncol Biol Phys. 1981;7:1655-1659.
- Wollina U, Hansel G, Schönlebe J, et al. Cutaneous angiosarcoma is a rare aggressive malignant vascular tumour of the skin. J Eur Acad Dermatol Venereol. 2011;25:964-968.
- Wollina U, Koch A, Hansel G, et al. A 10-year analysis of cutaneous mesenchymal tumors (sarcomas and related entities) in a skin cancer center. Int J Dermatol. 2013;52:1189-1197.
- Bien E, Stachowicz-Stencel T, Balcerska A, et al. Angiosarcoma in children - still uncontrollable oncological problem. The report of the Polish Paediatric Rare Tumours Study. Eur J Cancer Care (Engl). 2009;18:411-420.
- Suzuki G, Yamazaki H, Takenaka H, et al. Definitive radiation therapy for angiosarcoma of the face and scalp. In Vivo. 2016;30:921-926.
- Miki Y, Tada T, Kamo R, et al. Single institutional experience of the treatment of angiosarcoma of the face and scalp. Br J Radiol. 2013;86:20130439.
- Ohguri T, Imada H, Nomoto S, et al. Angiosarcoma of the scalp treated with curative radiotherapy plus recombinant interleukin-2 immunotherapy. Int J Radiat Oncol Biol Phys. 2005;61:1446-1453.
- Clayton BD, Leshin B, Hitchcock MG, et al. Utility of rush paraffin-embedded tangential sections in the management of cutaneous neoplasms. Dermatol Surg. 2000;26:671-678.
- Goldberg DJ, Kim YA. Angiosarcoma of the scalp treated with Mohs micrographic surgery. J Dermatol Surg Oncol. 1993;19:156-158.
- Mikhail GR, Kelly AP Jr. Malignant angioendothelioma of the face. J Dermatol Surg Oncol. 1977;3:181-183.
- Muscarella VA. Angiosarcoma treated by Mohs micrographic surgery. J Dermatol Surg Oncol. 1993;19:1132-1133.
- Bullen R, Larson PO, Landeck AE, et al. Angiosarcoma of the head and neck managed by a combination of multiple biopsies to determine tumor margin and radiation therapy. report of three cases and review of the literature. Dermatol Surg. 1998;24:1105-1110.
- Wiwatwongwana D, White VA, Dolman PJ. Two cases of periocular cutaneous angiosarcoma. Ophthalmic Plast Reconstr Surg. 2010;26:365-366.
- Morrison WH, Byers RM, Garden AS, et al. Cutaneous angiosarcoma of the head and neck. A therapeutic dilemma. Cancer. 1995;76:319-327.
- Hata M, Wada H, Ogino I, et al. Radiation therapy for angiosarcoma of the scalp: treatment outcomes of total scalp irradiation with X-rays and electrons. Strahlenther Onkol. 2014;190:899-904.
- Hwang K, Kim MY, Lee SH. Recommendations for therapeutic decisions of angiosarcoma of the scalp and face. J Craniofac Surg. 2015;26:E253-E256.
Cutaneous angiosarcoma (cAS) is a rare malignancy arising from vascular or lymphatic tissue. It classically presents during the sixth or seventh decades of life as a raised purple papule or plaque on the head and neck areas.1 Primary cAS frequently mimics benign conditions, leading to delays in care. Such delays coupled with the aggressive nature of angiosarcomas leads to a poor prognosis. Five-year survival rates range from 11% to 50%, and more than half of patients die within 1 year of diagnosis.2-7
Currently, there is no consensus on the most effective treatments, as the rare nature of cAS has made the development of controlled clinical trials difficult. Wide local excision (WLE) is most frequently employed; however, the tumor’s infiltrative growth makes complete resection and negative surgical margins difficult to achieve.8 Recently, Mohs micrographic surgery (MMS) has been postulated as a treatment option. The tissue-sparing nature and intraoperative margin control of MMS may provide tumor eradication and cosmesis benefits reported with other cutaneous malignancies.9
Nearly all localized cASs are treated with surgical excision with or without adjuvant treatment modalities; however, it is unclear which of these modalities provide a survival benefit. We conducted a systematic review of the literature to compare treatment modalities for localized cAS of the head and neck regions and to compare treatments based on tumor stage.
METHODS
A literature search was performed to identify published studies indexed by MEDLINE, Cochrane Central Register of Controlled Trials (CENTRAL), Embase, and PubMed from January 1, 1977, to May 8, 2020, reporting on cAS and treatment modalities used. The search was conducted in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analysis guidelines.5 Data extracted included patient demographics, tumor characteristics (including T1 [≤5 cm] and T2 [>5 cm and ≤10 cm] based on the American Joint Committee on Cancer soft tissue sarcoma staging criteria), treatments used, follow-up time, overall survival (OS) rates, and complications.10,11
Studies were required to (1) include participants with head and neck cAS; (2) report original patient data following cAS treatment with surgical (WLE or MMS) and/or nonsurgical modalities (chemotherapy [CT], radiotherapy [RT], immunotherapy [IT]); (3) report outcome data related to OS rates following treatment; and (4) have articles published in English. Given the rare nature of cAS, there was no limitation on the number of participants needed.
The Newcastle-Ottawa scale for observational studies was used to assess the quality of studies.12 Higher scores indicate low risk of bias, while lower scores represent high risk of bias.
Continuous data were reported with means and SDs, while categorical variables were reported as percentages. Overall survival means and SDs were compared between treatment modalities using an independent sample t test with P<.05 considered statistically significant. Due to the heterogeneity of the data, a meta-analysis was not reported.
RESULTS
Literature Search and Risk of Bias Assessment
There were 283 manuscripts identified, 56 articles read in full, and 40 articles included in the review (Figure). Among the 16 studies not meeting inclusion criteria, 7 did not provide enough data to isolate head and neck cAS cases,1,13-18 6 did not report outcomes related to the current review,19-24 and 3 did not provide enough data to isolate different treatment outcomes.25-27 Among the included studies, 32 reported use of WLE: WLE alone (n=21)2,7,11,28-45; WLE with RT (n=24)2,3,11,28-31,33-36,38-41,43-51; WLE with CT (n=7)2,31,35,39,41,48,52; WLE with RT and CT (n=11)2,29,31,33-35,39,40,48,52,53; WLE with RT and IT (n=3)35,54,55; and WLE with RT, CT, and IT (n=1).53 Nine studies reported MMS: MMS alone (n=5)39,56-59; MMS with RT (n=3)32,50,60,61; and MMS with RT and CT (n=1).51
Risk of bias assessment identified low risk in 3 articles. High risk was identified in 5 case reports,57-61 and 1 study did not describe patient selection.43 Clayton et al56 showed intermediate risk, given the study controlled for 1 factor.
Patient Demographics
A total of 1295 patients were included. The pooled mean age of the patients was 67.5 years (range, 3–88 years), and 64.7% were male. There were 79 cases identified as T1 and 105 as T2. A total of 825 cases were treated using WLE with or without adjuvant therapy, while a total of 9 cases were treated using MMS with and without adjuvant therapies (Table). There were 461 cases treated without surgical excision: RT alone (n=261), CT alone (n=38), IT alone (n=35), RT with CT (n=81), RT with IT (n=34), and RT with CT and IT (n=12)(Table). The median follow-up period across all studies was 23.5 months (range, 1–228 months).
Comparison Between Surgical and Nonsurgical Modalities
Wide Local Excision—Wide local excision (n=825; 63.7%) alone or in combination with other therapies was the most frequently used treatment modality. The mean (SD) OS was longest for WLE with RT, CT, and IT (n=3; 39.3 [24.1]), followed by WLE with RT (n=447; 35.9 [34.3] months), WLE with CT (n=13; 32.4 [30.2] months), WLE alone (n=324; 29.6 [34.1] months), WLE with RT and IT (n=11; 23.5 [4.9] months), and WLE with RT and CT (n=27; 20.7 [13.1] months).
Nonsurgical Modalities—Nonsurgical methods were used less frequently than surgical methods (n=461; 35.6%). The mean (SD) OS time in descending order was as follows: RT with CT and IT (n=12; 34.9 [1.2] months), RT with CT (n=81; 30.4 [37.8] months), IT alone (n=35; 25.7 [no SD reported] months), RT with IT (n=34; 20.5 [8.6] months), CT alone (n=38; 20.1 [15.9] months), and RT alone (n=261; 12.8 [8.3] months).
When comparing mean (SD) OS outcomes between surgical and nonsurgical treatment modalities, only the addition of WLE to RT significantly increased OS when compared with RT alone (WLE, 35.9 [34.3] months; RT alone, 12.8 [8.3] months; P=.001). When WLE was added to CT or both RT and CT, there was no significant difference with OS when compared with CT alone (WLE with CT, 32.4 [30.2] months; CT alone, 20.1 [15.9] months; P=.065); or both RT and CT in combination (WLE with RT and CT, 20.7 [13.1] months; RT and CT, 30.4 [37.8] months; P=.204).
Comparison Between T1 and T2 cAS
T1 Angiosarcoma—There were 79 patients identified as having T1 tumors across 16 studies.2,31,32,34,39-41,46,48-50,53,58-60,62 The mean (SD) OS was longest for WLE with RT, CT, and IT (n=2; 56.0 [6.0] months), followed by WLE with CT (n=4; 54.5 [41.0] months); WLE with RT (n=30; 39.7 [41.2] months); WLE alone (n=22; 37.2 [37.3] months); WLE with both RT and CT (n=7; 25.5 [18.7] months); RT with IT (n=2; 20.0 [11.0] months); RT with CT (n=6; 15.7 [6.8] months); and RT alone (n=1; 13 [no SD]) months)(eTable).
T2 Angiosarcoma—There were 105 patients with T2 tumors in 15 studies.2,31,32,34,39-41,46,48-50,52,53,57,62 The mean (SD) OS for each treatment modality in descending order was as follows: RT with CT and IT (n=1; 36 [no SD reported] months); RT with CT (n=23; 34.3 [46.3] months); WLE with RT (n=21; 26.3 [23.8] months); WLE with CT (n=8; 21.5 [16.6] months); WLE alone (n=16; 19.8 [15.6] months); WLE with RT and CT (n=14; 19.2 [10.5] months); RT alone (n=17; 10.1 [5.5] months); CT alone (n=2; 6.7 [3.7] months); and WLE with RT, CT, and IT (n=1; 6.0 [no SD] months)(eTable).
Mohs Micrographic Surgery—The use of MMS was only identified in case reports or small observational studies for a total of 9 patients. Five cASs were treated with MMS alone for a mean (SD) OS of 37 (21.5) months, with 4 reporting cAS staging: 2 were T158,59 (mean [SD] OS, 37.0 [17.0] months) and 2 were T2 tumors39,57 (mean [SD] OS, 44.5 [26.5] months). Mohs micrographic surgery with RT was used for 3 tumors (mean [SD] OS, 34.0 [26.9] months); 2 were T150,60 (mean [SD] OS, 42.0 [30.0] months) and 1 unreported staging (eTable).56 Mohs micrographic surgery with both RT and CT was used in 1 patient (unreported staging; OS, 82 months).51
Complications
Complications were rare and mainly associated with CT and RT. Four studies reported radiation dermatitis with RT.53,55,62,63 Two studies reported peripheral neuropathy and myelotoxicity with CT.35,51 Only 1 study reported poor wound healing due to surgical complications.29
COMMENT
Cutaneous angiosarcomas are rare and have limited treatment guidelines. Surgical excision does appear to be an effective adjunct to nonsurgical treatments, particularly WLE combined with RT, CT, and IT. Although MMS ultimately may be useful for cAS, the limited number and substantial heterogeneity of reported cases precludes definitive conclusions at this time.
Achieving margin control during WLE is associated with higher OS when treating angiosarcoma,36,46 which is particularly true for T1 tumors where margin control is imperative, and many cases are treated with a combination of WLE and RT. Overall survival times are lower for T2 tumors, as these tumors are larger and most likely have spread; therefore, more aggressive combination treatments were more prevalent. In these cases, complete margin control may be difficult to achieve and may not be as critical to the outcome if another form of adjuvant therapy can be administered promptly.24,64
When surgery is contraindicated, RT with or without CT was the most commonly reported treatment modality. However, these treatments were notably less effective than when used in combination with surgical resection. The use of RT alone has a recurrence rate reported up to 100% in certain studies, suggesting the need to utilize RT in combination with other modalities.23,39 It is important to note that RT often is used as monotherapy in palliative treatment, which may indirectly skew survival rates.2
Limitations of the study include a lack of randomized controlled trials. Most reports were retrospective reviews or case series, and tumor staging was sparsely reported. Finally, although MMS may provide utility in the treatment of cAS, the sample size of 9 precluded definitive conclusions from being formed about its efficacy.
CONCLUSION
Cutaneous angiosarcoma is rare and has limited data comparing different treatment modalities. The paucity of data currently limits definitive recommendations; however, both surgical and nonsurgical modalities have demonstrated potential efficacy in the treatment of cAS and may benefit from additional research. Clinicians should consider a multidisciplinary approach for patients with a diagnosis of cAS to tailor treatments on a case-by-case basis.
Cutaneous angiosarcoma (cAS) is a rare malignancy arising from vascular or lymphatic tissue. It classically presents during the sixth or seventh decades of life as a raised purple papule or plaque on the head and neck areas.1 Primary cAS frequently mimics benign conditions, leading to delays in care. Such delays coupled with the aggressive nature of angiosarcomas leads to a poor prognosis. Five-year survival rates range from 11% to 50%, and more than half of patients die within 1 year of diagnosis.2-7
Currently, there is no consensus on the most effective treatments, as the rare nature of cAS has made the development of controlled clinical trials difficult. Wide local excision (WLE) is most frequently employed; however, the tumor’s infiltrative growth makes complete resection and negative surgical margins difficult to achieve.8 Recently, Mohs micrographic surgery (MMS) has been postulated as a treatment option. The tissue-sparing nature and intraoperative margin control of MMS may provide tumor eradication and cosmesis benefits reported with other cutaneous malignancies.9
Nearly all localized cASs are treated with surgical excision with or without adjuvant treatment modalities; however, it is unclear which of these modalities provide a survival benefit. We conducted a systematic review of the literature to compare treatment modalities for localized cAS of the head and neck regions and to compare treatments based on tumor stage.
METHODS
A literature search was performed to identify published studies indexed by MEDLINE, Cochrane Central Register of Controlled Trials (CENTRAL), Embase, and PubMed from January 1, 1977, to May 8, 2020, reporting on cAS and treatment modalities used. The search was conducted in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analysis guidelines.5 Data extracted included patient demographics, tumor characteristics (including T1 [≤5 cm] and T2 [>5 cm and ≤10 cm] based on the American Joint Committee on Cancer soft tissue sarcoma staging criteria), treatments used, follow-up time, overall survival (OS) rates, and complications.10,11
Studies were required to (1) include participants with head and neck cAS; (2) report original patient data following cAS treatment with surgical (WLE or MMS) and/or nonsurgical modalities (chemotherapy [CT], radiotherapy [RT], immunotherapy [IT]); (3) report outcome data related to OS rates following treatment; and (4) have articles published in English. Given the rare nature of cAS, there was no limitation on the number of participants needed.
The Newcastle-Ottawa scale for observational studies was used to assess the quality of studies.12 Higher scores indicate low risk of bias, while lower scores represent high risk of bias.
Continuous data were reported with means and SDs, while categorical variables were reported as percentages. Overall survival means and SDs were compared between treatment modalities using an independent sample t test with P<.05 considered statistically significant. Due to the heterogeneity of the data, a meta-analysis was not reported.
RESULTS
Literature Search and Risk of Bias Assessment
There were 283 manuscripts identified, 56 articles read in full, and 40 articles included in the review (Figure). Among the 16 studies not meeting inclusion criteria, 7 did not provide enough data to isolate head and neck cAS cases,1,13-18 6 did not report outcomes related to the current review,19-24 and 3 did not provide enough data to isolate different treatment outcomes.25-27 Among the included studies, 32 reported use of WLE: WLE alone (n=21)2,7,11,28-45; WLE with RT (n=24)2,3,11,28-31,33-36,38-41,43-51; WLE with CT (n=7)2,31,35,39,41,48,52; WLE with RT and CT (n=11)2,29,31,33-35,39,40,48,52,53; WLE with RT and IT (n=3)35,54,55; and WLE with RT, CT, and IT (n=1).53 Nine studies reported MMS: MMS alone (n=5)39,56-59; MMS with RT (n=3)32,50,60,61; and MMS with RT and CT (n=1).51
Risk of bias assessment identified low risk in 3 articles. High risk was identified in 5 case reports,57-61 and 1 study did not describe patient selection.43 Clayton et al56 showed intermediate risk, given the study controlled for 1 factor.
Patient Demographics
A total of 1295 patients were included. The pooled mean age of the patients was 67.5 years (range, 3–88 years), and 64.7% were male. There were 79 cases identified as T1 and 105 as T2. A total of 825 cases were treated using WLE with or without adjuvant therapy, while a total of 9 cases were treated using MMS with and without adjuvant therapies (Table). There were 461 cases treated without surgical excision: RT alone (n=261), CT alone (n=38), IT alone (n=35), RT with CT (n=81), RT with IT (n=34), and RT with CT and IT (n=12)(Table). The median follow-up period across all studies was 23.5 months (range, 1–228 months).
Comparison Between Surgical and Nonsurgical Modalities
Wide Local Excision—Wide local excision (n=825; 63.7%) alone or in combination with other therapies was the most frequently used treatment modality. The mean (SD) OS was longest for WLE with RT, CT, and IT (n=3; 39.3 [24.1]), followed by WLE with RT (n=447; 35.9 [34.3] months), WLE with CT (n=13; 32.4 [30.2] months), WLE alone (n=324; 29.6 [34.1] months), WLE with RT and IT (n=11; 23.5 [4.9] months), and WLE with RT and CT (n=27; 20.7 [13.1] months).
Nonsurgical Modalities—Nonsurgical methods were used less frequently than surgical methods (n=461; 35.6%). The mean (SD) OS time in descending order was as follows: RT with CT and IT (n=12; 34.9 [1.2] months), RT with CT (n=81; 30.4 [37.8] months), IT alone (n=35; 25.7 [no SD reported] months), RT with IT (n=34; 20.5 [8.6] months), CT alone (n=38; 20.1 [15.9] months), and RT alone (n=261; 12.8 [8.3] months).
When comparing mean (SD) OS outcomes between surgical and nonsurgical treatment modalities, only the addition of WLE to RT significantly increased OS when compared with RT alone (WLE, 35.9 [34.3] months; RT alone, 12.8 [8.3] months; P=.001). When WLE was added to CT or both RT and CT, there was no significant difference with OS when compared with CT alone (WLE with CT, 32.4 [30.2] months; CT alone, 20.1 [15.9] months; P=.065); or both RT and CT in combination (WLE with RT and CT, 20.7 [13.1] months; RT and CT, 30.4 [37.8] months; P=.204).
Comparison Between T1 and T2 cAS
T1 Angiosarcoma—There were 79 patients identified as having T1 tumors across 16 studies.2,31,32,34,39-41,46,48-50,53,58-60,62 The mean (SD) OS was longest for WLE with RT, CT, and IT (n=2; 56.0 [6.0] months), followed by WLE with CT (n=4; 54.5 [41.0] months); WLE with RT (n=30; 39.7 [41.2] months); WLE alone (n=22; 37.2 [37.3] months); WLE with both RT and CT (n=7; 25.5 [18.7] months); RT with IT (n=2; 20.0 [11.0] months); RT with CT (n=6; 15.7 [6.8] months); and RT alone (n=1; 13 [no SD]) months)(eTable).
T2 Angiosarcoma—There were 105 patients with T2 tumors in 15 studies.2,31,32,34,39-41,46,48-50,52,53,57,62 The mean (SD) OS for each treatment modality in descending order was as follows: RT with CT and IT (n=1; 36 [no SD reported] months); RT with CT (n=23; 34.3 [46.3] months); WLE with RT (n=21; 26.3 [23.8] months); WLE with CT (n=8; 21.5 [16.6] months); WLE alone (n=16; 19.8 [15.6] months); WLE with RT and CT (n=14; 19.2 [10.5] months); RT alone (n=17; 10.1 [5.5] months); CT alone (n=2; 6.7 [3.7] months); and WLE with RT, CT, and IT (n=1; 6.0 [no SD] months)(eTable).
Mohs Micrographic Surgery—The use of MMS was only identified in case reports or small observational studies for a total of 9 patients. Five cASs were treated with MMS alone for a mean (SD) OS of 37 (21.5) months, with 4 reporting cAS staging: 2 were T158,59 (mean [SD] OS, 37.0 [17.0] months) and 2 were T2 tumors39,57 (mean [SD] OS, 44.5 [26.5] months). Mohs micrographic surgery with RT was used for 3 tumors (mean [SD] OS, 34.0 [26.9] months); 2 were T150,60 (mean [SD] OS, 42.0 [30.0] months) and 1 unreported staging (eTable).56 Mohs micrographic surgery with both RT and CT was used in 1 patient (unreported staging; OS, 82 months).51
Complications
Complications were rare and mainly associated with CT and RT. Four studies reported radiation dermatitis with RT.53,55,62,63 Two studies reported peripheral neuropathy and myelotoxicity with CT.35,51 Only 1 study reported poor wound healing due to surgical complications.29
COMMENT
Cutaneous angiosarcomas are rare and have limited treatment guidelines. Surgical excision does appear to be an effective adjunct to nonsurgical treatments, particularly WLE combined with RT, CT, and IT. Although MMS ultimately may be useful for cAS, the limited number and substantial heterogeneity of reported cases precludes definitive conclusions at this time.
Achieving margin control during WLE is associated with higher OS when treating angiosarcoma,36,46 which is particularly true for T1 tumors where margin control is imperative, and many cases are treated with a combination of WLE and RT. Overall survival times are lower for T2 tumors, as these tumors are larger and most likely have spread; therefore, more aggressive combination treatments were more prevalent. In these cases, complete margin control may be difficult to achieve and may not be as critical to the outcome if another form of adjuvant therapy can be administered promptly.24,64
When surgery is contraindicated, RT with or without CT was the most commonly reported treatment modality. However, these treatments were notably less effective than when used in combination with surgical resection. The use of RT alone has a recurrence rate reported up to 100% in certain studies, suggesting the need to utilize RT in combination with other modalities.23,39 It is important to note that RT often is used as monotherapy in palliative treatment, which may indirectly skew survival rates.2
Limitations of the study include a lack of randomized controlled trials. Most reports were retrospective reviews or case series, and tumor staging was sparsely reported. Finally, although MMS may provide utility in the treatment of cAS, the sample size of 9 precluded definitive conclusions from being formed about its efficacy.
CONCLUSION
Cutaneous angiosarcoma is rare and has limited data comparing different treatment modalities. The paucity of data currently limits definitive recommendations; however, both surgical and nonsurgical modalities have demonstrated potential efficacy in the treatment of cAS and may benefit from additional research. Clinicians should consider a multidisciplinary approach for patients with a diagnosis of cAS to tailor treatments on a case-by-case basis.
- Rodríguez-Jiménez P, Jimenez YD, Reolid A, et al. State of the art of Mohs surgery for rare cutaneous tumors in the Spanish Registry of Mohs Surgery (REGESMOHS). Int J Dermatol. 2020;59:321-325.
- Alqumber NA, Choi JW, Kang MK. The management and prognosis of facial and scalp angiosarcoma: a retrospective analysis of 15 patients. Ann Plast Surg. 2019;83:55-62.
- Pawlik TM, Paulino AF, McGinn CJ, et al. Cutaneous angiosarcoma of the scalp: a multidisciplinary approach. Cancer. 2003;98:1716-1726.
- Deyrup AT, McKenney JK, Tighiouart M, et al. Sporadic cutaneous angiosarcomas: a proposal for risk stratification based on 69 cases. Am J Surg Pathol. 2008;32:72-77.
- Meis-Kindblom JM, Kindblom LG. Angiosarcoma of soft tissue: a study of 80 cases. Am J Surg Pathol. 1998;22:683-697.
- Harbour P, Song DH. The skin and subcutaneous tissue. In: Brunicardi FC, Andersen DK, Billiar TR, et al, eds. Schwartz’s Principles of Surgery. 11th ed. McGraw-Hill Education; 2019. Accessed April 24, 2023. https://accesssurgery.mhmedical.com/content.aspx?bookid=2576§ionid=216206374
- Oashi K, Namikawa K, Tsutsumida A, et al. Surgery with curative intent is associated with prolonged survival in patients with cutaneous angiosarcoma of the scalp and face—a retrospective study of 38 untreated cases in the Japanese population. Eur J Surg Oncol. 2018;44:823-829.
- Young RJ, Brown NJ, Reed MW, et al. Angiosarcoma. Lancet Oncol. 2010;11:983-991.
- Tolkachjov SN, Brodland DG, Coldiron BM, et al. Understanding Mohs micrographic surgery: a review and practical guide for the nondermatologist. Mayo Clin Proc. 2017;92:1261-1271.
- Amin M, Edge SB, Greene FL, et al, eds. AJCC Cancer Staging Manual. 8th ed. Springer; 2017.
- Holden CA, Spittle MF, Jones EW. Angiosarcoma of the face and scalp, prognosis and treatment. Cancer. 1987;59:1046-1057.
- Cook DA, Reed DA. Appraising the quality of medical education research methods: the Medical Education Research Study Quality Instrument and the Newcastle-Ottawa Scale-Education. Acad Med. 2015;90:1067-1076.
- Lee BL, Chen CF, Chen PC, et al. Investigation of prognostic features in primary cutaneous and soft tissue angiosarcoma after surgical resection: a retrospective study. Ann Plast Surg. 2017;78(3 suppl 2):S41-S46.
- Shen CJ, Parzuchowski AS, Kummerlowe MN, et al. Combined modality therapy improves overall survival for angiosarcoma. Acta Oncol. 2017;56:1235-1238.
- Breakey RW, Crowley TP, Anderson IB, et al. The surgical management of head and neck sarcoma: the Newcastle experience. J Plast Reconstr Aesthet Surg. 2017;70:78-84.
- Singla S, Papavasiliou P, Powers B, et al. Challenges in the treatment of angiosarcoma: a single institution experience. Am J Surg. 2014;208:254-259.
- Sasaki R, Soejima T, Kishi K, et al. Angiosarcoma treated with radiotherapy: impact of tumor type and size on outcome. Int J Radiat Oncol Biol Phys. 2002;52:1032-1040.
- Naka N, Ohsawa M, Tomita Y, et al. Angiosarcoma in Japan. A review of 99 cases. Cancer. 1995;75:989-996.
- DeMartelaere SL, Roberts D, Burgess MA, et al. Neoadjuvant chemotherapy-specific and overall treatment outcomes in patients with cutaneous angiosarcoma of the face with periorbital involvement. Head Neck. 2008;30:639-646.
- Ward JR, Feigenberg SJ, Mendenhall NP, et al. Radiation therapy for angiosarcoma. Head Neck. 2003;25:873-878.
- Letsa I, Benson C, Al-Muderis O, et al. Angiosarcoma of the face and scalp: effective systemic treatment in the older patient. J Geriatr Oncol. 2014;5:276-280.
- Buehler D, Rice SR, Moody JS, et al. Angiosarcoma outcomes and prognostic factors: a 25-year single institution experience. Am J Clin Oncol. 2014;37:473-479.
- Patel SH, Hayden RE, Hinni ML, et al. Angiosarcoma of the scalp and face: the Mayo Clinic experience. JAMA Otolaryngol Head Neck Surg. 2015;141:335-340.
- Guadagnolo BA, Zagars GK, Araujo D, et al. Outcomes after definitive treatment for cutaneous angiosarcoma of the face and scalp. Head Neck. 2011;33:661-667.
- Zhang Y, Yan Y, Zhu M, et al. Clinical outcomes in primary scalp angiosarcoma. Oncol Lett. 2019;18:5091-5096.
- Kamo R, Ishii M. Histological differentiation, histogenesis and prognosis of cutaneous angiosarcoma. Osaka City Med J. 2011;57:31-44.
- Ito T, Uchi H, Nakahara T, et al. Cutaneous angiosarcoma of the head and face: a single-center analysis of treatment outcomes in 43 patients in Japan. J Cancer Res Clin Oncol. 2016;142:1387-1394.
- Aust MR, Olsen KD, Lewis JE, et al. Angiosarcomas of the head and neck: clinical and pathologic characteristics. Ann Otol Rhinol Laryngol. 1997;106:943-951.
- Buschmann A, Lehnhardt M, Toman N, et al. Surgical treatment of angiosarcoma of the scalp: less is more. Ann Plast Surg. 2008;61:399-403.
- Cassidy RJ, Switchenko JM, Yushak ML, et al. The importance of surgery in scalp angiosarcomas. Surg Oncol. 2018;27:A3-A8.
- Choi JH, Ahn KC, Chang H, et al. Surgical treatment and prognosis of angiosarcoma of the scalp: a retrospective analysis of 14 patients in a single institution. Biomed Res Int. 2015;2015:321896.
- Chow TL, Kwan WW, Kwan CK. Treatment of cutaneous angiosarcoma of the scalp and face in Chinese patients: local experience at a regional hospital in Hong Kong. Hong Kong Med J. 2018;24:25-31.
- Donghi D, Kerl K, Dummer R, et al. Cutaneous angiosarcoma: own experience over 13 years. clinical features, disease course and immunohistochemical profile. J Eur Acad Dermatol Venereol. 2010;24:1230-1234.
- Ferrari A, Casanova M, Bisogno G, et al. Malignant vascular tumors in children and adolescents: a report from the Italian and German Soft Tissue Sarcoma Cooperative Group. Med Pediatr Oncol. 2002;39:109-114.
- Fujisawa Y, Nakamura Y, Kawachi Y, et al. Comparison between taxane-based chemotherapy with conventional surgery-based therapy for cutaneous angiosarcoma: a single-center experience. J Dermatolog Treat. 2014;25:419-423.
- Hodgkinson DJ, Soule EH, Woods JE. Cutaneous angiosarcoma of the head and neck. Cancer. 1979;44:1106-1113.
- Lim SY, Pyon JK, Mun GH, et al. Surgical treatment of angiosarcoma of the scalp with superficial parotidectomy. Ann Plast Surg. 2010;64:180-182.
- Maddox JC, Evans HL. Angiosarcoma of skin and soft tissue: a study of forty-four cases. Cancer. 1981;48:1907-1921.
- Mark RJ, Tran LM, Sercarz J, et al. Angiosarcoma of the head and neck. The UCLA experience 1955 through 1990. Arch Otolaryngol Head Neck Surg. 1993;119:973-978.
- Morgan MB, Swann M, Somach S, et al. Cutaneous angiosarcoma: a case series with prognostic correlation. J Am Acad Dermatol. 2004;50:867-874.
- Mullins B, Hackman T. Angiosarcoma of the head and neck. Int Arch Otorhinolaryngol. 2015;19:191-195.
- Ogawa K, Takahashi K, Asato Y, et al. Treatment and prognosis of angiosarcoma of the scalp and face: a retrospective analysis of 48 patients. Br J Radiol. 2012;85:E1127-E1133.
- Panje WR, Moran WJ, Bostwick DG, et al. Angiosarcoma of the head and neck: review of 11 cases. Laryngoscope. 1986;96:1381-1384.
- Perez MC, Padhya TA, Messina JL, et al. Cutaneous angiosarcoma: a single-institution experience. Ann Surg Oncol. 2013;20:3391-3397.
- Veness M, Cooper S. Treatment of cutaneous angiosarcomas of the head and neck. Australas Radiol. 1995;39:277-281.
- Barttelbort SW, Stahl R, Ariyan S. Cutaneous angiosarcoma of the face and scalp. Plast Reconstr Surg. 1989;84:55-59.
- Bernstein JM, Irish JC, Brown DH, et al. Survival outcomes for cutaneous angiosarcoma of the scalp versus face. Head Neck. 2017;39:1205-1211.
- Köhler HF, Neves RI, Brechtbühl ER, et al. Cutaneous angiosarcoma of the head and neck: report of 23 cases from a single institution. Otolaryngol Head Neck Surg. 2008;139:519-524.
- Morales PH, Lindberg RD, Barkley HT Jr. Soft tissue angiosarcomas. Int J Radiat Oncol Biol Phys. 1981;7:1655-1659.
- Wollina U, Hansel G, Schönlebe J, et al. Cutaneous angiosarcoma is a rare aggressive malignant vascular tumour of the skin. J Eur Acad Dermatol Venereol. 2011;25:964-968.
- Wollina U, Koch A, Hansel G, et al. A 10-year analysis of cutaneous mesenchymal tumors (sarcomas and related entities) in a skin cancer center. Int J Dermatol. 2013;52:1189-1197.
- Bien E, Stachowicz-Stencel T, Balcerska A, et al. Angiosarcoma in children - still uncontrollable oncological problem. The report of the Polish Paediatric Rare Tumours Study. Eur J Cancer Care (Engl). 2009;18:411-420.
- Suzuki G, Yamazaki H, Takenaka H, et al. Definitive radiation therapy for angiosarcoma of the face and scalp. In Vivo. 2016;30:921-926.
- Miki Y, Tada T, Kamo R, et al. Single institutional experience of the treatment of angiosarcoma of the face and scalp. Br J Radiol. 2013;86:20130439.
- Ohguri T, Imada H, Nomoto S, et al. Angiosarcoma of the scalp treated with curative radiotherapy plus recombinant interleukin-2 immunotherapy. Int J Radiat Oncol Biol Phys. 2005;61:1446-1453.
- Clayton BD, Leshin B, Hitchcock MG, et al. Utility of rush paraffin-embedded tangential sections in the management of cutaneous neoplasms. Dermatol Surg. 2000;26:671-678.
- Goldberg DJ, Kim YA. Angiosarcoma of the scalp treated with Mohs micrographic surgery. J Dermatol Surg Oncol. 1993;19:156-158.
- Mikhail GR, Kelly AP Jr. Malignant angioendothelioma of the face. J Dermatol Surg Oncol. 1977;3:181-183.
- Muscarella VA. Angiosarcoma treated by Mohs micrographic surgery. J Dermatol Surg Oncol. 1993;19:1132-1133.
- Bullen R, Larson PO, Landeck AE, et al. Angiosarcoma of the head and neck managed by a combination of multiple biopsies to determine tumor margin and radiation therapy. report of three cases and review of the literature. Dermatol Surg. 1998;24:1105-1110.
- Wiwatwongwana D, White VA, Dolman PJ. Two cases of periocular cutaneous angiosarcoma. Ophthalmic Plast Reconstr Surg. 2010;26:365-366.
- Morrison WH, Byers RM, Garden AS, et al. Cutaneous angiosarcoma of the head and neck. A therapeutic dilemma. Cancer. 1995;76:319-327.
- Hata M, Wada H, Ogino I, et al. Radiation therapy for angiosarcoma of the scalp: treatment outcomes of total scalp irradiation with X-rays and electrons. Strahlenther Onkol. 2014;190:899-904.
- Hwang K, Kim MY, Lee SH. Recommendations for therapeutic decisions of angiosarcoma of the scalp and face. J Craniofac Surg. 2015;26:E253-E256.
- Rodríguez-Jiménez P, Jimenez YD, Reolid A, et al. State of the art of Mohs surgery for rare cutaneous tumors in the Spanish Registry of Mohs Surgery (REGESMOHS). Int J Dermatol. 2020;59:321-325.
- Alqumber NA, Choi JW, Kang MK. The management and prognosis of facial and scalp angiosarcoma: a retrospective analysis of 15 patients. Ann Plast Surg. 2019;83:55-62.
- Pawlik TM, Paulino AF, McGinn CJ, et al. Cutaneous angiosarcoma of the scalp: a multidisciplinary approach. Cancer. 2003;98:1716-1726.
- Deyrup AT, McKenney JK, Tighiouart M, et al. Sporadic cutaneous angiosarcomas: a proposal for risk stratification based on 69 cases. Am J Surg Pathol. 2008;32:72-77.
- Meis-Kindblom JM, Kindblom LG. Angiosarcoma of soft tissue: a study of 80 cases. Am J Surg Pathol. 1998;22:683-697.
- Harbour P, Song DH. The skin and subcutaneous tissue. In: Brunicardi FC, Andersen DK, Billiar TR, et al, eds. Schwartz’s Principles of Surgery. 11th ed. McGraw-Hill Education; 2019. Accessed April 24, 2023. https://accesssurgery.mhmedical.com/content.aspx?bookid=2576§ionid=216206374
- Oashi K, Namikawa K, Tsutsumida A, et al. Surgery with curative intent is associated with prolonged survival in patients with cutaneous angiosarcoma of the scalp and face—a retrospective study of 38 untreated cases in the Japanese population. Eur J Surg Oncol. 2018;44:823-829.
- Young RJ, Brown NJ, Reed MW, et al. Angiosarcoma. Lancet Oncol. 2010;11:983-991.
- Tolkachjov SN, Brodland DG, Coldiron BM, et al. Understanding Mohs micrographic surgery: a review and practical guide for the nondermatologist. Mayo Clin Proc. 2017;92:1261-1271.
- Amin M, Edge SB, Greene FL, et al, eds. AJCC Cancer Staging Manual. 8th ed. Springer; 2017.
- Holden CA, Spittle MF, Jones EW. Angiosarcoma of the face and scalp, prognosis and treatment. Cancer. 1987;59:1046-1057.
- Cook DA, Reed DA. Appraising the quality of medical education research methods: the Medical Education Research Study Quality Instrument and the Newcastle-Ottawa Scale-Education. Acad Med. 2015;90:1067-1076.
- Lee BL, Chen CF, Chen PC, et al. Investigation of prognostic features in primary cutaneous and soft tissue angiosarcoma after surgical resection: a retrospective study. Ann Plast Surg. 2017;78(3 suppl 2):S41-S46.
- Shen CJ, Parzuchowski AS, Kummerlowe MN, et al. Combined modality therapy improves overall survival for angiosarcoma. Acta Oncol. 2017;56:1235-1238.
- Breakey RW, Crowley TP, Anderson IB, et al. The surgical management of head and neck sarcoma: the Newcastle experience. J Plast Reconstr Aesthet Surg. 2017;70:78-84.
- Singla S, Papavasiliou P, Powers B, et al. Challenges in the treatment of angiosarcoma: a single institution experience. Am J Surg. 2014;208:254-259.
- Sasaki R, Soejima T, Kishi K, et al. Angiosarcoma treated with radiotherapy: impact of tumor type and size on outcome. Int J Radiat Oncol Biol Phys. 2002;52:1032-1040.
- Naka N, Ohsawa M, Tomita Y, et al. Angiosarcoma in Japan. A review of 99 cases. Cancer. 1995;75:989-996.
- DeMartelaere SL, Roberts D, Burgess MA, et al. Neoadjuvant chemotherapy-specific and overall treatment outcomes in patients with cutaneous angiosarcoma of the face with periorbital involvement. Head Neck. 2008;30:639-646.
- Ward JR, Feigenberg SJ, Mendenhall NP, et al. Radiation therapy for angiosarcoma. Head Neck. 2003;25:873-878.
- Letsa I, Benson C, Al-Muderis O, et al. Angiosarcoma of the face and scalp: effective systemic treatment in the older patient. J Geriatr Oncol. 2014;5:276-280.
- Buehler D, Rice SR, Moody JS, et al. Angiosarcoma outcomes and prognostic factors: a 25-year single institution experience. Am J Clin Oncol. 2014;37:473-479.
- Patel SH, Hayden RE, Hinni ML, et al. Angiosarcoma of the scalp and face: the Mayo Clinic experience. JAMA Otolaryngol Head Neck Surg. 2015;141:335-340.
- Guadagnolo BA, Zagars GK, Araujo D, et al. Outcomes after definitive treatment for cutaneous angiosarcoma of the face and scalp. Head Neck. 2011;33:661-667.
- Zhang Y, Yan Y, Zhu M, et al. Clinical outcomes in primary scalp angiosarcoma. Oncol Lett. 2019;18:5091-5096.
- Kamo R, Ishii M. Histological differentiation, histogenesis and prognosis of cutaneous angiosarcoma. Osaka City Med J. 2011;57:31-44.
- Ito T, Uchi H, Nakahara T, et al. Cutaneous angiosarcoma of the head and face: a single-center analysis of treatment outcomes in 43 patients in Japan. J Cancer Res Clin Oncol. 2016;142:1387-1394.
- Aust MR, Olsen KD, Lewis JE, et al. Angiosarcomas of the head and neck: clinical and pathologic characteristics. Ann Otol Rhinol Laryngol. 1997;106:943-951.
- Buschmann A, Lehnhardt M, Toman N, et al. Surgical treatment of angiosarcoma of the scalp: less is more. Ann Plast Surg. 2008;61:399-403.
- Cassidy RJ, Switchenko JM, Yushak ML, et al. The importance of surgery in scalp angiosarcomas. Surg Oncol. 2018;27:A3-A8.
- Choi JH, Ahn KC, Chang H, et al. Surgical treatment and prognosis of angiosarcoma of the scalp: a retrospective analysis of 14 patients in a single institution. Biomed Res Int. 2015;2015:321896.
- Chow TL, Kwan WW, Kwan CK. Treatment of cutaneous angiosarcoma of the scalp and face in Chinese patients: local experience at a regional hospital in Hong Kong. Hong Kong Med J. 2018;24:25-31.
- Donghi D, Kerl K, Dummer R, et al. Cutaneous angiosarcoma: own experience over 13 years. clinical features, disease course and immunohistochemical profile. J Eur Acad Dermatol Venereol. 2010;24:1230-1234.
- Ferrari A, Casanova M, Bisogno G, et al. Malignant vascular tumors in children and adolescents: a report from the Italian and German Soft Tissue Sarcoma Cooperative Group. Med Pediatr Oncol. 2002;39:109-114.
- Fujisawa Y, Nakamura Y, Kawachi Y, et al. Comparison between taxane-based chemotherapy with conventional surgery-based therapy for cutaneous angiosarcoma: a single-center experience. J Dermatolog Treat. 2014;25:419-423.
- Hodgkinson DJ, Soule EH, Woods JE. Cutaneous angiosarcoma of the head and neck. Cancer. 1979;44:1106-1113.
- Lim SY, Pyon JK, Mun GH, et al. Surgical treatment of angiosarcoma of the scalp with superficial parotidectomy. Ann Plast Surg. 2010;64:180-182.
- Maddox JC, Evans HL. Angiosarcoma of skin and soft tissue: a study of forty-four cases. Cancer. 1981;48:1907-1921.
- Mark RJ, Tran LM, Sercarz J, et al. Angiosarcoma of the head and neck. The UCLA experience 1955 through 1990. Arch Otolaryngol Head Neck Surg. 1993;119:973-978.
- Morgan MB, Swann M, Somach S, et al. Cutaneous angiosarcoma: a case series with prognostic correlation. J Am Acad Dermatol. 2004;50:867-874.
- Mullins B, Hackman T. Angiosarcoma of the head and neck. Int Arch Otorhinolaryngol. 2015;19:191-195.
- Ogawa K, Takahashi K, Asato Y, et al. Treatment and prognosis of angiosarcoma of the scalp and face: a retrospective analysis of 48 patients. Br J Radiol. 2012;85:E1127-E1133.
- Panje WR, Moran WJ, Bostwick DG, et al. Angiosarcoma of the head and neck: review of 11 cases. Laryngoscope. 1986;96:1381-1384.
- Perez MC, Padhya TA, Messina JL, et al. Cutaneous angiosarcoma: a single-institution experience. Ann Surg Oncol. 2013;20:3391-3397.
- Veness M, Cooper S. Treatment of cutaneous angiosarcomas of the head and neck. Australas Radiol. 1995;39:277-281.
- Barttelbort SW, Stahl R, Ariyan S. Cutaneous angiosarcoma of the face and scalp. Plast Reconstr Surg. 1989;84:55-59.
- Bernstein JM, Irish JC, Brown DH, et al. Survival outcomes for cutaneous angiosarcoma of the scalp versus face. Head Neck. 2017;39:1205-1211.
- Köhler HF, Neves RI, Brechtbühl ER, et al. Cutaneous angiosarcoma of the head and neck: report of 23 cases from a single institution. Otolaryngol Head Neck Surg. 2008;139:519-524.
- Morales PH, Lindberg RD, Barkley HT Jr. Soft tissue angiosarcomas. Int J Radiat Oncol Biol Phys. 1981;7:1655-1659.
- Wollina U, Hansel G, Schönlebe J, et al. Cutaneous angiosarcoma is a rare aggressive malignant vascular tumour of the skin. J Eur Acad Dermatol Venereol. 2011;25:964-968.
- Wollina U, Koch A, Hansel G, et al. A 10-year analysis of cutaneous mesenchymal tumors (sarcomas and related entities) in a skin cancer center. Int J Dermatol. 2013;52:1189-1197.
- Bien E, Stachowicz-Stencel T, Balcerska A, et al. Angiosarcoma in children - still uncontrollable oncological problem. The report of the Polish Paediatric Rare Tumours Study. Eur J Cancer Care (Engl). 2009;18:411-420.
- Suzuki G, Yamazaki H, Takenaka H, et al. Definitive radiation therapy for angiosarcoma of the face and scalp. In Vivo. 2016;30:921-926.
- Miki Y, Tada T, Kamo R, et al. Single institutional experience of the treatment of angiosarcoma of the face and scalp. Br J Radiol. 2013;86:20130439.
- Ohguri T, Imada H, Nomoto S, et al. Angiosarcoma of the scalp treated with curative radiotherapy plus recombinant interleukin-2 immunotherapy. Int J Radiat Oncol Biol Phys. 2005;61:1446-1453.
- Clayton BD, Leshin B, Hitchcock MG, et al. Utility of rush paraffin-embedded tangential sections in the management of cutaneous neoplasms. Dermatol Surg. 2000;26:671-678.
- Goldberg DJ, Kim YA. Angiosarcoma of the scalp treated with Mohs micrographic surgery. J Dermatol Surg Oncol. 1993;19:156-158.
- Mikhail GR, Kelly AP Jr. Malignant angioendothelioma of the face. J Dermatol Surg Oncol. 1977;3:181-183.
- Muscarella VA. Angiosarcoma treated by Mohs micrographic surgery. J Dermatol Surg Oncol. 1993;19:1132-1133.
- Bullen R, Larson PO, Landeck AE, et al. Angiosarcoma of the head and neck managed by a combination of multiple biopsies to determine tumor margin and radiation therapy. report of three cases and review of the literature. Dermatol Surg. 1998;24:1105-1110.
- Wiwatwongwana D, White VA, Dolman PJ. Two cases of periocular cutaneous angiosarcoma. Ophthalmic Plast Reconstr Surg. 2010;26:365-366.
- Morrison WH, Byers RM, Garden AS, et al. Cutaneous angiosarcoma of the head and neck. A therapeutic dilemma. Cancer. 1995;76:319-327.
- Hata M, Wada H, Ogino I, et al. Radiation therapy for angiosarcoma of the scalp: treatment outcomes of total scalp irradiation with X-rays and electrons. Strahlenther Onkol. 2014;190:899-904.
- Hwang K, Kim MY, Lee SH. Recommendations for therapeutic decisions of angiosarcoma of the scalp and face. J Craniofac Surg. 2015;26:E253-E256.
Practice Points
- Angiosarcoma is a rare tumor that is difficult to treat, with multiple treatment options being utilized.
- Within this systematic review, wide local excision (WLE) combined with radiotherapy (RT), chemotherapy, and immunotherapy, as well as Mohs micrographic surgery (MMS), offered the longest mean (SD) overall survival time.
- When clinicians are tasked with treating primary cutaneous angiosarcoma of the head and neck, they should consider MMS or WLE combined with RT.
ObGyn’s steady progress toward going green in the OR—but gaps persist
Have you ever looked at the operating room (OR) trash bin at the end of a case and wondered if all that waste is necessary? Since I started my residency, not a day goes by that I have not asked myself this question.
In the mid-1990s, John Elkington introduced the concept of the triple bottom line—that is, people, planet, and profit—for implementation and measurement of sustainability in businesses.1 The health care sector is no exception when it comes to the bottom line! However, “people” remain the priority. What is our role, as ObGyns, in protecting the “planet” while keeping the “people” safe?
According to the World Health Organization (WHO), climate change remains the single biggest health threat to humanity.2 The health care system is both the victim and the culprit. Studies suggest that the health care system, second to the food industry, is the biggest contributor to waste production in the United States. This sector generates more than 6,000 metric tons of waste each day and nearly 4 million tons (3.6 million metric tons) of solid waste each year.3 The health care system is responsible for an estimated 8% to 10% of total greenhouse gas emissions in the United States; the US health care system alone contributes to more than one-fourth of the global health care carbon footprint. If it were a country, the US health care system would rank 13th among all countries in emissions.4In turn, pollution produced by the health sector negatively impacts population health, further burdening the health care system. According to 2013 study data, the annual health damage caused by health care pollution was comparable to that of the deaths caused by preventable medical error.4
Aside from the environmental aspects, hospital waste disposal is expensive; reducing this cost is a potential area of interest for institutions.
As ObGyns, what is our role in reducing our waste generation and carbon footprint while keeping patients safe?
Defining health care waste, and disposal considerations
The WHO defines health care waste as including “the waste generated by health-care establishments, research facilities, and laboratories” as well as waste from scattered sources such as home dialysis and insulin injections.5 Despite representing a relatively small physical area of hospitals, labor and delivery units combined with ORs account for approximately 70% of all hospital waste.3 Operating room waste consists of disposable surgical supplies, personal protective equipment, drapes, plastic wrappers, sterile blue wraps, glass, cardboard, packaging material, medications, fluids, and other materials (FIGURE 1).
The WHO also notes that of all the waste generated by health care activities, about 85% is general, nonhazardous waste that is comparable to domestic waste.6 Hazardous waste is any material that poses a health risk, including potentially infectious materials, such as blood-soaked gauze, sharps, pharmaceuticals, or radioactive materials.6
Disposal of hazardous waste is expensiveand energy consuming as it is typically incinerated rather than disposed of in a landfill. This process produces substantial greenhouse gases, about 3 kg of carbon dioxide for every 1 kg of hazardous waste.7
Red bags are used for hazardous waste disposal, while clear bags are used for general waste. Operating rooms produce about two-thirds of the hospital red-bag waste.8 Waste segregation unfortunately is not accurate, and as much as 90% of OR general waste is improperly designated as hazardous waste.3 Drapes and uncontaminated, needleless syringes, for example, should be disposed of in clear bags, but often they are instead directed to the red-bag and sharps container (FIGURE 2).
Obstetrics and gynecology has an important role to play in accurate waste segregation given the specialty’s frequent interaction with bodily fluids. Clinicians and other staff need to recognize and appropriately separate hazardous waste from general waste. For instance, not all fabrics involved in a case should be disposed of in the red bin, only those saturated with blood or body fluids. Educating health care staff and placing instruction posters on the red trash bins potentially could aid in accurate waste segregation and reduce regulated waste while decreasing disposal costs.
Recycling in the OR
Recycling has become an established practice in many health care facilities and ORs. Studies suggest that introducing recycling programs in ORs not only reduces carbon footprints but also reduces costs.3 One study reported that US academic medical centers consume 2 million lb ($15 million) each year of recoverable medical supplies.9
Single-stream recycling, a system in which all recyclable material—including plastics, paper, metal, and glass—are placed in a single bin without segregation at the collection site, has gained in popularity. Recycling can be implemented both in ORs and in other perioperative areas where regular trash bins are located.
In a study done at Oxford University Hospitals in the United Kingdom, introducing recycling bins in every OR, as well as in recovery and staff rest areas, helped improve waste segregation such that approximately 22% of OR waste was recycled.10 Studies show that recycling programs not only decrease the health care carbon footprint but also have a considerable financial impact. Albert and colleagues demonstrated that introducing a single-stream recycling program to a 9-OR day (or ambulatory) surgery center could redirect more than 4 tons of waste each month and saved thousands of dollars.11
Despite continued improvement in recycling programs, the segregation process is still far from optimal. In a survey done at the Mayo Clinic by Azouz and colleagues, more than half of the staff reported being unclear about which OR items are recyclable and nearly half reported that lack of knowledge was the barrier to proper recycling.12 That study also showed that after implementation of a recycling education program, costs decreased 10% relative to the same time period in prior years.12
Blue wraps. One example of recycling optimization is blue wraps, the polypropylene (No. 5 plastic) material used for wrapping surgical instruments. Blue wraps account for approximately 19% of OR waste and 5% of all hospital waste.11 Blue wraps are not biodegradable and also are not widely recycled. In recent years, a resale market has emerged for blue wraps, as they can be used for production of other No. 5 plastic items.9 By reselling blue wraps, revenue can be generated by recycling a necessary packing material that would otherwise require payment for disposal.
Sterility considerations. While recycling in ORs may raise concern due to the absolute sterility required in procedural settings, technologic developments have been promising in advancing safe recycling to reduce carbon footprints and health care costs without compromising patients’ safety. Segregation of waste from recyclable packaging material prior to the case, as well as directing trash to the correct bin (regular vs red bin), is one example. Moreover, because about 80% of all OR waste is generated during the set up before the patient arrives in the OR, it is not contaminated and can be safely recycled.13
Continue to: Packaging material...
Packaging material
A substantial part of OR waste consists of packaging material; of all OR waste, 26% consists of plastics and 7%, paper and cartons.14 Increasing use of disposable or “single use” medical products in ORs, along with the intention to safeguard sterility, contributes significantly to the generation of medical waste in operating units. Containers, wraps and overwraps, cardboard, and plastic packaging are all composed of materials that when clean, can be recycled; however, these items often end up in the landfill (FIGURE 3).
Although the segregation of packaging material to recycling versus regular trash versus red bin is of paramount importance, packaging design plays a significant role as well. In 2018, Boston Scientific introduced a new packaging design for ureteral stents that reduced plastic use in packaging by 120,000 lb each year.15 Despite the advances in the medical packaging industry to increase sustainability while safeguarding sterility for medical devices, there is still room for innovation in this area.
Reducing overage by judicious selection of surgical devices, instruments, and supplies
Overage is the term used to describe surgical inventory that is opened and prepared for surgery but ultimately not used and therefore discarded. Design of surgical carts and instrument and supply selection requires direct input from ObGyns. Opening only the needed instruments while ensuring ready availability of potentially needed supplies can significantly reduce OR waste generation as well as decrease chemical pollution generated by instrument sterilization. Decreasing OR overage reduces overall costs as well (FIGURE 4).
In a pilot study at the University of Massachusetts, Albert and colleagues examined the sets of disposable items and instruments designated for common plastic and hand surgery procedures.11 They identified the supplies and instruments that are routinely opened and wasted, based on surgeons’ interview responses, and redesigned the sets. Fifteen items were removed from disposable plastic surgery packs and 7 items from hand surgery packs. The authors reported saving thousands of dollars per year with these changes alone, as well as reducing waste.11 This same concept easily could be implemented in obstetrics and gynecology. We must ask ourselves: Do we always need, for example, a complete dilation and curettage kit to place the uterine manipulator prior to a minimally invasive hysterectomy?
In another pilot study, Greenberg and colleagues investigated whether cesarean deliveries consistently could be performed in a safe manner with only 20 instruments in the surgical kit.16 Obstetricians rated the 20-instrument kit an 8.7 out of 10 for performing cesarean deliveries safely.16
In addition to instrument selection, surgeons have a role in other supply use and waste generation: for instance, opening multiple pairs of surgical gloves and surgical gowns in advance when most of them will not be used during the case. Furthermore, many ObGyn surgeons routinely change gloves or even gowns during gynecologic procedures when they go back and forth between the vaginal and abdominal fields. Is the perineum “dirty” after application of a surgical prep solution?
In an observational study, Shockley and colleagues investigated the type and quantity of bacteria found intraoperatively on the abdomen, vagina, surgical gloves, instrument tips, and uterus at distinct time points during total laparoscopic hysterectomy.17 They showed that in 98.9% of cultures, the overall bacterial concentrations did not exceed the threshold for infection. There was no bacterial growth from vaginal cultures, and the only samples with some bacterial growth belonged to the surgeon’s gloves after specimen extraction; about one-third of samples showed growth after specimen extraction, but only 1 sample had a bacterial load above the infectious threshold of 5,000 colony-forming units per mL. The authors therefore suggested that if a surgeon changes gloves, doing so after specimen extraction and before turning attention back to the abdomen for vaginal cuff closure may be most effective in reducing bacterial load.17
Surgical site infection contributes to medical cost and likely medical waste as well. For example, surgical site infection may require prolonged treatments, tests, and medical instruments. In severe cases with abscesses, treatment entails hospitalization with prolonged antibiotic therapy with or without procedures to drain the collections. Further research therefore is warranted to investigate safe and environmentally friendly practices.
Myriad products are introduced to the medical system each day, some of which replace conventional tools. For instance, low-density polyethylene, or LDPE, transfer sheet is advertised for lateral patient transfer from the OR table to the bed or stretcher. This No. 4–coded plastic, while recyclable, is routinely discarded as trash in ORs. One ergonomic study found that reusable slide boards are as effective for reducing friction and staff muscle activities and are noninferior to the plastic sheets.18
Steps to making an impact
Operating rooms and labor and delivery units are responsible for a large proportion of hospital waste, and therefore they are of paramount importance in reducing waste and carbon footprint at the individual and institutional level. Reduction of OR waste not only is environmentally conscious but also decreases cost. Steps as small as individual practices to as big as changing infrastructures can make an impact. For instance:
- redesigning surgical carts
- reformulating surgeon-specific supply lists
- raising awareness about surgical overage
- encouraging recycling through education and audit
- optimizing surgical waste segregation through educational posters.
These are all simple steps that could significantly reduce waste and carbon footprint.
Bottom line
Although waste reduction is the responsibility of all health care providers, as leaders in their workplace physicians can serve as role models by implementing “green” practices in procedural units. Raising awareness and using a team approach is critical to succeed in our endeavors to move toward an environmentally friendly future. ●
- Elkington J. Towards the sustainable corporation: win-winwin business strategies for sustainable development. Calif Manage Rev. 1994;36:90-100.
- Climate change and health. October 30, 2021. World Health Organization. Accessed October 10, 2022. https://www.who .int/news-room/fact-sheets/detail/climate-change-and -health
- Kwakye G, Brat GA, Makary MA. Green surgical practices for health care. Arch Surg. 2011;146:131-136.
- Eckelman MJ, Sherman J. Environmental impacts of the US health care system and effects on public health. PloS One. 2016;11:e0157014.
- Pruss A, Giroult E, Rushbrook P. Safe management of wastes from health-care activities. World Health Organization; 1999.
- Health-care waste. February 8, 2018. World Health Organization. Accessed October 4, 2022. https://www.who. int/news-room/fact-sheets/detail/health-care-waste2
- Southorn T, Norrish AR, Gardner K, et al. Reducing the carbon footprint of the operating theatre: a multicentre quality improvement report. J Perioper Pract. 2013;23:144-146.
- Greening the OR. Practice Greenhealth. Accessed October 24, 2022. https://practicegreenhealth.org/topics/greening -operating-room/greening-or
- Babu MA, Dalenberg AK, Goodsell G, et al. Greening the operating room: results of a scalable initiative to reduce waste and recover supply costs. Neurosurgery. 2019;85:432-437.
- Oxford University Hospitals NHS Trust. Introducing recycling into the operating theatres. Mapping Greener Healthcare. Accessed October 14, 2022. https://map .sustainablehealthcare.org.uk/oxford-radcliffe-hospitals -nhs-trust/introducing-recycling-operating-theatres
- Albert MG, Rothkopf DM. Operating room waste reduction in plastic and hand surgery. Plast Surg. 2015;23:235-238.
- Azouz S, Boyll P, Swanson M, et al. Managing barriers to recycling in the operating room. Am J Surg. 2019;217:634-638.
- Wyssusek KH, Keys MT, van Zundert AAJ. Operating room greening initiatives—the old, the new, and the way forward: a narrative review. Waste Manag Res. 2019;37:3-19.
- Tieszen ME, Gruenberg JC. A quantitative, qualitative, and critical assessment of surgical waste: surgeons venture through the trash can. JAMA. 1992;267:2765-2768.
- Boston Scientific 2018 Performance Report. Boston Scientific. Accessed November 19, 2022. https://www.bostonscientific. com/content/dam/bostonscientific/corporate/citizenship /sustainability/Boston_Scientific_Performance _Report_2018.pdf
- Greenberg JA, Wylie B, Robinson JN. A pilot study to assess the adequacy of the Brigham 20 Kit for cesarean delivery. Int J Gynaecol Obstet. 2012;117:157-159.
- Shockley ME, Beran B, Nutting H, et al. Sterility of selected operative sites during total laparoscopic hysterectomy. J Minim Invasive Gynecol. 2017;24:990-997.
- Al-Qaisi SK, El Tannir A, Younan LA, et al. An ergonomic assessment of using laterally-tilting operating room tables and friction reducing devices for patient lateral transfers. Appl Ergon. 2020;87:103122.
Have you ever looked at the operating room (OR) trash bin at the end of a case and wondered if all that waste is necessary? Since I started my residency, not a day goes by that I have not asked myself this question.
In the mid-1990s, John Elkington introduced the concept of the triple bottom line—that is, people, planet, and profit—for implementation and measurement of sustainability in businesses.1 The health care sector is no exception when it comes to the bottom line! However, “people” remain the priority. What is our role, as ObGyns, in protecting the “planet” while keeping the “people” safe?
According to the World Health Organization (WHO), climate change remains the single biggest health threat to humanity.2 The health care system is both the victim and the culprit. Studies suggest that the health care system, second to the food industry, is the biggest contributor to waste production in the United States. This sector generates more than 6,000 metric tons of waste each day and nearly 4 million tons (3.6 million metric tons) of solid waste each year.3 The health care system is responsible for an estimated 8% to 10% of total greenhouse gas emissions in the United States; the US health care system alone contributes to more than one-fourth of the global health care carbon footprint. If it were a country, the US health care system would rank 13th among all countries in emissions.4In turn, pollution produced by the health sector negatively impacts population health, further burdening the health care system. According to 2013 study data, the annual health damage caused by health care pollution was comparable to that of the deaths caused by preventable medical error.4
Aside from the environmental aspects, hospital waste disposal is expensive; reducing this cost is a potential area of interest for institutions.
As ObGyns, what is our role in reducing our waste generation and carbon footprint while keeping patients safe?
Defining health care waste, and disposal considerations
The WHO defines health care waste as including “the waste generated by health-care establishments, research facilities, and laboratories” as well as waste from scattered sources such as home dialysis and insulin injections.5 Despite representing a relatively small physical area of hospitals, labor and delivery units combined with ORs account for approximately 70% of all hospital waste.3 Operating room waste consists of disposable surgical supplies, personal protective equipment, drapes, plastic wrappers, sterile blue wraps, glass, cardboard, packaging material, medications, fluids, and other materials (FIGURE 1).
The WHO also notes that of all the waste generated by health care activities, about 85% is general, nonhazardous waste that is comparable to domestic waste.6 Hazardous waste is any material that poses a health risk, including potentially infectious materials, such as blood-soaked gauze, sharps, pharmaceuticals, or radioactive materials.6
Disposal of hazardous waste is expensiveand energy consuming as it is typically incinerated rather than disposed of in a landfill. This process produces substantial greenhouse gases, about 3 kg of carbon dioxide for every 1 kg of hazardous waste.7
Red bags are used for hazardous waste disposal, while clear bags are used for general waste. Operating rooms produce about two-thirds of the hospital red-bag waste.8 Waste segregation unfortunately is not accurate, and as much as 90% of OR general waste is improperly designated as hazardous waste.3 Drapes and uncontaminated, needleless syringes, for example, should be disposed of in clear bags, but often they are instead directed to the red-bag and sharps container (FIGURE 2).
Obstetrics and gynecology has an important role to play in accurate waste segregation given the specialty’s frequent interaction with bodily fluids. Clinicians and other staff need to recognize and appropriately separate hazardous waste from general waste. For instance, not all fabrics involved in a case should be disposed of in the red bin, only those saturated with blood or body fluids. Educating health care staff and placing instruction posters on the red trash bins potentially could aid in accurate waste segregation and reduce regulated waste while decreasing disposal costs.
Recycling in the OR
Recycling has become an established practice in many health care facilities and ORs. Studies suggest that introducing recycling programs in ORs not only reduces carbon footprints but also reduces costs.3 One study reported that US academic medical centers consume 2 million lb ($15 million) each year of recoverable medical supplies.9
Single-stream recycling, a system in which all recyclable material—including plastics, paper, metal, and glass—are placed in a single bin without segregation at the collection site, has gained in popularity. Recycling can be implemented both in ORs and in other perioperative areas where regular trash bins are located.
In a study done at Oxford University Hospitals in the United Kingdom, introducing recycling bins in every OR, as well as in recovery and staff rest areas, helped improve waste segregation such that approximately 22% of OR waste was recycled.10 Studies show that recycling programs not only decrease the health care carbon footprint but also have a considerable financial impact. Albert and colleagues demonstrated that introducing a single-stream recycling program to a 9-OR day (or ambulatory) surgery center could redirect more than 4 tons of waste each month and saved thousands of dollars.11
Despite continued improvement in recycling programs, the segregation process is still far from optimal. In a survey done at the Mayo Clinic by Azouz and colleagues, more than half of the staff reported being unclear about which OR items are recyclable and nearly half reported that lack of knowledge was the barrier to proper recycling.12 That study also showed that after implementation of a recycling education program, costs decreased 10% relative to the same time period in prior years.12
Blue wraps. One example of recycling optimization is blue wraps, the polypropylene (No. 5 plastic) material used for wrapping surgical instruments. Blue wraps account for approximately 19% of OR waste and 5% of all hospital waste.11 Blue wraps are not biodegradable and also are not widely recycled. In recent years, a resale market has emerged for blue wraps, as they can be used for production of other No. 5 plastic items.9 By reselling blue wraps, revenue can be generated by recycling a necessary packing material that would otherwise require payment for disposal.
Sterility considerations. While recycling in ORs may raise concern due to the absolute sterility required in procedural settings, technologic developments have been promising in advancing safe recycling to reduce carbon footprints and health care costs without compromising patients’ safety. Segregation of waste from recyclable packaging material prior to the case, as well as directing trash to the correct bin (regular vs red bin), is one example. Moreover, because about 80% of all OR waste is generated during the set up before the patient arrives in the OR, it is not contaminated and can be safely recycled.13
Continue to: Packaging material...
Packaging material
A substantial part of OR waste consists of packaging material; of all OR waste, 26% consists of plastics and 7%, paper and cartons.14 Increasing use of disposable or “single use” medical products in ORs, along with the intention to safeguard sterility, contributes significantly to the generation of medical waste in operating units. Containers, wraps and overwraps, cardboard, and plastic packaging are all composed of materials that when clean, can be recycled; however, these items often end up in the landfill (FIGURE 3).
Although the segregation of packaging material to recycling versus regular trash versus red bin is of paramount importance, packaging design plays a significant role as well. In 2018, Boston Scientific introduced a new packaging design for ureteral stents that reduced plastic use in packaging by 120,000 lb each year.15 Despite the advances in the medical packaging industry to increase sustainability while safeguarding sterility for medical devices, there is still room for innovation in this area.
Reducing overage by judicious selection of surgical devices, instruments, and supplies
Overage is the term used to describe surgical inventory that is opened and prepared for surgery but ultimately not used and therefore discarded. Design of surgical carts and instrument and supply selection requires direct input from ObGyns. Opening only the needed instruments while ensuring ready availability of potentially needed supplies can significantly reduce OR waste generation as well as decrease chemical pollution generated by instrument sterilization. Decreasing OR overage reduces overall costs as well (FIGURE 4).
In a pilot study at the University of Massachusetts, Albert and colleagues examined the sets of disposable items and instruments designated for common plastic and hand surgery procedures.11 They identified the supplies and instruments that are routinely opened and wasted, based on surgeons’ interview responses, and redesigned the sets. Fifteen items were removed from disposable plastic surgery packs and 7 items from hand surgery packs. The authors reported saving thousands of dollars per year with these changes alone, as well as reducing waste.11 This same concept easily could be implemented in obstetrics and gynecology. We must ask ourselves: Do we always need, for example, a complete dilation and curettage kit to place the uterine manipulator prior to a minimally invasive hysterectomy?
In another pilot study, Greenberg and colleagues investigated whether cesarean deliveries consistently could be performed in a safe manner with only 20 instruments in the surgical kit.16 Obstetricians rated the 20-instrument kit an 8.7 out of 10 for performing cesarean deliveries safely.16
In addition to instrument selection, surgeons have a role in other supply use and waste generation: for instance, opening multiple pairs of surgical gloves and surgical gowns in advance when most of them will not be used during the case. Furthermore, many ObGyn surgeons routinely change gloves or even gowns during gynecologic procedures when they go back and forth between the vaginal and abdominal fields. Is the perineum “dirty” after application of a surgical prep solution?
In an observational study, Shockley and colleagues investigated the type and quantity of bacteria found intraoperatively on the abdomen, vagina, surgical gloves, instrument tips, and uterus at distinct time points during total laparoscopic hysterectomy.17 They showed that in 98.9% of cultures, the overall bacterial concentrations did not exceed the threshold for infection. There was no bacterial growth from vaginal cultures, and the only samples with some bacterial growth belonged to the surgeon’s gloves after specimen extraction; about one-third of samples showed growth after specimen extraction, but only 1 sample had a bacterial load above the infectious threshold of 5,000 colony-forming units per mL. The authors therefore suggested that if a surgeon changes gloves, doing so after specimen extraction and before turning attention back to the abdomen for vaginal cuff closure may be most effective in reducing bacterial load.17
Surgical site infection contributes to medical cost and likely medical waste as well. For example, surgical site infection may require prolonged treatments, tests, and medical instruments. In severe cases with abscesses, treatment entails hospitalization with prolonged antibiotic therapy with or without procedures to drain the collections. Further research therefore is warranted to investigate safe and environmentally friendly practices.
Myriad products are introduced to the medical system each day, some of which replace conventional tools. For instance, low-density polyethylene, or LDPE, transfer sheet is advertised for lateral patient transfer from the OR table to the bed or stretcher. This No. 4–coded plastic, while recyclable, is routinely discarded as trash in ORs. One ergonomic study found that reusable slide boards are as effective for reducing friction and staff muscle activities and are noninferior to the plastic sheets.18
Steps to making an impact
Operating rooms and labor and delivery units are responsible for a large proportion of hospital waste, and therefore they are of paramount importance in reducing waste and carbon footprint at the individual and institutional level. Reduction of OR waste not only is environmentally conscious but also decreases cost. Steps as small as individual practices to as big as changing infrastructures can make an impact. For instance:
- redesigning surgical carts
- reformulating surgeon-specific supply lists
- raising awareness about surgical overage
- encouraging recycling through education and audit
- optimizing surgical waste segregation through educational posters.
These are all simple steps that could significantly reduce waste and carbon footprint.
Bottom line
Although waste reduction is the responsibility of all health care providers, as leaders in their workplace physicians can serve as role models by implementing “green” practices in procedural units. Raising awareness and using a team approach is critical to succeed in our endeavors to move toward an environmentally friendly future. ●
Have you ever looked at the operating room (OR) trash bin at the end of a case and wondered if all that waste is necessary? Since I started my residency, not a day goes by that I have not asked myself this question.
In the mid-1990s, John Elkington introduced the concept of the triple bottom line—that is, people, planet, and profit—for implementation and measurement of sustainability in businesses.1 The health care sector is no exception when it comes to the bottom line! However, “people” remain the priority. What is our role, as ObGyns, in protecting the “planet” while keeping the “people” safe?
According to the World Health Organization (WHO), climate change remains the single biggest health threat to humanity.2 The health care system is both the victim and the culprit. Studies suggest that the health care system, second to the food industry, is the biggest contributor to waste production in the United States. This sector generates more than 6,000 metric tons of waste each day and nearly 4 million tons (3.6 million metric tons) of solid waste each year.3 The health care system is responsible for an estimated 8% to 10% of total greenhouse gas emissions in the United States; the US health care system alone contributes to more than one-fourth of the global health care carbon footprint. If it were a country, the US health care system would rank 13th among all countries in emissions.4In turn, pollution produced by the health sector negatively impacts population health, further burdening the health care system. According to 2013 study data, the annual health damage caused by health care pollution was comparable to that of the deaths caused by preventable medical error.4
Aside from the environmental aspects, hospital waste disposal is expensive; reducing this cost is a potential area of interest for institutions.
As ObGyns, what is our role in reducing our waste generation and carbon footprint while keeping patients safe?
Defining health care waste, and disposal considerations
The WHO defines health care waste as including “the waste generated by health-care establishments, research facilities, and laboratories” as well as waste from scattered sources such as home dialysis and insulin injections.5 Despite representing a relatively small physical area of hospitals, labor and delivery units combined with ORs account for approximately 70% of all hospital waste.3 Operating room waste consists of disposable surgical supplies, personal protective equipment, drapes, plastic wrappers, sterile blue wraps, glass, cardboard, packaging material, medications, fluids, and other materials (FIGURE 1).
The WHO also notes that of all the waste generated by health care activities, about 85% is general, nonhazardous waste that is comparable to domestic waste.6 Hazardous waste is any material that poses a health risk, including potentially infectious materials, such as blood-soaked gauze, sharps, pharmaceuticals, or radioactive materials.6
Disposal of hazardous waste is expensiveand energy consuming as it is typically incinerated rather than disposed of in a landfill. This process produces substantial greenhouse gases, about 3 kg of carbon dioxide for every 1 kg of hazardous waste.7
Red bags are used for hazardous waste disposal, while clear bags are used for general waste. Operating rooms produce about two-thirds of the hospital red-bag waste.8 Waste segregation unfortunately is not accurate, and as much as 90% of OR general waste is improperly designated as hazardous waste.3 Drapes and uncontaminated, needleless syringes, for example, should be disposed of in clear bags, but often they are instead directed to the red-bag and sharps container (FIGURE 2).
Obstetrics and gynecology has an important role to play in accurate waste segregation given the specialty’s frequent interaction with bodily fluids. Clinicians and other staff need to recognize and appropriately separate hazardous waste from general waste. For instance, not all fabrics involved in a case should be disposed of in the red bin, only those saturated with blood or body fluids. Educating health care staff and placing instruction posters on the red trash bins potentially could aid in accurate waste segregation and reduce regulated waste while decreasing disposal costs.
Recycling in the OR
Recycling has become an established practice in many health care facilities and ORs. Studies suggest that introducing recycling programs in ORs not only reduces carbon footprints but also reduces costs.3 One study reported that US academic medical centers consume 2 million lb ($15 million) each year of recoverable medical supplies.9
Single-stream recycling, a system in which all recyclable material—including plastics, paper, metal, and glass—are placed in a single bin without segregation at the collection site, has gained in popularity. Recycling can be implemented both in ORs and in other perioperative areas where regular trash bins are located.
In a study done at Oxford University Hospitals in the United Kingdom, introducing recycling bins in every OR, as well as in recovery and staff rest areas, helped improve waste segregation such that approximately 22% of OR waste was recycled.10 Studies show that recycling programs not only decrease the health care carbon footprint but also have a considerable financial impact. Albert and colleagues demonstrated that introducing a single-stream recycling program to a 9-OR day (or ambulatory) surgery center could redirect more than 4 tons of waste each month and saved thousands of dollars.11
Despite continued improvement in recycling programs, the segregation process is still far from optimal. In a survey done at the Mayo Clinic by Azouz and colleagues, more than half of the staff reported being unclear about which OR items are recyclable and nearly half reported that lack of knowledge was the barrier to proper recycling.12 That study also showed that after implementation of a recycling education program, costs decreased 10% relative to the same time period in prior years.12
Blue wraps. One example of recycling optimization is blue wraps, the polypropylene (No. 5 plastic) material used for wrapping surgical instruments. Blue wraps account for approximately 19% of OR waste and 5% of all hospital waste.11 Blue wraps are not biodegradable and also are not widely recycled. In recent years, a resale market has emerged for blue wraps, as they can be used for production of other No. 5 plastic items.9 By reselling blue wraps, revenue can be generated by recycling a necessary packing material that would otherwise require payment for disposal.
Sterility considerations. While recycling in ORs may raise concern due to the absolute sterility required in procedural settings, technologic developments have been promising in advancing safe recycling to reduce carbon footprints and health care costs without compromising patients’ safety. Segregation of waste from recyclable packaging material prior to the case, as well as directing trash to the correct bin (regular vs red bin), is one example. Moreover, because about 80% of all OR waste is generated during the set up before the patient arrives in the OR, it is not contaminated and can be safely recycled.13
Continue to: Packaging material...
Packaging material
A substantial part of OR waste consists of packaging material; of all OR waste, 26% consists of plastics and 7%, paper and cartons.14 Increasing use of disposable or “single use” medical products in ORs, along with the intention to safeguard sterility, contributes significantly to the generation of medical waste in operating units. Containers, wraps and overwraps, cardboard, and plastic packaging are all composed of materials that when clean, can be recycled; however, these items often end up in the landfill (FIGURE 3).
Although the segregation of packaging material to recycling versus regular trash versus red bin is of paramount importance, packaging design plays a significant role as well. In 2018, Boston Scientific introduced a new packaging design for ureteral stents that reduced plastic use in packaging by 120,000 lb each year.15 Despite the advances in the medical packaging industry to increase sustainability while safeguarding sterility for medical devices, there is still room for innovation in this area.
Reducing overage by judicious selection of surgical devices, instruments, and supplies
Overage is the term used to describe surgical inventory that is opened and prepared for surgery but ultimately not used and therefore discarded. Design of surgical carts and instrument and supply selection requires direct input from ObGyns. Opening only the needed instruments while ensuring ready availability of potentially needed supplies can significantly reduce OR waste generation as well as decrease chemical pollution generated by instrument sterilization. Decreasing OR overage reduces overall costs as well (FIGURE 4).
In a pilot study at the University of Massachusetts, Albert and colleagues examined the sets of disposable items and instruments designated for common plastic and hand surgery procedures.11 They identified the supplies and instruments that are routinely opened and wasted, based on surgeons’ interview responses, and redesigned the sets. Fifteen items were removed from disposable plastic surgery packs and 7 items from hand surgery packs. The authors reported saving thousands of dollars per year with these changes alone, as well as reducing waste.11 This same concept easily could be implemented in obstetrics and gynecology. We must ask ourselves: Do we always need, for example, a complete dilation and curettage kit to place the uterine manipulator prior to a minimally invasive hysterectomy?
In another pilot study, Greenberg and colleagues investigated whether cesarean deliveries consistently could be performed in a safe manner with only 20 instruments in the surgical kit.16 Obstetricians rated the 20-instrument kit an 8.7 out of 10 for performing cesarean deliveries safely.16
In addition to instrument selection, surgeons have a role in other supply use and waste generation: for instance, opening multiple pairs of surgical gloves and surgical gowns in advance when most of them will not be used during the case. Furthermore, many ObGyn surgeons routinely change gloves or even gowns during gynecologic procedures when they go back and forth between the vaginal and abdominal fields. Is the perineum “dirty” after application of a surgical prep solution?
In an observational study, Shockley and colleagues investigated the type and quantity of bacteria found intraoperatively on the abdomen, vagina, surgical gloves, instrument tips, and uterus at distinct time points during total laparoscopic hysterectomy.17 They showed that in 98.9% of cultures, the overall bacterial concentrations did not exceed the threshold for infection. There was no bacterial growth from vaginal cultures, and the only samples with some bacterial growth belonged to the surgeon’s gloves after specimen extraction; about one-third of samples showed growth after specimen extraction, but only 1 sample had a bacterial load above the infectious threshold of 5,000 colony-forming units per mL. The authors therefore suggested that if a surgeon changes gloves, doing so after specimen extraction and before turning attention back to the abdomen for vaginal cuff closure may be most effective in reducing bacterial load.17
Surgical site infection contributes to medical cost and likely medical waste as well. For example, surgical site infection may require prolonged treatments, tests, and medical instruments. In severe cases with abscesses, treatment entails hospitalization with prolonged antibiotic therapy with or without procedures to drain the collections. Further research therefore is warranted to investigate safe and environmentally friendly practices.
Myriad products are introduced to the medical system each day, some of which replace conventional tools. For instance, low-density polyethylene, or LDPE, transfer sheet is advertised for lateral patient transfer from the OR table to the bed or stretcher. This No. 4–coded plastic, while recyclable, is routinely discarded as trash in ORs. One ergonomic study found that reusable slide boards are as effective for reducing friction and staff muscle activities and are noninferior to the plastic sheets.18
Steps to making an impact
Operating rooms and labor and delivery units are responsible for a large proportion of hospital waste, and therefore they are of paramount importance in reducing waste and carbon footprint at the individual and institutional level. Reduction of OR waste not only is environmentally conscious but also decreases cost. Steps as small as individual practices to as big as changing infrastructures can make an impact. For instance:
- redesigning surgical carts
- reformulating surgeon-specific supply lists
- raising awareness about surgical overage
- encouraging recycling through education and audit
- optimizing surgical waste segregation through educational posters.
These are all simple steps that could significantly reduce waste and carbon footprint.
Bottom line
Although waste reduction is the responsibility of all health care providers, as leaders in their workplace physicians can serve as role models by implementing “green” practices in procedural units. Raising awareness and using a team approach is critical to succeed in our endeavors to move toward an environmentally friendly future. ●
- Elkington J. Towards the sustainable corporation: win-winwin business strategies for sustainable development. Calif Manage Rev. 1994;36:90-100.
- Climate change and health. October 30, 2021. World Health Organization. Accessed October 10, 2022. https://www.who .int/news-room/fact-sheets/detail/climate-change-and -health
- Kwakye G, Brat GA, Makary MA. Green surgical practices for health care. Arch Surg. 2011;146:131-136.
- Eckelman MJ, Sherman J. Environmental impacts of the US health care system and effects on public health. PloS One. 2016;11:e0157014.
- Pruss A, Giroult E, Rushbrook P. Safe management of wastes from health-care activities. World Health Organization; 1999.
- Health-care waste. February 8, 2018. World Health Organization. Accessed October 4, 2022. https://www.who. int/news-room/fact-sheets/detail/health-care-waste2
- Southorn T, Norrish AR, Gardner K, et al. Reducing the carbon footprint of the operating theatre: a multicentre quality improvement report. J Perioper Pract. 2013;23:144-146.
- Greening the OR. Practice Greenhealth. Accessed October 24, 2022. https://practicegreenhealth.org/topics/greening -operating-room/greening-or
- Babu MA, Dalenberg AK, Goodsell G, et al. Greening the operating room: results of a scalable initiative to reduce waste and recover supply costs. Neurosurgery. 2019;85:432-437.
- Oxford University Hospitals NHS Trust. Introducing recycling into the operating theatres. Mapping Greener Healthcare. Accessed October 14, 2022. https://map .sustainablehealthcare.org.uk/oxford-radcliffe-hospitals -nhs-trust/introducing-recycling-operating-theatres
- Albert MG, Rothkopf DM. Operating room waste reduction in plastic and hand surgery. Plast Surg. 2015;23:235-238.
- Azouz S, Boyll P, Swanson M, et al. Managing barriers to recycling in the operating room. Am J Surg. 2019;217:634-638.
- Wyssusek KH, Keys MT, van Zundert AAJ. Operating room greening initiatives—the old, the new, and the way forward: a narrative review. Waste Manag Res. 2019;37:3-19.
- Tieszen ME, Gruenberg JC. A quantitative, qualitative, and critical assessment of surgical waste: surgeons venture through the trash can. JAMA. 1992;267:2765-2768.
- Boston Scientific 2018 Performance Report. Boston Scientific. Accessed November 19, 2022. https://www.bostonscientific. com/content/dam/bostonscientific/corporate/citizenship /sustainability/Boston_Scientific_Performance _Report_2018.pdf
- Greenberg JA, Wylie B, Robinson JN. A pilot study to assess the adequacy of the Brigham 20 Kit for cesarean delivery. Int J Gynaecol Obstet. 2012;117:157-159.
- Shockley ME, Beran B, Nutting H, et al. Sterility of selected operative sites during total laparoscopic hysterectomy. J Minim Invasive Gynecol. 2017;24:990-997.
- Al-Qaisi SK, El Tannir A, Younan LA, et al. An ergonomic assessment of using laterally-tilting operating room tables and friction reducing devices for patient lateral transfers. Appl Ergon. 2020;87:103122.
- Elkington J. Towards the sustainable corporation: win-winwin business strategies for sustainable development. Calif Manage Rev. 1994;36:90-100.
- Climate change and health. October 30, 2021. World Health Organization. Accessed October 10, 2022. https://www.who .int/news-room/fact-sheets/detail/climate-change-and -health
- Kwakye G, Brat GA, Makary MA. Green surgical practices for health care. Arch Surg. 2011;146:131-136.
- Eckelman MJ, Sherman J. Environmental impacts of the US health care system and effects on public health. PloS One. 2016;11:e0157014.
- Pruss A, Giroult E, Rushbrook P. Safe management of wastes from health-care activities. World Health Organization; 1999.
- Health-care waste. February 8, 2018. World Health Organization. Accessed October 4, 2022. https://www.who. int/news-room/fact-sheets/detail/health-care-waste2
- Southorn T, Norrish AR, Gardner K, et al. Reducing the carbon footprint of the operating theatre: a multicentre quality improvement report. J Perioper Pract. 2013;23:144-146.
- Greening the OR. Practice Greenhealth. Accessed October 24, 2022. https://practicegreenhealth.org/topics/greening -operating-room/greening-or
- Babu MA, Dalenberg AK, Goodsell G, et al. Greening the operating room: results of a scalable initiative to reduce waste and recover supply costs. Neurosurgery. 2019;85:432-437.
- Oxford University Hospitals NHS Trust. Introducing recycling into the operating theatres. Mapping Greener Healthcare. Accessed October 14, 2022. https://map .sustainablehealthcare.org.uk/oxford-radcliffe-hospitals -nhs-trust/introducing-recycling-operating-theatres
- Albert MG, Rothkopf DM. Operating room waste reduction in plastic and hand surgery. Plast Surg. 2015;23:235-238.
- Azouz S, Boyll P, Swanson M, et al. Managing barriers to recycling in the operating room. Am J Surg. 2019;217:634-638.
- Wyssusek KH, Keys MT, van Zundert AAJ. Operating room greening initiatives—the old, the new, and the way forward: a narrative review. Waste Manag Res. 2019;37:3-19.
- Tieszen ME, Gruenberg JC. A quantitative, qualitative, and critical assessment of surgical waste: surgeons venture through the trash can. JAMA. 1992;267:2765-2768.
- Boston Scientific 2018 Performance Report. Boston Scientific. Accessed November 19, 2022. https://www.bostonscientific. com/content/dam/bostonscientific/corporate/citizenship /sustainability/Boston_Scientific_Performance _Report_2018.pdf
- Greenberg JA, Wylie B, Robinson JN. A pilot study to assess the adequacy of the Brigham 20 Kit for cesarean delivery. Int J Gynaecol Obstet. 2012;117:157-159.
- Shockley ME, Beran B, Nutting H, et al. Sterility of selected operative sites during total laparoscopic hysterectomy. J Minim Invasive Gynecol. 2017;24:990-997.
- Al-Qaisi SK, El Tannir A, Younan LA, et al. An ergonomic assessment of using laterally-tilting operating room tables and friction reducing devices for patient lateral transfers. Appl Ergon. 2020;87:103122.
Erythema Ab Igne: A Clinical Review
Erythema ab igne (EAI)(also known as toasted skin syndrome) was first described in the British Journal of Dermatology in the 20th century, 1 though it was known by physicians long before. Reticular netlike skin changes were seen in association with patients who spent extended time directly next to a heat source. This association led to the name of this condition, which literally means “redness by fire.” Indeed, EAI induced by chronic heat exposure has been described across the world for centuries. For example, in the cold regions of northern China, people used to sleep on beds of hot bricks called kang to stay warm at night. The people of India’s Kashmir district carried pots of hot coals called kangri next to the skin under large woven shawls to stay warm. In the past, Irish women often spent much time by a turf- or peat-burning fire. Chronic heat exposure in these cases can lead not only to EAI but also to aggressive types of cancer, often with a latency of 30 years or more. 2
More recently, the invention of home central heating led to a stark decrease in the number of cases associated with combustion-based heat, with a transition to etiologies such as use of hot water bottles, electric blankets, and electric space heaters. Over time, technological advances led to ever-increasing potential causes for EAI, such as laptops or cell phones, car heaters and heated seats, heated blankets,3,4 infrared lamps for food, and even medical devices such as ultrasound-based heating products and convective temperature management systems for hospitalized patients. As technology evolves, so do the potential causes of EAI, requiring clinicians to diagnose and deduce the cause through a thorough social and medical history as well as a workup on the present illness with considerations for the anatomical location.5-7 Herein, we describe the etiology of EAI, diagnosis, and treatment options.
Clinical Characteristics
Erythema ab igne begins as mild, transient, and erythematous macules and patches in a reticular pattern that resolve minutes to hours after removal of the heat source. With weeks to months of continued or repeated application of the heat source, the affected area eventually becomes hyperpigmented where there once was erythema (Figures 1 and 2). Sometimes papules, bullae, telangiectasia, and hyperkeratosis also form. The rash usually is asymptomatic, though pain, pruritus, and dysesthesia have been reported.7 Dermoscopy of EAI in the hyperpigmented stage can reveal diffuse superficial dark pigmentation, telangiectasia, and mild whitish scaling.8 Although the pathogenesis has remained elusive over the years, lesions do seem to be mostly associated with cumulative exposure to heat rather than length of exposure.7
Etiology of EAI
Anatomic Location—The affected site depends on the source of heat (Table). Classic examples of this condition include a patient with EAI presenting on the anterior thighs after working in front of a hot oven or a patient with chronic back pain presenting with lower-back EAI secondary to frequent use of a hot water bottle or heating pad.7 With evolving technology over the last few decades, new etiologies have become more common—teenagers are presenting with anterior thigh EAI secondary to frequent laptop use2-29; patients are holding warm cell phones in their pant pockets, leading to unilateral geometric EAI on the anterior thigh (front pocket) or buttock (back pocket)30; plug-in radiators under computer desks are causing EAI on the lower legs31-34; and automobile seat heaters have been shown to cause EAI on the posterior legs.5,35-37 Clinicians should consider anatomic location a critical clue for etiology.
Social History—There are rarer and more highly specific causes of EAI than simple heat exposure that can be parsed from a patient’s social history. Occupational exposure has been documented, such as bakers with exposure to ovens, foundry workers with exposure to heated metals, or fast-food workers with chronic exposure to infrared food lamps.6,7 There also are cultural practices that can cause EAI. For example, the practice of cupping with moxibustion was shown to create a specific pattern in the shape of the cultural tool used.38 When footbaths with Chinese herbal remedies are performed frequently with high heat, they can lead to EAI on the feet with a linear border at the ankles. There also have been reports of kotatsu (heated tables in Japan) leading to lower-body EAI.39,40 These cultural practices also are more common in patients with darker skin types, which can lead to hyperpigmentation that is difficult to treat, making early diagnosis important.7
Medical History—Case reports have shown EAI caused by patients attempting to use heat-based methods for pain relief of an underlying serious disease such as cancer, bowel pathology (abdominal EAI), spinal disc prolapse (midline back EAI),41 sickle cell anemia, and renal pathology (posterior upper flank EAI).6,7,40-49 Patients with hypothyroidism or anorexia have been noted to have generalized EAI sparing the face secondary to repeated and extended hot baths or showers.50-53 One patient with schizophrenia was shown to have associated thermophilia due to a delusion that led the patient to soak in hot baths for long periods of time, leading to EAI.54 Finally, all physicians should be aware of iatrogenic causes of EAI, such as use of warming devices, ultrasound-based warming techniques, and laser therapy for lipolysis. Inquire about the patient’s surgical history or intensive care unit stays as well as alternative medicine or chiropractic visits. Obtaining a history of medical procedures can be enlightening when an etiology is not immediately clear.7,55,56
Diagnosis
Erythema ab igne is a clinical diagnosis based on recognizable cutaneous findings and a clear history of moderate heat exposure. However, when a clinical diagnosis of EAI is not certain (eg, when unable to obtain a clear history from the patient) or when malignant transformation is suspected, a biopsy can be performed. Pathologically, hematoxylin and eosin staining of EAI classically reveals dilated small vascular channels in the superficial dermis, hence a clinically reticular rash; interface dermatitis clinically manifesting as erythema; and pigment incontinence with melanin-laden macrophages consistent with clinical hyperpigmentation. Finally, for unclear reasons, increased numbers of elastic fibers classically are seen in biopsies of EAI.7
Differential Diagnosis
The differential diagnosis for a reticular patch includes livedo reticularis (Figure 3), which usually manifests as a more generalized rash in patients with chronic disease or coagulopathy such as systemic lupus erythematosus, cryoglobulinemia, or Raynaud phenomenon. When differentiating EAI from livedo reticularis or cutis marmorata, consider that both alternative diagnoses are more vascular appearing and are associated with cold exposure rather than heat exposure. In cases that are less reticular, livedo racemosa can be considered in the differential diagnosis. Finally, poikiloderma of Civatte can be reticular, particularly on dermoscopy, but the distribution on the neck with submental sparing should help to distinguish it from EAI unless a heat source around the neck is identified while taking the patient’s history.7
In babies, a reticular generalized rash is most likely to be cutis marmorata (Figure 4), which is a physiologic response to cold exposure that resolves with rewarming of the skin. A more serious condition—cutis marmorata telangiectatica congenita (Figure 5)—usually is present at birth, most frequently involves a single extremity, and notably does not resolve with rewarming. This is an important differential for EAI in children because it can be associated with vascular and neurologic anomalies as well as limb asymmetry. Finally, port-wine stains can sometimes be reticular in appearance and can mimic the early erythematous stages of EAI. However, unlike the erythematous stage of EAI, the port-wine stains will be present at birth.7
Emerging in 2020, an important differential diagnosis to consider is a cutaneous manifestation of COVID-19 infection. An erythematous, reticular, chilblainlike or transient livedo reticularis–like rash has been described as a cutaneous manifestation of COVID-19. Although the pathophysiology is still being elucidated, it is suspected that this is caused by a major vaso-occlusive crisis secondary to COVID-19–induced thrombotic vasculopathy. Interestingly, the majority of patients with this COVID-related exanthem also displayed symptoms of COVID-19 (eg, fever, cough) at the time of presentation,57-60 but there also have been cases in patients who were asymptomatic or mildly symptomatic.60
In some cases, EAI is an indication to screen for an underlying disease. For example, uncontrolled pain is an opportunity to improve interventions such as modifying the patient’s pain-control regimen, placing a palliative care pain consultation, or checking if the patient has had age-appropriate screenings for malignancy. New focal pain in a patient with a prior diagnosis of cancer may be a sign of a new metastasis. A thermophilic patient leaves opportunity to assess for underlying medical causes such as thyroid abnormalities or social/psychological issues. Geriatric patients who are diagnosed with EAI may need to be assessed for dementia or home safety issues. Patients with a history of diabetes mellitus can unknowingly develop EAI on the lower extremities, which may signal a need to assess the patient for peripheral neuropathy. Patients with gastroparesis secondary to diabetes also may develop EAI on the abdomen secondary to heating pad use for discomfort. These examples are a reminder to consider possible secondary comorbidities in all diagnoses of EAI.7
Prognosis
Although the prognosis of EAI is excellent if caught early, failure to diagnose this condition can lead to permanent discoloration of the skin and even malignancy.6 A rare sequela includes squamous cell carcinoma, most commonly seen in chronic cases of the lower leg, which is likely related to chronic inflammation of the skin.61-65 Rare cases of poorly differentiated carcinoma,66 cutaneous marginal zone lymphoma,67 and Merkel cell carcinoma68 have been reported. Patients diagnosed with EAI should receive normal periodic surveillance of the skin based on their medical history, though the physician should have an increased suspicion and plan for biopsy of any nodules or ulcerations found on the skin of the affected area.7
Treatments
Once the diagnosis of EAI is made, treatment starts with removal of the heat source causing the rash. Because the rash usually is asymptomatic, further treatment typically is not required. The discoloration can resolve over months or years, but permanent hyperpigmentation is not uncommon. If hyperpigmentation persists despite removal of the heat source and the patient desires further treatment for discoloration, there are few treatment options, none of which are approved by the US Food and Drug Administration for this condition.7 There is some evidence for the use of Nd:YAG lasers to reduce hyperpigmentation in EAI.69 There have been some reports of treatment using topical hydroquinone and topical tretinoin in an attempt to lighten the skin. If associated hyperkeratosis or other epithelial atypia is present, the use of 5-fluorouracil may show some improvement.70 One case report has been published of successful treatment with systemic mesoglycan and topical bioflavonoids.71 It also is conceivable that medications used to treat postinflammatory hyperpigmentation may be helpful in this condition (eg, kojic acid, arbutin, mild topical steroids, azelaic acid). Patients with darker skin may experience permanent discoloration and may not be good candidates for alternative treatments such as laser therapy due to the risk for inducible hyperpigmentation.7
Conclusion
No matter the etiology, EAI usually is a benign skin condition that is treated by removal of the causative heat source. Once a diagnosis is made, the clinician must work with the patient to determine the etiology. Care must be taken to ensure that there are no underlying signs, such as chronic pain or psychiatric illness, that could point to associated conditions. Rarely, sequalae such as cancers have been documented in areas of chronic EAI. Once the heat source is identified and removed, any remaining hyperpigmentation usually will self-resolve over months to years, though this may take longer in patients with darker skin types. If more aggressive treatment is preferred by the patient, laser therapy, topical medications, and oral over-the-counter vitamins have been tried with minimal responses.
- Perry. Case of erythema ab igne. Br J Dermatol. 1900;xxiii:375.
- Bose S, Ortonee JP. Diseases affected by heat. In: Parish LC, Millikan LE, Amer M, et al. Global Dermatology Diagnosis and Management According to Geography, Climate, and Culture. Springer-Varlag; 1994:83-92.
- Leal-Lobato MM, Blasco-Morente G. Electric blanket induced erythema ab igne [in Spanish]. Semergen. 2015;41:456-457. doi:10.1016/j.semerg.2014.12.008
- Huynh N, Sarma D, Huerter C. Erythema ab igne: a case report and review of the literature. Cutis. 2011;88:290-292.
- Kesty K, Feldman SR. Erythema ab igne: evolving technology, evolving presentation. Dermatol Online J. 2014;20. doi:10.5070/D32011024689
- Miller K, Hunt R, Chu J, et al. Erythema ab igne. Dermatol Online J. 2011;17:28.
- Smith ML. Environmental and sports-related skin diseases. In: Bolognia JL, Schaffer JV, Cerroni L, eds. Dermatology. 4th ed. Elsevier; 2018:1569-1594.
- Errichetti E, Stinco G. Dermoscopy in general dermatology: a practical overview. Dermatol Ther (Heidelb). 2016;6:471-507. doi:10.1007/s13555-016-0141-6
- Guarneri C, Tchernev G, Wollina U, et al. Erythema ab igne caused by laptop computer. Open Access Maced J Med Sci. 2017;5:490-492. doi:10.3889/oamjms.2017.137
- Arnold AW, Itin PH. Laptop computer-induced erythema ab igne in a child and review of the literature. Pediatrics. 2010;126:E1227-E1230. doi:10.1542/peds.2010-1390
- Dickman J, Kessler S. Unilateral reticulated patch localized to the anterior thigh. JAAD Case Rep. 2018;4:746-748. doi:10.1016/j.jdcr.2018.06.007
- Boffa MJ. Laptop computer-induced erythema ab igne on the left breast. Cutis. 2011;87:175-176.
- Li K, Barankin B. Cutaneous manifestations of modern technology use. J Cutan Med Surg. 2011;15:347-353. doi:10.2310/7750.2011.10053
- Riahi RR, Cohen PR. Laptop-induced erythema ab igne: report and review of literature. Dermatol Online J. 2012;18:5.
- Andersen F. Laptop-thighs--laptop-induced erythema ab igne [in Danish]. Ugeskr Laeger. 2010;172:635.
- Jagtman BA. Erythema ab igne due to a laptop computer. Contact Dermatitis. 2004;50:105. doi:10.1111/j.0105-1873.2004.0295g.x
- Olechowska M, Kisiel K, Ruszkowska L, et al. Erythema ab igne (EAI) induced by a laptop computer: report of two cases. J Dtsch Dermatol Ges. doi:10.1111/j.1610-0387.2014.12387
- Nayak SUK, Shenoi SD, Prabhu S. Laptop induced erythema ab igne. Indian J Dermatol. 2012;57:131-132. doi:10.4103/0019-5154.94284
- Salvio AG, Nunes AJ, Angarita DPR. Laptop computer induced erythema ab igne: a new presentation of an old disease. An Bras Dermatol. 2016;91:79-80. doi:10.1590/abd1806-4841.20165139
- Schummer C, Tittelbach J, Elsner P. Right-sided laptop dermatitis [in German]. Dtsch Med Wochenschr. 2015;140:1376-1377. doi:10.1055/s-0041-103615
- Manoharan D. Erythema ab igne: usual site, unusual cause. J Pharm Bioallied Sci. 2015;7(suppl 1):S74-S75. doi:10.4103/0975-7406.155811
- Giraldi S, Diettrich F, Abbage KT, et al. Erythema ab igne induced by a laptop computer in an adolescent. An Bras Dermatol. 2011;86:128-130. doi:10.1590/S0365-05962011000100018
- Secher LLS, Vind-Kezunovic D, Zachariae COC. Side-effects to the use of laptop computers: erythema ab igne. Dermatol Reports. 2010;31:E11. doi:10.4081/dr.2010.e11
- Botten D, Langley RGB, Webb A. Academic branding: erythema ab igne and use of laptop computers. CMAJ. 2010;182:E857. doi:10.1503/cmaj.091868
- Bilic M, Adams BB. Erythema ab igne induced by a laptop computer. J Am Acad Dermatol. 2004;50:973-974. doi:10.1016/j.jaad.2003.08.007
- Fu LW, Vender R. Erythema ab igne caused by laptop computer gaming - a case report. Int J Dermatol. 2012;51:716-717. doi:10.1111/j.1365-4632.2011.05033.x
- Levinbook WS, Mallett J, Grant-Kels JM. Laptop computer-associated erythema ab igne. Cutis. 2007;80:319-320.
- Mohr MR, Scott KA, Pariser RM, et al. Laptop computer-induced erythema ab igne: a case report. Cutis. 2007;79:59-60.
- Cantor AS, Bartling SJ. Laptop computer-induced hyperpigmentation. Dermatol Online J. 2018;24:13030/qt6k37r9wm.
- Kaptanog˘lu AF, Mullaaziz D. Erythema ab igne in the palmar area induced by smart phone: case report. Turkiye Klin J Med Sci. 2015;35:284-286. doi:10.5336/medsci.2015-46976
- Redding KS, Watts AN, Lee J, et al. Space heater-induced bullous erythema ab igne. Cutis. 2017;100:E9-E10.
- Goorland J, Edens MA, Baudoin TD. An emergency department presentation of erythema ab igne caused by repeated heater exposure. J La State Med Soc. 2016;168:33-34.
- Kokturk A, Kaya TI, Baz K, et al. Bullous erythema ab igne. Dermatol Online J. 2003;9:18.
- Brzezinski P, Ismail S, Chiriac A. Radiator-induced erythema ab igne in 8-year-old girl. Rev Chil Pediatr. 2014;85:239-240. doi:10.4067/S0370-41062014000200015
- Adams BB. Heated car seat-induced erythema ab igne. Arch Dermatol. 2012;148:265-266. doi:10.1001/archdermatol.2011.2207
- Helm TN, Spigel GT, Helm KF. Erythema ab igne caused by a car heater. Cutis. 1997;59:81-82.
- Gregory JF, Beute TC. Erythema ab igne. J Spec Oper Med. 2013;13:115-119. doi:10.55460/5AVH-NZHY
- Chua S, Chen Q, Lee HY. Erythema ab igne and dermal scarring caused by cupping and moxibustion treatment. J Dtsch Dermatol Ges. 2015;13:337-338. doi:10.1111/ddg.12581
- Chen JF, Liu YC, Chen YF, et al. Erythema ab igne after footbath with Chinese herbal remedies. J Chinese Med Assoc. 2011;74:51-53. doi:10.1016/j.jcma.2011.01.009
- Baltazar D, Brockman R, Simpson E. Kotatsu-induced erythema ab igne. An Bras Dermatol. 2019;94:253-254. doi:10.1590/abd1806-4841.20198792
- Baig M, Byrne F. Erythema ab igne and its relation to spinal pathology. Cureus. 2018;10:e2914. doi:10.7759/cureus.2914
- Aria AB, Chen L, Silapunt S. Erythema ab igne from heating pad use: a report of three clinical cases and a differential diagnosis. Cureus. 2018;10:e2635. doi:10.7759/cureus.2635
- Milchak M, Smucker J, Chung CG, et al. Erythema ab igne due to heating pad use: a case report and review of clinical presentation, prevention, and complications. Case Rep Med. 2016;1862480. doi:10.1155/2016/1862480
- Gmuca S, Yu J, Weiss PF, et al. Erythema ab igne in an adolescent with chronic pain: an alarming cutaneous eruption from heat exposure. Pediatr Emerg Care. 2020;36:e236-e238. doi:10.1097/PEC.0000000000001460
- Dizdarevic A, Karim OA, Bygum A. A reddish brown reticulated hyperpigmented erythema on the abdomen of a girl. Erythema ab igne, also known as toasted skin syndrome, caused by a heating pad onthe abdomen. Acta Derm Venereol. 2014;94:365-367. doi:10.2340/00015555-1722
- Chatterjee S. Erythema ab igne from prolonged use of a heating pad. Mayo Clin Proc. 2005;80:1500. doi:10.4065/80.11.1500
- Waldorf DS, Rast MF, Garofalo VJ. Heating-pad erythematous dermatitis “erythema ab igne.” JAMA. 1971;218:1704. doi:10.1001/jama.1971.03190240056023
- South AM, Crispin MK, Marqueling AL, et al. A hyperpigmented reticular rash in a patient on peritoneal dialysis. Perit Dial Int. 2016;36:677-700. doi:10.3747/pdi.2016.00042
- Ravindran R. Erythema ab igne in an individual with diabetes and gastroparesis. BMJ Case Rep. 2017;2017:bcr2014203856. doi:10.1136/bcr-2014-203856
- Dessinioti C, Katsambas A, Tzavela E, et al. Erythema ab igne in three girls with anorexia nervosa. Pediatr Dermatol. 2016;33:e149-e150. doi:10.1111/pde.12770
- Fischer J, Rein K, Erfurt-Berge C, et al. Three cases of erythema ab igne (EAI) in patients with eating disorders. Neuropsychiatr. 2010;24:141-143.
- Docx MKF, Simons A, Ramet J, et al. Erythema ab igne in an adolescent with anorexia nervosa. Int J Eat Disord. 2013;46:381-383. doi:10.1002/eat.22075
- Turan E, Cimen V, Haytoglu NSK, et al. A case of bullous erythema ab igne accompanied by anemia and subclinical hypothyroidism. Dermatol Online J. 2014;20:223366.
- Pavithran K. Erythema ab igne, schizophrenia and thermophilia. Indian J Dermatol Venereol Leprol. 1987;53:181-182.
- Dellavelle R, Gillum P. Erythema ab igne following heating/cooling blanket use in the intensive care unit. Cutis. 2000;66:136-138.
- Park SY, Kim SM, Yoon TJ. Erythema ab igne caused by weight loss heating pad. Korean J Dermatol. 2007;45:489-491.
- Sachdeva M, Gianotti R, Shah M, et al. Cutaneous manifestations of COVID-19: report of three cases and a review of literature. J Dermatol Sci. 2020;98:75-81. doi:10.1016/j.jdermsci.2020.04.011
- Gisondi P, Plaserico S, Bordin C, et al. Cutaneous manifestations of SARS‐CoV‐2 infection: a clinical update. J Eur Acad Dermatol Venereol. 2020;34:2499-2504. doi:10.1111/jdv.16774
- Manalo IF, Smith MK, Cheeley J, et al. A dermatologic manifestation of COVID-19: transient livedo reticularis. J Am Acad Dermatol. 2020;83:700. doi:10.1016/j.jaad.2020.04.018
- Zhao Q, Fang X, Pang Z, et al. COVID‐19 and cutaneous manifestations: a systematic review. J Eur Acad Dermatol Venereol. 2020;34:2505-2510. doi:10.1111/jdv.16778
- Akasaka T, Kon S. Two cases of squamous cell carcinoma arising from erythema ab igne. Nihon Hifuka Gakkai Zasshi. 1989;99:735-742.
- Arrington JH 3rd, Lockman DS. Thermal keratoses and squamous cell carcinoma in situ associated with erythema ab igne. Arch Dermatol. 1979;115:1226-1228.
- Wharton JB, Sheehan DJ, Lesher JL Jr. Squamous cell carcinoma in situ arising in the setting of erythema ab igne. J Drugs Dermatol. 2008;7:488-489.
- Wollina U, Helm C, Hansel G, et al. Two cases of erythema ab igne, one with a squamous cell carcinoma. G Ital Dermatol Venereol. 2007;142:415-418.
- Rudolph CM, Soyer HP, Wolf P, et al. Squamous cell carcinoma arising in erythema ab igne. Hautarzt. 2000;51:260-263. doi:10.1007/s001050051115
- Sigmon JR, Cantrell J, Teague D, et al. Poorly differentiated carcinoma arising in the setting of erythema ab igne. Am J Dermatopathol. 2013;35:676-678. doi:10.1097/DAD.0b013e3182871648
- Wharton J, Roffwarg D, Miller J, et al. Cutaneous marginal zone lymphoma arising in the setting of erythema ab igne. J Am Acad Dermatol. 2010;62:1080-1081. doi:10.1016/j.jaad.2009.08.005
- Jones CS, Tyring SK, Lee PC, et al. Development of neuroendocrine (Merkel cell) carcinoma mixed with squamous cell carcinoma in erythema ab igne. Arch Dermatol. 1988;124:110-113.
- Kim HW, Kim EJ, Park HC, et al. Erythema ab igne successfully treated with low fluenced 1,064-nm Q-switched neodymium-doped yttrium aluminum garnet laser. J Cosmet Laser Ther. 2014;16:147-148. doi:10.3109/14764172.2013.854623
- Tan S, Bertucci V. Erythema ab igne: an old condition new again. CMAJ. 2000;62:77-78.
- Gianfaldoni S, Gianfaldoni R, Tchernev G, et al. Erythema ab igne successfully treated with mesoglycan and bioflavonoids: a case-report. Open Access Maced J Med Sci. 2017;5:432-435. doi:10.3889/oamjms.2017.123
Erythema ab igne (EAI)(also known as toasted skin syndrome) was first described in the British Journal of Dermatology in the 20th century, 1 though it was known by physicians long before. Reticular netlike skin changes were seen in association with patients who spent extended time directly next to a heat source. This association led to the name of this condition, which literally means “redness by fire.” Indeed, EAI induced by chronic heat exposure has been described across the world for centuries. For example, in the cold regions of northern China, people used to sleep on beds of hot bricks called kang to stay warm at night. The people of India’s Kashmir district carried pots of hot coals called kangri next to the skin under large woven shawls to stay warm. In the past, Irish women often spent much time by a turf- or peat-burning fire. Chronic heat exposure in these cases can lead not only to EAI but also to aggressive types of cancer, often with a latency of 30 years or more. 2
More recently, the invention of home central heating led to a stark decrease in the number of cases associated with combustion-based heat, with a transition to etiologies such as use of hot water bottles, electric blankets, and electric space heaters. Over time, technological advances led to ever-increasing potential causes for EAI, such as laptops or cell phones, car heaters and heated seats, heated blankets,3,4 infrared lamps for food, and even medical devices such as ultrasound-based heating products and convective temperature management systems for hospitalized patients. As technology evolves, so do the potential causes of EAI, requiring clinicians to diagnose and deduce the cause through a thorough social and medical history as well as a workup on the present illness with considerations for the anatomical location.5-7 Herein, we describe the etiology of EAI, diagnosis, and treatment options.
Clinical Characteristics
Erythema ab igne begins as mild, transient, and erythematous macules and patches in a reticular pattern that resolve minutes to hours after removal of the heat source. With weeks to months of continued or repeated application of the heat source, the affected area eventually becomes hyperpigmented where there once was erythema (Figures 1 and 2). Sometimes papules, bullae, telangiectasia, and hyperkeratosis also form. The rash usually is asymptomatic, though pain, pruritus, and dysesthesia have been reported.7 Dermoscopy of EAI in the hyperpigmented stage can reveal diffuse superficial dark pigmentation, telangiectasia, and mild whitish scaling.8 Although the pathogenesis has remained elusive over the years, lesions do seem to be mostly associated with cumulative exposure to heat rather than length of exposure.7
Etiology of EAI
Anatomic Location—The affected site depends on the source of heat (Table). Classic examples of this condition include a patient with EAI presenting on the anterior thighs after working in front of a hot oven or a patient with chronic back pain presenting with lower-back EAI secondary to frequent use of a hot water bottle or heating pad.7 With evolving technology over the last few decades, new etiologies have become more common—teenagers are presenting with anterior thigh EAI secondary to frequent laptop use2-29; patients are holding warm cell phones in their pant pockets, leading to unilateral geometric EAI on the anterior thigh (front pocket) or buttock (back pocket)30; plug-in radiators under computer desks are causing EAI on the lower legs31-34; and automobile seat heaters have been shown to cause EAI on the posterior legs.5,35-37 Clinicians should consider anatomic location a critical clue for etiology.
Social History—There are rarer and more highly specific causes of EAI than simple heat exposure that can be parsed from a patient’s social history. Occupational exposure has been documented, such as bakers with exposure to ovens, foundry workers with exposure to heated metals, or fast-food workers with chronic exposure to infrared food lamps.6,7 There also are cultural practices that can cause EAI. For example, the practice of cupping with moxibustion was shown to create a specific pattern in the shape of the cultural tool used.38 When footbaths with Chinese herbal remedies are performed frequently with high heat, they can lead to EAI on the feet with a linear border at the ankles. There also have been reports of kotatsu (heated tables in Japan) leading to lower-body EAI.39,40 These cultural practices also are more common in patients with darker skin types, which can lead to hyperpigmentation that is difficult to treat, making early diagnosis important.7
Medical History—Case reports have shown EAI caused by patients attempting to use heat-based methods for pain relief of an underlying serious disease such as cancer, bowel pathology (abdominal EAI), spinal disc prolapse (midline back EAI),41 sickle cell anemia, and renal pathology (posterior upper flank EAI).6,7,40-49 Patients with hypothyroidism or anorexia have been noted to have generalized EAI sparing the face secondary to repeated and extended hot baths or showers.50-53 One patient with schizophrenia was shown to have associated thermophilia due to a delusion that led the patient to soak in hot baths for long periods of time, leading to EAI.54 Finally, all physicians should be aware of iatrogenic causes of EAI, such as use of warming devices, ultrasound-based warming techniques, and laser therapy for lipolysis. Inquire about the patient’s surgical history or intensive care unit stays as well as alternative medicine or chiropractic visits. Obtaining a history of medical procedures can be enlightening when an etiology is not immediately clear.7,55,56
Diagnosis
Erythema ab igne is a clinical diagnosis based on recognizable cutaneous findings and a clear history of moderate heat exposure. However, when a clinical diagnosis of EAI is not certain (eg, when unable to obtain a clear history from the patient) or when malignant transformation is suspected, a biopsy can be performed. Pathologically, hematoxylin and eosin staining of EAI classically reveals dilated small vascular channels in the superficial dermis, hence a clinically reticular rash; interface dermatitis clinically manifesting as erythema; and pigment incontinence with melanin-laden macrophages consistent with clinical hyperpigmentation. Finally, for unclear reasons, increased numbers of elastic fibers classically are seen in biopsies of EAI.7
Differential Diagnosis
The differential diagnosis for a reticular patch includes livedo reticularis (Figure 3), which usually manifests as a more generalized rash in patients with chronic disease or coagulopathy such as systemic lupus erythematosus, cryoglobulinemia, or Raynaud phenomenon. When differentiating EAI from livedo reticularis or cutis marmorata, consider that both alternative diagnoses are more vascular appearing and are associated with cold exposure rather than heat exposure. In cases that are less reticular, livedo racemosa can be considered in the differential diagnosis. Finally, poikiloderma of Civatte can be reticular, particularly on dermoscopy, but the distribution on the neck with submental sparing should help to distinguish it from EAI unless a heat source around the neck is identified while taking the patient’s history.7
In babies, a reticular generalized rash is most likely to be cutis marmorata (Figure 4), which is a physiologic response to cold exposure that resolves with rewarming of the skin. A more serious condition—cutis marmorata telangiectatica congenita (Figure 5)—usually is present at birth, most frequently involves a single extremity, and notably does not resolve with rewarming. This is an important differential for EAI in children because it can be associated with vascular and neurologic anomalies as well as limb asymmetry. Finally, port-wine stains can sometimes be reticular in appearance and can mimic the early erythematous stages of EAI. However, unlike the erythematous stage of EAI, the port-wine stains will be present at birth.7
Emerging in 2020, an important differential diagnosis to consider is a cutaneous manifestation of COVID-19 infection. An erythematous, reticular, chilblainlike or transient livedo reticularis–like rash has been described as a cutaneous manifestation of COVID-19. Although the pathophysiology is still being elucidated, it is suspected that this is caused by a major vaso-occlusive crisis secondary to COVID-19–induced thrombotic vasculopathy. Interestingly, the majority of patients with this COVID-related exanthem also displayed symptoms of COVID-19 (eg, fever, cough) at the time of presentation,57-60 but there also have been cases in patients who were asymptomatic or mildly symptomatic.60
In some cases, EAI is an indication to screen for an underlying disease. For example, uncontrolled pain is an opportunity to improve interventions such as modifying the patient’s pain-control regimen, placing a palliative care pain consultation, or checking if the patient has had age-appropriate screenings for malignancy. New focal pain in a patient with a prior diagnosis of cancer may be a sign of a new metastasis. A thermophilic patient leaves opportunity to assess for underlying medical causes such as thyroid abnormalities or social/psychological issues. Geriatric patients who are diagnosed with EAI may need to be assessed for dementia or home safety issues. Patients with a history of diabetes mellitus can unknowingly develop EAI on the lower extremities, which may signal a need to assess the patient for peripheral neuropathy. Patients with gastroparesis secondary to diabetes also may develop EAI on the abdomen secondary to heating pad use for discomfort. These examples are a reminder to consider possible secondary comorbidities in all diagnoses of EAI.7
Prognosis
Although the prognosis of EAI is excellent if caught early, failure to diagnose this condition can lead to permanent discoloration of the skin and even malignancy.6 A rare sequela includes squamous cell carcinoma, most commonly seen in chronic cases of the lower leg, which is likely related to chronic inflammation of the skin.61-65 Rare cases of poorly differentiated carcinoma,66 cutaneous marginal zone lymphoma,67 and Merkel cell carcinoma68 have been reported. Patients diagnosed with EAI should receive normal periodic surveillance of the skin based on their medical history, though the physician should have an increased suspicion and plan for biopsy of any nodules or ulcerations found on the skin of the affected area.7
Treatments
Once the diagnosis of EAI is made, treatment starts with removal of the heat source causing the rash. Because the rash usually is asymptomatic, further treatment typically is not required. The discoloration can resolve over months or years, but permanent hyperpigmentation is not uncommon. If hyperpigmentation persists despite removal of the heat source and the patient desires further treatment for discoloration, there are few treatment options, none of which are approved by the US Food and Drug Administration for this condition.7 There is some evidence for the use of Nd:YAG lasers to reduce hyperpigmentation in EAI.69 There have been some reports of treatment using topical hydroquinone and topical tretinoin in an attempt to lighten the skin. If associated hyperkeratosis or other epithelial atypia is present, the use of 5-fluorouracil may show some improvement.70 One case report has been published of successful treatment with systemic mesoglycan and topical bioflavonoids.71 It also is conceivable that medications used to treat postinflammatory hyperpigmentation may be helpful in this condition (eg, kojic acid, arbutin, mild topical steroids, azelaic acid). Patients with darker skin may experience permanent discoloration and may not be good candidates for alternative treatments such as laser therapy due to the risk for inducible hyperpigmentation.7
Conclusion
No matter the etiology, EAI usually is a benign skin condition that is treated by removal of the causative heat source. Once a diagnosis is made, the clinician must work with the patient to determine the etiology. Care must be taken to ensure that there are no underlying signs, such as chronic pain or psychiatric illness, that could point to associated conditions. Rarely, sequalae such as cancers have been documented in areas of chronic EAI. Once the heat source is identified and removed, any remaining hyperpigmentation usually will self-resolve over months to years, though this may take longer in patients with darker skin types. If more aggressive treatment is preferred by the patient, laser therapy, topical medications, and oral over-the-counter vitamins have been tried with minimal responses.
Erythema ab igne (EAI)(also known as toasted skin syndrome) was first described in the British Journal of Dermatology in the 20th century, 1 though it was known by physicians long before. Reticular netlike skin changes were seen in association with patients who spent extended time directly next to a heat source. This association led to the name of this condition, which literally means “redness by fire.” Indeed, EAI induced by chronic heat exposure has been described across the world for centuries. For example, in the cold regions of northern China, people used to sleep on beds of hot bricks called kang to stay warm at night. The people of India’s Kashmir district carried pots of hot coals called kangri next to the skin under large woven shawls to stay warm. In the past, Irish women often spent much time by a turf- or peat-burning fire. Chronic heat exposure in these cases can lead not only to EAI but also to aggressive types of cancer, often with a latency of 30 years or more. 2
More recently, the invention of home central heating led to a stark decrease in the number of cases associated with combustion-based heat, with a transition to etiologies such as use of hot water bottles, electric blankets, and electric space heaters. Over time, technological advances led to ever-increasing potential causes for EAI, such as laptops or cell phones, car heaters and heated seats, heated blankets,3,4 infrared lamps for food, and even medical devices such as ultrasound-based heating products and convective temperature management systems for hospitalized patients. As technology evolves, so do the potential causes of EAI, requiring clinicians to diagnose and deduce the cause through a thorough social and medical history as well as a workup on the present illness with considerations for the anatomical location.5-7 Herein, we describe the etiology of EAI, diagnosis, and treatment options.
Clinical Characteristics
Erythema ab igne begins as mild, transient, and erythematous macules and patches in a reticular pattern that resolve minutes to hours after removal of the heat source. With weeks to months of continued or repeated application of the heat source, the affected area eventually becomes hyperpigmented where there once was erythema (Figures 1 and 2). Sometimes papules, bullae, telangiectasia, and hyperkeratosis also form. The rash usually is asymptomatic, though pain, pruritus, and dysesthesia have been reported.7 Dermoscopy of EAI in the hyperpigmented stage can reveal diffuse superficial dark pigmentation, telangiectasia, and mild whitish scaling.8 Although the pathogenesis has remained elusive over the years, lesions do seem to be mostly associated with cumulative exposure to heat rather than length of exposure.7
Etiology of EAI
Anatomic Location—The affected site depends on the source of heat (Table). Classic examples of this condition include a patient with EAI presenting on the anterior thighs after working in front of a hot oven or a patient with chronic back pain presenting with lower-back EAI secondary to frequent use of a hot water bottle or heating pad.7 With evolving technology over the last few decades, new etiologies have become more common—teenagers are presenting with anterior thigh EAI secondary to frequent laptop use2-29; patients are holding warm cell phones in their pant pockets, leading to unilateral geometric EAI on the anterior thigh (front pocket) or buttock (back pocket)30; plug-in radiators under computer desks are causing EAI on the lower legs31-34; and automobile seat heaters have been shown to cause EAI on the posterior legs.5,35-37 Clinicians should consider anatomic location a critical clue for etiology.
Social History—There are rarer and more highly specific causes of EAI than simple heat exposure that can be parsed from a patient’s social history. Occupational exposure has been documented, such as bakers with exposure to ovens, foundry workers with exposure to heated metals, or fast-food workers with chronic exposure to infrared food lamps.6,7 There also are cultural practices that can cause EAI. For example, the practice of cupping with moxibustion was shown to create a specific pattern in the shape of the cultural tool used.38 When footbaths with Chinese herbal remedies are performed frequently with high heat, they can lead to EAI on the feet with a linear border at the ankles. There also have been reports of kotatsu (heated tables in Japan) leading to lower-body EAI.39,40 These cultural practices also are more common in patients with darker skin types, which can lead to hyperpigmentation that is difficult to treat, making early diagnosis important.7
Medical History—Case reports have shown EAI caused by patients attempting to use heat-based methods for pain relief of an underlying serious disease such as cancer, bowel pathology (abdominal EAI), spinal disc prolapse (midline back EAI),41 sickle cell anemia, and renal pathology (posterior upper flank EAI).6,7,40-49 Patients with hypothyroidism or anorexia have been noted to have generalized EAI sparing the face secondary to repeated and extended hot baths or showers.50-53 One patient with schizophrenia was shown to have associated thermophilia due to a delusion that led the patient to soak in hot baths for long periods of time, leading to EAI.54 Finally, all physicians should be aware of iatrogenic causes of EAI, such as use of warming devices, ultrasound-based warming techniques, and laser therapy for lipolysis. Inquire about the patient’s surgical history or intensive care unit stays as well as alternative medicine or chiropractic visits. Obtaining a history of medical procedures can be enlightening when an etiology is not immediately clear.7,55,56
Diagnosis
Erythema ab igne is a clinical diagnosis based on recognizable cutaneous findings and a clear history of moderate heat exposure. However, when a clinical diagnosis of EAI is not certain (eg, when unable to obtain a clear history from the patient) or when malignant transformation is suspected, a biopsy can be performed. Pathologically, hematoxylin and eosin staining of EAI classically reveals dilated small vascular channels in the superficial dermis, hence a clinically reticular rash; interface dermatitis clinically manifesting as erythema; and pigment incontinence with melanin-laden macrophages consistent with clinical hyperpigmentation. Finally, for unclear reasons, increased numbers of elastic fibers classically are seen in biopsies of EAI.7
Differential Diagnosis
The differential diagnosis for a reticular patch includes livedo reticularis (Figure 3), which usually manifests as a more generalized rash in patients with chronic disease or coagulopathy such as systemic lupus erythematosus, cryoglobulinemia, or Raynaud phenomenon. When differentiating EAI from livedo reticularis or cutis marmorata, consider that both alternative diagnoses are more vascular appearing and are associated with cold exposure rather than heat exposure. In cases that are less reticular, livedo racemosa can be considered in the differential diagnosis. Finally, poikiloderma of Civatte can be reticular, particularly on dermoscopy, but the distribution on the neck with submental sparing should help to distinguish it from EAI unless a heat source around the neck is identified while taking the patient’s history.7
In babies, a reticular generalized rash is most likely to be cutis marmorata (Figure 4), which is a physiologic response to cold exposure that resolves with rewarming of the skin. A more serious condition—cutis marmorata telangiectatica congenita (Figure 5)—usually is present at birth, most frequently involves a single extremity, and notably does not resolve with rewarming. This is an important differential for EAI in children because it can be associated with vascular and neurologic anomalies as well as limb asymmetry. Finally, port-wine stains can sometimes be reticular in appearance and can mimic the early erythematous stages of EAI. However, unlike the erythematous stage of EAI, the port-wine stains will be present at birth.7
Emerging in 2020, an important differential diagnosis to consider is a cutaneous manifestation of COVID-19 infection. An erythematous, reticular, chilblainlike or transient livedo reticularis–like rash has been described as a cutaneous manifestation of COVID-19. Although the pathophysiology is still being elucidated, it is suspected that this is caused by a major vaso-occlusive crisis secondary to COVID-19–induced thrombotic vasculopathy. Interestingly, the majority of patients with this COVID-related exanthem also displayed symptoms of COVID-19 (eg, fever, cough) at the time of presentation,57-60 but there also have been cases in patients who were asymptomatic or mildly symptomatic.60
In some cases, EAI is an indication to screen for an underlying disease. For example, uncontrolled pain is an opportunity to improve interventions such as modifying the patient’s pain-control regimen, placing a palliative care pain consultation, or checking if the patient has had age-appropriate screenings for malignancy. New focal pain in a patient with a prior diagnosis of cancer may be a sign of a new metastasis. A thermophilic patient leaves opportunity to assess for underlying medical causes such as thyroid abnormalities or social/psychological issues. Geriatric patients who are diagnosed with EAI may need to be assessed for dementia or home safety issues. Patients with a history of diabetes mellitus can unknowingly develop EAI on the lower extremities, which may signal a need to assess the patient for peripheral neuropathy. Patients with gastroparesis secondary to diabetes also may develop EAI on the abdomen secondary to heating pad use for discomfort. These examples are a reminder to consider possible secondary comorbidities in all diagnoses of EAI.7
Prognosis
Although the prognosis of EAI is excellent if caught early, failure to diagnose this condition can lead to permanent discoloration of the skin and even malignancy.6 A rare sequela includes squamous cell carcinoma, most commonly seen in chronic cases of the lower leg, which is likely related to chronic inflammation of the skin.61-65 Rare cases of poorly differentiated carcinoma,66 cutaneous marginal zone lymphoma,67 and Merkel cell carcinoma68 have been reported. Patients diagnosed with EAI should receive normal periodic surveillance of the skin based on their medical history, though the physician should have an increased suspicion and plan for biopsy of any nodules or ulcerations found on the skin of the affected area.7
Treatments
Once the diagnosis of EAI is made, treatment starts with removal of the heat source causing the rash. Because the rash usually is asymptomatic, further treatment typically is not required. The discoloration can resolve over months or years, but permanent hyperpigmentation is not uncommon. If hyperpigmentation persists despite removal of the heat source and the patient desires further treatment for discoloration, there are few treatment options, none of which are approved by the US Food and Drug Administration for this condition.7 There is some evidence for the use of Nd:YAG lasers to reduce hyperpigmentation in EAI.69 There have been some reports of treatment using topical hydroquinone and topical tretinoin in an attempt to lighten the skin. If associated hyperkeratosis or other epithelial atypia is present, the use of 5-fluorouracil may show some improvement.70 One case report has been published of successful treatment with systemic mesoglycan and topical bioflavonoids.71 It also is conceivable that medications used to treat postinflammatory hyperpigmentation may be helpful in this condition (eg, kojic acid, arbutin, mild topical steroids, azelaic acid). Patients with darker skin may experience permanent discoloration and may not be good candidates for alternative treatments such as laser therapy due to the risk for inducible hyperpigmentation.7
Conclusion
No matter the etiology, EAI usually is a benign skin condition that is treated by removal of the causative heat source. Once a diagnosis is made, the clinician must work with the patient to determine the etiology. Care must be taken to ensure that there are no underlying signs, such as chronic pain or psychiatric illness, that could point to associated conditions. Rarely, sequalae such as cancers have been documented in areas of chronic EAI. Once the heat source is identified and removed, any remaining hyperpigmentation usually will self-resolve over months to years, though this may take longer in patients with darker skin types. If more aggressive treatment is preferred by the patient, laser therapy, topical medications, and oral over-the-counter vitamins have been tried with minimal responses.
- Perry. Case of erythema ab igne. Br J Dermatol. 1900;xxiii:375.
- Bose S, Ortonee JP. Diseases affected by heat. In: Parish LC, Millikan LE, Amer M, et al. Global Dermatology Diagnosis and Management According to Geography, Climate, and Culture. Springer-Varlag; 1994:83-92.
- Leal-Lobato MM, Blasco-Morente G. Electric blanket induced erythema ab igne [in Spanish]. Semergen. 2015;41:456-457. doi:10.1016/j.semerg.2014.12.008
- Huynh N, Sarma D, Huerter C. Erythema ab igne: a case report and review of the literature. Cutis. 2011;88:290-292.
- Kesty K, Feldman SR. Erythema ab igne: evolving technology, evolving presentation. Dermatol Online J. 2014;20. doi:10.5070/D32011024689
- Miller K, Hunt R, Chu J, et al. Erythema ab igne. Dermatol Online J. 2011;17:28.
- Smith ML. Environmental and sports-related skin diseases. In: Bolognia JL, Schaffer JV, Cerroni L, eds. Dermatology. 4th ed. Elsevier; 2018:1569-1594.
- Errichetti E, Stinco G. Dermoscopy in general dermatology: a practical overview. Dermatol Ther (Heidelb). 2016;6:471-507. doi:10.1007/s13555-016-0141-6
- Guarneri C, Tchernev G, Wollina U, et al. Erythema ab igne caused by laptop computer. Open Access Maced J Med Sci. 2017;5:490-492. doi:10.3889/oamjms.2017.137
- Arnold AW, Itin PH. Laptop computer-induced erythema ab igne in a child and review of the literature. Pediatrics. 2010;126:E1227-E1230. doi:10.1542/peds.2010-1390
- Dickman J, Kessler S. Unilateral reticulated patch localized to the anterior thigh. JAAD Case Rep. 2018;4:746-748. doi:10.1016/j.jdcr.2018.06.007
- Boffa MJ. Laptop computer-induced erythema ab igne on the left breast. Cutis. 2011;87:175-176.
- Li K, Barankin B. Cutaneous manifestations of modern technology use. J Cutan Med Surg. 2011;15:347-353. doi:10.2310/7750.2011.10053
- Riahi RR, Cohen PR. Laptop-induced erythema ab igne: report and review of literature. Dermatol Online J. 2012;18:5.
- Andersen F. Laptop-thighs--laptop-induced erythema ab igne [in Danish]. Ugeskr Laeger. 2010;172:635.
- Jagtman BA. Erythema ab igne due to a laptop computer. Contact Dermatitis. 2004;50:105. doi:10.1111/j.0105-1873.2004.0295g.x
- Olechowska M, Kisiel K, Ruszkowska L, et al. Erythema ab igne (EAI) induced by a laptop computer: report of two cases. J Dtsch Dermatol Ges. doi:10.1111/j.1610-0387.2014.12387
- Nayak SUK, Shenoi SD, Prabhu S. Laptop induced erythema ab igne. Indian J Dermatol. 2012;57:131-132. doi:10.4103/0019-5154.94284
- Salvio AG, Nunes AJ, Angarita DPR. Laptop computer induced erythema ab igne: a new presentation of an old disease. An Bras Dermatol. 2016;91:79-80. doi:10.1590/abd1806-4841.20165139
- Schummer C, Tittelbach J, Elsner P. Right-sided laptop dermatitis [in German]. Dtsch Med Wochenschr. 2015;140:1376-1377. doi:10.1055/s-0041-103615
- Manoharan D. Erythema ab igne: usual site, unusual cause. J Pharm Bioallied Sci. 2015;7(suppl 1):S74-S75. doi:10.4103/0975-7406.155811
- Giraldi S, Diettrich F, Abbage KT, et al. Erythema ab igne induced by a laptop computer in an adolescent. An Bras Dermatol. 2011;86:128-130. doi:10.1590/S0365-05962011000100018
- Secher LLS, Vind-Kezunovic D, Zachariae COC. Side-effects to the use of laptop computers: erythema ab igne. Dermatol Reports. 2010;31:E11. doi:10.4081/dr.2010.e11
- Botten D, Langley RGB, Webb A. Academic branding: erythema ab igne and use of laptop computers. CMAJ. 2010;182:E857. doi:10.1503/cmaj.091868
- Bilic M, Adams BB. Erythema ab igne induced by a laptop computer. J Am Acad Dermatol. 2004;50:973-974. doi:10.1016/j.jaad.2003.08.007
- Fu LW, Vender R. Erythema ab igne caused by laptop computer gaming - a case report. Int J Dermatol. 2012;51:716-717. doi:10.1111/j.1365-4632.2011.05033.x
- Levinbook WS, Mallett J, Grant-Kels JM. Laptop computer-associated erythema ab igne. Cutis. 2007;80:319-320.
- Mohr MR, Scott KA, Pariser RM, et al. Laptop computer-induced erythema ab igne: a case report. Cutis. 2007;79:59-60.
- Cantor AS, Bartling SJ. Laptop computer-induced hyperpigmentation. Dermatol Online J. 2018;24:13030/qt6k37r9wm.
- Kaptanog˘lu AF, Mullaaziz D. Erythema ab igne in the palmar area induced by smart phone: case report. Turkiye Klin J Med Sci. 2015;35:284-286. doi:10.5336/medsci.2015-46976
- Redding KS, Watts AN, Lee J, et al. Space heater-induced bullous erythema ab igne. Cutis. 2017;100:E9-E10.
- Goorland J, Edens MA, Baudoin TD. An emergency department presentation of erythema ab igne caused by repeated heater exposure. J La State Med Soc. 2016;168:33-34.
- Kokturk A, Kaya TI, Baz K, et al. Bullous erythema ab igne. Dermatol Online J. 2003;9:18.
- Brzezinski P, Ismail S, Chiriac A. Radiator-induced erythema ab igne in 8-year-old girl. Rev Chil Pediatr. 2014;85:239-240. doi:10.4067/S0370-41062014000200015
- Adams BB. Heated car seat-induced erythema ab igne. Arch Dermatol. 2012;148:265-266. doi:10.1001/archdermatol.2011.2207
- Helm TN, Spigel GT, Helm KF. Erythema ab igne caused by a car heater. Cutis. 1997;59:81-82.
- Gregory JF, Beute TC. Erythema ab igne. J Spec Oper Med. 2013;13:115-119. doi:10.55460/5AVH-NZHY
- Chua S, Chen Q, Lee HY. Erythema ab igne and dermal scarring caused by cupping and moxibustion treatment. J Dtsch Dermatol Ges. 2015;13:337-338. doi:10.1111/ddg.12581
- Chen JF, Liu YC, Chen YF, et al. Erythema ab igne after footbath with Chinese herbal remedies. J Chinese Med Assoc. 2011;74:51-53. doi:10.1016/j.jcma.2011.01.009
- Baltazar D, Brockman R, Simpson E. Kotatsu-induced erythema ab igne. An Bras Dermatol. 2019;94:253-254. doi:10.1590/abd1806-4841.20198792
- Baig M, Byrne F. Erythema ab igne and its relation to spinal pathology. Cureus. 2018;10:e2914. doi:10.7759/cureus.2914
- Aria AB, Chen L, Silapunt S. Erythema ab igne from heating pad use: a report of three clinical cases and a differential diagnosis. Cureus. 2018;10:e2635. doi:10.7759/cureus.2635
- Milchak M, Smucker J, Chung CG, et al. Erythema ab igne due to heating pad use: a case report and review of clinical presentation, prevention, and complications. Case Rep Med. 2016;1862480. doi:10.1155/2016/1862480
- Gmuca S, Yu J, Weiss PF, et al. Erythema ab igne in an adolescent with chronic pain: an alarming cutaneous eruption from heat exposure. Pediatr Emerg Care. 2020;36:e236-e238. doi:10.1097/PEC.0000000000001460
- Dizdarevic A, Karim OA, Bygum A. A reddish brown reticulated hyperpigmented erythema on the abdomen of a girl. Erythema ab igne, also known as toasted skin syndrome, caused by a heating pad onthe abdomen. Acta Derm Venereol. 2014;94:365-367. doi:10.2340/00015555-1722
- Chatterjee S. Erythema ab igne from prolonged use of a heating pad. Mayo Clin Proc. 2005;80:1500. doi:10.4065/80.11.1500
- Waldorf DS, Rast MF, Garofalo VJ. Heating-pad erythematous dermatitis “erythema ab igne.” JAMA. 1971;218:1704. doi:10.1001/jama.1971.03190240056023
- South AM, Crispin MK, Marqueling AL, et al. A hyperpigmented reticular rash in a patient on peritoneal dialysis. Perit Dial Int. 2016;36:677-700. doi:10.3747/pdi.2016.00042
- Ravindran R. Erythema ab igne in an individual with diabetes and gastroparesis. BMJ Case Rep. 2017;2017:bcr2014203856. doi:10.1136/bcr-2014-203856
- Dessinioti C, Katsambas A, Tzavela E, et al. Erythema ab igne in three girls with anorexia nervosa. Pediatr Dermatol. 2016;33:e149-e150. doi:10.1111/pde.12770
- Fischer J, Rein K, Erfurt-Berge C, et al. Three cases of erythema ab igne (EAI) in patients with eating disorders. Neuropsychiatr. 2010;24:141-143.
- Docx MKF, Simons A, Ramet J, et al. Erythema ab igne in an adolescent with anorexia nervosa. Int J Eat Disord. 2013;46:381-383. doi:10.1002/eat.22075
- Turan E, Cimen V, Haytoglu NSK, et al. A case of bullous erythema ab igne accompanied by anemia and subclinical hypothyroidism. Dermatol Online J. 2014;20:223366.
- Pavithran K. Erythema ab igne, schizophrenia and thermophilia. Indian J Dermatol Venereol Leprol. 1987;53:181-182.
- Dellavelle R, Gillum P. Erythema ab igne following heating/cooling blanket use in the intensive care unit. Cutis. 2000;66:136-138.
- Park SY, Kim SM, Yoon TJ. Erythema ab igne caused by weight loss heating pad. Korean J Dermatol. 2007;45:489-491.
- Sachdeva M, Gianotti R, Shah M, et al. Cutaneous manifestations of COVID-19: report of three cases and a review of literature. J Dermatol Sci. 2020;98:75-81. doi:10.1016/j.jdermsci.2020.04.011
- Gisondi P, Plaserico S, Bordin C, et al. Cutaneous manifestations of SARS‐CoV‐2 infection: a clinical update. J Eur Acad Dermatol Venereol. 2020;34:2499-2504. doi:10.1111/jdv.16774
- Manalo IF, Smith MK, Cheeley J, et al. A dermatologic manifestation of COVID-19: transient livedo reticularis. J Am Acad Dermatol. 2020;83:700. doi:10.1016/j.jaad.2020.04.018
- Zhao Q, Fang X, Pang Z, et al. COVID‐19 and cutaneous manifestations: a systematic review. J Eur Acad Dermatol Venereol. 2020;34:2505-2510. doi:10.1111/jdv.16778
- Akasaka T, Kon S. Two cases of squamous cell carcinoma arising from erythema ab igne. Nihon Hifuka Gakkai Zasshi. 1989;99:735-742.
- Arrington JH 3rd, Lockman DS. Thermal keratoses and squamous cell carcinoma in situ associated with erythema ab igne. Arch Dermatol. 1979;115:1226-1228.
- Wharton JB, Sheehan DJ, Lesher JL Jr. Squamous cell carcinoma in situ arising in the setting of erythema ab igne. J Drugs Dermatol. 2008;7:488-489.
- Wollina U, Helm C, Hansel G, et al. Two cases of erythema ab igne, one with a squamous cell carcinoma. G Ital Dermatol Venereol. 2007;142:415-418.
- Rudolph CM, Soyer HP, Wolf P, et al. Squamous cell carcinoma arising in erythema ab igne. Hautarzt. 2000;51:260-263. doi:10.1007/s001050051115
- Sigmon JR, Cantrell J, Teague D, et al. Poorly differentiated carcinoma arising in the setting of erythema ab igne. Am J Dermatopathol. 2013;35:676-678. doi:10.1097/DAD.0b013e3182871648
- Wharton J, Roffwarg D, Miller J, et al. Cutaneous marginal zone lymphoma arising in the setting of erythema ab igne. J Am Acad Dermatol. 2010;62:1080-1081. doi:10.1016/j.jaad.2009.08.005
- Jones CS, Tyring SK, Lee PC, et al. Development of neuroendocrine (Merkel cell) carcinoma mixed with squamous cell carcinoma in erythema ab igne. Arch Dermatol. 1988;124:110-113.
- Kim HW, Kim EJ, Park HC, et al. Erythema ab igne successfully treated with low fluenced 1,064-nm Q-switched neodymium-doped yttrium aluminum garnet laser. J Cosmet Laser Ther. 2014;16:147-148. doi:10.3109/14764172.2013.854623
- Tan S, Bertucci V. Erythema ab igne: an old condition new again. CMAJ. 2000;62:77-78.
- Gianfaldoni S, Gianfaldoni R, Tchernev G, et al. Erythema ab igne successfully treated with mesoglycan and bioflavonoids: a case-report. Open Access Maced J Med Sci. 2017;5:432-435. doi:10.3889/oamjms.2017.123
- Perry. Case of erythema ab igne. Br J Dermatol. 1900;xxiii:375.
- Bose S, Ortonee JP. Diseases affected by heat. In: Parish LC, Millikan LE, Amer M, et al. Global Dermatology Diagnosis and Management According to Geography, Climate, and Culture. Springer-Varlag; 1994:83-92.
- Leal-Lobato MM, Blasco-Morente G. Electric blanket induced erythema ab igne [in Spanish]. Semergen. 2015;41:456-457. doi:10.1016/j.semerg.2014.12.008
- Huynh N, Sarma D, Huerter C. Erythema ab igne: a case report and review of the literature. Cutis. 2011;88:290-292.
- Kesty K, Feldman SR. Erythema ab igne: evolving technology, evolving presentation. Dermatol Online J. 2014;20. doi:10.5070/D32011024689
- Miller K, Hunt R, Chu J, et al. Erythema ab igne. Dermatol Online J. 2011;17:28.
- Smith ML. Environmental and sports-related skin diseases. In: Bolognia JL, Schaffer JV, Cerroni L, eds. Dermatology. 4th ed. Elsevier; 2018:1569-1594.
- Errichetti E, Stinco G. Dermoscopy in general dermatology: a practical overview. Dermatol Ther (Heidelb). 2016;6:471-507. doi:10.1007/s13555-016-0141-6
- Guarneri C, Tchernev G, Wollina U, et al. Erythema ab igne caused by laptop computer. Open Access Maced J Med Sci. 2017;5:490-492. doi:10.3889/oamjms.2017.137
- Arnold AW, Itin PH. Laptop computer-induced erythema ab igne in a child and review of the literature. Pediatrics. 2010;126:E1227-E1230. doi:10.1542/peds.2010-1390
- Dickman J, Kessler S. Unilateral reticulated patch localized to the anterior thigh. JAAD Case Rep. 2018;4:746-748. doi:10.1016/j.jdcr.2018.06.007
- Boffa MJ. Laptop computer-induced erythema ab igne on the left breast. Cutis. 2011;87:175-176.
- Li K, Barankin B. Cutaneous manifestations of modern technology use. J Cutan Med Surg. 2011;15:347-353. doi:10.2310/7750.2011.10053
- Riahi RR, Cohen PR. Laptop-induced erythema ab igne: report and review of literature. Dermatol Online J. 2012;18:5.
- Andersen F. Laptop-thighs--laptop-induced erythema ab igne [in Danish]. Ugeskr Laeger. 2010;172:635.
- Jagtman BA. Erythema ab igne due to a laptop computer. Contact Dermatitis. 2004;50:105. doi:10.1111/j.0105-1873.2004.0295g.x
- Olechowska M, Kisiel K, Ruszkowska L, et al. Erythema ab igne (EAI) induced by a laptop computer: report of two cases. J Dtsch Dermatol Ges. doi:10.1111/j.1610-0387.2014.12387
- Nayak SUK, Shenoi SD, Prabhu S. Laptop induced erythema ab igne. Indian J Dermatol. 2012;57:131-132. doi:10.4103/0019-5154.94284
- Salvio AG, Nunes AJ, Angarita DPR. Laptop computer induced erythema ab igne: a new presentation of an old disease. An Bras Dermatol. 2016;91:79-80. doi:10.1590/abd1806-4841.20165139
- Schummer C, Tittelbach J, Elsner P. Right-sided laptop dermatitis [in German]. Dtsch Med Wochenschr. 2015;140:1376-1377. doi:10.1055/s-0041-103615
- Manoharan D. Erythema ab igne: usual site, unusual cause. J Pharm Bioallied Sci. 2015;7(suppl 1):S74-S75. doi:10.4103/0975-7406.155811
- Giraldi S, Diettrich F, Abbage KT, et al. Erythema ab igne induced by a laptop computer in an adolescent. An Bras Dermatol. 2011;86:128-130. doi:10.1590/S0365-05962011000100018
- Secher LLS, Vind-Kezunovic D, Zachariae COC. Side-effects to the use of laptop computers: erythema ab igne. Dermatol Reports. 2010;31:E11. doi:10.4081/dr.2010.e11
- Botten D, Langley RGB, Webb A. Academic branding: erythema ab igne and use of laptop computers. CMAJ. 2010;182:E857. doi:10.1503/cmaj.091868
- Bilic M, Adams BB. Erythema ab igne induced by a laptop computer. J Am Acad Dermatol. 2004;50:973-974. doi:10.1016/j.jaad.2003.08.007
- Fu LW, Vender R. Erythema ab igne caused by laptop computer gaming - a case report. Int J Dermatol. 2012;51:716-717. doi:10.1111/j.1365-4632.2011.05033.x
- Levinbook WS, Mallett J, Grant-Kels JM. Laptop computer-associated erythema ab igne. Cutis. 2007;80:319-320.
- Mohr MR, Scott KA, Pariser RM, et al. Laptop computer-induced erythema ab igne: a case report. Cutis. 2007;79:59-60.
- Cantor AS, Bartling SJ. Laptop computer-induced hyperpigmentation. Dermatol Online J. 2018;24:13030/qt6k37r9wm.
- Kaptanog˘lu AF, Mullaaziz D. Erythema ab igne in the palmar area induced by smart phone: case report. Turkiye Klin J Med Sci. 2015;35:284-286. doi:10.5336/medsci.2015-46976
- Redding KS, Watts AN, Lee J, et al. Space heater-induced bullous erythema ab igne. Cutis. 2017;100:E9-E10.
- Goorland J, Edens MA, Baudoin TD. An emergency department presentation of erythema ab igne caused by repeated heater exposure. J La State Med Soc. 2016;168:33-34.
- Kokturk A, Kaya TI, Baz K, et al. Bullous erythema ab igne. Dermatol Online J. 2003;9:18.
- Brzezinski P, Ismail S, Chiriac A. Radiator-induced erythema ab igne in 8-year-old girl. Rev Chil Pediatr. 2014;85:239-240. doi:10.4067/S0370-41062014000200015
- Adams BB. Heated car seat-induced erythema ab igne. Arch Dermatol. 2012;148:265-266. doi:10.1001/archdermatol.2011.2207
- Helm TN, Spigel GT, Helm KF. Erythema ab igne caused by a car heater. Cutis. 1997;59:81-82.
- Gregory JF, Beute TC. Erythema ab igne. J Spec Oper Med. 2013;13:115-119. doi:10.55460/5AVH-NZHY
- Chua S, Chen Q, Lee HY. Erythema ab igne and dermal scarring caused by cupping and moxibustion treatment. J Dtsch Dermatol Ges. 2015;13:337-338. doi:10.1111/ddg.12581
- Chen JF, Liu YC, Chen YF, et al. Erythema ab igne after footbath with Chinese herbal remedies. J Chinese Med Assoc. 2011;74:51-53. doi:10.1016/j.jcma.2011.01.009
- Baltazar D, Brockman R, Simpson E. Kotatsu-induced erythema ab igne. An Bras Dermatol. 2019;94:253-254. doi:10.1590/abd1806-4841.20198792
- Baig M, Byrne F. Erythema ab igne and its relation to spinal pathology. Cureus. 2018;10:e2914. doi:10.7759/cureus.2914
- Aria AB, Chen L, Silapunt S. Erythema ab igne from heating pad use: a report of three clinical cases and a differential diagnosis. Cureus. 2018;10:e2635. doi:10.7759/cureus.2635
- Milchak M, Smucker J, Chung CG, et al. Erythema ab igne due to heating pad use: a case report and review of clinical presentation, prevention, and complications. Case Rep Med. 2016;1862480. doi:10.1155/2016/1862480
- Gmuca S, Yu J, Weiss PF, et al. Erythema ab igne in an adolescent with chronic pain: an alarming cutaneous eruption from heat exposure. Pediatr Emerg Care. 2020;36:e236-e238. doi:10.1097/PEC.0000000000001460
- Dizdarevic A, Karim OA, Bygum A. A reddish brown reticulated hyperpigmented erythema on the abdomen of a girl. Erythema ab igne, also known as toasted skin syndrome, caused by a heating pad onthe abdomen. Acta Derm Venereol. 2014;94:365-367. doi:10.2340/00015555-1722
- Chatterjee S. Erythema ab igne from prolonged use of a heating pad. Mayo Clin Proc. 2005;80:1500. doi:10.4065/80.11.1500
- Waldorf DS, Rast MF, Garofalo VJ. Heating-pad erythematous dermatitis “erythema ab igne.” JAMA. 1971;218:1704. doi:10.1001/jama.1971.03190240056023
- South AM, Crispin MK, Marqueling AL, et al. A hyperpigmented reticular rash in a patient on peritoneal dialysis. Perit Dial Int. 2016;36:677-700. doi:10.3747/pdi.2016.00042
- Ravindran R. Erythema ab igne in an individual with diabetes and gastroparesis. BMJ Case Rep. 2017;2017:bcr2014203856. doi:10.1136/bcr-2014-203856
- Dessinioti C, Katsambas A, Tzavela E, et al. Erythema ab igne in three girls with anorexia nervosa. Pediatr Dermatol. 2016;33:e149-e150. doi:10.1111/pde.12770
- Fischer J, Rein K, Erfurt-Berge C, et al. Three cases of erythema ab igne (EAI) in patients with eating disorders. Neuropsychiatr. 2010;24:141-143.
- Docx MKF, Simons A, Ramet J, et al. Erythema ab igne in an adolescent with anorexia nervosa. Int J Eat Disord. 2013;46:381-383. doi:10.1002/eat.22075
- Turan E, Cimen V, Haytoglu NSK, et al. A case of bullous erythema ab igne accompanied by anemia and subclinical hypothyroidism. Dermatol Online J. 2014;20:223366.
- Pavithran K. Erythema ab igne, schizophrenia and thermophilia. Indian J Dermatol Venereol Leprol. 1987;53:181-182.
- Dellavelle R, Gillum P. Erythema ab igne following heating/cooling blanket use in the intensive care unit. Cutis. 2000;66:136-138.
- Park SY, Kim SM, Yoon TJ. Erythema ab igne caused by weight loss heating pad. Korean J Dermatol. 2007;45:489-491.
- Sachdeva M, Gianotti R, Shah M, et al. Cutaneous manifestations of COVID-19: report of three cases and a review of literature. J Dermatol Sci. 2020;98:75-81. doi:10.1016/j.jdermsci.2020.04.011
- Gisondi P, Plaserico S, Bordin C, et al. Cutaneous manifestations of SARS‐CoV‐2 infection: a clinical update. J Eur Acad Dermatol Venereol. 2020;34:2499-2504. doi:10.1111/jdv.16774
- Manalo IF, Smith MK, Cheeley J, et al. A dermatologic manifestation of COVID-19: transient livedo reticularis. J Am Acad Dermatol. 2020;83:700. doi:10.1016/j.jaad.2020.04.018
- Zhao Q, Fang X, Pang Z, et al. COVID‐19 and cutaneous manifestations: a systematic review. J Eur Acad Dermatol Venereol. 2020;34:2505-2510. doi:10.1111/jdv.16778
- Akasaka T, Kon S. Two cases of squamous cell carcinoma arising from erythema ab igne. Nihon Hifuka Gakkai Zasshi. 1989;99:735-742.
- Arrington JH 3rd, Lockman DS. Thermal keratoses and squamous cell carcinoma in situ associated with erythema ab igne. Arch Dermatol. 1979;115:1226-1228.
- Wharton JB, Sheehan DJ, Lesher JL Jr. Squamous cell carcinoma in situ arising in the setting of erythema ab igne. J Drugs Dermatol. 2008;7:488-489.
- Wollina U, Helm C, Hansel G, et al. Two cases of erythema ab igne, one with a squamous cell carcinoma. G Ital Dermatol Venereol. 2007;142:415-418.
- Rudolph CM, Soyer HP, Wolf P, et al. Squamous cell carcinoma arising in erythema ab igne. Hautarzt. 2000;51:260-263. doi:10.1007/s001050051115
- Sigmon JR, Cantrell J, Teague D, et al. Poorly differentiated carcinoma arising in the setting of erythema ab igne. Am J Dermatopathol. 2013;35:676-678. doi:10.1097/DAD.0b013e3182871648
- Wharton J, Roffwarg D, Miller J, et al. Cutaneous marginal zone lymphoma arising in the setting of erythema ab igne. J Am Acad Dermatol. 2010;62:1080-1081. doi:10.1016/j.jaad.2009.08.005
- Jones CS, Tyring SK, Lee PC, et al. Development of neuroendocrine (Merkel cell) carcinoma mixed with squamous cell carcinoma in erythema ab igne. Arch Dermatol. 1988;124:110-113.
- Kim HW, Kim EJ, Park HC, et al. Erythema ab igne successfully treated with low fluenced 1,064-nm Q-switched neodymium-doped yttrium aluminum garnet laser. J Cosmet Laser Ther. 2014;16:147-148. doi:10.3109/14764172.2013.854623
- Tan S, Bertucci V. Erythema ab igne: an old condition new again. CMAJ. 2000;62:77-78.
- Gianfaldoni S, Gianfaldoni R, Tchernev G, et al. Erythema ab igne successfully treated with mesoglycan and bioflavonoids: a case-report. Open Access Maced J Med Sci. 2017;5:432-435. doi:10.3889/oamjms.2017.123
Practice Points
- Erythema ab igne (EAI) is a skin condition caused by chronic exposure to heat; removal of the heat source often will result in self-resolution of the rash.
- Erythema ab igne can be a sign of underlying illness in patients self-treating chronic pain with application of heat.
- Recognition and discontinuation of the exposure with close observation are key components in the treatment of EAI.
Skin Diseases Associated With COVID-19: A Narrative Review
COVID-19 is a potentially severe systemic disease caused by SARS-CoV-2. SARS-CoV and Middle East respiratory syndrome (MERS-CoV) caused fatal epidemics in Asia in 2002 to 2003 and in the Arabian Peninsula in 2012, respectively. In 2019, SARS-CoV-2 was detected in patients with severe, sometimes fatal pneumonia of previously unknown origin; it rapidly spread around the world, and the World Health Organization declared the disease a pandemic on March 11, 2020. SARS-CoV-2 is a β-coronavirus that is genetically related to the bat coronavirus and SARS-CoV; it is a single-stranded RNA virus of which several variants and subvariants exist. The SARS-CoV-2 viral particles bind via their surface spike protein (S protein) to the angiotensin-converting enzyme 2 receptor present on the membrane of several cell types, including epidermal and adnexal keratinocytes.1,2 The α and δ variants, predominant from 2020 to 2021, mainly affected the lower respiratory tract and caused severe, potentially fatal pneumonia, especially in patients older than 65 years and/or with comorbidities, such as obesity, hypertension, diabetes, and (iatrogenic) immunosuppression. The ο variant, which appeared in late 2021, is more contagious than the initial variants, but it causes a less severe disease preferentially affecting the upper respiratory airways.3 As of April 5, 2023, more than 762,000,000 confirmed cases of COVID-19 have been recorded worldwide, causing more than 6,800,000 deaths.4
Early studies from China describing the symptoms of COVID-19 reported a low frequency of skin manifestations (0.2%), probably because they were focused on the most severe disease symptoms.5 Subsequently, when COVID-19 spread to the rest of the world, an increasing number of skin manifestations were reported in association with the disease. After the first publication from northern Italy in spring 2020, which was specifically devoted to skin manifestations of COVID-19,6 an explosive number of publications reported a large number of skin manifestations, and national registries were established in several countries to record these manifestations, such as the American Academy of Dermatology and the International League of Dermatological Societies registry,7,8 the COVIDSKIN registry of the French Dermatology Society,9 and the Italian registry.10 Highlighting the unprecedented number of scientific articles published on this new disease, a PubMed search of articles indexed for MEDLINE search using the terms SARS-CoV-2 or COVID-19, on April 6, 2023, revealed 351,596 articles; that is more than 300 articles published every day in this database alone, with a large number of them concerning the skin.
SKIN DISEASSES ASSOCIATED WITH COVID-19
There are several types of COVID-19–related skin manifestations, depending on the circumstances of onset and the evolution of the pandemic.
Skin Manifestations Associated With SARS-CoV-2 Infection
The estimated incidence varies greatly according to the published series of patients, possibly depending on the geographic location. The estimated incidence seems lower in Asian countries, such as China (0.2%)5 and Japan (0.56%),11 compared with Europe (up to 20%).6 Skin manifestations associated with SARS-CoV-2 infection affect individuals of all ages, slightly more females, and are clinically polymorphous; some of them are associated with the severity of the infection.12 They may precede, accompany, or appear after the symptoms of COVID-19, most often within a month of the infection, of which they rarely are the only manifestation; however, their precise relationship to SARS-CoV-2 is not always well known. They have been classified according to their clinical presentation into several forms.13-15
Morbilliform Maculopapular Eruption—Representing 16% to 53% of skin manifestations, morbilliform and maculopapular eruptions usually appear within 15 days of infection; they manifest with more or less confluent erythematous macules that may be hemorrhagic/petechial, and usually are asymptomatic and rarely pruritic. The rash mainly affects the trunk and limbs, sparing the face, palmoplantar regions, and mucous membranes; it appears concomitantly with or a few days after the first symptoms of COVID-19 (eg, fever, respiratory symptoms), regresses within a few days, and does not appear to be associated with disease severity. The distinction from maculopapular drug eruptions may be subtle. Histologically, the rash manifests with a spongiform dermatitis (ie, variable parakeratosis; spongiosis; and a mixed dermal perivascular infiltrate of lymphocytes, eosinophils and histiocytes, depending on the lesion age)(Figure 1). The etiopathogenesis is unknown; it may involve immune complexes to SARS-CoV-2 deposited on skin vessels. Treatment is not mandatory; if necessary, local or systemic corticosteroids may be used.
Vesicular (Pseudovaricella) Rash—This rash accounts for 11% to 18% of all skin manifestations and usually appears within 15 days of COVID-19 onset. It manifests with small monomorphous or varicellalike (pseudopolymorphic) vesicles appearing on the trunk, usually in young patients. The vesicles may be herpetiform, hemorrhagic, or pruritic, and appear before or within 3 days of the onset of mild COVID-19 symptoms; they regress within a few days without scarring. Histologically, the lesions show basal cell vacuolization; multinucleated, dyskeratotic/apoptotic or ballooning/acantholytic epidermal keratinocytes; reticular degeneration of the epidermis; intraepidermal vesicles sometimes resembling herpetic vesicular infections or Grover disease; and mild dermal inflammation. There is no specific treatment.
Urticaria—Urticarial rash, or urticaria, represents 5% to 16% of skin manifestations; usually appears within 15 days of disease onset; and manifests with pruritic, migratory, edematous papules appearing mainly on the trunk and occasionally the face and limbs. The urticarial rash tends to be associated with more severe forms of the disease and regresses within a week, responding to antihistamines. Of note, clinically similar rashes can be caused by drugs. Histologically, the lesions show dermal edema and a mild perivascular lymphocytic infiltrate, sometimes admixed with eosinophils.
Chilblainlike Lesions—Chilblainlike lesions (CBLLs) account for 19% of skin manifestations associated with COVID-1913 and present as erythematous-purplish, edematous lesions that can be mildly pruritic or painful, appearing on the toes—COVID toes—and more rarely the fingers (Figure 2). They were seen epidemically during the first pandemic wave (2020 lockdown) in several countries, and clinically are very similar to, if not indistinguishable from, idiopathic chilblains, but are not necessarily associated with cold exposure. They appear in young, generally healthy patients or those with mild COVID-19 symptoms 2 to 4 weeks after symptom onset. They regress spontaneously or under local corticosteroid treatment within a few days or weeks. Histologically, CBLLs are indistinguishable from chilblains of other origins, namely idiopathic (seasonal) ones. They manifest with necrosis of epidermal keratinocytes; dermal edema that may be severe, leading to the development of subepidermal pseudobullae; a rather dense perivascular and perieccrine gland lymphocytic infiltrate; and sometimes with vascular lesions (eg, edema of endothelial cells, microthromboses of dermal capillaries and venules, fibrinoid deposits within the wall of dermal venules)(Figure 3).16-18 Most patients (>80%) with CBLLs have negative serologic or polymerase chain reaction tests for SARS-CoV-2,19 which generated a lively debate about the role of SARS-CoV-2 in the genesis of CBLLs. According to some authors, SARS-CoV-2 plays no direct role, and CBLLs would occur in young people who sit or walk barefoot on cold floors at home during confinement.20-23 Remarkably, CBLLs appeared in patients with no history of chilblains during a season that was not particularly cold, namely in France or in southern California, where their incidence was much higher compared to the same time period of prior years. Some reports have supported a direct role for the virus based on questionable observations of the virus within skin lesions (eg, sweat glands, endothelial cells) by immunohistochemistry, electron microscopy, and/or in situ hybridization.17,24,25 A more satisfactory hypothesis would involve the role of a strong innate immunity leading to elimination of the virus before the development of specific antibodies via the increased production of type 1 interferon (IFN-1); this would affect the vessels, causing CBLLs. This mechanism would be similar to the one observed in some interferonopathies (eg, Aicardi-Goutières syndrome), also characterized by IFN-1 hypersecretion and chilblains.26-29 According to this hypothesis, CBLLs should be considered a paraviral rash similar to other skin manifestations associated with COVID-19.30
Acro-ischemia—Acro-ischemia livedoid lesions account for 1% to 6% of skin manifestations and comprise lesions of livedo (either reticulated or racemosa); necrotic acral bullae; and gangrenous necrosis of the extremities, especially the toes. The livedoid lesions most often appear within 15 days of COVID-19 symptom onset, and the purpuric lesions somewhat later (2–4 weeks); they mainly affect adult patients, last about 10 days, and are the hallmark of severe infection, presumably related to microthromboses of the cutaneous capillaries (endothelial dysfunction, prothrombotic state, elevated D-dimers). Histologically, they show capillary thrombosis and dermoepidermal necrosis (Figure 4).
Other Reported Polymorphic or Atypical Rashes—Erythema multiforme–like eruptions may appear before other COVID-19 symptoms and manifest as reddish-purple, nearly symmetric, diffuse, occasionally targetoid bullous or necrotic macules. The eruptions mainly affect adults and most often are seen on the palms, elbows, knees, and sometimes the mucous membranes. The rash regresses in 1 to 3 weeks without scarring and represents a delayed cutaneous hypersensitivity reaction. Histologically, the lesions show vacuolization of basal epidermal keratinocytes, keratinocyte necrosis, dermoepidermal detachment, a variably dense dermal T-lymphocytic infiltrate, and red blood cell extravasation (Figure 5).
Leukocytoclastic vasculitis may be generalized or localized. It manifests clinically by petechial/purpuric maculopapules, especially on the legs, mainly in elderly patients with COVID-19. Histologically, the lesions show necrotizing changes of dermal postcapillary venules, neutrophilic perivascular inflammation, red blood cell extravasation, and occasionally vascular IgA deposits by direct immunofluorescence examination. The course usually is benign.
The incidence of pityriasis rosea and of clinically similar rashes (referred to as “pityriasis rosea–like”) increased 5-fold during the COVID-19 pandemic.31,32 These dermatoses manifest with erythematous, scaly, circinate plaques, typically with an initial herald lesion followed a few days later by smaller erythematous macules. Histologically, the lesions comprise a spongiform dermatitis with intraepidermal exocytosis of red blood cells and a mild to moderate dermal lymphocytic infiltrate.
Erythrodysesthesia, or hand-foot syndrome, manifests with edematous erythema and palmoplantar desquamation accompanied by a burning sensation or pain. This syndrome is known as an adverse effect of some chemotherapies because of the associated drug toxicity and sweat gland inflammation; it was observed in 40% of 666 COVID-19–positive patients with mild to moderate pneumonitis.33
“COVID nose” is a rare cutaneous manifestation characterized by nasal pigmentation comprising multiple coalescent frecklelike macules on the tip and wings of the nose and sometimes the malar areas. These lesions predominantly appear in women aged 25 to 65 years and show on average 23 days after onset of COVID-19, which is usually mild. This pigmentation is similar to pigmentary changes after infection with chikungunya; it can be treated with depigmenting products such as azelaic acid and hydroquinone cream with sunscreen use, and it regresses in 2 to 4 months.34
Telogen effluvium (excessive and temporary shedding of normal telogen club hairs of the entire scalp due to the disturbance of the hair cycle) is reportedly frequent in patients (48%) 1 month after COVID-19 infection, but it may appear later (after 12 weeks).35 Alopecia also is frequently reported during long (or postacute) COVID-19 (ie, the symptomatic disease phase past the acute 4 weeks’ stage of the infection) and shows a female predominance36; it likely represents the telogen effluvium seen 90 days after a severe illness. Trichodynia (pruritus, burning, pain, or paresthesia of the scalp) also is reportedly common (developing in more than 58% of patients) and is associated with telogen effluvium in 44% of cases. Several cases of alopecia areata (AA) triggered or aggravated by COVID-19 also have been reported37,38; they could be explained by the “cytokine storm” triggered by the infection, involving T and B lymphocytes; plasmacytoid dendritic cells; natural killer cells with oversecretion of IL-6, IL-4, tumor necrosis factor α, and IFN type I; and a cytotoxic reaction associated with loss of the immune privilege of hair follicles.
Nail Manifestations
The red half-moon nail sign is an asymptomatic purplish-red band around the distal margin of the lunula that affects some adult patients with COVID-19.39 It appears shortly after onset of symptoms, likely the manifestation of vascular inflammation in the nail bed, and regresses slowly after approximately 1 week.40 Beau lines are transverse grooves in the nail plate due to the temporary arrest of the proximal nail matrix growth accompanying systemic illnesses; they appear approximately 2 to 3 weeks after the onset of COVID-19.41 Furthermore, nail alterations can be caused by drugs used to treat COVID-19, such as longitudinal melanonychia due to treatment with hydroxychloroquine or fluorescence of the lunula or nail plate due to treatment with favipiravir.42
Multisystem Inflammatory Syndrome
Multisystem inflammatory syndrome (MIS) is clinically similar to Kawasaki disease; it typically affects children43 and more rarely adults with COVID-19. It manifests with fever, weakness, and biological inflammation and also frequently with skin lesions (72%), which are polymorphous and include morbilliform rash (27%); urticaria (24%); periorbital edema (24%); nonspecific erythema (21.2%); retiform purpura (18%); targetoid lesions (15%); malar rash (15.2%); and periareolar erythema (6%).44 Compared to Kawasaki disease, MIS affects slightly older children (mean age, 8.5 vs 3 years) and more frequently includes cardiac and gastrointestinal manifestations; the mortality rate also is slightly higher (2% vs 0.17%).45
Confirmed COVID-19 Infection
At the beginning of the pandemic, skin manifestations were reported in patients who were suspected of having COVID-19 but did not always have biological confirmation of SARS-CoV-2 infection due to the unavailability of diagnostic tests or the physical impossibility of testing. However, subsequent studies have confirmed that most of these dermatoses were indeed associated with COVID-19 infection.9,46 For example, a study of 655 patients with confirmed COVID-19 infection reported maculopapular (38%), vascular (22%), urticarial (15%), and vesicular (15%) rashes; erythema multiforme or Stevens-Johnson–like syndrome (3%, often related to the use of hydroxychloroquine); generalized pruritus (1%); and MIS (0.5%). The study confirmed that CBLLs were mostly seen in young patients with mild disease, whereas livedo (fixed rash) and retiform purpura occurred in older patients with a guarded prognosis.46
Remarkably, most dermatoses associated with SARS-CoV-2 infection were reported during the initial waves of the pandemic, which were due to the α and δ viral variants. These manifestations were reported more rarely when the ο variant was predominant, even though most patients (63%) who developed CBLLs in the first wave also developed them during the second pandemic wave.47 This decrease in the incidence of COVID-19–associated dermatoses could be because of the lower pathogenicity of the o variant,3 a lower tropism for the skin, and variations in SARS-CoV-2 antigenicity that would induce a different immunologic response, combined with an increasingly stronger herd immunity compared to the first pandemic waves achieved through vaccination and spontaneous infections in the population. Additional reasons may include different baseline characteristics in patients hospitalized with COVID-19 (regarding comorbidities, disease severity, and received treatments), and the possibility that some of the initially reported COVID-19–associated skin manifestations could have been produced by different etiologic agents.48 In the last 2 years, COVID-19–related skin manifestations have been reported mainly as adverse events to COVID-19 vaccination.
CUTANEOUS ADVERSE EFFECTS OF DRUGS USED TO TREAT COVID-19
Prior to the advent of vaccines and specific treatments for SARS-CoV-2, various drugs were used—namely hydroxychloroquine, ivermectin, and tocilizumab—that did not prove efficacious and caused diverse adverse effects, including cutaneous eruptions such as urticaria, maculopapular eruptions, erythema multiforme or Stevens-Johnson syndrome, vasculitis, longitudinal melanonychia, and acute generalized exanthematous pustulosis.49,50 Nirmatrelvir 150 mg–ritonavir 100 mg, which was authorized for emergency use by the US Food and Drug Administration for the treatment of COVID-19, is a viral protease inhibitor blocking the replication of the virus. Ritonavir can induce pruritus, maculopapular rash, acne, Stevens-Johnson syndrome, and toxic epidermal necrolysis; of note, these effects have been observed following administration of ritonavir for treatment of HIV at higher daily doses and for much longer periods of time compared with treatment of COVID-19 (600–1200 mg vs 200 mg/d, respectively). These cutaneous drug side effects are clinically similar to the manifestations caused either directly or indirectly by SARS-CoV-2 infection; therefore, it may be difficult to differentiate them.
DERMATOSES DUE TO PROTECTIVE DEVICES
Dermatoses due to personal protective equipment such as masks or face shields affected the general population and mostly health care professionals51; 54.4% of 879 health care professionals in one study reported such events.52 These dermatoses mainly include contact dermatitis of the face (nose, forehead, and cheeks) of irritant or allergic nature (eg, from preservatives releasing formaldehyde contained in masks and protective goggles). They manifest with skin dryness; desquamation; maceration; fissures; or erosions or ulcerations of the cheeks, forehead, and nose. Cases of pressure urticaria also have been reported. Irritant dermatitis induced by the frequent use of disinfectants (eg, soaps, hydroalcoholic sanitizing gels) also can affect the hands. Allergic hand dermatitis can be caused by medical gloves.
The term maskne (or mask acne) refers to a variety of mechanical acne due to the prolonged use of surgical masks (>4 hours per day for ≥6 weeks); it includes cases of de novo acne and cases of pre-existing acne aggravated by wearing a mask. Maskne is characterized by acne lesions located on the facial area covered by the mask (Figure 6). It is caused by follicular occlusion; increased sebum secretion; mechanical stress (pressure, friction); and dysbiosis of the microbiome induced by changes in heat, pH, and humidity. Preventive measures include application of noncomedogenic moisturizers or gauze before wearing the mask as well as facial cleansing with appropriate nonalcoholic products. Similar to acne, rosacea often is aggravated by prolonged wearing of surgical masks (mask rosacea).53,54
DERMATOSES REVEALED OR AGGRAVATED BY COVID-19
Exacerbation of various skin diseases has been reported after infection with SARS-CoV-2.55 Psoriasis and acrodermatitis continua of Hallopeau,56 which may progress into generalized, pustular, or erythrodermic forms,57 have been reported; the role of hydroxychloroquine and oral corticosteroids used for the treatment of COVID-19 has been suspected.57 Atopic dermatitis patients—26% to 43%—have experienced worsening of their disease after symptomatic COVID-19 infection.58 The incidence of herpesvirus infections, including herpes zoster, increased during the pandemic.59 Alopecia areata relapses occurred in 42.5% of 392 patients with preexisting disease within 2 months of COVID-19 onset in one study,60 possibly favored by the psychological stress; however, some studies have not confirmed the aggravating role of COVID-19 on alopecia areata.61 Lupus erythematosus, which may relapse in the form of Rowell syndrome,62 and livedoid vasculopathy63 also have been reported following COVID-19 infection.
SKIN MANIFESTATIONS ASSOCIATED WITH COVID-19 VACCINES
In parallel with the rapid spread of COVID-19 vaccination,4 an increasing number of skin manifestations has been observed following vaccination; these dermatoses now are more frequently reported than those related to natural SARS-CoV-2 infection.64-70 Vaccine-induced skin manifestations have a reported incidence of approximately 4% and show a female predominance.65 Most of them (79%) have been reported in association with messenger RNA (mRNA)–based vaccines, which have been the most widely used; however, the frequency of side effects would be lower after mRNA vaccines than after inactivated virus-based vaccines. Eighteen percent occurred after the adenoviral vector vaccine, and 3% after the inactivated virus vaccine.70 Fifty-nine percent were observed after the first dose. They are clinically polymorphous and generally benign, regressing spontaneously after a few days, and they should not constitute a contraindication to vaccination.Interestingly, many skin manifestations are similar to those associated with natural SARS-CoV-2 infection; however, their frequency and severity does not seem to depend on whether the patients had developed skin reactions during prior SARS-CoV-2 infection. These reactions have been classified into several types:
• Immediate local reactions at the injection site: pain, erythema, or edema represent the vast majority (96%) of reactions to vaccines. They appear within 7 days after vaccination (average, 1 day), slightly more frequently (59%) after the first dose. They concern mostly young patients and are benign, regressing in 2 to 3 days.70
• Delayed local reactions: characterized by pain or pruritus, erythema, and skin induration mimicking cellulitis (COVID arm) and represent 1.7% of postvaccination reactions. They correspond to a delayed hypersensitivity reaction and appear approximately 7 days after vaccination, most often after the first vaccine dose (75% of cases), which is almost invariably mRNA based.70
• Urticarial reactions corresponding to an immediate (type 1) hypersensitivity reaction: constitute 1% of postvaccination reactions, probably due to an allergy to vaccine ingredients. They appear on average 1 day after vaccination, almost always with mRNA vaccines.70
• Angioedema: characterized by mucosal or subcutaneous edema and constitutes 0.5% of postvaccination reactions. It is a potentially serious reaction that appears on average 12 hours after vaccination, always with an mRNA-based vaccine.70
• Morbilliform rash: represents delayed hypersensitivity reactions (0.1% of postvaccination reactions) that appear mostly after the first dose (72%), on average 3 days after vaccination, always with an mRNA-based vaccine.70
• Herpes zoster: usually develops after the first vaccine dose in elderly patients (69% of cases) on average 4 days after vaccination and constitutes 0.1% of postvaccination reactions.71
• Bullous diseases: mainly bullous pemphigoid (90%) and more rarely pemphigus (5%) or bullous erythema pigmentosum (5%). They appear in elderly patients on average 7 days after vaccination and constitute 0.04% of postvaccination reactions.72
• Chilblainlike lesions: several such cases have been reported so far73; they constitute 0.03% of postvaccination reactions.70 Clinically, they are similar to those associated with natural COVID-19; they appear mostly after the first dose (64%), on average 5 days after vaccination with the mRNA or adenovirus vaccine, and show a female predominance. The appearance of these lesions in vaccinated patients, who are a priori not carriers of the virus, strongly suggests that CBLLs are due to the immune reaction against SARS-CoV-2 rather than to a direct effect of this virus on the skin, which also is a likely scenario with regards to other skin manifestations seen during the successive COVID-19 epidemic waves.73-75
• Reactions to hyaluronic acid–containing cosmetic fillers: erythema, edema, and potentially painful induration at the filler injection sites. They constitute 0.04% of postvaccination skin reactions and appear 24 hours after vaccination with mRNA-based vaccines, equally after the first or second dose.76
• Pityriasis rosea–like rash: most occur after the second dose of mRNA-based vaccines (0.023% of postvaccination skin reactions).70
• Severe reactions: these include acute generalized exanthematous pustulosis77 and Stevens-Johnson syndrome.78 One case of each has been reported after the adenoviral vector vaccine 3 days after vaccination.
Other more rarely observed manifestations include reactivation/aggravation or de novo appearance of inflammatory dermatoses such as psoriasis,79,80 leukocytoclastic vasculitis,81,82 lymphocytic83 or urticarial84 vasculitis, Sweet syndrome,85 lupus erythematosus, dermatomyositis,86,87 alopecia,37,88 infection with Trichophyton rubrum,89 Grover disease,90 and lymphomatoid reactions (such as recurrences of cutaneous T-cell lymphomas [CD30+], and de novo development of lymphomatoid papulosis).91
FINAL THOUGHTS
COVID-19 is associated with several skin manifestations, even though the causative role of SARS-CoV-2 has remained elusive. These dermatoses are highly polymorphous, mostly benign, and usually spontaneously regressive, but some of them reflect severe infection. They mostly were described during the first pandemic waves, reported in several national and international registries, which allowed for their morphological classification. Currently, cutaneous adverse effects of vaccines are the most frequently reported dermatoses associated with SARS-CoV-2, and it is likely that they will continue to be observed while COVID-19 vaccination lasts. Hopefully the end of the COVID-19 pandemic is near. In January 2023, the International Health Regulations Emergency Committee of the World Health Organization acknowledged that the COVID-19 pandemic may be approaching an inflexion point, and even though the event continues to constitute a public health emergency of international concern, the higher levels of population immunity achieved globally through infection and/or vaccination may limit the impact of SARS-CoV-2 on morbidity and mortality. However, there is little doubt that this virus will remain a permanently established pathogen in humans and animals for the foreseeable future.92 Therefore, physicians—especially dermatologists—should be aware of the various skin manifestations associated with COVID-19 so they can more efficiently manage their patients.
- Ashraf UM, Abokor AA, Edwards JM, et al. SARS-CoV-2, ACE2 expression, and systemic organ invasion. Physiol Genomics. 2021;53:51-60.
- Ganier C, Harun N, Peplow I, et al. Angiotensin-converting enzyme 2 expression is detectable in keratinocytes, cutaneous appendages, and blood vessels by multiplex RNA in situ hybridization. Adv Skin Wound Care. 2022;35:219-223.
- Ulloa AC, Buchan SA, Daneman N, et al. Estimates of SARS-CoV-2 omicron variant severity in Ontario, Canada. JAMA. 2022;327:1286-1288.
- World Health Organization. Coronavirus (COVID-19) Dashboard. Accessed April 6, 2023. https://covid19.who.int
- Guan WJ, Ni ZY, Hu Y, et al; China Medical Treatment Expert Group for COVID-19. clinical characteristics of coronavirus disease 2019 in China. N Engl J Med. 2020;382:1708-1720.
- Recalcati S. Cutaneous manifestations in COVID-9: a first perspective. J Eur Acad Dermatol Venereol. 2020;34:E212-E213.
- Freeman EE, McMahon DE, Lipoff JB, et al. The spectrum of COVID-19-associated dermatologic manifestations: an international registry of 716 patients from 31 countries. J Am Acad Dermatol. 2020;83:1118-1129.
- Freeman EE, Chamberlin GC, McMahon DE, et al. Dermatology COVID-19 registries: updates and future directions. Dermatol Clin. 2021;39:575-585.
- Guelimi R, Salle R, Dousset L, et al. Non-acral skin manifestations during the COVID-19 epidemic: COVIDSKIN study by the French Society of Dermatology. J Eur Acad Dermatol Venereol. 2021;35:E539-E541.
- Marzano AV, Genovese G, Moltrasio C, et al; Italian Skin COVID-19 Network of the Italian Society of Dermatology and Sexually Transmitted Diseases. The clinical spectrum of COVID-19 associated cutaneous manifestations: an Italian multicenter study of 200 adult patients. J Am Acad Dermatol. 2021;84:1356-1363.
- Sugai T, Fujita Y, Inamura E, et al. Prevalence and patterns of cutaneous manifestations in 1245 COVID-19 patients in Japan: a single-centre study. J Eur Acad Dermatol Venereol. 2022;36:E522-E524.
- Holmes Z, Courtney A, Lincoln M, et al. Rash morphology as a predictor of COVID‐19 severity: a systematic review of the cutaneous manifestations of COVID‐19. Skin Health Dis. 2022;2:E120. doi:10.1002/ski2.120
- Galván Casas C, Català A, Carretero Hernández G, et al. Classification of the cutaneous manifestations of COVID-19: a rapid prospective nationwide consensus study in Spain with 375 cases. Br J Dermatol. 2020;183:71-77.
- Garduño‑Soto M, Choreño-Parra, Cazarin-Barrientos Dermatological aspects of SARS‑CoV‑2 infection: mechanisms and manifestations. Arch Dermatol Res. 2021;313:611-622.
- Huynh T, Sanchez-Flores X, Yau J, et al. Cutaneous manifestations of SARS-CoV-2 Infection. Am J Clin Dermatol. 2022;23:277-286.
- Kanitakis J, Lesort C, Danset M, et al.
Chilblain-like acral lesions during the COVID-19 pandemic (“COVID toes”): histologic, immunofluorescence, and immunohistochemical study of 17 cases. J Am Acad Dermatol. 2020; 83:870-875. - Kolivras A, Thompson C, Pastushenko I, et al. A clinicopathological description of COVID-19-induced chilblains (COVID-toes) correlated with a published literature review. J Cutan Pathol. 2022;49:17-28.
- Roca-Ginés J, Torres-Navarro I, Sánchez-Arráez J, et al. Assessment of acute acral lesions in a case series of children and adolescents during the COVID-19 pandemic. 2020;156:992-997.
- Le Cleach L, Dousset L, Assier H, et al; French Society of Dermatology. Most chilblains observed during the COVID-19 outbreak occur in patients who are negative for COVID-19 on polymerase chain reaction and serology testing. Br J Dermatol. 2020;183:866-874.
- Discepolo V, Catzola A, Pierri L, et al. Bilateral chilblain-like lesions of the toes characterized by microvascular remodeling in adolescents during the COVID-19 pandemic. JAMA Netw Open. 2021;4:E2111369.
- Gehlhausen JR, Little AJ, Ko CJ, et al. Lack of association between pandemic chilblains and SARS-CoV-2 infection. Proc Natl Acad Sci U S A. 2022;119:e2122090119.
- Neri, Virdi, Corsini, et al Major cluster of paediatric ‘true’ primary chilblains during the COVID-19 pandemic: a consequence of lifestyle changes due to lockdown. J Eur Acad Dermatol Venereol. 2020;34:2630-2635.
- De Greef A, Choteau M, Herman A, et al. Chilblains observed during the COVID-19 pandemic cannot be distinguished from the classic, cold-related chilblains. Eur J Dermatol. 2022;32:377-383.
- Colmenero I, Santonja C, Alonso-Riaño M, et al. SARS-CoV-2 endothelial infection causes COVID-19 chilblains: histopathological, immunohistochemical and ultrastructural study of seven paediatric cases. Br J Dermatol. 2020;183:729-737.
- Quintero-Bustos G, Aguilar-Leon D, Saeb-Lima M. Histopathological and immunohistochemical characterization of skin biopsies from 41 SARS-CoV-2 (+) patients: experience in a Mexican concentration institute: a case series and literature review. Am J Dermatopathol. 2022;44:327-337.
- Arkin LM, Moon JJ, Tran JM, et al; COVID Human Genetic Effort. From your nose to your toes: a review of severe acute respiratory syndrome coronavirus 2 pandemic-associated pernio. J Invest Dermatol. 2021;141:2791-2796.
- Frumholtz L, Bouaziz JD, Battistella M, et al; Saint-Louis CORE (COvid REsearch). Type I interferon response and vascular alteration in chilblain-like lesions during the COVID-19 outbreak. Br J Dermatol. 2021;185:1176-1185.
- Hubiche T, Cardot-Leccia N, Le Duff F, et al. Clinical, laboratory, and interferon-alpha response characteristics of patients with chilblain-like lesions during the COVID-19 pandemic. JAMA Dermatol. 2021;157:202-206.
- Lesort C, Kanitakis J, Villani A, et al. COVID-19 and outbreak of chilblains: are they related? J Eur Acad Dermatol Venereol. 2020;34:E757-E758.
- Sanchez A, Sohier P, Benghanem S, et al. Digitate papulosquamous eruption associated with severe acute respiratory syndrome coronavirus 2 infection. JAMA Dermatol. 2020;156:819-820.
- Drago F, Broccolo F, Ciccarese G. Pityriasis rosea, pityriasis rosea-like eruptions, and herpes zoster in the setting of COVID-19 and COVID-19 vaccination. Clin Dermatol. 2022;S0738-081X(22)00002-5.
- Dursun R, Temiz SA. The clinics of HHV-6 infection in COVID-19 pandemic: pityriasis rosea and Kawasaki disease. Dermatol Ther. 2020;33:E13730.
- Nuno-Gonzalez A, Magaletsky K, Feito Rodríguez M, et al. Palmoplantar erythrodysesthesia: a diagnostic sign of COVID-19. J Eur Acad Dermatol Venereol. 2021;35:e247-e249.
- Sil A, Panigrahi A, Chandra A, et al. “COVID nose”: a unique post-COVID pigmentary sequelae reminiscing Chik sign: a descriptive case series. J Eur Acad Dermatol Venereol. 2022;36:E419-E421.
- Starace M, Iorizzo M, Sechi A, et al. Trichodynia and telogen effluvium in COVID-19 patients: results of an international expert opinion survey on diagnosis and management. JAAD Int. 2021;5:11-18.
- Wong-Chew RM, Rodríguez Cabrera EX, Rodríguez Valdez CA, et al. Symptom cluster analysis of long COVID-19 in patients discharged from the Temporary COVID-19 Hospital in Mexico City. Ther Adv Infect Dis. 2022;9:20499361211069264.
- Bardazzi F, Guglielmo A, Abbenante D, et al. New insights into alopecia areata during COVID-19 pandemic: when infection or vaccination could play a role. J Cosmet Dermatol. 2022;21:1796-1798.
- Christensen RE, Jafferany M. Association between alopecia areata and COVID-19: a systematic review. JAAD Int. 2022;7:57-61.
- Wollina U, Kanitakis J, Baran R. Nails and COVID-19: a comprehensive review of clinical findings and treatment. Dermatol Ther. 2021;34:E15100.
- Méndez-Flores S, Zaladonis A, Valdes-Rodriguez R. COVID-19 and nail manifestation: be on the lookout for the red half-moon nail sign. Int J Dermatol. 2020;59:1414.
- Alobaida S, Lam JM. Beau lines associated with COVID-19. CMAJ. 2020;192:E1040.
- Durmaz EÖ, Demirciog˘lu D. Fluorescence in the sclera, nails, and teeth secondary to favipiravir use for COVID-19 infections. J Clin Aesthet Dermatol. 2022;15:35-37.
- Brumfiel CM, DiLorenzo AM, Petronic-Rosic VM. Dermatologic manifestations of COVID-19-associated multisystem inflammatory syndrome in children. Clin Dermatol. 2021;39:329-333.
- Akçay N, Topkarcı Z, Menentog˘lu ME, et al. New dermatological findings of MIS-C: can mucocutaneous involvement be associated with severe disease course? Australas J Dermatol. 2022;63:228-234. doi:10.1111/ajd.13819
- Vogel TP, Top KA, Karatzios C, et al. Multisystem inflammatory syndrome in children and adults (MIS-C/A): case definition & guidelines for data collection, analysis, and presentation of immunization safety data. Vaccine. 2021;39:3037-3049.
- Conforti C, Dianzani C, Agozzino M, et al. Cutaneous manifestations in confirmed COVID-19 patients: a systematic review. Biology (Basel). 2020;9:449.
- Hubiche T, Le Duff F, Fontas E, et al. Relapse of chilblain-like lesions during the second wave of the COVID-19 pandemic: a cohort follow-up. Br J Dermatol. 2021;185:858-859.
- Fernandez-Nieto, Ortega-Quijano, Suarez-Valle, et al Lack of skin manifestations in COVID-19 hospitalized patients during the second epidemic wave in Spain: a possible association with a novel SARS-CoV-2 variant: a cross-sectional study. J Eur Acad Dermatol Venereol. 2021;35:E183-E185.
- Martinez-LopezA, Cuenca-Barrales, Montero-Vilchezet al Review of adverse cutaneous reactions of pharmacologic interventions for COVID-19: a guide for the dermatologist. J Am Acad Dermatol. 2020;83:1738-1748.
- Cutaneous side-effects of the potential COVID-19 drugs. Dermatol Ther. 2020;33:E13476.
- Mawhirt SL, Frankel D, Diaz AM. Cutaneous manifestations in adult patients with COVID-19 and dermatologic conditions related to the COVID-19 pandemic in health care workers. Curr Allerg Asthma Rep. 2020;20:75.
- Nguyen C, Young FG, McElroy D, et al. Personal protective equipment and adverse dermatological reactions among healthcare workers: survey observations from the COVID-19 pandemic. Medicine (Baltimore). 2022;101:E29003.
- Rathi SK, Dsouza JM. Maskne: a new acne variant in COVID-19 era. Indian J Dermatol. 2022;67:552-555.
- Damiani G, Girono L, Grada A, et al. COVID-19 related masks increase severity of both acne (maskne) and rosacea (mask rosacea): multi-center, real-life, telemedical, and observational prospective study. Dermatol Ther. 2021;34:E14848.
- Aram K, Patil A, Goldust M, et al. COVID-19 and exacerbation of dermatological diseases: a review of the available literature. Dermatol Ther. 2021;34:E15113.
- Samotij D, Gawron E, Szcze˛ch J, et al. Acrodermatitis continua of Hallopeau evolving into generalized pustular psoriasis following COVID-19: a case report of a successful treatment with infliximab in combination with acitretin. Biologics. 2021;15:107-113.
- Demiri J, Abdo M, Tsianakas A. Erythrodermic psoriasis after COVID-19 [in German]. Hautarzt. 2022;73:156-159.
- de Wijs LEM, Joustra MM, Olydam JI, et al. COVID-19 in patients with cutaneous immune-mediated diseases in the Netherlands: real-world observational data. J Eur Acad Dermatol Venereol. 2021;35:E173-E176.
- Marques NP, Maia CMF, Marques NCT, et al. Continuous increase of herpes zoster cases in Brazil during the COVID-19 pandemic. Oral Surg Oral Med Oral Pathol Oral Radiol. 2022;133:612-614.
- Rinaldi F, Trink A, Giuliani G, et al. Italian survey for the evaluation of the effects of coronavirus disease 2019 (COVID-19) pandemic on alopecia areata recurrence. Dermatol Ther (Heidelb). 2021;11:339-345.
- Rudnicka L, Rakowska A, Waskiel-Burnat A, et al. Mild-to-moderate COVID-19 is not associated with worsening of alopecia areata: a retrospective analysis of 32 patients. J Am Acad Dermatol. 2021;85:723-725.
- Drenovska K, Shahid M, Mateeva V, et al. Case report: Rowell syndrome-like flare of cutaneous lupus erythematosus following COVID-19 infection. Front Med (Lausanne). 2022;9:815743.
- Kawabe R, Tonomura K, Kotobuki Y, et al. Exacerbation of livedoid vasculopathy after coronavirus disease 2019. Eur J Dermatol. 2022;32:129-131. doi:10.1684/ejd.2022.4200
- McMahon DE, Kovarik CL, Damsky W, et al. Clinical and pathologic correlation of cutaneous COVID-19 vaccine reactions including V-REPP: a registry-based study. J Am Acad Dermatol. 2022;86:113-121.
- Avallone G, Quaglino P, Cavallo F, et al. SARS-CoV-2 vaccine-related cutaneous manifestations: a systematic review. Int J Dermatol. 2022;61:1187-1204. doi:10.1111/ijd.16063
- Gambichler T, Boms S, Susok L, et al. Cutaneous findings following COVID-19 vaccination: review of world literature and own experience. J Eur Acad Dermatol Venereol. 2022;36:172-180.
- Kroumpouzos G, Paroikaki ME, Yumeen S, et al. Cutaneous complications of mRNA and AZD1222 COVID-19 vaccines: a worldwide review. Microorganisms. 2022;10:624.
- Robinson L,Fu X,Hashimoto D, et al. Incidence of cutaneous reactions after messenger RNA COVID-19 vaccines. 2021;
- Wollina U, Chiriac A, Kocic H, et al. Cutaneous and hypersensitivity reactions associated with COVID-19 vaccination: a narrative review. Wien Med Wochenschr. 2022;172:63-69.
- Wei TS. Cutaneous reactions to COVID-19 vaccines: a review. JAAD Int. 2022;7:178-186.
- Katsikas Triantafyllidis K, Giannos P, Mian IT, et al. Varicella zoster virus reactivation following COVID-19 vaccination: a systematic review of case reports. Vaccines (Basel). 2021;9:1013.
- Maronese CA, Caproni M, Moltrasio C, et al. Bullous pemphigoid associated with COVID-19 vaccines: an Italian multicentre study. Front Med (Lausanne). 2022;9:841506.
- Cavazos A, Deb A, Sharma U, et al. COVID toes following vaccination. Proc (Bayl Univ Med Cent). 2022;35:476-479.
- Lesort C, Kanitakis J, Danset M, et al. Chilblain-like lesions after BNT162b2 mRNA COVID-19 vaccine: a case report suggesting that ‘COVID toes’ are due to the immune reaction to SARS-CoV-2. J Eur Acad Dermatol Venereol. 2021;35:E630-E632.
- Russo R, Cozzani E, Micalizzi C, et al. Chilblain-like lesions after COVID-19 vaccination: a case series. Acta Derm Venereol. 2022;102:adv00711. doi:10.2340/actadv.v102.2076
- Ortigosa LCM, Lenzoni FC, Suárez MV, et al. Hypersensitivity reaction to hyaluronic acid dermal filler after COVID-19 vaccination: a series of cases in São Paulo, Brazil. Int J Infect Dis. 2022;116:268-270.
- Agaronov A, Makdesi C, Hall CS. Acute generalized exanthematous pustulosis induced by Moderna COVID-19 messenger RNA vaccine. JAAD Case Rep. 2021;16:96-97.
- Dash S, Sirka CS, Mishra S, et al. COVID-19 vaccine-induced Stevens-Johnson syndrome. Clin Exp Dermatol. 2021;46:1615-1617.
- Huang Y, Tsai TF. Exacerbation of psoriasis following COVID-19 vaccination: report from a single center. Front Med (Lausanne). 2021;8:812010.
- Elamin S, Hinds F, Tolland J. De novo generalized pustular psoriasis following Oxford-AstraZeneca COVID-19 vaccine. Clin Exp Dermatol 2022;47:153-155.
- Abdelmaksoud A, Wollina U, Temiz SA, et al. SARS-CoV-2 vaccination-induced cutaneous vasculitis: report of two new cases and literature review. Dermatol Ther. 2022;35:E15458.
- Fritzen M, Funchal GDG, Luiz MO, et al. Leukocytoclastic vasculitis after exposure to COVID-19 vaccine. An Bras Dermatol. 2022;97:118-121.
- Vassallo C, Boveri E, Brazzelli V, et al. Cutaneous lymphocytic vasculitis after administration of COVID-19 mRNA vaccine. Dermatol Ther. 2021;34:E15076.
- Nazzaro G, Maronese CA. Urticarial vasculitis following mRNA anti-COVID-19 vaccine. Dermatol Ther. 2022;35:E15282.
- Hoshina D, Orita A. Sweet syndrome after severe acute respiratory syndrome coronavirus 2 mRNA vaccine: a case report and literature review. J Dermatol. 2022;49:E175-E176.
- Lemoine C, Padilla C, Krampe N, et al. Systemic lupus erythematous after Pfizer COVID-19 vaccine: a case report. Clin Rheumatol. 2022;41:1597-1601.
- Nguyen B, Lalama MJ, Gamret AC, et al. Cutaneous symptoms of connective tissue diseases after COVID-19 vaccination: a systematic review. Int J Dermatol. 2022;61:E238-E241.
- Gallo G, Mastorino L, Tonella L, et al. Alopecia areata after COVID-19 vaccination. Clin Exp Vaccine Res. 2022;11:129-132.
- Norimatsu Y, Norimatsu Y. A severe case of Trichophyton rubrum-caused dermatomycosis exacerbated after COVID-19 vaccination that had to be differentiated from pustular psoriasis. Med Mycol Case Rep. 2022;36:19-22.
- Yang K, Prussick L, Hartman R, et al. Acantholytic dyskeratosis post-COVID vaccination. Am J Dermatopathol. 2022;44:E61-E63.
- Koumaki D, Marinos L, Nikolaou V, et al. Lymphomatoid papulosis (LyP) after AZD1222 and BNT162b2 COVID-19 vaccines. Int J Dermatol. 2022;61:900-902.
- World Health Organization. Statement on the fourteenth meeting of the International Health Regulations (2005) Emergency Committee regarding the coronavirus disease (COVID-19) pandemic. Published January 30, 2023. Accessed April 12, 2023. https://www.who.int/news/item/30-01-2023-statement-on-the-fourteenth-meeting-of-the-international-health-regulations-(2005)-emergency-committee-regarding-the-coronavirus-disease-(covid-19)-pandemic
COVID-19 is a potentially severe systemic disease caused by SARS-CoV-2. SARS-CoV and Middle East respiratory syndrome (MERS-CoV) caused fatal epidemics in Asia in 2002 to 2003 and in the Arabian Peninsula in 2012, respectively. In 2019, SARS-CoV-2 was detected in patients with severe, sometimes fatal pneumonia of previously unknown origin; it rapidly spread around the world, and the World Health Organization declared the disease a pandemic on March 11, 2020. SARS-CoV-2 is a β-coronavirus that is genetically related to the bat coronavirus and SARS-CoV; it is a single-stranded RNA virus of which several variants and subvariants exist. The SARS-CoV-2 viral particles bind via their surface spike protein (S protein) to the angiotensin-converting enzyme 2 receptor present on the membrane of several cell types, including epidermal and adnexal keratinocytes.1,2 The α and δ variants, predominant from 2020 to 2021, mainly affected the lower respiratory tract and caused severe, potentially fatal pneumonia, especially in patients older than 65 years and/or with comorbidities, such as obesity, hypertension, diabetes, and (iatrogenic) immunosuppression. The ο variant, which appeared in late 2021, is more contagious than the initial variants, but it causes a less severe disease preferentially affecting the upper respiratory airways.3 As of April 5, 2023, more than 762,000,000 confirmed cases of COVID-19 have been recorded worldwide, causing more than 6,800,000 deaths.4
Early studies from China describing the symptoms of COVID-19 reported a low frequency of skin manifestations (0.2%), probably because they were focused on the most severe disease symptoms.5 Subsequently, when COVID-19 spread to the rest of the world, an increasing number of skin manifestations were reported in association with the disease. After the first publication from northern Italy in spring 2020, which was specifically devoted to skin manifestations of COVID-19,6 an explosive number of publications reported a large number of skin manifestations, and national registries were established in several countries to record these manifestations, such as the American Academy of Dermatology and the International League of Dermatological Societies registry,7,8 the COVIDSKIN registry of the French Dermatology Society,9 and the Italian registry.10 Highlighting the unprecedented number of scientific articles published on this new disease, a PubMed search of articles indexed for MEDLINE search using the terms SARS-CoV-2 or COVID-19, on April 6, 2023, revealed 351,596 articles; that is more than 300 articles published every day in this database alone, with a large number of them concerning the skin.
SKIN DISEASSES ASSOCIATED WITH COVID-19
There are several types of COVID-19–related skin manifestations, depending on the circumstances of onset and the evolution of the pandemic.
Skin Manifestations Associated With SARS-CoV-2 Infection
The estimated incidence varies greatly according to the published series of patients, possibly depending on the geographic location. The estimated incidence seems lower in Asian countries, such as China (0.2%)5 and Japan (0.56%),11 compared with Europe (up to 20%).6 Skin manifestations associated with SARS-CoV-2 infection affect individuals of all ages, slightly more females, and are clinically polymorphous; some of them are associated with the severity of the infection.12 They may precede, accompany, or appear after the symptoms of COVID-19, most often within a month of the infection, of which they rarely are the only manifestation; however, their precise relationship to SARS-CoV-2 is not always well known. They have been classified according to their clinical presentation into several forms.13-15
Morbilliform Maculopapular Eruption—Representing 16% to 53% of skin manifestations, morbilliform and maculopapular eruptions usually appear within 15 days of infection; they manifest with more or less confluent erythematous macules that may be hemorrhagic/petechial, and usually are asymptomatic and rarely pruritic. The rash mainly affects the trunk and limbs, sparing the face, palmoplantar regions, and mucous membranes; it appears concomitantly with or a few days after the first symptoms of COVID-19 (eg, fever, respiratory symptoms), regresses within a few days, and does not appear to be associated with disease severity. The distinction from maculopapular drug eruptions may be subtle. Histologically, the rash manifests with a spongiform dermatitis (ie, variable parakeratosis; spongiosis; and a mixed dermal perivascular infiltrate of lymphocytes, eosinophils and histiocytes, depending on the lesion age)(Figure 1). The etiopathogenesis is unknown; it may involve immune complexes to SARS-CoV-2 deposited on skin vessels. Treatment is not mandatory; if necessary, local or systemic corticosteroids may be used.
Vesicular (Pseudovaricella) Rash—This rash accounts for 11% to 18% of all skin manifestations and usually appears within 15 days of COVID-19 onset. It manifests with small monomorphous or varicellalike (pseudopolymorphic) vesicles appearing on the trunk, usually in young patients. The vesicles may be herpetiform, hemorrhagic, or pruritic, and appear before or within 3 days of the onset of mild COVID-19 symptoms; they regress within a few days without scarring. Histologically, the lesions show basal cell vacuolization; multinucleated, dyskeratotic/apoptotic or ballooning/acantholytic epidermal keratinocytes; reticular degeneration of the epidermis; intraepidermal vesicles sometimes resembling herpetic vesicular infections or Grover disease; and mild dermal inflammation. There is no specific treatment.
Urticaria—Urticarial rash, or urticaria, represents 5% to 16% of skin manifestations; usually appears within 15 days of disease onset; and manifests with pruritic, migratory, edematous papules appearing mainly on the trunk and occasionally the face and limbs. The urticarial rash tends to be associated with more severe forms of the disease and regresses within a week, responding to antihistamines. Of note, clinically similar rashes can be caused by drugs. Histologically, the lesions show dermal edema and a mild perivascular lymphocytic infiltrate, sometimes admixed with eosinophils.
Chilblainlike Lesions—Chilblainlike lesions (CBLLs) account for 19% of skin manifestations associated with COVID-1913 and present as erythematous-purplish, edematous lesions that can be mildly pruritic or painful, appearing on the toes—COVID toes—and more rarely the fingers (Figure 2). They were seen epidemically during the first pandemic wave (2020 lockdown) in several countries, and clinically are very similar to, if not indistinguishable from, idiopathic chilblains, but are not necessarily associated with cold exposure. They appear in young, generally healthy patients or those with mild COVID-19 symptoms 2 to 4 weeks after symptom onset. They regress spontaneously or under local corticosteroid treatment within a few days or weeks. Histologically, CBLLs are indistinguishable from chilblains of other origins, namely idiopathic (seasonal) ones. They manifest with necrosis of epidermal keratinocytes; dermal edema that may be severe, leading to the development of subepidermal pseudobullae; a rather dense perivascular and perieccrine gland lymphocytic infiltrate; and sometimes with vascular lesions (eg, edema of endothelial cells, microthromboses of dermal capillaries and venules, fibrinoid deposits within the wall of dermal venules)(Figure 3).16-18 Most patients (>80%) with CBLLs have negative serologic or polymerase chain reaction tests for SARS-CoV-2,19 which generated a lively debate about the role of SARS-CoV-2 in the genesis of CBLLs. According to some authors, SARS-CoV-2 plays no direct role, and CBLLs would occur in young people who sit or walk barefoot on cold floors at home during confinement.20-23 Remarkably, CBLLs appeared in patients with no history of chilblains during a season that was not particularly cold, namely in France or in southern California, where their incidence was much higher compared to the same time period of prior years. Some reports have supported a direct role for the virus based on questionable observations of the virus within skin lesions (eg, sweat glands, endothelial cells) by immunohistochemistry, electron microscopy, and/or in situ hybridization.17,24,25 A more satisfactory hypothesis would involve the role of a strong innate immunity leading to elimination of the virus before the development of specific antibodies via the increased production of type 1 interferon (IFN-1); this would affect the vessels, causing CBLLs. This mechanism would be similar to the one observed in some interferonopathies (eg, Aicardi-Goutières syndrome), also characterized by IFN-1 hypersecretion and chilblains.26-29 According to this hypothesis, CBLLs should be considered a paraviral rash similar to other skin manifestations associated with COVID-19.30
Acro-ischemia—Acro-ischemia livedoid lesions account for 1% to 6% of skin manifestations and comprise lesions of livedo (either reticulated or racemosa); necrotic acral bullae; and gangrenous necrosis of the extremities, especially the toes. The livedoid lesions most often appear within 15 days of COVID-19 symptom onset, and the purpuric lesions somewhat later (2–4 weeks); they mainly affect adult patients, last about 10 days, and are the hallmark of severe infection, presumably related to microthromboses of the cutaneous capillaries (endothelial dysfunction, prothrombotic state, elevated D-dimers). Histologically, they show capillary thrombosis and dermoepidermal necrosis (Figure 4).
Other Reported Polymorphic or Atypical Rashes—Erythema multiforme–like eruptions may appear before other COVID-19 symptoms and manifest as reddish-purple, nearly symmetric, diffuse, occasionally targetoid bullous or necrotic macules. The eruptions mainly affect adults and most often are seen on the palms, elbows, knees, and sometimes the mucous membranes. The rash regresses in 1 to 3 weeks without scarring and represents a delayed cutaneous hypersensitivity reaction. Histologically, the lesions show vacuolization of basal epidermal keratinocytes, keratinocyte necrosis, dermoepidermal detachment, a variably dense dermal T-lymphocytic infiltrate, and red blood cell extravasation (Figure 5).
Leukocytoclastic vasculitis may be generalized or localized. It manifests clinically by petechial/purpuric maculopapules, especially on the legs, mainly in elderly patients with COVID-19. Histologically, the lesions show necrotizing changes of dermal postcapillary venules, neutrophilic perivascular inflammation, red blood cell extravasation, and occasionally vascular IgA deposits by direct immunofluorescence examination. The course usually is benign.
The incidence of pityriasis rosea and of clinically similar rashes (referred to as “pityriasis rosea–like”) increased 5-fold during the COVID-19 pandemic.31,32 These dermatoses manifest with erythematous, scaly, circinate plaques, typically with an initial herald lesion followed a few days later by smaller erythematous macules. Histologically, the lesions comprise a spongiform dermatitis with intraepidermal exocytosis of red blood cells and a mild to moderate dermal lymphocytic infiltrate.
Erythrodysesthesia, or hand-foot syndrome, manifests with edematous erythema and palmoplantar desquamation accompanied by a burning sensation or pain. This syndrome is known as an adverse effect of some chemotherapies because of the associated drug toxicity and sweat gland inflammation; it was observed in 40% of 666 COVID-19–positive patients with mild to moderate pneumonitis.33
“COVID nose” is a rare cutaneous manifestation characterized by nasal pigmentation comprising multiple coalescent frecklelike macules on the tip and wings of the nose and sometimes the malar areas. These lesions predominantly appear in women aged 25 to 65 years and show on average 23 days after onset of COVID-19, which is usually mild. This pigmentation is similar to pigmentary changes after infection with chikungunya; it can be treated with depigmenting products such as azelaic acid and hydroquinone cream with sunscreen use, and it regresses in 2 to 4 months.34
Telogen effluvium (excessive and temporary shedding of normal telogen club hairs of the entire scalp due to the disturbance of the hair cycle) is reportedly frequent in patients (48%) 1 month after COVID-19 infection, but it may appear later (after 12 weeks).35 Alopecia also is frequently reported during long (or postacute) COVID-19 (ie, the symptomatic disease phase past the acute 4 weeks’ stage of the infection) and shows a female predominance36; it likely represents the telogen effluvium seen 90 days after a severe illness. Trichodynia (pruritus, burning, pain, or paresthesia of the scalp) also is reportedly common (developing in more than 58% of patients) and is associated with telogen effluvium in 44% of cases. Several cases of alopecia areata (AA) triggered or aggravated by COVID-19 also have been reported37,38; they could be explained by the “cytokine storm” triggered by the infection, involving T and B lymphocytes; plasmacytoid dendritic cells; natural killer cells with oversecretion of IL-6, IL-4, tumor necrosis factor α, and IFN type I; and a cytotoxic reaction associated with loss of the immune privilege of hair follicles.
Nail Manifestations
The red half-moon nail sign is an asymptomatic purplish-red band around the distal margin of the lunula that affects some adult patients with COVID-19.39 It appears shortly after onset of symptoms, likely the manifestation of vascular inflammation in the nail bed, and regresses slowly after approximately 1 week.40 Beau lines are transverse grooves in the nail plate due to the temporary arrest of the proximal nail matrix growth accompanying systemic illnesses; they appear approximately 2 to 3 weeks after the onset of COVID-19.41 Furthermore, nail alterations can be caused by drugs used to treat COVID-19, such as longitudinal melanonychia due to treatment with hydroxychloroquine or fluorescence of the lunula or nail plate due to treatment with favipiravir.42
Multisystem Inflammatory Syndrome
Multisystem inflammatory syndrome (MIS) is clinically similar to Kawasaki disease; it typically affects children43 and more rarely adults with COVID-19. It manifests with fever, weakness, and biological inflammation and also frequently with skin lesions (72%), which are polymorphous and include morbilliform rash (27%); urticaria (24%); periorbital edema (24%); nonspecific erythema (21.2%); retiform purpura (18%); targetoid lesions (15%); malar rash (15.2%); and periareolar erythema (6%).44 Compared to Kawasaki disease, MIS affects slightly older children (mean age, 8.5 vs 3 years) and more frequently includes cardiac and gastrointestinal manifestations; the mortality rate also is slightly higher (2% vs 0.17%).45
Confirmed COVID-19 Infection
At the beginning of the pandemic, skin manifestations were reported in patients who were suspected of having COVID-19 but did not always have biological confirmation of SARS-CoV-2 infection due to the unavailability of diagnostic tests or the physical impossibility of testing. However, subsequent studies have confirmed that most of these dermatoses were indeed associated with COVID-19 infection.9,46 For example, a study of 655 patients with confirmed COVID-19 infection reported maculopapular (38%), vascular (22%), urticarial (15%), and vesicular (15%) rashes; erythema multiforme or Stevens-Johnson–like syndrome (3%, often related to the use of hydroxychloroquine); generalized pruritus (1%); and MIS (0.5%). The study confirmed that CBLLs were mostly seen in young patients with mild disease, whereas livedo (fixed rash) and retiform purpura occurred in older patients with a guarded prognosis.46
Remarkably, most dermatoses associated with SARS-CoV-2 infection were reported during the initial waves of the pandemic, which were due to the α and δ viral variants. These manifestations were reported more rarely when the ο variant was predominant, even though most patients (63%) who developed CBLLs in the first wave also developed them during the second pandemic wave.47 This decrease in the incidence of COVID-19–associated dermatoses could be because of the lower pathogenicity of the o variant,3 a lower tropism for the skin, and variations in SARS-CoV-2 antigenicity that would induce a different immunologic response, combined with an increasingly stronger herd immunity compared to the first pandemic waves achieved through vaccination and spontaneous infections in the population. Additional reasons may include different baseline characteristics in patients hospitalized with COVID-19 (regarding comorbidities, disease severity, and received treatments), and the possibility that some of the initially reported COVID-19–associated skin manifestations could have been produced by different etiologic agents.48 In the last 2 years, COVID-19–related skin manifestations have been reported mainly as adverse events to COVID-19 vaccination.
CUTANEOUS ADVERSE EFFECTS OF DRUGS USED TO TREAT COVID-19
Prior to the advent of vaccines and specific treatments for SARS-CoV-2, various drugs were used—namely hydroxychloroquine, ivermectin, and tocilizumab—that did not prove efficacious and caused diverse adverse effects, including cutaneous eruptions such as urticaria, maculopapular eruptions, erythema multiforme or Stevens-Johnson syndrome, vasculitis, longitudinal melanonychia, and acute generalized exanthematous pustulosis.49,50 Nirmatrelvir 150 mg–ritonavir 100 mg, which was authorized for emergency use by the US Food and Drug Administration for the treatment of COVID-19, is a viral protease inhibitor blocking the replication of the virus. Ritonavir can induce pruritus, maculopapular rash, acne, Stevens-Johnson syndrome, and toxic epidermal necrolysis; of note, these effects have been observed following administration of ritonavir for treatment of HIV at higher daily doses and for much longer periods of time compared with treatment of COVID-19 (600–1200 mg vs 200 mg/d, respectively). These cutaneous drug side effects are clinically similar to the manifestations caused either directly or indirectly by SARS-CoV-2 infection; therefore, it may be difficult to differentiate them.
DERMATOSES DUE TO PROTECTIVE DEVICES
Dermatoses due to personal protective equipment such as masks or face shields affected the general population and mostly health care professionals51; 54.4% of 879 health care professionals in one study reported such events.52 These dermatoses mainly include contact dermatitis of the face (nose, forehead, and cheeks) of irritant or allergic nature (eg, from preservatives releasing formaldehyde contained in masks and protective goggles). They manifest with skin dryness; desquamation; maceration; fissures; or erosions or ulcerations of the cheeks, forehead, and nose. Cases of pressure urticaria also have been reported. Irritant dermatitis induced by the frequent use of disinfectants (eg, soaps, hydroalcoholic sanitizing gels) also can affect the hands. Allergic hand dermatitis can be caused by medical gloves.
The term maskne (or mask acne) refers to a variety of mechanical acne due to the prolonged use of surgical masks (>4 hours per day for ≥6 weeks); it includes cases of de novo acne and cases of pre-existing acne aggravated by wearing a mask. Maskne is characterized by acne lesions located on the facial area covered by the mask (Figure 6). It is caused by follicular occlusion; increased sebum secretion; mechanical stress (pressure, friction); and dysbiosis of the microbiome induced by changes in heat, pH, and humidity. Preventive measures include application of noncomedogenic moisturizers or gauze before wearing the mask as well as facial cleansing with appropriate nonalcoholic products. Similar to acne, rosacea often is aggravated by prolonged wearing of surgical masks (mask rosacea).53,54
DERMATOSES REVEALED OR AGGRAVATED BY COVID-19
Exacerbation of various skin diseases has been reported after infection with SARS-CoV-2.55 Psoriasis and acrodermatitis continua of Hallopeau,56 which may progress into generalized, pustular, or erythrodermic forms,57 have been reported; the role of hydroxychloroquine and oral corticosteroids used for the treatment of COVID-19 has been suspected.57 Atopic dermatitis patients—26% to 43%—have experienced worsening of their disease after symptomatic COVID-19 infection.58 The incidence of herpesvirus infections, including herpes zoster, increased during the pandemic.59 Alopecia areata relapses occurred in 42.5% of 392 patients with preexisting disease within 2 months of COVID-19 onset in one study,60 possibly favored by the psychological stress; however, some studies have not confirmed the aggravating role of COVID-19 on alopecia areata.61 Lupus erythematosus, which may relapse in the form of Rowell syndrome,62 and livedoid vasculopathy63 also have been reported following COVID-19 infection.
SKIN MANIFESTATIONS ASSOCIATED WITH COVID-19 VACCINES
In parallel with the rapid spread of COVID-19 vaccination,4 an increasing number of skin manifestations has been observed following vaccination; these dermatoses now are more frequently reported than those related to natural SARS-CoV-2 infection.64-70 Vaccine-induced skin manifestations have a reported incidence of approximately 4% and show a female predominance.65 Most of them (79%) have been reported in association with messenger RNA (mRNA)–based vaccines, which have been the most widely used; however, the frequency of side effects would be lower after mRNA vaccines than after inactivated virus-based vaccines. Eighteen percent occurred after the adenoviral vector vaccine, and 3% after the inactivated virus vaccine.70 Fifty-nine percent were observed after the first dose. They are clinically polymorphous and generally benign, regressing spontaneously after a few days, and they should not constitute a contraindication to vaccination.Interestingly, many skin manifestations are similar to those associated with natural SARS-CoV-2 infection; however, their frequency and severity does not seem to depend on whether the patients had developed skin reactions during prior SARS-CoV-2 infection. These reactions have been classified into several types:
• Immediate local reactions at the injection site: pain, erythema, or edema represent the vast majority (96%) of reactions to vaccines. They appear within 7 days after vaccination (average, 1 day), slightly more frequently (59%) after the first dose. They concern mostly young patients and are benign, regressing in 2 to 3 days.70
• Delayed local reactions: characterized by pain or pruritus, erythema, and skin induration mimicking cellulitis (COVID arm) and represent 1.7% of postvaccination reactions. They correspond to a delayed hypersensitivity reaction and appear approximately 7 days after vaccination, most often after the first vaccine dose (75% of cases), which is almost invariably mRNA based.70
• Urticarial reactions corresponding to an immediate (type 1) hypersensitivity reaction: constitute 1% of postvaccination reactions, probably due to an allergy to vaccine ingredients. They appear on average 1 day after vaccination, almost always with mRNA vaccines.70
• Angioedema: characterized by mucosal or subcutaneous edema and constitutes 0.5% of postvaccination reactions. It is a potentially serious reaction that appears on average 12 hours after vaccination, always with an mRNA-based vaccine.70
• Morbilliform rash: represents delayed hypersensitivity reactions (0.1% of postvaccination reactions) that appear mostly after the first dose (72%), on average 3 days after vaccination, always with an mRNA-based vaccine.70
• Herpes zoster: usually develops after the first vaccine dose in elderly patients (69% of cases) on average 4 days after vaccination and constitutes 0.1% of postvaccination reactions.71
• Bullous diseases: mainly bullous pemphigoid (90%) and more rarely pemphigus (5%) or bullous erythema pigmentosum (5%). They appear in elderly patients on average 7 days after vaccination and constitute 0.04% of postvaccination reactions.72
• Chilblainlike lesions: several such cases have been reported so far73; they constitute 0.03% of postvaccination reactions.70 Clinically, they are similar to those associated with natural COVID-19; they appear mostly after the first dose (64%), on average 5 days after vaccination with the mRNA or adenovirus vaccine, and show a female predominance. The appearance of these lesions in vaccinated patients, who are a priori not carriers of the virus, strongly suggests that CBLLs are due to the immune reaction against SARS-CoV-2 rather than to a direct effect of this virus on the skin, which also is a likely scenario with regards to other skin manifestations seen during the successive COVID-19 epidemic waves.73-75
• Reactions to hyaluronic acid–containing cosmetic fillers: erythema, edema, and potentially painful induration at the filler injection sites. They constitute 0.04% of postvaccination skin reactions and appear 24 hours after vaccination with mRNA-based vaccines, equally after the first or second dose.76
• Pityriasis rosea–like rash: most occur after the second dose of mRNA-based vaccines (0.023% of postvaccination skin reactions).70
• Severe reactions: these include acute generalized exanthematous pustulosis77 and Stevens-Johnson syndrome.78 One case of each has been reported after the adenoviral vector vaccine 3 days after vaccination.
Other more rarely observed manifestations include reactivation/aggravation or de novo appearance of inflammatory dermatoses such as psoriasis,79,80 leukocytoclastic vasculitis,81,82 lymphocytic83 or urticarial84 vasculitis, Sweet syndrome,85 lupus erythematosus, dermatomyositis,86,87 alopecia,37,88 infection with Trichophyton rubrum,89 Grover disease,90 and lymphomatoid reactions (such as recurrences of cutaneous T-cell lymphomas [CD30+], and de novo development of lymphomatoid papulosis).91
FINAL THOUGHTS
COVID-19 is associated with several skin manifestations, even though the causative role of SARS-CoV-2 has remained elusive. These dermatoses are highly polymorphous, mostly benign, and usually spontaneously regressive, but some of them reflect severe infection. They mostly were described during the first pandemic waves, reported in several national and international registries, which allowed for their morphological classification. Currently, cutaneous adverse effects of vaccines are the most frequently reported dermatoses associated with SARS-CoV-2, and it is likely that they will continue to be observed while COVID-19 vaccination lasts. Hopefully the end of the COVID-19 pandemic is near. In January 2023, the International Health Regulations Emergency Committee of the World Health Organization acknowledged that the COVID-19 pandemic may be approaching an inflexion point, and even though the event continues to constitute a public health emergency of international concern, the higher levels of population immunity achieved globally through infection and/or vaccination may limit the impact of SARS-CoV-2 on morbidity and mortality. However, there is little doubt that this virus will remain a permanently established pathogen in humans and animals for the foreseeable future.92 Therefore, physicians—especially dermatologists—should be aware of the various skin manifestations associated with COVID-19 so they can more efficiently manage their patients.
COVID-19 is a potentially severe systemic disease caused by SARS-CoV-2. SARS-CoV and Middle East respiratory syndrome (MERS-CoV) caused fatal epidemics in Asia in 2002 to 2003 and in the Arabian Peninsula in 2012, respectively. In 2019, SARS-CoV-2 was detected in patients with severe, sometimes fatal pneumonia of previously unknown origin; it rapidly spread around the world, and the World Health Organization declared the disease a pandemic on March 11, 2020. SARS-CoV-2 is a β-coronavirus that is genetically related to the bat coronavirus and SARS-CoV; it is a single-stranded RNA virus of which several variants and subvariants exist. The SARS-CoV-2 viral particles bind via their surface spike protein (S protein) to the angiotensin-converting enzyme 2 receptor present on the membrane of several cell types, including epidermal and adnexal keratinocytes.1,2 The α and δ variants, predominant from 2020 to 2021, mainly affected the lower respiratory tract and caused severe, potentially fatal pneumonia, especially in patients older than 65 years and/or with comorbidities, such as obesity, hypertension, diabetes, and (iatrogenic) immunosuppression. The ο variant, which appeared in late 2021, is more contagious than the initial variants, but it causes a less severe disease preferentially affecting the upper respiratory airways.3 As of April 5, 2023, more than 762,000,000 confirmed cases of COVID-19 have been recorded worldwide, causing more than 6,800,000 deaths.4
Early studies from China describing the symptoms of COVID-19 reported a low frequency of skin manifestations (0.2%), probably because they were focused on the most severe disease symptoms.5 Subsequently, when COVID-19 spread to the rest of the world, an increasing number of skin manifestations were reported in association with the disease. After the first publication from northern Italy in spring 2020, which was specifically devoted to skin manifestations of COVID-19,6 an explosive number of publications reported a large number of skin manifestations, and national registries were established in several countries to record these manifestations, such as the American Academy of Dermatology and the International League of Dermatological Societies registry,7,8 the COVIDSKIN registry of the French Dermatology Society,9 and the Italian registry.10 Highlighting the unprecedented number of scientific articles published on this new disease, a PubMed search of articles indexed for MEDLINE search using the terms SARS-CoV-2 or COVID-19, on April 6, 2023, revealed 351,596 articles; that is more than 300 articles published every day in this database alone, with a large number of them concerning the skin.
SKIN DISEASSES ASSOCIATED WITH COVID-19
There are several types of COVID-19–related skin manifestations, depending on the circumstances of onset and the evolution of the pandemic.
Skin Manifestations Associated With SARS-CoV-2 Infection
The estimated incidence varies greatly according to the published series of patients, possibly depending on the geographic location. The estimated incidence seems lower in Asian countries, such as China (0.2%)5 and Japan (0.56%),11 compared with Europe (up to 20%).6 Skin manifestations associated with SARS-CoV-2 infection affect individuals of all ages, slightly more females, and are clinically polymorphous; some of them are associated with the severity of the infection.12 They may precede, accompany, or appear after the symptoms of COVID-19, most often within a month of the infection, of which they rarely are the only manifestation; however, their precise relationship to SARS-CoV-2 is not always well known. They have been classified according to their clinical presentation into several forms.13-15
Morbilliform Maculopapular Eruption—Representing 16% to 53% of skin manifestations, morbilliform and maculopapular eruptions usually appear within 15 days of infection; they manifest with more or less confluent erythematous macules that may be hemorrhagic/petechial, and usually are asymptomatic and rarely pruritic. The rash mainly affects the trunk and limbs, sparing the face, palmoplantar regions, and mucous membranes; it appears concomitantly with or a few days after the first symptoms of COVID-19 (eg, fever, respiratory symptoms), regresses within a few days, and does not appear to be associated with disease severity. The distinction from maculopapular drug eruptions may be subtle. Histologically, the rash manifests with a spongiform dermatitis (ie, variable parakeratosis; spongiosis; and a mixed dermal perivascular infiltrate of lymphocytes, eosinophils and histiocytes, depending on the lesion age)(Figure 1). The etiopathogenesis is unknown; it may involve immune complexes to SARS-CoV-2 deposited on skin vessels. Treatment is not mandatory; if necessary, local or systemic corticosteroids may be used.
Vesicular (Pseudovaricella) Rash—This rash accounts for 11% to 18% of all skin manifestations and usually appears within 15 days of COVID-19 onset. It manifests with small monomorphous or varicellalike (pseudopolymorphic) vesicles appearing on the trunk, usually in young patients. The vesicles may be herpetiform, hemorrhagic, or pruritic, and appear before or within 3 days of the onset of mild COVID-19 symptoms; they regress within a few days without scarring. Histologically, the lesions show basal cell vacuolization; multinucleated, dyskeratotic/apoptotic or ballooning/acantholytic epidermal keratinocytes; reticular degeneration of the epidermis; intraepidermal vesicles sometimes resembling herpetic vesicular infections or Grover disease; and mild dermal inflammation. There is no specific treatment.
Urticaria—Urticarial rash, or urticaria, represents 5% to 16% of skin manifestations; usually appears within 15 days of disease onset; and manifests with pruritic, migratory, edematous papules appearing mainly on the trunk and occasionally the face and limbs. The urticarial rash tends to be associated with more severe forms of the disease and regresses within a week, responding to antihistamines. Of note, clinically similar rashes can be caused by drugs. Histologically, the lesions show dermal edema and a mild perivascular lymphocytic infiltrate, sometimes admixed with eosinophils.
Chilblainlike Lesions—Chilblainlike lesions (CBLLs) account for 19% of skin manifestations associated with COVID-1913 and present as erythematous-purplish, edematous lesions that can be mildly pruritic or painful, appearing on the toes—COVID toes—and more rarely the fingers (Figure 2). They were seen epidemically during the first pandemic wave (2020 lockdown) in several countries, and clinically are very similar to, if not indistinguishable from, idiopathic chilblains, but are not necessarily associated with cold exposure. They appear in young, generally healthy patients or those with mild COVID-19 symptoms 2 to 4 weeks after symptom onset. They regress spontaneously or under local corticosteroid treatment within a few days or weeks. Histologically, CBLLs are indistinguishable from chilblains of other origins, namely idiopathic (seasonal) ones. They manifest with necrosis of epidermal keratinocytes; dermal edema that may be severe, leading to the development of subepidermal pseudobullae; a rather dense perivascular and perieccrine gland lymphocytic infiltrate; and sometimes with vascular lesions (eg, edema of endothelial cells, microthromboses of dermal capillaries and venules, fibrinoid deposits within the wall of dermal venules)(Figure 3).16-18 Most patients (>80%) with CBLLs have negative serologic or polymerase chain reaction tests for SARS-CoV-2,19 which generated a lively debate about the role of SARS-CoV-2 in the genesis of CBLLs. According to some authors, SARS-CoV-2 plays no direct role, and CBLLs would occur in young people who sit or walk barefoot on cold floors at home during confinement.20-23 Remarkably, CBLLs appeared in patients with no history of chilblains during a season that was not particularly cold, namely in France or in southern California, where their incidence was much higher compared to the same time period of prior years. Some reports have supported a direct role for the virus based on questionable observations of the virus within skin lesions (eg, sweat glands, endothelial cells) by immunohistochemistry, electron microscopy, and/or in situ hybridization.17,24,25 A more satisfactory hypothesis would involve the role of a strong innate immunity leading to elimination of the virus before the development of specific antibodies via the increased production of type 1 interferon (IFN-1); this would affect the vessels, causing CBLLs. This mechanism would be similar to the one observed in some interferonopathies (eg, Aicardi-Goutières syndrome), also characterized by IFN-1 hypersecretion and chilblains.26-29 According to this hypothesis, CBLLs should be considered a paraviral rash similar to other skin manifestations associated with COVID-19.30
Acro-ischemia—Acro-ischemia livedoid lesions account for 1% to 6% of skin manifestations and comprise lesions of livedo (either reticulated or racemosa); necrotic acral bullae; and gangrenous necrosis of the extremities, especially the toes. The livedoid lesions most often appear within 15 days of COVID-19 symptom onset, and the purpuric lesions somewhat later (2–4 weeks); they mainly affect adult patients, last about 10 days, and are the hallmark of severe infection, presumably related to microthromboses of the cutaneous capillaries (endothelial dysfunction, prothrombotic state, elevated D-dimers). Histologically, they show capillary thrombosis and dermoepidermal necrosis (Figure 4).
Other Reported Polymorphic or Atypical Rashes—Erythema multiforme–like eruptions may appear before other COVID-19 symptoms and manifest as reddish-purple, nearly symmetric, diffuse, occasionally targetoid bullous or necrotic macules. The eruptions mainly affect adults and most often are seen on the palms, elbows, knees, and sometimes the mucous membranes. The rash regresses in 1 to 3 weeks without scarring and represents a delayed cutaneous hypersensitivity reaction. Histologically, the lesions show vacuolization of basal epidermal keratinocytes, keratinocyte necrosis, dermoepidermal detachment, a variably dense dermal T-lymphocytic infiltrate, and red blood cell extravasation (Figure 5).
Leukocytoclastic vasculitis may be generalized or localized. It manifests clinically by petechial/purpuric maculopapules, especially on the legs, mainly in elderly patients with COVID-19. Histologically, the lesions show necrotizing changes of dermal postcapillary venules, neutrophilic perivascular inflammation, red blood cell extravasation, and occasionally vascular IgA deposits by direct immunofluorescence examination. The course usually is benign.
The incidence of pityriasis rosea and of clinically similar rashes (referred to as “pityriasis rosea–like”) increased 5-fold during the COVID-19 pandemic.31,32 These dermatoses manifest with erythematous, scaly, circinate plaques, typically with an initial herald lesion followed a few days later by smaller erythematous macules. Histologically, the lesions comprise a spongiform dermatitis with intraepidermal exocytosis of red blood cells and a mild to moderate dermal lymphocytic infiltrate.
Erythrodysesthesia, or hand-foot syndrome, manifests with edematous erythema and palmoplantar desquamation accompanied by a burning sensation or pain. This syndrome is known as an adverse effect of some chemotherapies because of the associated drug toxicity and sweat gland inflammation; it was observed in 40% of 666 COVID-19–positive patients with mild to moderate pneumonitis.33
“COVID nose” is a rare cutaneous manifestation characterized by nasal pigmentation comprising multiple coalescent frecklelike macules on the tip and wings of the nose and sometimes the malar areas. These lesions predominantly appear in women aged 25 to 65 years and show on average 23 days after onset of COVID-19, which is usually mild. This pigmentation is similar to pigmentary changes after infection with chikungunya; it can be treated with depigmenting products such as azelaic acid and hydroquinone cream with sunscreen use, and it regresses in 2 to 4 months.34
Telogen effluvium (excessive and temporary shedding of normal telogen club hairs of the entire scalp due to the disturbance of the hair cycle) is reportedly frequent in patients (48%) 1 month after COVID-19 infection, but it may appear later (after 12 weeks).35 Alopecia also is frequently reported during long (or postacute) COVID-19 (ie, the symptomatic disease phase past the acute 4 weeks’ stage of the infection) and shows a female predominance36; it likely represents the telogen effluvium seen 90 days after a severe illness. Trichodynia (pruritus, burning, pain, or paresthesia of the scalp) also is reportedly common (developing in more than 58% of patients) and is associated with telogen effluvium in 44% of cases. Several cases of alopecia areata (AA) triggered or aggravated by COVID-19 also have been reported37,38; they could be explained by the “cytokine storm” triggered by the infection, involving T and B lymphocytes; plasmacytoid dendritic cells; natural killer cells with oversecretion of IL-6, IL-4, tumor necrosis factor α, and IFN type I; and a cytotoxic reaction associated with loss of the immune privilege of hair follicles.
Nail Manifestations
The red half-moon nail sign is an asymptomatic purplish-red band around the distal margin of the lunula that affects some adult patients with COVID-19.39 It appears shortly after onset of symptoms, likely the manifestation of vascular inflammation in the nail bed, and regresses slowly after approximately 1 week.40 Beau lines are transverse grooves in the nail plate due to the temporary arrest of the proximal nail matrix growth accompanying systemic illnesses; they appear approximately 2 to 3 weeks after the onset of COVID-19.41 Furthermore, nail alterations can be caused by drugs used to treat COVID-19, such as longitudinal melanonychia due to treatment with hydroxychloroquine or fluorescence of the lunula or nail plate due to treatment with favipiravir.42
Multisystem Inflammatory Syndrome
Multisystem inflammatory syndrome (MIS) is clinically similar to Kawasaki disease; it typically affects children43 and more rarely adults with COVID-19. It manifests with fever, weakness, and biological inflammation and also frequently with skin lesions (72%), which are polymorphous and include morbilliform rash (27%); urticaria (24%); periorbital edema (24%); nonspecific erythema (21.2%); retiform purpura (18%); targetoid lesions (15%); malar rash (15.2%); and periareolar erythema (6%).44 Compared to Kawasaki disease, MIS affects slightly older children (mean age, 8.5 vs 3 years) and more frequently includes cardiac and gastrointestinal manifestations; the mortality rate also is slightly higher (2% vs 0.17%).45
Confirmed COVID-19 Infection
At the beginning of the pandemic, skin manifestations were reported in patients who were suspected of having COVID-19 but did not always have biological confirmation of SARS-CoV-2 infection due to the unavailability of diagnostic tests or the physical impossibility of testing. However, subsequent studies have confirmed that most of these dermatoses were indeed associated with COVID-19 infection.9,46 For example, a study of 655 patients with confirmed COVID-19 infection reported maculopapular (38%), vascular (22%), urticarial (15%), and vesicular (15%) rashes; erythema multiforme or Stevens-Johnson–like syndrome (3%, often related to the use of hydroxychloroquine); generalized pruritus (1%); and MIS (0.5%). The study confirmed that CBLLs were mostly seen in young patients with mild disease, whereas livedo (fixed rash) and retiform purpura occurred in older patients with a guarded prognosis.46
Remarkably, most dermatoses associated with SARS-CoV-2 infection were reported during the initial waves of the pandemic, which were due to the α and δ viral variants. These manifestations were reported more rarely when the ο variant was predominant, even though most patients (63%) who developed CBLLs in the first wave also developed them during the second pandemic wave.47 This decrease in the incidence of COVID-19–associated dermatoses could be because of the lower pathogenicity of the o variant,3 a lower tropism for the skin, and variations in SARS-CoV-2 antigenicity that would induce a different immunologic response, combined with an increasingly stronger herd immunity compared to the first pandemic waves achieved through vaccination and spontaneous infections in the population. Additional reasons may include different baseline characteristics in patients hospitalized with COVID-19 (regarding comorbidities, disease severity, and received treatments), and the possibility that some of the initially reported COVID-19–associated skin manifestations could have been produced by different etiologic agents.48 In the last 2 years, COVID-19–related skin manifestations have been reported mainly as adverse events to COVID-19 vaccination.
CUTANEOUS ADVERSE EFFECTS OF DRUGS USED TO TREAT COVID-19
Prior to the advent of vaccines and specific treatments for SARS-CoV-2, various drugs were used—namely hydroxychloroquine, ivermectin, and tocilizumab—that did not prove efficacious and caused diverse adverse effects, including cutaneous eruptions such as urticaria, maculopapular eruptions, erythema multiforme or Stevens-Johnson syndrome, vasculitis, longitudinal melanonychia, and acute generalized exanthematous pustulosis.49,50 Nirmatrelvir 150 mg–ritonavir 100 mg, which was authorized for emergency use by the US Food and Drug Administration for the treatment of COVID-19, is a viral protease inhibitor blocking the replication of the virus. Ritonavir can induce pruritus, maculopapular rash, acne, Stevens-Johnson syndrome, and toxic epidermal necrolysis; of note, these effects have been observed following administration of ritonavir for treatment of HIV at higher daily doses and for much longer periods of time compared with treatment of COVID-19 (600–1200 mg vs 200 mg/d, respectively). These cutaneous drug side effects are clinically similar to the manifestations caused either directly or indirectly by SARS-CoV-2 infection; therefore, it may be difficult to differentiate them.
DERMATOSES DUE TO PROTECTIVE DEVICES
Dermatoses due to personal protective equipment such as masks or face shields affected the general population and mostly health care professionals51; 54.4% of 879 health care professionals in one study reported such events.52 These dermatoses mainly include contact dermatitis of the face (nose, forehead, and cheeks) of irritant or allergic nature (eg, from preservatives releasing formaldehyde contained in masks and protective goggles). They manifest with skin dryness; desquamation; maceration; fissures; or erosions or ulcerations of the cheeks, forehead, and nose. Cases of pressure urticaria also have been reported. Irritant dermatitis induced by the frequent use of disinfectants (eg, soaps, hydroalcoholic sanitizing gels) also can affect the hands. Allergic hand dermatitis can be caused by medical gloves.
The term maskne (or mask acne) refers to a variety of mechanical acne due to the prolonged use of surgical masks (>4 hours per day for ≥6 weeks); it includes cases of de novo acne and cases of pre-existing acne aggravated by wearing a mask. Maskne is characterized by acne lesions located on the facial area covered by the mask (Figure 6). It is caused by follicular occlusion; increased sebum secretion; mechanical stress (pressure, friction); and dysbiosis of the microbiome induced by changes in heat, pH, and humidity. Preventive measures include application of noncomedogenic moisturizers or gauze before wearing the mask as well as facial cleansing with appropriate nonalcoholic products. Similar to acne, rosacea often is aggravated by prolonged wearing of surgical masks (mask rosacea).53,54
DERMATOSES REVEALED OR AGGRAVATED BY COVID-19
Exacerbation of various skin diseases has been reported after infection with SARS-CoV-2.55 Psoriasis and acrodermatitis continua of Hallopeau,56 which may progress into generalized, pustular, or erythrodermic forms,57 have been reported; the role of hydroxychloroquine and oral corticosteroids used for the treatment of COVID-19 has been suspected.57 Atopic dermatitis patients—26% to 43%—have experienced worsening of their disease after symptomatic COVID-19 infection.58 The incidence of herpesvirus infections, including herpes zoster, increased during the pandemic.59 Alopecia areata relapses occurred in 42.5% of 392 patients with preexisting disease within 2 months of COVID-19 onset in one study,60 possibly favored by the psychological stress; however, some studies have not confirmed the aggravating role of COVID-19 on alopecia areata.61 Lupus erythematosus, which may relapse in the form of Rowell syndrome,62 and livedoid vasculopathy63 also have been reported following COVID-19 infection.
SKIN MANIFESTATIONS ASSOCIATED WITH COVID-19 VACCINES
In parallel with the rapid spread of COVID-19 vaccination,4 an increasing number of skin manifestations has been observed following vaccination; these dermatoses now are more frequently reported than those related to natural SARS-CoV-2 infection.64-70 Vaccine-induced skin manifestations have a reported incidence of approximately 4% and show a female predominance.65 Most of them (79%) have been reported in association with messenger RNA (mRNA)–based vaccines, which have been the most widely used; however, the frequency of side effects would be lower after mRNA vaccines than after inactivated virus-based vaccines. Eighteen percent occurred after the adenoviral vector vaccine, and 3% after the inactivated virus vaccine.70 Fifty-nine percent were observed after the first dose. They are clinically polymorphous and generally benign, regressing spontaneously after a few days, and they should not constitute a contraindication to vaccination.Interestingly, many skin manifestations are similar to those associated with natural SARS-CoV-2 infection; however, their frequency and severity does not seem to depend on whether the patients had developed skin reactions during prior SARS-CoV-2 infection. These reactions have been classified into several types:
• Immediate local reactions at the injection site: pain, erythema, or edema represent the vast majority (96%) of reactions to vaccines. They appear within 7 days after vaccination (average, 1 day), slightly more frequently (59%) after the first dose. They concern mostly young patients and are benign, regressing in 2 to 3 days.70
• Delayed local reactions: characterized by pain or pruritus, erythema, and skin induration mimicking cellulitis (COVID arm) and represent 1.7% of postvaccination reactions. They correspond to a delayed hypersensitivity reaction and appear approximately 7 days after vaccination, most often after the first vaccine dose (75% of cases), which is almost invariably mRNA based.70
• Urticarial reactions corresponding to an immediate (type 1) hypersensitivity reaction: constitute 1% of postvaccination reactions, probably due to an allergy to vaccine ingredients. They appear on average 1 day after vaccination, almost always with mRNA vaccines.70
• Angioedema: characterized by mucosal or subcutaneous edema and constitutes 0.5% of postvaccination reactions. It is a potentially serious reaction that appears on average 12 hours after vaccination, always with an mRNA-based vaccine.70
• Morbilliform rash: represents delayed hypersensitivity reactions (0.1% of postvaccination reactions) that appear mostly after the first dose (72%), on average 3 days after vaccination, always with an mRNA-based vaccine.70
• Herpes zoster: usually develops after the first vaccine dose in elderly patients (69% of cases) on average 4 days after vaccination and constitutes 0.1% of postvaccination reactions.71
• Bullous diseases: mainly bullous pemphigoid (90%) and more rarely pemphigus (5%) or bullous erythema pigmentosum (5%). They appear in elderly patients on average 7 days after vaccination and constitute 0.04% of postvaccination reactions.72
• Chilblainlike lesions: several such cases have been reported so far73; they constitute 0.03% of postvaccination reactions.70 Clinically, they are similar to those associated with natural COVID-19; they appear mostly after the first dose (64%), on average 5 days after vaccination with the mRNA or adenovirus vaccine, and show a female predominance. The appearance of these lesions in vaccinated patients, who are a priori not carriers of the virus, strongly suggests that CBLLs are due to the immune reaction against SARS-CoV-2 rather than to a direct effect of this virus on the skin, which also is a likely scenario with regards to other skin manifestations seen during the successive COVID-19 epidemic waves.73-75
• Reactions to hyaluronic acid–containing cosmetic fillers: erythema, edema, and potentially painful induration at the filler injection sites. They constitute 0.04% of postvaccination skin reactions and appear 24 hours after vaccination with mRNA-based vaccines, equally after the first or second dose.76
• Pityriasis rosea–like rash: most occur after the second dose of mRNA-based vaccines (0.023% of postvaccination skin reactions).70
• Severe reactions: these include acute generalized exanthematous pustulosis77 and Stevens-Johnson syndrome.78 One case of each has been reported after the adenoviral vector vaccine 3 days after vaccination.
Other more rarely observed manifestations include reactivation/aggravation or de novo appearance of inflammatory dermatoses such as psoriasis,79,80 leukocytoclastic vasculitis,81,82 lymphocytic83 or urticarial84 vasculitis, Sweet syndrome,85 lupus erythematosus, dermatomyositis,86,87 alopecia,37,88 infection with Trichophyton rubrum,89 Grover disease,90 and lymphomatoid reactions (such as recurrences of cutaneous T-cell lymphomas [CD30+], and de novo development of lymphomatoid papulosis).91
FINAL THOUGHTS
COVID-19 is associated with several skin manifestations, even though the causative role of SARS-CoV-2 has remained elusive. These dermatoses are highly polymorphous, mostly benign, and usually spontaneously regressive, but some of them reflect severe infection. They mostly were described during the first pandemic waves, reported in several national and international registries, which allowed for their morphological classification. Currently, cutaneous adverse effects of vaccines are the most frequently reported dermatoses associated with SARS-CoV-2, and it is likely that they will continue to be observed while COVID-19 vaccination lasts. Hopefully the end of the COVID-19 pandemic is near. In January 2023, the International Health Regulations Emergency Committee of the World Health Organization acknowledged that the COVID-19 pandemic may be approaching an inflexion point, and even though the event continues to constitute a public health emergency of international concern, the higher levels of population immunity achieved globally through infection and/or vaccination may limit the impact of SARS-CoV-2 on morbidity and mortality. However, there is little doubt that this virus will remain a permanently established pathogen in humans and animals for the foreseeable future.92 Therefore, physicians—especially dermatologists—should be aware of the various skin manifestations associated with COVID-19 so they can more efficiently manage their patients.
- Ashraf UM, Abokor AA, Edwards JM, et al. SARS-CoV-2, ACE2 expression, and systemic organ invasion. Physiol Genomics. 2021;53:51-60.
- Ganier C, Harun N, Peplow I, et al. Angiotensin-converting enzyme 2 expression is detectable in keratinocytes, cutaneous appendages, and blood vessels by multiplex RNA in situ hybridization. Adv Skin Wound Care. 2022;35:219-223.
- Ulloa AC, Buchan SA, Daneman N, et al. Estimates of SARS-CoV-2 omicron variant severity in Ontario, Canada. JAMA. 2022;327:1286-1288.
- World Health Organization. Coronavirus (COVID-19) Dashboard. Accessed April 6, 2023. https://covid19.who.int
- Guan WJ, Ni ZY, Hu Y, et al; China Medical Treatment Expert Group for COVID-19. clinical characteristics of coronavirus disease 2019 in China. N Engl J Med. 2020;382:1708-1720.
- Recalcati S. Cutaneous manifestations in COVID-9: a first perspective. J Eur Acad Dermatol Venereol. 2020;34:E212-E213.
- Freeman EE, McMahon DE, Lipoff JB, et al. The spectrum of COVID-19-associated dermatologic manifestations: an international registry of 716 patients from 31 countries. J Am Acad Dermatol. 2020;83:1118-1129.
- Freeman EE, Chamberlin GC, McMahon DE, et al. Dermatology COVID-19 registries: updates and future directions. Dermatol Clin. 2021;39:575-585.
- Guelimi R, Salle R, Dousset L, et al. Non-acral skin manifestations during the COVID-19 epidemic: COVIDSKIN study by the French Society of Dermatology. J Eur Acad Dermatol Venereol. 2021;35:E539-E541.
- Marzano AV, Genovese G, Moltrasio C, et al; Italian Skin COVID-19 Network of the Italian Society of Dermatology and Sexually Transmitted Diseases. The clinical spectrum of COVID-19 associated cutaneous manifestations: an Italian multicenter study of 200 adult patients. J Am Acad Dermatol. 2021;84:1356-1363.
- Sugai T, Fujita Y, Inamura E, et al. Prevalence and patterns of cutaneous manifestations in 1245 COVID-19 patients in Japan: a single-centre study. J Eur Acad Dermatol Venereol. 2022;36:E522-E524.
- Holmes Z, Courtney A, Lincoln M, et al. Rash morphology as a predictor of COVID‐19 severity: a systematic review of the cutaneous manifestations of COVID‐19. Skin Health Dis. 2022;2:E120. doi:10.1002/ski2.120
- Galván Casas C, Català A, Carretero Hernández G, et al. Classification of the cutaneous manifestations of COVID-19: a rapid prospective nationwide consensus study in Spain with 375 cases. Br J Dermatol. 2020;183:71-77.
- Garduño‑Soto M, Choreño-Parra, Cazarin-Barrientos Dermatological aspects of SARS‑CoV‑2 infection: mechanisms and manifestations. Arch Dermatol Res. 2021;313:611-622.
- Huynh T, Sanchez-Flores X, Yau J, et al. Cutaneous manifestations of SARS-CoV-2 Infection. Am J Clin Dermatol. 2022;23:277-286.
- Kanitakis J, Lesort C, Danset M, et al.
Chilblain-like acral lesions during the COVID-19 pandemic (“COVID toes”): histologic, immunofluorescence, and immunohistochemical study of 17 cases. J Am Acad Dermatol. 2020; 83:870-875. - Kolivras A, Thompson C, Pastushenko I, et al. A clinicopathological description of COVID-19-induced chilblains (COVID-toes) correlated with a published literature review. J Cutan Pathol. 2022;49:17-28.
- Roca-Ginés J, Torres-Navarro I, Sánchez-Arráez J, et al. Assessment of acute acral lesions in a case series of children and adolescents during the COVID-19 pandemic. 2020;156:992-997.
- Le Cleach L, Dousset L, Assier H, et al; French Society of Dermatology. Most chilblains observed during the COVID-19 outbreak occur in patients who are negative for COVID-19 on polymerase chain reaction and serology testing. Br J Dermatol. 2020;183:866-874.
- Discepolo V, Catzola A, Pierri L, et al. Bilateral chilblain-like lesions of the toes characterized by microvascular remodeling in adolescents during the COVID-19 pandemic. JAMA Netw Open. 2021;4:E2111369.
- Gehlhausen JR, Little AJ, Ko CJ, et al. Lack of association between pandemic chilblains and SARS-CoV-2 infection. Proc Natl Acad Sci U S A. 2022;119:e2122090119.
- Neri, Virdi, Corsini, et al Major cluster of paediatric ‘true’ primary chilblains during the COVID-19 pandemic: a consequence of lifestyle changes due to lockdown. J Eur Acad Dermatol Venereol. 2020;34:2630-2635.
- De Greef A, Choteau M, Herman A, et al. Chilblains observed during the COVID-19 pandemic cannot be distinguished from the classic, cold-related chilblains. Eur J Dermatol. 2022;32:377-383.
- Colmenero I, Santonja C, Alonso-Riaño M, et al. SARS-CoV-2 endothelial infection causes COVID-19 chilblains: histopathological, immunohistochemical and ultrastructural study of seven paediatric cases. Br J Dermatol. 2020;183:729-737.
- Quintero-Bustos G, Aguilar-Leon D, Saeb-Lima M. Histopathological and immunohistochemical characterization of skin biopsies from 41 SARS-CoV-2 (+) patients: experience in a Mexican concentration institute: a case series and literature review. Am J Dermatopathol. 2022;44:327-337.
- Arkin LM, Moon JJ, Tran JM, et al; COVID Human Genetic Effort. From your nose to your toes: a review of severe acute respiratory syndrome coronavirus 2 pandemic-associated pernio. J Invest Dermatol. 2021;141:2791-2796.
- Frumholtz L, Bouaziz JD, Battistella M, et al; Saint-Louis CORE (COvid REsearch). Type I interferon response and vascular alteration in chilblain-like lesions during the COVID-19 outbreak. Br J Dermatol. 2021;185:1176-1185.
- Hubiche T, Cardot-Leccia N, Le Duff F, et al. Clinical, laboratory, and interferon-alpha response characteristics of patients with chilblain-like lesions during the COVID-19 pandemic. JAMA Dermatol. 2021;157:202-206.
- Lesort C, Kanitakis J, Villani A, et al. COVID-19 and outbreak of chilblains: are they related? J Eur Acad Dermatol Venereol. 2020;34:E757-E758.
- Sanchez A, Sohier P, Benghanem S, et al. Digitate papulosquamous eruption associated with severe acute respiratory syndrome coronavirus 2 infection. JAMA Dermatol. 2020;156:819-820.
- Drago F, Broccolo F, Ciccarese G. Pityriasis rosea, pityriasis rosea-like eruptions, and herpes zoster in the setting of COVID-19 and COVID-19 vaccination. Clin Dermatol. 2022;S0738-081X(22)00002-5.
- Dursun R, Temiz SA. The clinics of HHV-6 infection in COVID-19 pandemic: pityriasis rosea and Kawasaki disease. Dermatol Ther. 2020;33:E13730.
- Nuno-Gonzalez A, Magaletsky K, Feito Rodríguez M, et al. Palmoplantar erythrodysesthesia: a diagnostic sign of COVID-19. J Eur Acad Dermatol Venereol. 2021;35:e247-e249.
- Sil A, Panigrahi A, Chandra A, et al. “COVID nose”: a unique post-COVID pigmentary sequelae reminiscing Chik sign: a descriptive case series. J Eur Acad Dermatol Venereol. 2022;36:E419-E421.
- Starace M, Iorizzo M, Sechi A, et al. Trichodynia and telogen effluvium in COVID-19 patients: results of an international expert opinion survey on diagnosis and management. JAAD Int. 2021;5:11-18.
- Wong-Chew RM, Rodríguez Cabrera EX, Rodríguez Valdez CA, et al. Symptom cluster analysis of long COVID-19 in patients discharged from the Temporary COVID-19 Hospital in Mexico City. Ther Adv Infect Dis. 2022;9:20499361211069264.
- Bardazzi F, Guglielmo A, Abbenante D, et al. New insights into alopecia areata during COVID-19 pandemic: when infection or vaccination could play a role. J Cosmet Dermatol. 2022;21:1796-1798.
- Christensen RE, Jafferany M. Association between alopecia areata and COVID-19: a systematic review. JAAD Int. 2022;7:57-61.
- Wollina U, Kanitakis J, Baran R. Nails and COVID-19: a comprehensive review of clinical findings and treatment. Dermatol Ther. 2021;34:E15100.
- Méndez-Flores S, Zaladonis A, Valdes-Rodriguez R. COVID-19 and nail manifestation: be on the lookout for the red half-moon nail sign. Int J Dermatol. 2020;59:1414.
- Alobaida S, Lam JM. Beau lines associated with COVID-19. CMAJ. 2020;192:E1040.
- Durmaz EÖ, Demirciog˘lu D. Fluorescence in the sclera, nails, and teeth secondary to favipiravir use for COVID-19 infections. J Clin Aesthet Dermatol. 2022;15:35-37.
- Brumfiel CM, DiLorenzo AM, Petronic-Rosic VM. Dermatologic manifestations of COVID-19-associated multisystem inflammatory syndrome in children. Clin Dermatol. 2021;39:329-333.
- Akçay N, Topkarcı Z, Menentog˘lu ME, et al. New dermatological findings of MIS-C: can mucocutaneous involvement be associated with severe disease course? Australas J Dermatol. 2022;63:228-234. doi:10.1111/ajd.13819
- Vogel TP, Top KA, Karatzios C, et al. Multisystem inflammatory syndrome in children and adults (MIS-C/A): case definition & guidelines for data collection, analysis, and presentation of immunization safety data. Vaccine. 2021;39:3037-3049.
- Conforti C, Dianzani C, Agozzino M, et al. Cutaneous manifestations in confirmed COVID-19 patients: a systematic review. Biology (Basel). 2020;9:449.
- Hubiche T, Le Duff F, Fontas E, et al. Relapse of chilblain-like lesions during the second wave of the COVID-19 pandemic: a cohort follow-up. Br J Dermatol. 2021;185:858-859.
- Fernandez-Nieto, Ortega-Quijano, Suarez-Valle, et al Lack of skin manifestations in COVID-19 hospitalized patients during the second epidemic wave in Spain: a possible association with a novel SARS-CoV-2 variant: a cross-sectional study. J Eur Acad Dermatol Venereol. 2021;35:E183-E185.
- Martinez-LopezA, Cuenca-Barrales, Montero-Vilchezet al Review of adverse cutaneous reactions of pharmacologic interventions for COVID-19: a guide for the dermatologist. J Am Acad Dermatol. 2020;83:1738-1748.
- Cutaneous side-effects of the potential COVID-19 drugs. Dermatol Ther. 2020;33:E13476.
- Mawhirt SL, Frankel D, Diaz AM. Cutaneous manifestations in adult patients with COVID-19 and dermatologic conditions related to the COVID-19 pandemic in health care workers. Curr Allerg Asthma Rep. 2020;20:75.
- Nguyen C, Young FG, McElroy D, et al. Personal protective equipment and adverse dermatological reactions among healthcare workers: survey observations from the COVID-19 pandemic. Medicine (Baltimore). 2022;101:E29003.
- Rathi SK, Dsouza JM. Maskne: a new acne variant in COVID-19 era. Indian J Dermatol. 2022;67:552-555.
- Damiani G, Girono L, Grada A, et al. COVID-19 related masks increase severity of both acne (maskne) and rosacea (mask rosacea): multi-center, real-life, telemedical, and observational prospective study. Dermatol Ther. 2021;34:E14848.
- Aram K, Patil A, Goldust M, et al. COVID-19 and exacerbation of dermatological diseases: a review of the available literature. Dermatol Ther. 2021;34:E15113.
- Samotij D, Gawron E, Szcze˛ch J, et al. Acrodermatitis continua of Hallopeau evolving into generalized pustular psoriasis following COVID-19: a case report of a successful treatment with infliximab in combination with acitretin. Biologics. 2021;15:107-113.
- Demiri J, Abdo M, Tsianakas A. Erythrodermic psoriasis after COVID-19 [in German]. Hautarzt. 2022;73:156-159.
- de Wijs LEM, Joustra MM, Olydam JI, et al. COVID-19 in patients with cutaneous immune-mediated diseases in the Netherlands: real-world observational data. J Eur Acad Dermatol Venereol. 2021;35:E173-E176.
- Marques NP, Maia CMF, Marques NCT, et al. Continuous increase of herpes zoster cases in Brazil during the COVID-19 pandemic. Oral Surg Oral Med Oral Pathol Oral Radiol. 2022;133:612-614.
- Rinaldi F, Trink A, Giuliani G, et al. Italian survey for the evaluation of the effects of coronavirus disease 2019 (COVID-19) pandemic on alopecia areata recurrence. Dermatol Ther (Heidelb). 2021;11:339-345.
- Rudnicka L, Rakowska A, Waskiel-Burnat A, et al. Mild-to-moderate COVID-19 is not associated with worsening of alopecia areata: a retrospective analysis of 32 patients. J Am Acad Dermatol. 2021;85:723-725.
- Drenovska K, Shahid M, Mateeva V, et al. Case report: Rowell syndrome-like flare of cutaneous lupus erythematosus following COVID-19 infection. Front Med (Lausanne). 2022;9:815743.
- Kawabe R, Tonomura K, Kotobuki Y, et al. Exacerbation of livedoid vasculopathy after coronavirus disease 2019. Eur J Dermatol. 2022;32:129-131. doi:10.1684/ejd.2022.4200
- McMahon DE, Kovarik CL, Damsky W, et al. Clinical and pathologic correlation of cutaneous COVID-19 vaccine reactions including V-REPP: a registry-based study. J Am Acad Dermatol. 2022;86:113-121.
- Avallone G, Quaglino P, Cavallo F, et al. SARS-CoV-2 vaccine-related cutaneous manifestations: a systematic review. Int J Dermatol. 2022;61:1187-1204. doi:10.1111/ijd.16063
- Gambichler T, Boms S, Susok L, et al. Cutaneous findings following COVID-19 vaccination: review of world literature and own experience. J Eur Acad Dermatol Venereol. 2022;36:172-180.
- Kroumpouzos G, Paroikaki ME, Yumeen S, et al. Cutaneous complications of mRNA and AZD1222 COVID-19 vaccines: a worldwide review. Microorganisms. 2022;10:624.
- Robinson L,Fu X,Hashimoto D, et al. Incidence of cutaneous reactions after messenger RNA COVID-19 vaccines. 2021;
- Wollina U, Chiriac A, Kocic H, et al. Cutaneous and hypersensitivity reactions associated with COVID-19 vaccination: a narrative review. Wien Med Wochenschr. 2022;172:63-69.
- Wei TS. Cutaneous reactions to COVID-19 vaccines: a review. JAAD Int. 2022;7:178-186.
- Katsikas Triantafyllidis K, Giannos P, Mian IT, et al. Varicella zoster virus reactivation following COVID-19 vaccination: a systematic review of case reports. Vaccines (Basel). 2021;9:1013.
- Maronese CA, Caproni M, Moltrasio C, et al. Bullous pemphigoid associated with COVID-19 vaccines: an Italian multicentre study. Front Med (Lausanne). 2022;9:841506.
- Cavazos A, Deb A, Sharma U, et al. COVID toes following vaccination. Proc (Bayl Univ Med Cent). 2022;35:476-479.
- Lesort C, Kanitakis J, Danset M, et al. Chilblain-like lesions after BNT162b2 mRNA COVID-19 vaccine: a case report suggesting that ‘COVID toes’ are due to the immune reaction to SARS-CoV-2. J Eur Acad Dermatol Venereol. 2021;35:E630-E632.
- Russo R, Cozzani E, Micalizzi C, et al. Chilblain-like lesions after COVID-19 vaccination: a case series. Acta Derm Venereol. 2022;102:adv00711. doi:10.2340/actadv.v102.2076
- Ortigosa LCM, Lenzoni FC, Suárez MV, et al. Hypersensitivity reaction to hyaluronic acid dermal filler after COVID-19 vaccination: a series of cases in São Paulo, Brazil. Int J Infect Dis. 2022;116:268-270.
- Agaronov A, Makdesi C, Hall CS. Acute generalized exanthematous pustulosis induced by Moderna COVID-19 messenger RNA vaccine. JAAD Case Rep. 2021;16:96-97.
- Dash S, Sirka CS, Mishra S, et al. COVID-19 vaccine-induced Stevens-Johnson syndrome. Clin Exp Dermatol. 2021;46:1615-1617.
- Huang Y, Tsai TF. Exacerbation of psoriasis following COVID-19 vaccination: report from a single center. Front Med (Lausanne). 2021;8:812010.
- Elamin S, Hinds F, Tolland J. De novo generalized pustular psoriasis following Oxford-AstraZeneca COVID-19 vaccine. Clin Exp Dermatol 2022;47:153-155.
- Abdelmaksoud A, Wollina U, Temiz SA, et al. SARS-CoV-2 vaccination-induced cutaneous vasculitis: report of two new cases and literature review. Dermatol Ther. 2022;35:E15458.
- Fritzen M, Funchal GDG, Luiz MO, et al. Leukocytoclastic vasculitis after exposure to COVID-19 vaccine. An Bras Dermatol. 2022;97:118-121.
- Vassallo C, Boveri E, Brazzelli V, et al. Cutaneous lymphocytic vasculitis after administration of COVID-19 mRNA vaccine. Dermatol Ther. 2021;34:E15076.
- Nazzaro G, Maronese CA. Urticarial vasculitis following mRNA anti-COVID-19 vaccine. Dermatol Ther. 2022;35:E15282.
- Hoshina D, Orita A. Sweet syndrome after severe acute respiratory syndrome coronavirus 2 mRNA vaccine: a case report and literature review. J Dermatol. 2022;49:E175-E176.
- Lemoine C, Padilla C, Krampe N, et al. Systemic lupus erythematous after Pfizer COVID-19 vaccine: a case report. Clin Rheumatol. 2022;41:1597-1601.
- Nguyen B, Lalama MJ, Gamret AC, et al. Cutaneous symptoms of connective tissue diseases after COVID-19 vaccination: a systematic review. Int J Dermatol. 2022;61:E238-E241.
- Gallo G, Mastorino L, Tonella L, et al. Alopecia areata after COVID-19 vaccination. Clin Exp Vaccine Res. 2022;11:129-132.
- Norimatsu Y, Norimatsu Y. A severe case of Trichophyton rubrum-caused dermatomycosis exacerbated after COVID-19 vaccination that had to be differentiated from pustular psoriasis. Med Mycol Case Rep. 2022;36:19-22.
- Yang K, Prussick L, Hartman R, et al. Acantholytic dyskeratosis post-COVID vaccination. Am J Dermatopathol. 2022;44:E61-E63.
- Koumaki D, Marinos L, Nikolaou V, et al. Lymphomatoid papulosis (LyP) after AZD1222 and BNT162b2 COVID-19 vaccines. Int J Dermatol. 2022;61:900-902.
- World Health Organization. Statement on the fourteenth meeting of the International Health Regulations (2005) Emergency Committee regarding the coronavirus disease (COVID-19) pandemic. Published January 30, 2023. Accessed April 12, 2023. https://www.who.int/news/item/30-01-2023-statement-on-the-fourteenth-meeting-of-the-international-health-regulations-(2005)-emergency-committee-regarding-the-coronavirus-disease-(covid-19)-pandemic
- Ashraf UM, Abokor AA, Edwards JM, et al. SARS-CoV-2, ACE2 expression, and systemic organ invasion. Physiol Genomics. 2021;53:51-60.
- Ganier C, Harun N, Peplow I, et al. Angiotensin-converting enzyme 2 expression is detectable in keratinocytes, cutaneous appendages, and blood vessels by multiplex RNA in situ hybridization. Adv Skin Wound Care. 2022;35:219-223.
- Ulloa AC, Buchan SA, Daneman N, et al. Estimates of SARS-CoV-2 omicron variant severity in Ontario, Canada. JAMA. 2022;327:1286-1288.
- World Health Organization. Coronavirus (COVID-19) Dashboard. Accessed April 6, 2023. https://covid19.who.int
- Guan WJ, Ni ZY, Hu Y, et al; China Medical Treatment Expert Group for COVID-19. clinical characteristics of coronavirus disease 2019 in China. N Engl J Med. 2020;382:1708-1720.
- Recalcati S. Cutaneous manifestations in COVID-9: a first perspective. J Eur Acad Dermatol Venereol. 2020;34:E212-E213.
- Freeman EE, McMahon DE, Lipoff JB, et al. The spectrum of COVID-19-associated dermatologic manifestations: an international registry of 716 patients from 31 countries. J Am Acad Dermatol. 2020;83:1118-1129.
- Freeman EE, Chamberlin GC, McMahon DE, et al. Dermatology COVID-19 registries: updates and future directions. Dermatol Clin. 2021;39:575-585.
- Guelimi R, Salle R, Dousset L, et al. Non-acral skin manifestations during the COVID-19 epidemic: COVIDSKIN study by the French Society of Dermatology. J Eur Acad Dermatol Venereol. 2021;35:E539-E541.
- Marzano AV, Genovese G, Moltrasio C, et al; Italian Skin COVID-19 Network of the Italian Society of Dermatology and Sexually Transmitted Diseases. The clinical spectrum of COVID-19 associated cutaneous manifestations: an Italian multicenter study of 200 adult patients. J Am Acad Dermatol. 2021;84:1356-1363.
- Sugai T, Fujita Y, Inamura E, et al. Prevalence and patterns of cutaneous manifestations in 1245 COVID-19 patients in Japan: a single-centre study. J Eur Acad Dermatol Venereol. 2022;36:E522-E524.
- Holmes Z, Courtney A, Lincoln M, et al. Rash morphology as a predictor of COVID‐19 severity: a systematic review of the cutaneous manifestations of COVID‐19. Skin Health Dis. 2022;2:E120. doi:10.1002/ski2.120
- Galván Casas C, Català A, Carretero Hernández G, et al. Classification of the cutaneous manifestations of COVID-19: a rapid prospective nationwide consensus study in Spain with 375 cases. Br J Dermatol. 2020;183:71-77.
- Garduño‑Soto M, Choreño-Parra, Cazarin-Barrientos Dermatological aspects of SARS‑CoV‑2 infection: mechanisms and manifestations. Arch Dermatol Res. 2021;313:611-622.
- Huynh T, Sanchez-Flores X, Yau J, et al. Cutaneous manifestations of SARS-CoV-2 Infection. Am J Clin Dermatol. 2022;23:277-286.
- Kanitakis J, Lesort C, Danset M, et al.
Chilblain-like acral lesions during the COVID-19 pandemic (“COVID toes”): histologic, immunofluorescence, and immunohistochemical study of 17 cases. J Am Acad Dermatol. 2020; 83:870-875. - Kolivras A, Thompson C, Pastushenko I, et al. A clinicopathological description of COVID-19-induced chilblains (COVID-toes) correlated with a published literature review. J Cutan Pathol. 2022;49:17-28.
- Roca-Ginés J, Torres-Navarro I, Sánchez-Arráez J, et al. Assessment of acute acral lesions in a case series of children and adolescents during the COVID-19 pandemic. 2020;156:992-997.
- Le Cleach L, Dousset L, Assier H, et al; French Society of Dermatology. Most chilblains observed during the COVID-19 outbreak occur in patients who are negative for COVID-19 on polymerase chain reaction and serology testing. Br J Dermatol. 2020;183:866-874.
- Discepolo V, Catzola A, Pierri L, et al. Bilateral chilblain-like lesions of the toes characterized by microvascular remodeling in adolescents during the COVID-19 pandemic. JAMA Netw Open. 2021;4:E2111369.
- Gehlhausen JR, Little AJ, Ko CJ, et al. Lack of association between pandemic chilblains and SARS-CoV-2 infection. Proc Natl Acad Sci U S A. 2022;119:e2122090119.
- Neri, Virdi, Corsini, et al Major cluster of paediatric ‘true’ primary chilblains during the COVID-19 pandemic: a consequence of lifestyle changes due to lockdown. J Eur Acad Dermatol Venereol. 2020;34:2630-2635.
- De Greef A, Choteau M, Herman A, et al. Chilblains observed during the COVID-19 pandemic cannot be distinguished from the classic, cold-related chilblains. Eur J Dermatol. 2022;32:377-383.
- Colmenero I, Santonja C, Alonso-Riaño M, et al. SARS-CoV-2 endothelial infection causes COVID-19 chilblains: histopathological, immunohistochemical and ultrastructural study of seven paediatric cases. Br J Dermatol. 2020;183:729-737.
- Quintero-Bustos G, Aguilar-Leon D, Saeb-Lima M. Histopathological and immunohistochemical characterization of skin biopsies from 41 SARS-CoV-2 (+) patients: experience in a Mexican concentration institute: a case series and literature review. Am J Dermatopathol. 2022;44:327-337.
- Arkin LM, Moon JJ, Tran JM, et al; COVID Human Genetic Effort. From your nose to your toes: a review of severe acute respiratory syndrome coronavirus 2 pandemic-associated pernio. J Invest Dermatol. 2021;141:2791-2796.
- Frumholtz L, Bouaziz JD, Battistella M, et al; Saint-Louis CORE (COvid REsearch). Type I interferon response and vascular alteration in chilblain-like lesions during the COVID-19 outbreak. Br J Dermatol. 2021;185:1176-1185.
- Hubiche T, Cardot-Leccia N, Le Duff F, et al. Clinical, laboratory, and interferon-alpha response characteristics of patients with chilblain-like lesions during the COVID-19 pandemic. JAMA Dermatol. 2021;157:202-206.
- Lesort C, Kanitakis J, Villani A, et al. COVID-19 and outbreak of chilblains: are they related? J Eur Acad Dermatol Venereol. 2020;34:E757-E758.
- Sanchez A, Sohier P, Benghanem S, et al. Digitate papulosquamous eruption associated with severe acute respiratory syndrome coronavirus 2 infection. JAMA Dermatol. 2020;156:819-820.
- Drago F, Broccolo F, Ciccarese G. Pityriasis rosea, pityriasis rosea-like eruptions, and herpes zoster in the setting of COVID-19 and COVID-19 vaccination. Clin Dermatol. 2022;S0738-081X(22)00002-5.
- Dursun R, Temiz SA. The clinics of HHV-6 infection in COVID-19 pandemic: pityriasis rosea and Kawasaki disease. Dermatol Ther. 2020;33:E13730.
- Nuno-Gonzalez A, Magaletsky K, Feito Rodríguez M, et al. Palmoplantar erythrodysesthesia: a diagnostic sign of COVID-19. J Eur Acad Dermatol Venereol. 2021;35:e247-e249.
- Sil A, Panigrahi A, Chandra A, et al. “COVID nose”: a unique post-COVID pigmentary sequelae reminiscing Chik sign: a descriptive case series. J Eur Acad Dermatol Venereol. 2022;36:E419-E421.
- Starace M, Iorizzo M, Sechi A, et al. Trichodynia and telogen effluvium in COVID-19 patients: results of an international expert opinion survey on diagnosis and management. JAAD Int. 2021;5:11-18.
- Wong-Chew RM, Rodríguez Cabrera EX, Rodríguez Valdez CA, et al. Symptom cluster analysis of long COVID-19 in patients discharged from the Temporary COVID-19 Hospital in Mexico City. Ther Adv Infect Dis. 2022;9:20499361211069264.
- Bardazzi F, Guglielmo A, Abbenante D, et al. New insights into alopecia areata during COVID-19 pandemic: when infection or vaccination could play a role. J Cosmet Dermatol. 2022;21:1796-1798.
- Christensen RE, Jafferany M. Association between alopecia areata and COVID-19: a systematic review. JAAD Int. 2022;7:57-61.
- Wollina U, Kanitakis J, Baran R. Nails and COVID-19: a comprehensive review of clinical findings and treatment. Dermatol Ther. 2021;34:E15100.
- Méndez-Flores S, Zaladonis A, Valdes-Rodriguez R. COVID-19 and nail manifestation: be on the lookout for the red half-moon nail sign. Int J Dermatol. 2020;59:1414.
- Alobaida S, Lam JM. Beau lines associated with COVID-19. CMAJ. 2020;192:E1040.
- Durmaz EÖ, Demirciog˘lu D. Fluorescence in the sclera, nails, and teeth secondary to favipiravir use for COVID-19 infections. J Clin Aesthet Dermatol. 2022;15:35-37.
- Brumfiel CM, DiLorenzo AM, Petronic-Rosic VM. Dermatologic manifestations of COVID-19-associated multisystem inflammatory syndrome in children. Clin Dermatol. 2021;39:329-333.
- Akçay N, Topkarcı Z, Menentog˘lu ME, et al. New dermatological findings of MIS-C: can mucocutaneous involvement be associated with severe disease course? Australas J Dermatol. 2022;63:228-234. doi:10.1111/ajd.13819
- Vogel TP, Top KA, Karatzios C, et al. Multisystem inflammatory syndrome in children and adults (MIS-C/A): case definition & guidelines for data collection, analysis, and presentation of immunization safety data. Vaccine. 2021;39:3037-3049.
- Conforti C, Dianzani C, Agozzino M, et al. Cutaneous manifestations in confirmed COVID-19 patients: a systematic review. Biology (Basel). 2020;9:449.
- Hubiche T, Le Duff F, Fontas E, et al. Relapse of chilblain-like lesions during the second wave of the COVID-19 pandemic: a cohort follow-up. Br J Dermatol. 2021;185:858-859.
- Fernandez-Nieto, Ortega-Quijano, Suarez-Valle, et al Lack of skin manifestations in COVID-19 hospitalized patients during the second epidemic wave in Spain: a possible association with a novel SARS-CoV-2 variant: a cross-sectional study. J Eur Acad Dermatol Venereol. 2021;35:E183-E185.
- Martinez-LopezA, Cuenca-Barrales, Montero-Vilchezet al Review of adverse cutaneous reactions of pharmacologic interventions for COVID-19: a guide for the dermatologist. J Am Acad Dermatol. 2020;83:1738-1748.
- Cutaneous side-effects of the potential COVID-19 drugs. Dermatol Ther. 2020;33:E13476.
- Mawhirt SL, Frankel D, Diaz AM. Cutaneous manifestations in adult patients with COVID-19 and dermatologic conditions related to the COVID-19 pandemic in health care workers. Curr Allerg Asthma Rep. 2020;20:75.
- Nguyen C, Young FG, McElroy D, et al. Personal protective equipment and adverse dermatological reactions among healthcare workers: survey observations from the COVID-19 pandemic. Medicine (Baltimore). 2022;101:E29003.
- Rathi SK, Dsouza JM. Maskne: a new acne variant in COVID-19 era. Indian J Dermatol. 2022;67:552-555.
- Damiani G, Girono L, Grada A, et al. COVID-19 related masks increase severity of both acne (maskne) and rosacea (mask rosacea): multi-center, real-life, telemedical, and observational prospective study. Dermatol Ther. 2021;34:E14848.
- Aram K, Patil A, Goldust M, et al. COVID-19 and exacerbation of dermatological diseases: a review of the available literature. Dermatol Ther. 2021;34:E15113.
- Samotij D, Gawron E, Szcze˛ch J, et al. Acrodermatitis continua of Hallopeau evolving into generalized pustular psoriasis following COVID-19: a case report of a successful treatment with infliximab in combination with acitretin. Biologics. 2021;15:107-113.
- Demiri J, Abdo M, Tsianakas A. Erythrodermic psoriasis after COVID-19 [in German]. Hautarzt. 2022;73:156-159.
- de Wijs LEM, Joustra MM, Olydam JI, et al. COVID-19 in patients with cutaneous immune-mediated diseases in the Netherlands: real-world observational data. J Eur Acad Dermatol Venereol. 2021;35:E173-E176.
- Marques NP, Maia CMF, Marques NCT, et al. Continuous increase of herpes zoster cases in Brazil during the COVID-19 pandemic. Oral Surg Oral Med Oral Pathol Oral Radiol. 2022;133:612-614.
- Rinaldi F, Trink A, Giuliani G, et al. Italian survey for the evaluation of the effects of coronavirus disease 2019 (COVID-19) pandemic on alopecia areata recurrence. Dermatol Ther (Heidelb). 2021;11:339-345.
- Rudnicka L, Rakowska A, Waskiel-Burnat A, et al. Mild-to-moderate COVID-19 is not associated with worsening of alopecia areata: a retrospective analysis of 32 patients. J Am Acad Dermatol. 2021;85:723-725.
- Drenovska K, Shahid M, Mateeva V, et al. Case report: Rowell syndrome-like flare of cutaneous lupus erythematosus following COVID-19 infection. Front Med (Lausanne). 2022;9:815743.
- Kawabe R, Tonomura K, Kotobuki Y, et al. Exacerbation of livedoid vasculopathy after coronavirus disease 2019. Eur J Dermatol. 2022;32:129-131. doi:10.1684/ejd.2022.4200
- McMahon DE, Kovarik CL, Damsky W, et al. Clinical and pathologic correlation of cutaneous COVID-19 vaccine reactions including V-REPP: a registry-based study. J Am Acad Dermatol. 2022;86:113-121.
- Avallone G, Quaglino P, Cavallo F, et al. SARS-CoV-2 vaccine-related cutaneous manifestations: a systematic review. Int J Dermatol. 2022;61:1187-1204. doi:10.1111/ijd.16063
- Gambichler T, Boms S, Susok L, et al. Cutaneous findings following COVID-19 vaccination: review of world literature and own experience. J Eur Acad Dermatol Venereol. 2022;36:172-180.
- Kroumpouzos G, Paroikaki ME, Yumeen S, et al. Cutaneous complications of mRNA and AZD1222 COVID-19 vaccines: a worldwide review. Microorganisms. 2022;10:624.
- Robinson L,Fu X,Hashimoto D, et al. Incidence of cutaneous reactions after messenger RNA COVID-19 vaccines. 2021;
- Wollina U, Chiriac A, Kocic H, et al. Cutaneous and hypersensitivity reactions associated with COVID-19 vaccination: a narrative review. Wien Med Wochenschr. 2022;172:63-69.
- Wei TS. Cutaneous reactions to COVID-19 vaccines: a review. JAAD Int. 2022;7:178-186.
- Katsikas Triantafyllidis K, Giannos P, Mian IT, et al. Varicella zoster virus reactivation following COVID-19 vaccination: a systematic review of case reports. Vaccines (Basel). 2021;9:1013.
- Maronese CA, Caproni M, Moltrasio C, et al. Bullous pemphigoid associated with COVID-19 vaccines: an Italian multicentre study. Front Med (Lausanne). 2022;9:841506.
- Cavazos A, Deb A, Sharma U, et al. COVID toes following vaccination. Proc (Bayl Univ Med Cent). 2022;35:476-479.
- Lesort C, Kanitakis J, Danset M, et al. Chilblain-like lesions after BNT162b2 mRNA COVID-19 vaccine: a case report suggesting that ‘COVID toes’ are due to the immune reaction to SARS-CoV-2. J Eur Acad Dermatol Venereol. 2021;35:E630-E632.
- Russo R, Cozzani E, Micalizzi C, et al. Chilblain-like lesions after COVID-19 vaccination: a case series. Acta Derm Venereol. 2022;102:adv00711. doi:10.2340/actadv.v102.2076
- Ortigosa LCM, Lenzoni FC, Suárez MV, et al. Hypersensitivity reaction to hyaluronic acid dermal filler after COVID-19 vaccination: a series of cases in São Paulo, Brazil. Int J Infect Dis. 2022;116:268-270.
- Agaronov A, Makdesi C, Hall CS. Acute generalized exanthematous pustulosis induced by Moderna COVID-19 messenger RNA vaccine. JAAD Case Rep. 2021;16:96-97.
- Dash S, Sirka CS, Mishra S, et al. COVID-19 vaccine-induced Stevens-Johnson syndrome. Clin Exp Dermatol. 2021;46:1615-1617.
- Huang Y, Tsai TF. Exacerbation of psoriasis following COVID-19 vaccination: report from a single center. Front Med (Lausanne). 2021;8:812010.
- Elamin S, Hinds F, Tolland J. De novo generalized pustular psoriasis following Oxford-AstraZeneca COVID-19 vaccine. Clin Exp Dermatol 2022;47:153-155.
- Abdelmaksoud A, Wollina U, Temiz SA, et al. SARS-CoV-2 vaccination-induced cutaneous vasculitis: report of two new cases and literature review. Dermatol Ther. 2022;35:E15458.
- Fritzen M, Funchal GDG, Luiz MO, et al. Leukocytoclastic vasculitis after exposure to COVID-19 vaccine. An Bras Dermatol. 2022;97:118-121.
- Vassallo C, Boveri E, Brazzelli V, et al. Cutaneous lymphocytic vasculitis after administration of COVID-19 mRNA vaccine. Dermatol Ther. 2021;34:E15076.
- Nazzaro G, Maronese CA. Urticarial vasculitis following mRNA anti-COVID-19 vaccine. Dermatol Ther. 2022;35:E15282.
- Hoshina D, Orita A. Sweet syndrome after severe acute respiratory syndrome coronavirus 2 mRNA vaccine: a case report and literature review. J Dermatol. 2022;49:E175-E176.
- Lemoine C, Padilla C, Krampe N, et al. Systemic lupus erythematous after Pfizer COVID-19 vaccine: a case report. Clin Rheumatol. 2022;41:1597-1601.
- Nguyen B, Lalama MJ, Gamret AC, et al. Cutaneous symptoms of connective tissue diseases after COVID-19 vaccination: a systematic review. Int J Dermatol. 2022;61:E238-E241.
- Gallo G, Mastorino L, Tonella L, et al. Alopecia areata after COVID-19 vaccination. Clin Exp Vaccine Res. 2022;11:129-132.
- Norimatsu Y, Norimatsu Y. A severe case of Trichophyton rubrum-caused dermatomycosis exacerbated after COVID-19 vaccination that had to be differentiated from pustular psoriasis. Med Mycol Case Rep. 2022;36:19-22.
- Yang K, Prussick L, Hartman R, et al. Acantholytic dyskeratosis post-COVID vaccination. Am J Dermatopathol. 2022;44:E61-E63.
- Koumaki D, Marinos L, Nikolaou V, et al. Lymphomatoid papulosis (LyP) after AZD1222 and BNT162b2 COVID-19 vaccines. Int J Dermatol. 2022;61:900-902.
- World Health Organization. Statement on the fourteenth meeting of the International Health Regulations (2005) Emergency Committee regarding the coronavirus disease (COVID-19) pandemic. Published January 30, 2023. Accessed April 12, 2023. https://www.who.int/news/item/30-01-2023-statement-on-the-fourteenth-meeting-of-the-international-health-regulations-(2005)-emergency-committee-regarding-the-coronavirus-disease-(covid-19)-pandemic
Practice Points
- During the COVID-19 pandemic, several skin diseases were reported in association with this new infectious disease and were classified mainly according to their morphologic aspect. However, the pathogenetic mechanisms often are unclear and the causal link of the culprit virus (SARS-CoV-2) not always well established.
- Currently, most skin manifestations related to COVID-19 are reported after vaccination against COVID-19; remarkably, many of them are similar to those attributed to the natural infection.
Should antenatal testing be performed in patients with a pre-pregnancy BMI ≥ 35?
Possibly. Elevated body mass index (BMI) is associated with an increased risk for stillbirth (strength of recommendation (SOR), B; Cohort studies and meta-analysis of cohort studies). Three studies found an association between elevated BMI and stillbirth and one did not. However, no studies demonstrate that antenatal testing in pregnant people with higher BMIs decreases stillbirth rates, or that no harm is caused by unnecessary testing or resultant interventions.
Still, in 2021, the American College of Obstetricians and Gynecologists (ACOG) suggested weekly antenatal testing may be considered from 34 weeks' 0 days' gestation for pregnant people with a BMI ≥ 40.0 kg/m2 and from 37 weeks' 0 days' gestation for pregnant people with a BMI between 35.0 and 39.9 kg/m2 (SOR, C; consensus guideline). Thus, doing the antenatal testing recommended in the ACOG guideline in an attempt to prevent stillbirth is reasonable, given evidence that elevated BMI is associated with stillbirth.
Evidence summary
Association between higher maternal BMI and increased risk for stillbirth
The purpose of antenatal testing is to decrease the risk for stillbirth between visits. Because of the resources involved and the risk for false-positives when testing low-risk patients, antenatal testing is reserved for pregnant people with higher risk for stillbirth.
In a retrospective cohort study of more than 2.8 million singleton births including 9,030 stillbirths, pregnant people with an elevated BMI had an increased risk for stillbirth compared with those with a normal BMI. The adjusted hazard ratio was 1.71 (95% confidence interval (CI), 1.62-1.83) for those with a BMI of 30.0 to 34.9 kg/m2; 2.04 (95% CI, 1.8-2.21) for those with a BMI of 35.0 to 39.9 kg/m2; and 2.50 (95% CI, 2.28-2.74) for those with a BMI ≥ 40 kg/m2.1
A meta-analysis of 38 studies, which included data on 16,274 stillbirths, found that a 5-unit increase in BMI was associated with an increased risk for stillbirth (relative risk, 1.24; 95% CI, 1.18-1.30).2
Another meta-analysis included 6 cohort studies involving more than 1 million pregnancies and 3 case-control studies involving 2,530 stillbirths and 2,837 controls from 1980-2005. There was an association between increasing BMI and stillbirth: the odds ratio (OR) was 1.47 (95% CI, 1.08-1.94) for those with a BMI of 25.0 to 29.9 kg/m2 and 2.07 (95% CI, 1.59-2.74) for those with a BMI ≥ 30.0, compared with those with a normal BMI.3
However, a retrospective cohort study of 182,362 singleton births including 442 stillbirths found no association between stillbirth and increasing BMI. The OR was 1.10 (95% CI, 0.90-1.36) for those with a BMI of 25.0 to 29.9 and 1.09 (95% CI, 0.87-1.37) for those with a BMI ≥ 30.0 kg/m2, compared with those with a normal BMI.4 However, this cohort study may have been underpowered to detect an association between stillbirth and BMI.
Recommendations from others
In 2021, ACOG suggested that weekly antenatal testing may be considered from 34 weeks' and 0 days' gestation for pregnant people with a BMI ≥ 40.0 kg/m2 and from 37 weeks' and 0 days' gestation for pregnant people with a BMI between 35.0 and 39.9 kg/m2.5 The 2021 ACOG Practice Bulletin on obesity in pregnancy rates this recommendation as Level C—based primarily on consensus and expert opinion.6
A 2018 Royal College of Obstetricians and Gynecologists Green-top Guideline recognizes “definitive recommendations for fetal surveillance are hampered by the lack of randomized controlled trials demonstrating that antepartum fetal surveillance decreases perinatal morbidity or mortality in late-term and post-term gestations…. There are no definitive studies determining the optimal type or frequency of such testing and no evidence specific for women with obesity.”7
A 2019 Society of Obstetricians and Gynecologists of Canada practice guideline states “stillbirth is more common with maternal obesity” and recommends “increased fetal surveillance … in the third trimester if reduced fetal movements are reported.” The guideline notes “the role for non-stress tests … in surveillance of well-being in this population is uncertain.” Also, for pregnant people with a BMI > 30 kg/m2, “assessment of fetal well-being is … recommended weekly from 37 weeks until delivery.” Finally, increased fetal surveillance is recommended in the setting of increased BMI and an abnormal pulsatility index of the umbilical artery and/or maternal uterine artery.8
Editor’s takeaway
Evidence demonstrates that increased maternal BMI is associated with increased stillbirths. However, evidence has not shown that third-trimester antenatal testing decreases this morbidity and mortality. Expert opinion varies, with ACOG recommending weekly antenatal testing from 34 and 37 weeks’ gestation, respectively, for pregnant people with BMIs of ≥ 40 kg/m2 and of 35 to 39.9 kg/m2. ●
- Yao R, Ananth C, Park B, et al; Perinatal Research Consortium. Obesity and the risk of stillbirth: a population-based cohort study. Am J Obstet Gynecol. 2014;210:e1-e9. doi: 10.1016/j. ajog. 2014.01.044
- Aune D, Saugstad O, Henriksen T, et al. Maternal body mass index and the risk of fetal death, stillbirth, and infant death: a systematic review and meta-analysis. JAMA. 2014;311:15361546. doi: 10.1001/jama.2014.2269
- Chu S, Kim S, Lau J, et al. Maternal obesity and risk of stillbirth: a meta-analysis. Am J Obstet Gynecol. 2007;197:223-228. doi: 10.1016/j.ajog.2007.03.027
- Mahomed K, Chan G, Norton M. Obesity and the risk of stillbirth—a reappraisal—a retrospective cohort study. Eur J Obstet Gynecol Reprod Biol. 2020;255:25-28. doi: 10.1016/j. ejogrb. 2020.09.044
- American College of Obstetricians and Gynecologists’ Committee on Obstetric Practice, Society for MaternalFetal Medicine. Indications for outpatient antenatal fetal surveillance: ACOG committee opinion, number 828. Obstet Gynecol. 2021;137:e177-e197. doi: 10.1097/ AOG.0000000000004407
- American College of Obstetricians and Gynecologists’ Committee on Practice Bulletins–Obstetrics. Obesity in pregnancy: ACOG practice bulletin, number 230. Obstet Gynecol. 2021;137:e128-e144. doi: 10.1097/ AOG.0000000000004395
- Denison F, Aedla N, Keag O, et al; Royal College of Obstetricians and Gynaecologists. Care of women with obesity in pregnancy: Green-top Guideline No. 72. BJOG. 2019;126:e62-e106. doi: 10.1111/1471-0528.15386
- Maxwell C, Gaudet L, Cassir G, et al. Guideline No. 391Pregnancy and maternal obesity part 1: pre-conception and prenatal care. J Obstet Gynaecol Can. 2019;41:1623-1640. doi: 10.1016/j.jogc. 2019.03.026
Possibly. Elevated body mass index (BMI) is associated with an increased risk for stillbirth (strength of recommendation (SOR), B; Cohort studies and meta-analysis of cohort studies). Three studies found an association between elevated BMI and stillbirth and one did not. However, no studies demonstrate that antenatal testing in pregnant people with higher BMIs decreases stillbirth rates, or that no harm is caused by unnecessary testing or resultant interventions.
Still, in 2021, the American College of Obstetricians and Gynecologists (ACOG) suggested weekly antenatal testing may be considered from 34 weeks' 0 days' gestation for pregnant people with a BMI ≥ 40.0 kg/m2 and from 37 weeks' 0 days' gestation for pregnant people with a BMI between 35.0 and 39.9 kg/m2 (SOR, C; consensus guideline). Thus, doing the antenatal testing recommended in the ACOG guideline in an attempt to prevent stillbirth is reasonable, given evidence that elevated BMI is associated with stillbirth.
Evidence summary
Association between higher maternal BMI and increased risk for stillbirth
The purpose of antenatal testing is to decrease the risk for stillbirth between visits. Because of the resources involved and the risk for false-positives when testing low-risk patients, antenatal testing is reserved for pregnant people with higher risk for stillbirth.
In a retrospective cohort study of more than 2.8 million singleton births including 9,030 stillbirths, pregnant people with an elevated BMI had an increased risk for stillbirth compared with those with a normal BMI. The adjusted hazard ratio was 1.71 (95% confidence interval (CI), 1.62-1.83) for those with a BMI of 30.0 to 34.9 kg/m2; 2.04 (95% CI, 1.8-2.21) for those with a BMI of 35.0 to 39.9 kg/m2; and 2.50 (95% CI, 2.28-2.74) for those with a BMI ≥ 40 kg/m2.1
A meta-analysis of 38 studies, which included data on 16,274 stillbirths, found that a 5-unit increase in BMI was associated with an increased risk for stillbirth (relative risk, 1.24; 95% CI, 1.18-1.30).2
Another meta-analysis included 6 cohort studies involving more than 1 million pregnancies and 3 case-control studies involving 2,530 stillbirths and 2,837 controls from 1980-2005. There was an association between increasing BMI and stillbirth: the odds ratio (OR) was 1.47 (95% CI, 1.08-1.94) for those with a BMI of 25.0 to 29.9 kg/m2 and 2.07 (95% CI, 1.59-2.74) for those with a BMI ≥ 30.0, compared with those with a normal BMI.3
However, a retrospective cohort study of 182,362 singleton births including 442 stillbirths found no association between stillbirth and increasing BMI. The OR was 1.10 (95% CI, 0.90-1.36) for those with a BMI of 25.0 to 29.9 and 1.09 (95% CI, 0.87-1.37) for those with a BMI ≥ 30.0 kg/m2, compared with those with a normal BMI.4 However, this cohort study may have been underpowered to detect an association between stillbirth and BMI.
Recommendations from others
In 2021, ACOG suggested that weekly antenatal testing may be considered from 34 weeks' and 0 days' gestation for pregnant people with a BMI ≥ 40.0 kg/m2 and from 37 weeks' and 0 days' gestation for pregnant people with a BMI between 35.0 and 39.9 kg/m2.5 The 2021 ACOG Practice Bulletin on obesity in pregnancy rates this recommendation as Level C—based primarily on consensus and expert opinion.6
A 2018 Royal College of Obstetricians and Gynecologists Green-top Guideline recognizes “definitive recommendations for fetal surveillance are hampered by the lack of randomized controlled trials demonstrating that antepartum fetal surveillance decreases perinatal morbidity or mortality in late-term and post-term gestations…. There are no definitive studies determining the optimal type or frequency of such testing and no evidence specific for women with obesity.”7
A 2019 Society of Obstetricians and Gynecologists of Canada practice guideline states “stillbirth is more common with maternal obesity” and recommends “increased fetal surveillance … in the third trimester if reduced fetal movements are reported.” The guideline notes “the role for non-stress tests … in surveillance of well-being in this population is uncertain.” Also, for pregnant people with a BMI > 30 kg/m2, “assessment of fetal well-being is … recommended weekly from 37 weeks until delivery.” Finally, increased fetal surveillance is recommended in the setting of increased BMI and an abnormal pulsatility index of the umbilical artery and/or maternal uterine artery.8
Editor’s takeaway
Evidence demonstrates that increased maternal BMI is associated with increased stillbirths. However, evidence has not shown that third-trimester antenatal testing decreases this morbidity and mortality. Expert opinion varies, with ACOG recommending weekly antenatal testing from 34 and 37 weeks’ gestation, respectively, for pregnant people with BMIs of ≥ 40 kg/m2 and of 35 to 39.9 kg/m2. ●
Possibly. Elevated body mass index (BMI) is associated with an increased risk for stillbirth (strength of recommendation (SOR), B; Cohort studies and meta-analysis of cohort studies). Three studies found an association between elevated BMI and stillbirth and one did not. However, no studies demonstrate that antenatal testing in pregnant people with higher BMIs decreases stillbirth rates, or that no harm is caused by unnecessary testing or resultant interventions.
Still, in 2021, the American College of Obstetricians and Gynecologists (ACOG) suggested weekly antenatal testing may be considered from 34 weeks' 0 days' gestation for pregnant people with a BMI ≥ 40.0 kg/m2 and from 37 weeks' 0 days' gestation for pregnant people with a BMI between 35.0 and 39.9 kg/m2 (SOR, C; consensus guideline). Thus, doing the antenatal testing recommended in the ACOG guideline in an attempt to prevent stillbirth is reasonable, given evidence that elevated BMI is associated with stillbirth.
Evidence summary
Association between higher maternal BMI and increased risk for stillbirth
The purpose of antenatal testing is to decrease the risk for stillbirth between visits. Because of the resources involved and the risk for false-positives when testing low-risk patients, antenatal testing is reserved for pregnant people with higher risk for stillbirth.
In a retrospective cohort study of more than 2.8 million singleton births including 9,030 stillbirths, pregnant people with an elevated BMI had an increased risk for stillbirth compared with those with a normal BMI. The adjusted hazard ratio was 1.71 (95% confidence interval (CI), 1.62-1.83) for those with a BMI of 30.0 to 34.9 kg/m2; 2.04 (95% CI, 1.8-2.21) for those with a BMI of 35.0 to 39.9 kg/m2; and 2.50 (95% CI, 2.28-2.74) for those with a BMI ≥ 40 kg/m2.1
A meta-analysis of 38 studies, which included data on 16,274 stillbirths, found that a 5-unit increase in BMI was associated with an increased risk for stillbirth (relative risk, 1.24; 95% CI, 1.18-1.30).2
Another meta-analysis included 6 cohort studies involving more than 1 million pregnancies and 3 case-control studies involving 2,530 stillbirths and 2,837 controls from 1980-2005. There was an association between increasing BMI and stillbirth: the odds ratio (OR) was 1.47 (95% CI, 1.08-1.94) for those with a BMI of 25.0 to 29.9 kg/m2 and 2.07 (95% CI, 1.59-2.74) for those with a BMI ≥ 30.0, compared with those with a normal BMI.3
However, a retrospective cohort study of 182,362 singleton births including 442 stillbirths found no association between stillbirth and increasing BMI. The OR was 1.10 (95% CI, 0.90-1.36) for those with a BMI of 25.0 to 29.9 and 1.09 (95% CI, 0.87-1.37) for those with a BMI ≥ 30.0 kg/m2, compared with those with a normal BMI.4 However, this cohort study may have been underpowered to detect an association between stillbirth and BMI.
Recommendations from others
In 2021, ACOG suggested that weekly antenatal testing may be considered from 34 weeks' and 0 days' gestation for pregnant people with a BMI ≥ 40.0 kg/m2 and from 37 weeks' and 0 days' gestation for pregnant people with a BMI between 35.0 and 39.9 kg/m2.5 The 2021 ACOG Practice Bulletin on obesity in pregnancy rates this recommendation as Level C—based primarily on consensus and expert opinion.6
A 2018 Royal College of Obstetricians and Gynecologists Green-top Guideline recognizes “definitive recommendations for fetal surveillance are hampered by the lack of randomized controlled trials demonstrating that antepartum fetal surveillance decreases perinatal morbidity or mortality in late-term and post-term gestations…. There are no definitive studies determining the optimal type or frequency of such testing and no evidence specific for women with obesity.”7
A 2019 Society of Obstetricians and Gynecologists of Canada practice guideline states “stillbirth is more common with maternal obesity” and recommends “increased fetal surveillance … in the third trimester if reduced fetal movements are reported.” The guideline notes “the role for non-stress tests … in surveillance of well-being in this population is uncertain.” Also, for pregnant people with a BMI > 30 kg/m2, “assessment of fetal well-being is … recommended weekly from 37 weeks until delivery.” Finally, increased fetal surveillance is recommended in the setting of increased BMI and an abnormal pulsatility index of the umbilical artery and/or maternal uterine artery.8
Editor’s takeaway
Evidence demonstrates that increased maternal BMI is associated with increased stillbirths. However, evidence has not shown that third-trimester antenatal testing decreases this morbidity and mortality. Expert opinion varies, with ACOG recommending weekly antenatal testing from 34 and 37 weeks’ gestation, respectively, for pregnant people with BMIs of ≥ 40 kg/m2 and of 35 to 39.9 kg/m2. ●
- Yao R, Ananth C, Park B, et al; Perinatal Research Consortium. Obesity and the risk of stillbirth: a population-based cohort study. Am J Obstet Gynecol. 2014;210:e1-e9. doi: 10.1016/j. ajog. 2014.01.044
- Aune D, Saugstad O, Henriksen T, et al. Maternal body mass index and the risk of fetal death, stillbirth, and infant death: a systematic review and meta-analysis. JAMA. 2014;311:15361546. doi: 10.1001/jama.2014.2269
- Chu S, Kim S, Lau J, et al. Maternal obesity and risk of stillbirth: a meta-analysis. Am J Obstet Gynecol. 2007;197:223-228. doi: 10.1016/j.ajog.2007.03.027
- Mahomed K, Chan G, Norton M. Obesity and the risk of stillbirth—a reappraisal—a retrospective cohort study. Eur J Obstet Gynecol Reprod Biol. 2020;255:25-28. doi: 10.1016/j. ejogrb. 2020.09.044
- American College of Obstetricians and Gynecologists’ Committee on Obstetric Practice, Society for MaternalFetal Medicine. Indications for outpatient antenatal fetal surveillance: ACOG committee opinion, number 828. Obstet Gynecol. 2021;137:e177-e197. doi: 10.1097/ AOG.0000000000004407
- American College of Obstetricians and Gynecologists’ Committee on Practice Bulletins–Obstetrics. Obesity in pregnancy: ACOG practice bulletin, number 230. Obstet Gynecol. 2021;137:e128-e144. doi: 10.1097/ AOG.0000000000004395
- Denison F, Aedla N, Keag O, et al; Royal College of Obstetricians and Gynaecologists. Care of women with obesity in pregnancy: Green-top Guideline No. 72. BJOG. 2019;126:e62-e106. doi: 10.1111/1471-0528.15386
- Maxwell C, Gaudet L, Cassir G, et al. Guideline No. 391Pregnancy and maternal obesity part 1: pre-conception and prenatal care. J Obstet Gynaecol Can. 2019;41:1623-1640. doi: 10.1016/j.jogc. 2019.03.026
- Yao R, Ananth C, Park B, et al; Perinatal Research Consortium. Obesity and the risk of stillbirth: a population-based cohort study. Am J Obstet Gynecol. 2014;210:e1-e9. doi: 10.1016/j. ajog. 2014.01.044
- Aune D, Saugstad O, Henriksen T, et al. Maternal body mass index and the risk of fetal death, stillbirth, and infant death: a systematic review and meta-analysis. JAMA. 2014;311:15361546. doi: 10.1001/jama.2014.2269
- Chu S, Kim S, Lau J, et al. Maternal obesity and risk of stillbirth: a meta-analysis. Am J Obstet Gynecol. 2007;197:223-228. doi: 10.1016/j.ajog.2007.03.027
- Mahomed K, Chan G, Norton M. Obesity and the risk of stillbirth—a reappraisal—a retrospective cohort study. Eur J Obstet Gynecol Reprod Biol. 2020;255:25-28. doi: 10.1016/j. ejogrb. 2020.09.044
- American College of Obstetricians and Gynecologists’ Committee on Obstetric Practice, Society for MaternalFetal Medicine. Indications for outpatient antenatal fetal surveillance: ACOG committee opinion, number 828. Obstet Gynecol. 2021;137:e177-e197. doi: 10.1097/ AOG.0000000000004407
- American College of Obstetricians and Gynecologists’ Committee on Practice Bulletins–Obstetrics. Obesity in pregnancy: ACOG practice bulletin, number 230. Obstet Gynecol. 2021;137:e128-e144. doi: 10.1097/ AOG.0000000000004395
- Denison F, Aedla N, Keag O, et al; Royal College of Obstetricians and Gynaecologists. Care of women with obesity in pregnancy: Green-top Guideline No. 72. BJOG. 2019;126:e62-e106. doi: 10.1111/1471-0528.15386
- Maxwell C, Gaudet L, Cassir G, et al. Guideline No. 391Pregnancy and maternal obesity part 1: pre-conception and prenatal care. J Obstet Gynaecol Can. 2019;41:1623-1640. doi: 10.1016/j.jogc. 2019.03.026
Evaluating patients with breast concerns: Lump, pain, and mastitis
The vast majority of symptomatic breast conditions are benign, with the most common symptoms being palpable mass and breast pain. Clinicians, including primary care clinicians and gynecologists, play a crucial role by performing the initial assessment and subsequent therapies and referrals and serve as the mediator between the specialists and by being the patient’s spokesperson. It is therefore important for clinicians to be aware of the various possible causes of these breast symptoms, to know which imaging tests to order, and also to understand the indications for biopsies and surgical referral.
Common types of breast lumps: Imaging workup and management
Accounting for 8% of women who present with breast symptoms, breast lump is the second most common symptom after breast pain.1 The positive likelihood ratio of finding breast cancer is highest among women with breast lumps compared with any other breast symptoms. Therefore, anxiety is related to this symptom, and a thorough evaluation is recommended.1 Cysts, fibroadenoma, and fat necrosis are 3 common benign causes of breast lumps.2
In this section, we review clinical presentation, imaging workup, and management strategies for common types of breast lumps.
CASE 1 Woman with tender breast lump
A 45-year-old woman presents with a breast lump of 6 months’ duration that is associated with a change in size with the menstrual cycle and pain. Clinical examination reveals a 4 x 4.5–cm mass in the right breast in the retroareolar region, which is smooth with some tenderness on palpation.
Breast cyst
According to the American College of Radiology appropriateness criteria for an adult woman 40 years of age or older who presents with a palpable breast mass, the initial imaging study is diagnostic mammography with or without digital tomosynthesis, usually followed by a directed ultrasound. If the mammogram is suspicious or highly suggestive of malignancy, or in cases where the mammogram does not show an abnormality, the next recommended step is breast ultrasonography. Any suspicious findings on ultrasound or mammogram should be followed by an image guided biopsy. Ultrasonography also may be appropriate if the mammogram findings are benign or probably benign.
For an adult woman younger than age 30 who presents with a palpable breast mass, breast ultrasonography is the appropriate initial imaging study. If the ultrasound is suspicious or highly suggestive of malignancy, then performing diagnostic mammography with or without digital tomosynthesis or ultrasound-guided core needle biopsy of the mass are both considered appropriate. However, no further imaging is recommended if the ultrasound is benign, probably benign, or negative. Breast ultrasonography or mammography is appropriate as the initial imaging test for adult women aged 30 to 39 years who present with a palpable breast mass.3,4
Approximately 50% of women after age 30 may develop fibrocystic breast disease, and 20% of them can present with pain or lump due to a macrocysts. Simple cysts must be distinguished from complex cysts with the help of ultrasound as the latter are associated with 23% to 31% increased risk of malignancy.
In this 45-year-old patient, the initial mammogram demonstrated a circumscribed mass underneath the area of palpable concern (FIGURE 1a, 1b). Targeted breast ultrasonography was performed for further assessment, which depicted the mass as a benign simple cyst (FIGURE 1c).
On ultrasound, a simple cyst is an anechoic, well-circumscribed mass with a thin capsule and with increased through transmission. Patients with small and asymptomatic simple cysts do not need imaging follow-up and can return for routine screening mammograms.
A breast surgeon, radiologist, or gynecologist can perform percutaneous aspiration if a cyst is large and symptomatic. A cyst with low-level internal echoes, fluid-fluid, or fluid-debris levels is considered a complicated cyst. Differential diagnosis also includes hematoma, fat necrosis, abscess, and galactocele, depending on the clinical presentation. Fine-needle aspiration or short-interval follow-up5,6 is appropriate for complicated cysts, while incision and drainage is indicated in patients with infected cysts and abscesses. A cyst with a solid component is considered a cystic, solid mass, and core needle biopsy is recommended. The differential diagnosis for cysts with solid components includes intracystic papilloma, papillary carcinoma, ductal carcinoma in situ, and necrotic cancers.5,6
Continue to: CASE 2 Painless breast mass in a young woman...
CASE 2 Painless breast mass in a young woman
A 22-year-old woman presents with a 2-month history of breast lump, which is not associated with pain or nipple discharge. On examination, there is a 2 x 2–cm mass in the right breast at 12 o’clock, 2 cm from the nipple, which is mobile, smooth, and nontender on palpation.
Fibroadenoma
In this 22-year-old, the initial imaging of choice is breast ultrasonography. Breast ultrasonography can differentiate a cystic mass from a solid mass, and it does not involve radiation. Right breast targeted ultrasound showed a circumscribed oval homogeneous hypoechoic mass that is wider than tall (FIGURE 2). The patient desired surgical removal, and a pre-lumpectomy core needle biopsy revealed a fibroadenoma.
Fibroadenoma is the most common benign tumor of the breast. It is most often encountered in premenopausal women. Patients present with a painless breast lump, which is smooth and mobile on palpation. Fibroadenoma can be followed expectantly with repeat ultrasound (to assess over time for growth) if it is small and asymptomatic. No further action is needed if it remains stable. If a patient desires surgical excision, a core needle biopsy is usually performed before lumpectomy.
Excisional biopsy or removal of the mass is recommended if the mass is greater than 3 or 4 cm, is symptomatic, or if there is an increase in size that raises clinical concern for phyllodes tumor. Imaging features that are concerning for phyllodes tumors are size greater than 3 cm, indistinct or microlobulated margins, and heterogeneous echo pattern.7,8 In cases in which the imaging features are concerning for phyllodes tumor and a core needle biopsy is not definitive, wide surgical excision is recommended for definitive diagnosis.8
CASE 3 Patient develops breast mass post-surgery
A 45-year-old woman presents with a tender left breast mass that she noticed 2 months after breast reduction surgery. It has been increasing in size since. On clinical examination, a 4 x 4–cm mass is found at the surgical scar site, which is indurated on palpation and tender.
Fat necrosis
In this 45-year-old, the initial test of choice is diagnostic mammography, which showed a somewhat circumscribed area with fat under the palpable marker (FIGURE 3a). Breast ultrasonography was performed for further evaluation, which was inconclusive as the ultrasound showed ill-defined areas of mixed echogenicity (FIGURE 3b). Breast magnetic resonance imaging (MRI) clearly demonstrated fat necrosis in the area of the palpable lump (FIGURE 3c).
Fat necrosis of the breast is an inflammatory process that is seen after breast trauma or surgery. It can present as an incidental mammogram finding or a palpable mass. The patient may give a history of trauma, breast reduction surgery, or breast cancer surgery followed by radiation treatment. On clinical examination, fat necrosis occasionally can present as a firm mass with skin retraction or swelling concerning for cancer. Imaging features are variable depending on the stage of fat necrosis and inflammation.9-11
A mammogram may demonstrate a circumscribed fat-containing mass, an ill-defined mass, asymmetry or calcified oil cyst, and dystrophic calcifications. On ultrasound, fat necrosis can appear as anechoic or hypoechoic or as a complicated cyst or a mixed cystic, solid mass. MRI demonstrates a circumscribed or irregular fat-containing mass, with or without enhancement, and architectural distortion.
When the imaging features are clearly benign—for example, a circumscribed fat-containing mass on mammogram or on ultrasound or, on MRI, marked hypointensity of fat in the center of a circumscribed mass when compared with surrounding fat (keyhole sign)—no further follow-up is needed. When the imaging features are indeterminate, however, a short-interval follow-up can be considered. In cases with irregular fat-containing mass with enhancement, core needle biopsy is indicated to exclude cancer. If the workup remains inconclusive and the level of clinical suspicion is high, surgical excision can be performed for a definitive diagnosis.12
Continue to: Investigating breast pain: Imaging workup and management...
Investigating breast pain: Imaging workup and management
Breast pain, or mastalgia, is the most common concern of women presenting to a breast clinic and accounts for approximately half of such encounters.13 Causes of breast pain include hormonal changes, fibrocystic changes, musculoskeletal causes (such as costochondritis), lack of support, infection, and injury. While mastalgia often causes patient concern, the risk of malignancy in a woman presenting with breast pain alone is low. Still, it is essential to rule out other findings suspicious for cancer (mass, skin changes, or nipple discharge) with a thorough history and breast examination.
In this section, we review clinical presentation, imaging workup, and management for breast pain.
CASE 4 Woman with noncyclic breast pain
A 26-year-old woman presents to the clinic with mastalgia. The pain is noncyclic and primarily located in the upper outer quadrant of her left breast. There is no history of breast cancer in her family. She has no suspicious findings on the breast examination.
Mastalgia
The test of choice for this 26-year-old with focal left breast pain is targeted breast ultrasound. The patient’s ultrasound image showed no suspicious findings or solid or cystic mass (FIGURE 4).
Two important characteristics of breast pain are whether it is noncyclical and whether it is focal. According to the American College of Radiology, no breast imaging is recommended for clinically insignificant cyclical, nonfocal (greater than 1 quadrant)/diffuse pain, as this type of mastalgia is not associated with malignancy.14
For patients age 40 or older, if they are not up to date with their annual screening mammogram, then a mammogram should be performed. An imaging workup is warranted for clinically significant mastalgia that is noncyclical and focal. Even then, no malignancy is identified in most patients with clinically significant mastalgia; in patients with breast pain as their only symptom, the prevalence of breast cancer is 0% to 3.0%.15-19
The initial imaging modality differs by patient age: younger than 30 years, ultrasonography; between 30 and 40 years, mammography or ultrasonography; and older than 40 years, mammography first followed by ultrasonography.14
Treatment of breast pain is primarily symptomatic, and evidence for specific treatments is generally lacking. Cyclical breast pain resolves spontaneously in 20% to 30% of women, while noncyclical pain responds poorly to treatment but resolves spontaneously in half of women.20 Reassurance is important and wearing a supportive bra often can alleviate breast pain. In addition, reducing caffeine intake can be helpful.
As a first-line treatment, both topical (diclofenac) and oral nonsteroidal anti-inflammatory drugs effectively can relieve breast pain. Supplements and herbal remedies (for example, evening primrose oil, vitamin E, flaxseed) have varying effectiveness and are of questionable benefit as few have trials to support their effectiveness.4 Danazol and tamoxifen have been shown to have some benefits but they also have adverse effects.20 Surgery does not play a role in the treatment of mastalgia.
CASE 5 Breastfeeding woman with breast pain
A 27-year-old postpartum woman presents with concerns for redness and pain in the upper inner left breast. She has been breastfeeding for the past few months. Breast examination demonstrates a 5-cm area of erythema and warmth but no fluctuance or masses.
Lactational mastitis
Targeted ultrasonography is the test of choice for this 27-year-old patient with focal breast pain, and the imaging revealed edema of subcutaneous tissues and ill-defined hypoechoic areas, likely inflamed fat lobules (FIGURE 5). These findings suggest uncomplicated lactational mastitis, which can be treated with antibiotics. Generally, the mastitis will improve within days of starting the antibiotics; if it does not improve, repeat examination and ultrasound should be performed to look for formation of an abscess that may require aspiration.
Continue to: CASE 6 Woman with painful periareolar mass...
CASE 6 Woman with painful periareolar mass
A 42-year-old perimenopausal woman describes having pain near the nipple of her right breast. She is a smoker and has no history of breast cancer in her family. Examination demonstrates a palpable, erythematous, painful, 3-cm periareolar fluctuant mass.
Nonpuerperal periareolar abscess
Appropriate initial imaging for this 42-year-old patient with focal pain is a diagnostic mammogram, which showed skin thickening and a retroareolar mass (FIGURE 6a). Further evaluation with targeted ultrasound showed a thick-walled anechoic collection with echoes compatible with an abscess (FIGURE 6b).
Mammographic findings in a patient with mastitis may be normal or demonstrate skin and trabecular thickening. Ultrasound imaging may show dilated ducts and heterogeneous tissue secondary to inflammation and edema without a discrete fluid collection. In cases with breast abscess, in addition to the mammographic findings described above, a mass, or an asymmetry, may be seen, most commonly in a subareolar location. On ultrasound, a hypoechoic collection with mobile debris, no internal flow on Doppler, and thick hypervascular walls can be seen with abscess, occasionally giving the appearance of a complicated cyst or a mixed cystic, solid mass.
The most important differential for mastitis is inflammatory breast cancer. Most cancers appear solid but can have central necrosis, mimicking a complicated cystic mass on ultrasound. The location for mastitis or abscess is most frequently subareolar. The presence of microcalcifications in a mass indicates the possibility of cancer.
Contrast-enhanced MRI can be helpful to differentiate between infection and cancer, with cancers showing initial early enhancement and washout kinetics compared with infected collections that show no enhancement or peripheral enhancement with a plateau or persistent enhancement curves. When clinical and imaging findings are unchanged after treatment of mastitis and abscesses, a core needle biopsy should be performed.21,22
There are 2 categories of mastitis and breast abscess: lactational and nonpuerperal (all mastitis that occurs outside the lactational period). The World Health Organization definition of puerperal mastitis includes pain, local redness, warmth and swelling of the breast (usually unilateral), fever, and malaise.4 Concerning etiology, epithelial lesions in the nipple area caused by breastfeeding can allow pathogens to enter and cause infection. The most common microorganism is Staphylococcus aureus.4 Continued emptying of the breast is important, combined with early antibiotic therapy (dicloxacillin is often the first line; if the patient is penicillin allergic, use a macrolide such as clindamycin). If no improvement is seen in 48 to 72 hours, imaging should be performed.
In most cases, continuation of breastfeeding is possible. If mastitis has evolved into an abscess in a lactating woman, it can be aspirated under ultrasound guidance. Incision and drainage should be avoided unless the abscess persists after multiple aspiration attempts, it is large, or if the overlying skin is thin or otherwise appears nonviable.
Nonpuerperal mastitis includes peripheral, periductal, and idiopathic granulomatous mastitis (IGM). Peripheral mastitis behaves like infections/abscesses in other soft tissues, responds well to treatment (antibiotics and percutaneous drainage), and is less likely to recur than periductal mastitis and IGM.21,23
Periductal mastitis and abscess, also known as Zuska disease, has a pathogenesis distinct from other forms of mastitis. Squamous metaplasia of the usual cuboidal epithelium of the breast ducts leads to keratin plugging that can cause infection.23 Risk factors include obesity, smoking, and macromastia. The typical presentation of Zuska disease is a woman with a history of chronic smoking and/or a congenital cleft in the central nipple.23 Periareolar signs of inflammation (redness, swelling, warmth) may be accompanied by an abscess. These can recur and lead to chronic fistula formation, especially if there is a history of intervention (such as aspiration, incision, and drainage).
Treatment of Zuska disease includes symptom relief and antibiotics. If S aureus is present, infection with methicillin-resistant S aureus is likely, and treatment with clindamycin or amoxicillin/clavulanic acid is preferred. If abscess is present, aspiration (preferred, often under ultrasound guidance) or incision and drainage (if the skin is compromised) may be required. If disease is recurrent or associated with a chronically draining fistula, surgical intervention may be warranted, in which resolution requires removing the keratin-plugged ducts in and immediately below the central core of the nipple. Given the association between Zuska disease and smoking, cessation should be encouraged, although there is no guarantee that this will resolve the issue.23
Continue to: CASE 7 Patient with breast pain and swelling...
CASE 7 Patient with breast pain and swelling
A 39-year-old woman presents with left breast swelling and pain of 1 month’s duration. On examination, there is a 6-cm area of edema, induration, and erythema.
Granulomatous mastitis
A diagnostic mammogram and ultrasound demonstrated an ill-defined hypoechoic mass (FIGURE 7a). Ultrasound-guided biopsy was performed, which showed granulomatous mastitis, negative for fungus and acid-fast bacilli. The patient was treated with prednisone and gradually improved (FIGURE 7b).
Granulomatous mastitis (GM) is a rare benign inflammatory process, with etiologies that include fungal infections, tuberculosis, Wegener granulomatosis, sarcoidosis, and idiopathic causes. Imaging can be nonspecific and show variable features. Mammograms can appear normal or show asymmetry or mass and skin thickening. Ultrasound can show heterogeneous parenchyma, ill-defined hypoechoic collection, or a mass with margins that can be circumscribed or indistinct or with tubular extensions, with or without overlying skin thickening, fistulas, and reactive lymph nodes.24
In this clinical setting, the differential diagnosis includes infectious mastitis, inflammatory breast cancer, foreign body injection granulomas, and diabetic mastopathy. Treatment involves drainage and fluid culture if there is a collection on imaging. A core biopsy is performed if imaging demonstrates a solid mass or fluid culture is negative and symptoms persist or recur. Oral steroids represent the mainstay of treatment if a core biopsy shows GM. However, immunosuppressants, including methotrexate, and surgery are options if initial treatment is not helpful.25,26
Conclusion
Breast symptoms are common reasons for patient visits to obstetricians and gynecologists. With a good understanding of the various symptomatic breast diseases and conditions, and by having a close collaboration with radiologists and breast surgeons, clinicians can provide excellent care to these patients and thereby improve patient outcomes and satisfaction. ●
- Eberl MM, Phillips RL Jr, Lamberts H, et al. Characterizing breast symptoms in family practice. Ann Fam Med. 2008;6:528-533.
- Malherbe F, Nel D, Molabe H, et al. Palpable breast lumps: an age-based approach to evaluation and diagnosis. S Afr Fam Pract (2022). 2022;64:e1-e5.
- Expert Panel on Breast Imaging; Klein KA, Kocher M, Lourenco AP, et al. American College of Radiology ACR appropriateness criteria: palpable breast masses. Accessed February 15, 2023. https://acsearch.acr.org/docs/69495/Narrative/
- Stachs A, Stubert J, Reimer T, et al. Benign breast disease in women. Dtsch Arztebl Int. 2019;116:565574.
- Hines N, Slanetz PJ, Eisenberg RL. Cystic masses of the breast. AJR Am J Roentgenol. 2010;194:W122133.
- Berg WA. Reducing unnecessary biopsy and follow-up of benign cystic breast lesions. Radiology. 2020;295:52-53.
- Duman L, Gezer NS, Balcı P, et al. Differentiation between phyllodes tumors and fibroadenomas based on mammographic sonographic and MRI features. Breast Care. 2016;11:123-127.
- Lerwill MF, Lee AHS, Tan PH. Fibroepithelial tumours of the breast—a review. Virchows Arch. 2022;480:45-63.
- Vasei N, Shishegar A, Ghalkhani F, et al. Fat necrosis in the breast: a systematic review of clinical. Lipids Health Dis. 2019;18:139.
- Kerridge WD, Kryvenko ON, Thompson A, et al. Fat necrosis of the breast: a pictorial review of the mammographic, ultrasound, CT, and MRI findings with histopathologic correlation. Radiol Res Pract. 2015;2015:613139.
- Taboada JL, Stephens TW, Krishnamurthy S, et al. The many faces of fat necrosis in the breast. AJR Am J Roentgenol. 2009;192:815-825.
- Tan PH, Lai LM, Carrington EV, et al. Fat necrosis of the breast—a review. Breast. 2006;15:313-318.
- Holbrook AI. Breast pain, a common grievance: guidance to radiologists. AJR Am J Roentgenol. 2020;214:259-264.
- Expert Panel on Breast Imaging; Moy L, Heller SL, Bailey L, et al. ACR appropriateness criteria: palpable breast masses. J Am Coll Radiol. 2017;14:S203-S224.
- Chetlen AL, Kapoor MM, Watts MR. Mastalgia: imaging workup appropriateness. Acad Radiol. 2017;24:345-349.
- Arslan M, Kucukerdem HS, Can H, et al. Retrospective analysis of women with only mastalgia. J Breast Health. 2016;12:151-154.
- Fariselli G, Lepera P, Viganotti G, et al. Localized mastalgia as presenting symptom in breast cancer. Eur J Surg Oncol. 1988;14:213-215.
- Leddy R, Irshad A, Zerwas E, et al. Role of breast ultrasound and mammography in evaluating patients presenting with focal breast pain in the absence of a palpable lump. Breast J. 2013;19:582-589.
- Leung JW, Kornguth PJ, Gotway MB. Utility of targeted sonography in the evaluation of focal breast pain. J Ultrasound Med. 2002;21:521-526.
- Goyal A. Breast pain. BMJ Clin Evid. 2011; 2011:0812.
- Kasales CJ, Han B, Smith Jr JS, et al. Nonpuerperal mastitis and subareolar abscess of the breast. AJR Am J Roentgenol. 2014;202:W133-W139.
- Mahoney MC, Ingram AD. Breast emergencies: types, imaging features, and management. AJR Am J Roentgenol. 2014;202:W390-W399.
- Snider HC. Management of mastitis, abscess, and fistula. Surg Clin North Am. 2022;102:1103-1116.
- Oztekin PS, Durhan G, Kosar PN, et al. Imaging findings in patients with granulomatous mastitis. Iran J Radiol. 2016;13:e33900.
- Pluguez-Turull CW, Nanyes JE, Quintero CJ, et al. Idiopathic granulomatous mastitis: manifestations at multimodality imaging and pitfalls. Radiographics. 2018;38:330-356.
- Hovanessian-Larsen LJ, Peyvandi B, Klipfel N, et al. Granulomatous lobular mastitis: imaging, diagnosis, and treatment. AJR Am J Roentgenol. 2009;193:574-581.
The vast majority of symptomatic breast conditions are benign, with the most common symptoms being palpable mass and breast pain. Clinicians, including primary care clinicians and gynecologists, play a crucial role by performing the initial assessment and subsequent therapies and referrals and serve as the mediator between the specialists and by being the patient’s spokesperson. It is therefore important for clinicians to be aware of the various possible causes of these breast symptoms, to know which imaging tests to order, and also to understand the indications for biopsies and surgical referral.
Common types of breast lumps: Imaging workup and management
Accounting for 8% of women who present with breast symptoms, breast lump is the second most common symptom after breast pain.1 The positive likelihood ratio of finding breast cancer is highest among women with breast lumps compared with any other breast symptoms. Therefore, anxiety is related to this symptom, and a thorough evaluation is recommended.1 Cysts, fibroadenoma, and fat necrosis are 3 common benign causes of breast lumps.2
In this section, we review clinical presentation, imaging workup, and management strategies for common types of breast lumps.
CASE 1 Woman with tender breast lump
A 45-year-old woman presents with a breast lump of 6 months’ duration that is associated with a change in size with the menstrual cycle and pain. Clinical examination reveals a 4 x 4.5–cm mass in the right breast in the retroareolar region, which is smooth with some tenderness on palpation.
Breast cyst
According to the American College of Radiology appropriateness criteria for an adult woman 40 years of age or older who presents with a palpable breast mass, the initial imaging study is diagnostic mammography with or without digital tomosynthesis, usually followed by a directed ultrasound. If the mammogram is suspicious or highly suggestive of malignancy, or in cases where the mammogram does not show an abnormality, the next recommended step is breast ultrasonography. Any suspicious findings on ultrasound or mammogram should be followed by an image guided biopsy. Ultrasonography also may be appropriate if the mammogram findings are benign or probably benign.
For an adult woman younger than age 30 who presents with a palpable breast mass, breast ultrasonography is the appropriate initial imaging study. If the ultrasound is suspicious or highly suggestive of malignancy, then performing diagnostic mammography with or without digital tomosynthesis or ultrasound-guided core needle biopsy of the mass are both considered appropriate. However, no further imaging is recommended if the ultrasound is benign, probably benign, or negative. Breast ultrasonography or mammography is appropriate as the initial imaging test for adult women aged 30 to 39 years who present with a palpable breast mass.3,4
Approximately 50% of women after age 30 may develop fibrocystic breast disease, and 20% of them can present with pain or lump due to a macrocysts. Simple cysts must be distinguished from complex cysts with the help of ultrasound as the latter are associated with 23% to 31% increased risk of malignancy.
In this 45-year-old patient, the initial mammogram demonstrated a circumscribed mass underneath the area of palpable concern (FIGURE 1a, 1b). Targeted breast ultrasonography was performed for further assessment, which depicted the mass as a benign simple cyst (FIGURE 1c).
On ultrasound, a simple cyst is an anechoic, well-circumscribed mass with a thin capsule and with increased through transmission. Patients with small and asymptomatic simple cysts do not need imaging follow-up and can return for routine screening mammograms.
A breast surgeon, radiologist, or gynecologist can perform percutaneous aspiration if a cyst is large and symptomatic. A cyst with low-level internal echoes, fluid-fluid, or fluid-debris levels is considered a complicated cyst. Differential diagnosis also includes hematoma, fat necrosis, abscess, and galactocele, depending on the clinical presentation. Fine-needle aspiration or short-interval follow-up5,6 is appropriate for complicated cysts, while incision and drainage is indicated in patients with infected cysts and abscesses. A cyst with a solid component is considered a cystic, solid mass, and core needle biopsy is recommended. The differential diagnosis for cysts with solid components includes intracystic papilloma, papillary carcinoma, ductal carcinoma in situ, and necrotic cancers.5,6
Continue to: CASE 2 Painless breast mass in a young woman...
CASE 2 Painless breast mass in a young woman
A 22-year-old woman presents with a 2-month history of breast lump, which is not associated with pain or nipple discharge. On examination, there is a 2 x 2–cm mass in the right breast at 12 o’clock, 2 cm from the nipple, which is mobile, smooth, and nontender on palpation.
Fibroadenoma
In this 22-year-old, the initial imaging of choice is breast ultrasonography. Breast ultrasonography can differentiate a cystic mass from a solid mass, and it does not involve radiation. Right breast targeted ultrasound showed a circumscribed oval homogeneous hypoechoic mass that is wider than tall (FIGURE 2). The patient desired surgical removal, and a pre-lumpectomy core needle biopsy revealed a fibroadenoma.
Fibroadenoma is the most common benign tumor of the breast. It is most often encountered in premenopausal women. Patients present with a painless breast lump, which is smooth and mobile on palpation. Fibroadenoma can be followed expectantly with repeat ultrasound (to assess over time for growth) if it is small and asymptomatic. No further action is needed if it remains stable. If a patient desires surgical excision, a core needle biopsy is usually performed before lumpectomy.
Excisional biopsy or removal of the mass is recommended if the mass is greater than 3 or 4 cm, is symptomatic, or if there is an increase in size that raises clinical concern for phyllodes tumor. Imaging features that are concerning for phyllodes tumors are size greater than 3 cm, indistinct or microlobulated margins, and heterogeneous echo pattern.7,8 In cases in which the imaging features are concerning for phyllodes tumor and a core needle biopsy is not definitive, wide surgical excision is recommended for definitive diagnosis.8
CASE 3 Patient develops breast mass post-surgery
A 45-year-old woman presents with a tender left breast mass that she noticed 2 months after breast reduction surgery. It has been increasing in size since. On clinical examination, a 4 x 4–cm mass is found at the surgical scar site, which is indurated on palpation and tender.
Fat necrosis
In this 45-year-old, the initial test of choice is diagnostic mammography, which showed a somewhat circumscribed area with fat under the palpable marker (FIGURE 3a). Breast ultrasonography was performed for further evaluation, which was inconclusive as the ultrasound showed ill-defined areas of mixed echogenicity (FIGURE 3b). Breast magnetic resonance imaging (MRI) clearly demonstrated fat necrosis in the area of the palpable lump (FIGURE 3c).
Fat necrosis of the breast is an inflammatory process that is seen after breast trauma or surgery. It can present as an incidental mammogram finding or a palpable mass. The patient may give a history of trauma, breast reduction surgery, or breast cancer surgery followed by radiation treatment. On clinical examination, fat necrosis occasionally can present as a firm mass with skin retraction or swelling concerning for cancer. Imaging features are variable depending on the stage of fat necrosis and inflammation.9-11
A mammogram may demonstrate a circumscribed fat-containing mass, an ill-defined mass, asymmetry or calcified oil cyst, and dystrophic calcifications. On ultrasound, fat necrosis can appear as anechoic or hypoechoic or as a complicated cyst or a mixed cystic, solid mass. MRI demonstrates a circumscribed or irregular fat-containing mass, with or without enhancement, and architectural distortion.
When the imaging features are clearly benign—for example, a circumscribed fat-containing mass on mammogram or on ultrasound or, on MRI, marked hypointensity of fat in the center of a circumscribed mass when compared with surrounding fat (keyhole sign)—no further follow-up is needed. When the imaging features are indeterminate, however, a short-interval follow-up can be considered. In cases with irregular fat-containing mass with enhancement, core needle biopsy is indicated to exclude cancer. If the workup remains inconclusive and the level of clinical suspicion is high, surgical excision can be performed for a definitive diagnosis.12
Continue to: Investigating breast pain: Imaging workup and management...
Investigating breast pain: Imaging workup and management
Breast pain, or mastalgia, is the most common concern of women presenting to a breast clinic and accounts for approximately half of such encounters.13 Causes of breast pain include hormonal changes, fibrocystic changes, musculoskeletal causes (such as costochondritis), lack of support, infection, and injury. While mastalgia often causes patient concern, the risk of malignancy in a woman presenting with breast pain alone is low. Still, it is essential to rule out other findings suspicious for cancer (mass, skin changes, or nipple discharge) with a thorough history and breast examination.
In this section, we review clinical presentation, imaging workup, and management for breast pain.
CASE 4 Woman with noncyclic breast pain
A 26-year-old woman presents to the clinic with mastalgia. The pain is noncyclic and primarily located in the upper outer quadrant of her left breast. There is no history of breast cancer in her family. She has no suspicious findings on the breast examination.
Mastalgia
The test of choice for this 26-year-old with focal left breast pain is targeted breast ultrasound. The patient’s ultrasound image showed no suspicious findings or solid or cystic mass (FIGURE 4).
Two important characteristics of breast pain are whether it is noncyclical and whether it is focal. According to the American College of Radiology, no breast imaging is recommended for clinically insignificant cyclical, nonfocal (greater than 1 quadrant)/diffuse pain, as this type of mastalgia is not associated with malignancy.14
For patients age 40 or older, if they are not up to date with their annual screening mammogram, then a mammogram should be performed. An imaging workup is warranted for clinically significant mastalgia that is noncyclical and focal. Even then, no malignancy is identified in most patients with clinically significant mastalgia; in patients with breast pain as their only symptom, the prevalence of breast cancer is 0% to 3.0%.15-19
The initial imaging modality differs by patient age: younger than 30 years, ultrasonography; between 30 and 40 years, mammography or ultrasonography; and older than 40 years, mammography first followed by ultrasonography.14
Treatment of breast pain is primarily symptomatic, and evidence for specific treatments is generally lacking. Cyclical breast pain resolves spontaneously in 20% to 30% of women, while noncyclical pain responds poorly to treatment but resolves spontaneously in half of women.20 Reassurance is important and wearing a supportive bra often can alleviate breast pain. In addition, reducing caffeine intake can be helpful.
As a first-line treatment, both topical (diclofenac) and oral nonsteroidal anti-inflammatory drugs effectively can relieve breast pain. Supplements and herbal remedies (for example, evening primrose oil, vitamin E, flaxseed) have varying effectiveness and are of questionable benefit as few have trials to support their effectiveness.4 Danazol and tamoxifen have been shown to have some benefits but they also have adverse effects.20 Surgery does not play a role in the treatment of mastalgia.
CASE 5 Breastfeeding woman with breast pain
A 27-year-old postpartum woman presents with concerns for redness and pain in the upper inner left breast. She has been breastfeeding for the past few months. Breast examination demonstrates a 5-cm area of erythema and warmth but no fluctuance or masses.
Lactational mastitis
Targeted ultrasonography is the test of choice for this 27-year-old patient with focal breast pain, and the imaging revealed edema of subcutaneous tissues and ill-defined hypoechoic areas, likely inflamed fat lobules (FIGURE 5). These findings suggest uncomplicated lactational mastitis, which can be treated with antibiotics. Generally, the mastitis will improve within days of starting the antibiotics; if it does not improve, repeat examination and ultrasound should be performed to look for formation of an abscess that may require aspiration.
Continue to: CASE 6 Woman with painful periareolar mass...
CASE 6 Woman with painful periareolar mass
A 42-year-old perimenopausal woman describes having pain near the nipple of her right breast. She is a smoker and has no history of breast cancer in her family. Examination demonstrates a palpable, erythematous, painful, 3-cm periareolar fluctuant mass.
Nonpuerperal periareolar abscess
Appropriate initial imaging for this 42-year-old patient with focal pain is a diagnostic mammogram, which showed skin thickening and a retroareolar mass (FIGURE 6a). Further evaluation with targeted ultrasound showed a thick-walled anechoic collection with echoes compatible with an abscess (FIGURE 6b).
Mammographic findings in a patient with mastitis may be normal or demonstrate skin and trabecular thickening. Ultrasound imaging may show dilated ducts and heterogeneous tissue secondary to inflammation and edema without a discrete fluid collection. In cases with breast abscess, in addition to the mammographic findings described above, a mass, or an asymmetry, may be seen, most commonly in a subareolar location. On ultrasound, a hypoechoic collection with mobile debris, no internal flow on Doppler, and thick hypervascular walls can be seen with abscess, occasionally giving the appearance of a complicated cyst or a mixed cystic, solid mass.
The most important differential for mastitis is inflammatory breast cancer. Most cancers appear solid but can have central necrosis, mimicking a complicated cystic mass on ultrasound. The location for mastitis or abscess is most frequently subareolar. The presence of microcalcifications in a mass indicates the possibility of cancer.
Contrast-enhanced MRI can be helpful to differentiate between infection and cancer, with cancers showing initial early enhancement and washout kinetics compared with infected collections that show no enhancement or peripheral enhancement with a plateau or persistent enhancement curves. When clinical and imaging findings are unchanged after treatment of mastitis and abscesses, a core needle biopsy should be performed.21,22
There are 2 categories of mastitis and breast abscess: lactational and nonpuerperal (all mastitis that occurs outside the lactational period). The World Health Organization definition of puerperal mastitis includes pain, local redness, warmth and swelling of the breast (usually unilateral), fever, and malaise.4 Concerning etiology, epithelial lesions in the nipple area caused by breastfeeding can allow pathogens to enter and cause infection. The most common microorganism is Staphylococcus aureus.4 Continued emptying of the breast is important, combined with early antibiotic therapy (dicloxacillin is often the first line; if the patient is penicillin allergic, use a macrolide such as clindamycin). If no improvement is seen in 48 to 72 hours, imaging should be performed.
In most cases, continuation of breastfeeding is possible. If mastitis has evolved into an abscess in a lactating woman, it can be aspirated under ultrasound guidance. Incision and drainage should be avoided unless the abscess persists after multiple aspiration attempts, it is large, or if the overlying skin is thin or otherwise appears nonviable.
Nonpuerperal mastitis includes peripheral, periductal, and idiopathic granulomatous mastitis (IGM). Peripheral mastitis behaves like infections/abscesses in other soft tissues, responds well to treatment (antibiotics and percutaneous drainage), and is less likely to recur than periductal mastitis and IGM.21,23
Periductal mastitis and abscess, also known as Zuska disease, has a pathogenesis distinct from other forms of mastitis. Squamous metaplasia of the usual cuboidal epithelium of the breast ducts leads to keratin plugging that can cause infection.23 Risk factors include obesity, smoking, and macromastia. The typical presentation of Zuska disease is a woman with a history of chronic smoking and/or a congenital cleft in the central nipple.23 Periareolar signs of inflammation (redness, swelling, warmth) may be accompanied by an abscess. These can recur and lead to chronic fistula formation, especially if there is a history of intervention (such as aspiration, incision, and drainage).
Treatment of Zuska disease includes symptom relief and antibiotics. If S aureus is present, infection with methicillin-resistant S aureus is likely, and treatment with clindamycin or amoxicillin/clavulanic acid is preferred. If abscess is present, aspiration (preferred, often under ultrasound guidance) or incision and drainage (if the skin is compromised) may be required. If disease is recurrent or associated with a chronically draining fistula, surgical intervention may be warranted, in which resolution requires removing the keratin-plugged ducts in and immediately below the central core of the nipple. Given the association between Zuska disease and smoking, cessation should be encouraged, although there is no guarantee that this will resolve the issue.23
Continue to: CASE 7 Patient with breast pain and swelling...
CASE 7 Patient with breast pain and swelling
A 39-year-old woman presents with left breast swelling and pain of 1 month’s duration. On examination, there is a 6-cm area of edema, induration, and erythema.
Granulomatous mastitis
A diagnostic mammogram and ultrasound demonstrated an ill-defined hypoechoic mass (FIGURE 7a). Ultrasound-guided biopsy was performed, which showed granulomatous mastitis, negative for fungus and acid-fast bacilli. The patient was treated with prednisone and gradually improved (FIGURE 7b).
Granulomatous mastitis (GM) is a rare benign inflammatory process, with etiologies that include fungal infections, tuberculosis, Wegener granulomatosis, sarcoidosis, and idiopathic causes. Imaging can be nonspecific and show variable features. Mammograms can appear normal or show asymmetry or mass and skin thickening. Ultrasound can show heterogeneous parenchyma, ill-defined hypoechoic collection, or a mass with margins that can be circumscribed or indistinct or with tubular extensions, with or without overlying skin thickening, fistulas, and reactive lymph nodes.24
In this clinical setting, the differential diagnosis includes infectious mastitis, inflammatory breast cancer, foreign body injection granulomas, and diabetic mastopathy. Treatment involves drainage and fluid culture if there is a collection on imaging. A core biopsy is performed if imaging demonstrates a solid mass or fluid culture is negative and symptoms persist or recur. Oral steroids represent the mainstay of treatment if a core biopsy shows GM. However, immunosuppressants, including methotrexate, and surgery are options if initial treatment is not helpful.25,26
Conclusion
Breast symptoms are common reasons for patient visits to obstetricians and gynecologists. With a good understanding of the various symptomatic breast diseases and conditions, and by having a close collaboration with radiologists and breast surgeons, clinicians can provide excellent care to these patients and thereby improve patient outcomes and satisfaction. ●
The vast majority of symptomatic breast conditions are benign, with the most common symptoms being palpable mass and breast pain. Clinicians, including primary care clinicians and gynecologists, play a crucial role by performing the initial assessment and subsequent therapies and referrals and serve as the mediator between the specialists and by being the patient’s spokesperson. It is therefore important for clinicians to be aware of the various possible causes of these breast symptoms, to know which imaging tests to order, and also to understand the indications for biopsies and surgical referral.
Common types of breast lumps: Imaging workup and management
Accounting for 8% of women who present with breast symptoms, breast lump is the second most common symptom after breast pain.1 The positive likelihood ratio of finding breast cancer is highest among women with breast lumps compared with any other breast symptoms. Therefore, anxiety is related to this symptom, and a thorough evaluation is recommended.1 Cysts, fibroadenoma, and fat necrosis are 3 common benign causes of breast lumps.2
In this section, we review clinical presentation, imaging workup, and management strategies for common types of breast lumps.
CASE 1 Woman with tender breast lump
A 45-year-old woman presents with a breast lump of 6 months’ duration that is associated with a change in size with the menstrual cycle and pain. Clinical examination reveals a 4 x 4.5–cm mass in the right breast in the retroareolar region, which is smooth with some tenderness on palpation.
Breast cyst
According to the American College of Radiology appropriateness criteria for an adult woman 40 years of age or older who presents with a palpable breast mass, the initial imaging study is diagnostic mammography with or without digital tomosynthesis, usually followed by a directed ultrasound. If the mammogram is suspicious or highly suggestive of malignancy, or in cases where the mammogram does not show an abnormality, the next recommended step is breast ultrasonography. Any suspicious findings on ultrasound or mammogram should be followed by an image guided biopsy. Ultrasonography also may be appropriate if the mammogram findings are benign or probably benign.
For an adult woman younger than age 30 who presents with a palpable breast mass, breast ultrasonography is the appropriate initial imaging study. If the ultrasound is suspicious or highly suggestive of malignancy, then performing diagnostic mammography with or without digital tomosynthesis or ultrasound-guided core needle biopsy of the mass are both considered appropriate. However, no further imaging is recommended if the ultrasound is benign, probably benign, or negative. Breast ultrasonography or mammography is appropriate as the initial imaging test for adult women aged 30 to 39 years who present with a palpable breast mass.3,4
Approximately 50% of women after age 30 may develop fibrocystic breast disease, and 20% of them can present with pain or lump due to a macrocysts. Simple cysts must be distinguished from complex cysts with the help of ultrasound as the latter are associated with 23% to 31% increased risk of malignancy.
In this 45-year-old patient, the initial mammogram demonstrated a circumscribed mass underneath the area of palpable concern (FIGURE 1a, 1b). Targeted breast ultrasonography was performed for further assessment, which depicted the mass as a benign simple cyst (FIGURE 1c).
On ultrasound, a simple cyst is an anechoic, well-circumscribed mass with a thin capsule and with increased through transmission. Patients with small and asymptomatic simple cysts do not need imaging follow-up and can return for routine screening mammograms.
A breast surgeon, radiologist, or gynecologist can perform percutaneous aspiration if a cyst is large and symptomatic. A cyst with low-level internal echoes, fluid-fluid, or fluid-debris levels is considered a complicated cyst. Differential diagnosis also includes hematoma, fat necrosis, abscess, and galactocele, depending on the clinical presentation. Fine-needle aspiration or short-interval follow-up5,6 is appropriate for complicated cysts, while incision and drainage is indicated in patients with infected cysts and abscesses. A cyst with a solid component is considered a cystic, solid mass, and core needle biopsy is recommended. The differential diagnosis for cysts with solid components includes intracystic papilloma, papillary carcinoma, ductal carcinoma in situ, and necrotic cancers.5,6
Continue to: CASE 2 Painless breast mass in a young woman...
CASE 2 Painless breast mass in a young woman
A 22-year-old woman presents with a 2-month history of breast lump, which is not associated with pain or nipple discharge. On examination, there is a 2 x 2–cm mass in the right breast at 12 o’clock, 2 cm from the nipple, which is mobile, smooth, and nontender on palpation.
Fibroadenoma
In this 22-year-old, the initial imaging of choice is breast ultrasonography. Breast ultrasonography can differentiate a cystic mass from a solid mass, and it does not involve radiation. Right breast targeted ultrasound showed a circumscribed oval homogeneous hypoechoic mass that is wider than tall (FIGURE 2). The patient desired surgical removal, and a pre-lumpectomy core needle biopsy revealed a fibroadenoma.
Fibroadenoma is the most common benign tumor of the breast. It is most often encountered in premenopausal women. Patients present with a painless breast lump, which is smooth and mobile on palpation. Fibroadenoma can be followed expectantly with repeat ultrasound (to assess over time for growth) if it is small and asymptomatic. No further action is needed if it remains stable. If a patient desires surgical excision, a core needle biopsy is usually performed before lumpectomy.
Excisional biopsy or removal of the mass is recommended if the mass is greater than 3 or 4 cm, is symptomatic, or if there is an increase in size that raises clinical concern for phyllodes tumor. Imaging features that are concerning for phyllodes tumors are size greater than 3 cm, indistinct or microlobulated margins, and heterogeneous echo pattern.7,8 In cases in which the imaging features are concerning for phyllodes tumor and a core needle biopsy is not definitive, wide surgical excision is recommended for definitive diagnosis.8
CASE 3 Patient develops breast mass post-surgery
A 45-year-old woman presents with a tender left breast mass that she noticed 2 months after breast reduction surgery. It has been increasing in size since. On clinical examination, a 4 x 4–cm mass is found at the surgical scar site, which is indurated on palpation and tender.
Fat necrosis
In this 45-year-old, the initial test of choice is diagnostic mammography, which showed a somewhat circumscribed area with fat under the palpable marker (FIGURE 3a). Breast ultrasonography was performed for further evaluation, which was inconclusive as the ultrasound showed ill-defined areas of mixed echogenicity (FIGURE 3b). Breast magnetic resonance imaging (MRI) clearly demonstrated fat necrosis in the area of the palpable lump (FIGURE 3c).
Fat necrosis of the breast is an inflammatory process that is seen after breast trauma or surgery. It can present as an incidental mammogram finding or a palpable mass. The patient may give a history of trauma, breast reduction surgery, or breast cancer surgery followed by radiation treatment. On clinical examination, fat necrosis occasionally can present as a firm mass with skin retraction or swelling concerning for cancer. Imaging features are variable depending on the stage of fat necrosis and inflammation.9-11
A mammogram may demonstrate a circumscribed fat-containing mass, an ill-defined mass, asymmetry or calcified oil cyst, and dystrophic calcifications. On ultrasound, fat necrosis can appear as anechoic or hypoechoic or as a complicated cyst or a mixed cystic, solid mass. MRI demonstrates a circumscribed or irregular fat-containing mass, with or without enhancement, and architectural distortion.
When the imaging features are clearly benign—for example, a circumscribed fat-containing mass on mammogram or on ultrasound or, on MRI, marked hypointensity of fat in the center of a circumscribed mass when compared with surrounding fat (keyhole sign)—no further follow-up is needed. When the imaging features are indeterminate, however, a short-interval follow-up can be considered. In cases with irregular fat-containing mass with enhancement, core needle biopsy is indicated to exclude cancer. If the workup remains inconclusive and the level of clinical suspicion is high, surgical excision can be performed for a definitive diagnosis.12
Continue to: Investigating breast pain: Imaging workup and management...
Investigating breast pain: Imaging workup and management
Breast pain, or mastalgia, is the most common concern of women presenting to a breast clinic and accounts for approximately half of such encounters.13 Causes of breast pain include hormonal changes, fibrocystic changes, musculoskeletal causes (such as costochondritis), lack of support, infection, and injury. While mastalgia often causes patient concern, the risk of malignancy in a woman presenting with breast pain alone is low. Still, it is essential to rule out other findings suspicious for cancer (mass, skin changes, or nipple discharge) with a thorough history and breast examination.
In this section, we review clinical presentation, imaging workup, and management for breast pain.
CASE 4 Woman with noncyclic breast pain
A 26-year-old woman presents to the clinic with mastalgia. The pain is noncyclic and primarily located in the upper outer quadrant of her left breast. There is no history of breast cancer in her family. She has no suspicious findings on the breast examination.
Mastalgia
The test of choice for this 26-year-old with focal left breast pain is targeted breast ultrasound. The patient’s ultrasound image showed no suspicious findings or solid or cystic mass (FIGURE 4).
Two important characteristics of breast pain are whether it is noncyclical and whether it is focal. According to the American College of Radiology, no breast imaging is recommended for clinically insignificant cyclical, nonfocal (greater than 1 quadrant)/diffuse pain, as this type of mastalgia is not associated with malignancy.14
For patients age 40 or older, if they are not up to date with their annual screening mammogram, then a mammogram should be performed. An imaging workup is warranted for clinically significant mastalgia that is noncyclical and focal. Even then, no malignancy is identified in most patients with clinically significant mastalgia; in patients with breast pain as their only symptom, the prevalence of breast cancer is 0% to 3.0%.15-19
The initial imaging modality differs by patient age: younger than 30 years, ultrasonography; between 30 and 40 years, mammography or ultrasonography; and older than 40 years, mammography first followed by ultrasonography.14
Treatment of breast pain is primarily symptomatic, and evidence for specific treatments is generally lacking. Cyclical breast pain resolves spontaneously in 20% to 30% of women, while noncyclical pain responds poorly to treatment but resolves spontaneously in half of women.20 Reassurance is important and wearing a supportive bra often can alleviate breast pain. In addition, reducing caffeine intake can be helpful.
As a first-line treatment, both topical (diclofenac) and oral nonsteroidal anti-inflammatory drugs effectively can relieve breast pain. Supplements and herbal remedies (for example, evening primrose oil, vitamin E, flaxseed) have varying effectiveness and are of questionable benefit as few have trials to support their effectiveness.4 Danazol and tamoxifen have been shown to have some benefits but they also have adverse effects.20 Surgery does not play a role in the treatment of mastalgia.
CASE 5 Breastfeeding woman with breast pain
A 27-year-old postpartum woman presents with concerns for redness and pain in the upper inner left breast. She has been breastfeeding for the past few months. Breast examination demonstrates a 5-cm area of erythema and warmth but no fluctuance or masses.
Lactational mastitis
Targeted ultrasonography is the test of choice for this 27-year-old patient with focal breast pain, and the imaging revealed edema of subcutaneous tissues and ill-defined hypoechoic areas, likely inflamed fat lobules (FIGURE 5). These findings suggest uncomplicated lactational mastitis, which can be treated with antibiotics. Generally, the mastitis will improve within days of starting the antibiotics; if it does not improve, repeat examination and ultrasound should be performed to look for formation of an abscess that may require aspiration.
Continue to: CASE 6 Woman with painful periareolar mass...
CASE 6 Woman with painful periareolar mass
A 42-year-old perimenopausal woman describes having pain near the nipple of her right breast. She is a smoker and has no history of breast cancer in her family. Examination demonstrates a palpable, erythematous, painful, 3-cm periareolar fluctuant mass.
Nonpuerperal periareolar abscess
Appropriate initial imaging for this 42-year-old patient with focal pain is a diagnostic mammogram, which showed skin thickening and a retroareolar mass (FIGURE 6a). Further evaluation with targeted ultrasound showed a thick-walled anechoic collection with echoes compatible with an abscess (FIGURE 6b).
Mammographic findings in a patient with mastitis may be normal or demonstrate skin and trabecular thickening. Ultrasound imaging may show dilated ducts and heterogeneous tissue secondary to inflammation and edema without a discrete fluid collection. In cases with breast abscess, in addition to the mammographic findings described above, a mass, or an asymmetry, may be seen, most commonly in a subareolar location. On ultrasound, a hypoechoic collection with mobile debris, no internal flow on Doppler, and thick hypervascular walls can be seen with abscess, occasionally giving the appearance of a complicated cyst or a mixed cystic, solid mass.
The most important differential for mastitis is inflammatory breast cancer. Most cancers appear solid but can have central necrosis, mimicking a complicated cystic mass on ultrasound. The location for mastitis or abscess is most frequently subareolar. The presence of microcalcifications in a mass indicates the possibility of cancer.
Contrast-enhanced MRI can be helpful to differentiate between infection and cancer, with cancers showing initial early enhancement and washout kinetics compared with infected collections that show no enhancement or peripheral enhancement with a plateau or persistent enhancement curves. When clinical and imaging findings are unchanged after treatment of mastitis and abscesses, a core needle biopsy should be performed.21,22
There are 2 categories of mastitis and breast abscess: lactational and nonpuerperal (all mastitis that occurs outside the lactational period). The World Health Organization definition of puerperal mastitis includes pain, local redness, warmth and swelling of the breast (usually unilateral), fever, and malaise.4 Concerning etiology, epithelial lesions in the nipple area caused by breastfeeding can allow pathogens to enter and cause infection. The most common microorganism is Staphylococcus aureus.4 Continued emptying of the breast is important, combined with early antibiotic therapy (dicloxacillin is often the first line; if the patient is penicillin allergic, use a macrolide such as clindamycin). If no improvement is seen in 48 to 72 hours, imaging should be performed.
In most cases, continuation of breastfeeding is possible. If mastitis has evolved into an abscess in a lactating woman, it can be aspirated under ultrasound guidance. Incision and drainage should be avoided unless the abscess persists after multiple aspiration attempts, it is large, or if the overlying skin is thin or otherwise appears nonviable.
Nonpuerperal mastitis includes peripheral, periductal, and idiopathic granulomatous mastitis (IGM). Peripheral mastitis behaves like infections/abscesses in other soft tissues, responds well to treatment (antibiotics and percutaneous drainage), and is less likely to recur than periductal mastitis and IGM.21,23
Periductal mastitis and abscess, also known as Zuska disease, has a pathogenesis distinct from other forms of mastitis. Squamous metaplasia of the usual cuboidal epithelium of the breast ducts leads to keratin plugging that can cause infection.23 Risk factors include obesity, smoking, and macromastia. The typical presentation of Zuska disease is a woman with a history of chronic smoking and/or a congenital cleft in the central nipple.23 Periareolar signs of inflammation (redness, swelling, warmth) may be accompanied by an abscess. These can recur and lead to chronic fistula formation, especially if there is a history of intervention (such as aspiration, incision, and drainage).
Treatment of Zuska disease includes symptom relief and antibiotics. If S aureus is present, infection with methicillin-resistant S aureus is likely, and treatment with clindamycin or amoxicillin/clavulanic acid is preferred. If abscess is present, aspiration (preferred, often under ultrasound guidance) or incision and drainage (if the skin is compromised) may be required. If disease is recurrent or associated with a chronically draining fistula, surgical intervention may be warranted, in which resolution requires removing the keratin-plugged ducts in and immediately below the central core of the nipple. Given the association between Zuska disease and smoking, cessation should be encouraged, although there is no guarantee that this will resolve the issue.23
Continue to: CASE 7 Patient with breast pain and swelling...
CASE 7 Patient with breast pain and swelling
A 39-year-old woman presents with left breast swelling and pain of 1 month’s duration. On examination, there is a 6-cm area of edema, induration, and erythema.
Granulomatous mastitis
A diagnostic mammogram and ultrasound demonstrated an ill-defined hypoechoic mass (FIGURE 7a). Ultrasound-guided biopsy was performed, which showed granulomatous mastitis, negative for fungus and acid-fast bacilli. The patient was treated with prednisone and gradually improved (FIGURE 7b).
Granulomatous mastitis (GM) is a rare benign inflammatory process, with etiologies that include fungal infections, tuberculosis, Wegener granulomatosis, sarcoidosis, and idiopathic causes. Imaging can be nonspecific and show variable features. Mammograms can appear normal or show asymmetry or mass and skin thickening. Ultrasound can show heterogeneous parenchyma, ill-defined hypoechoic collection, or a mass with margins that can be circumscribed or indistinct or with tubular extensions, with or without overlying skin thickening, fistulas, and reactive lymph nodes.24
In this clinical setting, the differential diagnosis includes infectious mastitis, inflammatory breast cancer, foreign body injection granulomas, and diabetic mastopathy. Treatment involves drainage and fluid culture if there is a collection on imaging. A core biopsy is performed if imaging demonstrates a solid mass or fluid culture is negative and symptoms persist or recur. Oral steroids represent the mainstay of treatment if a core biopsy shows GM. However, immunosuppressants, including methotrexate, and surgery are options if initial treatment is not helpful.25,26
Conclusion
Breast symptoms are common reasons for patient visits to obstetricians and gynecologists. With a good understanding of the various symptomatic breast diseases and conditions, and by having a close collaboration with radiologists and breast surgeons, clinicians can provide excellent care to these patients and thereby improve patient outcomes and satisfaction. ●
- Eberl MM, Phillips RL Jr, Lamberts H, et al. Characterizing breast symptoms in family practice. Ann Fam Med. 2008;6:528-533.
- Malherbe F, Nel D, Molabe H, et al. Palpable breast lumps: an age-based approach to evaluation and diagnosis. S Afr Fam Pract (2022). 2022;64:e1-e5.
- Expert Panel on Breast Imaging; Klein KA, Kocher M, Lourenco AP, et al. American College of Radiology ACR appropriateness criteria: palpable breast masses. Accessed February 15, 2023. https://acsearch.acr.org/docs/69495/Narrative/
- Stachs A, Stubert J, Reimer T, et al. Benign breast disease in women. Dtsch Arztebl Int. 2019;116:565574.
- Hines N, Slanetz PJ, Eisenberg RL. Cystic masses of the breast. AJR Am J Roentgenol. 2010;194:W122133.
- Berg WA. Reducing unnecessary biopsy and follow-up of benign cystic breast lesions. Radiology. 2020;295:52-53.
- Duman L, Gezer NS, Balcı P, et al. Differentiation between phyllodes tumors and fibroadenomas based on mammographic sonographic and MRI features. Breast Care. 2016;11:123-127.
- Lerwill MF, Lee AHS, Tan PH. Fibroepithelial tumours of the breast—a review. Virchows Arch. 2022;480:45-63.
- Vasei N, Shishegar A, Ghalkhani F, et al. Fat necrosis in the breast: a systematic review of clinical. Lipids Health Dis. 2019;18:139.
- Kerridge WD, Kryvenko ON, Thompson A, et al. Fat necrosis of the breast: a pictorial review of the mammographic, ultrasound, CT, and MRI findings with histopathologic correlation. Radiol Res Pract. 2015;2015:613139.
- Taboada JL, Stephens TW, Krishnamurthy S, et al. The many faces of fat necrosis in the breast. AJR Am J Roentgenol. 2009;192:815-825.
- Tan PH, Lai LM, Carrington EV, et al. Fat necrosis of the breast—a review. Breast. 2006;15:313-318.
- Holbrook AI. Breast pain, a common grievance: guidance to radiologists. AJR Am J Roentgenol. 2020;214:259-264.
- Expert Panel on Breast Imaging; Moy L, Heller SL, Bailey L, et al. ACR appropriateness criteria: palpable breast masses. J Am Coll Radiol. 2017;14:S203-S224.
- Chetlen AL, Kapoor MM, Watts MR. Mastalgia: imaging workup appropriateness. Acad Radiol. 2017;24:345-349.
- Arslan M, Kucukerdem HS, Can H, et al. Retrospective analysis of women with only mastalgia. J Breast Health. 2016;12:151-154.
- Fariselli G, Lepera P, Viganotti G, et al. Localized mastalgia as presenting symptom in breast cancer. Eur J Surg Oncol. 1988;14:213-215.
- Leddy R, Irshad A, Zerwas E, et al. Role of breast ultrasound and mammography in evaluating patients presenting with focal breast pain in the absence of a palpable lump. Breast J. 2013;19:582-589.
- Leung JW, Kornguth PJ, Gotway MB. Utility of targeted sonography in the evaluation of focal breast pain. J Ultrasound Med. 2002;21:521-526.
- Goyal A. Breast pain. BMJ Clin Evid. 2011; 2011:0812.
- Kasales CJ, Han B, Smith Jr JS, et al. Nonpuerperal mastitis and subareolar abscess of the breast. AJR Am J Roentgenol. 2014;202:W133-W139.
- Mahoney MC, Ingram AD. Breast emergencies: types, imaging features, and management. AJR Am J Roentgenol. 2014;202:W390-W399.
- Snider HC. Management of mastitis, abscess, and fistula. Surg Clin North Am. 2022;102:1103-1116.
- Oztekin PS, Durhan G, Kosar PN, et al. Imaging findings in patients with granulomatous mastitis. Iran J Radiol. 2016;13:e33900.
- Pluguez-Turull CW, Nanyes JE, Quintero CJ, et al. Idiopathic granulomatous mastitis: manifestations at multimodality imaging and pitfalls. Radiographics. 2018;38:330-356.
- Hovanessian-Larsen LJ, Peyvandi B, Klipfel N, et al. Granulomatous lobular mastitis: imaging, diagnosis, and treatment. AJR Am J Roentgenol. 2009;193:574-581.
- Eberl MM, Phillips RL Jr, Lamberts H, et al. Characterizing breast symptoms in family practice. Ann Fam Med. 2008;6:528-533.
- Malherbe F, Nel D, Molabe H, et al. Palpable breast lumps: an age-based approach to evaluation and diagnosis. S Afr Fam Pract (2022). 2022;64:e1-e5.
- Expert Panel on Breast Imaging; Klein KA, Kocher M, Lourenco AP, et al. American College of Radiology ACR appropriateness criteria: palpable breast masses. Accessed February 15, 2023. https://acsearch.acr.org/docs/69495/Narrative/
- Stachs A, Stubert J, Reimer T, et al. Benign breast disease in women. Dtsch Arztebl Int. 2019;116:565574.
- Hines N, Slanetz PJ, Eisenberg RL. Cystic masses of the breast. AJR Am J Roentgenol. 2010;194:W122133.
- Berg WA. Reducing unnecessary biopsy and follow-up of benign cystic breast lesions. Radiology. 2020;295:52-53.
- Duman L, Gezer NS, Balcı P, et al. Differentiation between phyllodes tumors and fibroadenomas based on mammographic sonographic and MRI features. Breast Care. 2016;11:123-127.
- Lerwill MF, Lee AHS, Tan PH. Fibroepithelial tumours of the breast—a review. Virchows Arch. 2022;480:45-63.
- Vasei N, Shishegar A, Ghalkhani F, et al. Fat necrosis in the breast: a systematic review of clinical. Lipids Health Dis. 2019;18:139.
- Kerridge WD, Kryvenko ON, Thompson A, et al. Fat necrosis of the breast: a pictorial review of the mammographic, ultrasound, CT, and MRI findings with histopathologic correlation. Radiol Res Pract. 2015;2015:613139.
- Taboada JL, Stephens TW, Krishnamurthy S, et al. The many faces of fat necrosis in the breast. AJR Am J Roentgenol. 2009;192:815-825.
- Tan PH, Lai LM, Carrington EV, et al. Fat necrosis of the breast—a review. Breast. 2006;15:313-318.
- Holbrook AI. Breast pain, a common grievance: guidance to radiologists. AJR Am J Roentgenol. 2020;214:259-264.
- Expert Panel on Breast Imaging; Moy L, Heller SL, Bailey L, et al. ACR appropriateness criteria: palpable breast masses. J Am Coll Radiol. 2017;14:S203-S224.
- Chetlen AL, Kapoor MM, Watts MR. Mastalgia: imaging workup appropriateness. Acad Radiol. 2017;24:345-349.
- Arslan M, Kucukerdem HS, Can H, et al. Retrospective analysis of women with only mastalgia. J Breast Health. 2016;12:151-154.
- Fariselli G, Lepera P, Viganotti G, et al. Localized mastalgia as presenting symptom in breast cancer. Eur J Surg Oncol. 1988;14:213-215.
- Leddy R, Irshad A, Zerwas E, et al. Role of breast ultrasound and mammography in evaluating patients presenting with focal breast pain in the absence of a palpable lump. Breast J. 2013;19:582-589.
- Leung JW, Kornguth PJ, Gotway MB. Utility of targeted sonography in the evaluation of focal breast pain. J Ultrasound Med. 2002;21:521-526.
- Goyal A. Breast pain. BMJ Clin Evid. 2011; 2011:0812.
- Kasales CJ, Han B, Smith Jr JS, et al. Nonpuerperal mastitis and subareolar abscess of the breast. AJR Am J Roentgenol. 2014;202:W133-W139.
- Mahoney MC, Ingram AD. Breast emergencies: types, imaging features, and management. AJR Am J Roentgenol. 2014;202:W390-W399.
- Snider HC. Management of mastitis, abscess, and fistula. Surg Clin North Am. 2022;102:1103-1116.
- Oztekin PS, Durhan G, Kosar PN, et al. Imaging findings in patients with granulomatous mastitis. Iran J Radiol. 2016;13:e33900.
- Pluguez-Turull CW, Nanyes JE, Quintero CJ, et al. Idiopathic granulomatous mastitis: manifestations at multimodality imaging and pitfalls. Radiographics. 2018;38:330-356.
- Hovanessian-Larsen LJ, Peyvandi B, Klipfel N, et al. Granulomatous lobular mastitis: imaging, diagnosis, and treatment. AJR Am J Roentgenol. 2009;193:574-581.
Telemedicine: Medicolegal aspects in ObGyn
Telemedicine (or telehealth) originated in the early 1900s, when radios were used to communicate medical advice to clinics aboard ships.1 According to the American Telemedicine Association, telemedicine is namely “the use of medical information exchanged from one site to another via electronic communications to improve a patient’s clinical health status.”2 These communications use 2-way video, email, smartphones, wireless tools, and other forms of telecommunications technology.
During the COVID-19 pandemic, many ObGyns—encouraged and advised by professional organizations—began providing telemedicine services.3 The first reported case of COVID-19 was in late 2019; the use of telemedicine was 38 times higher in February 2021 than in February 2020,4 illustrating how many physicians quickly moved to telemedicine practices.
CASE Dr. TM’s telemedicine dream
Before COVID-19, Dr. TM (an ObGyn practi-tioner) practiced in-person medicine in his home state. With the onset of the pandemic, Dr. TM struggled to switch to primarily seeing patients online (generally using Zoom or Facebook Live), with 1 day per week in the office for essential in-person visits.
After several months, however, Dr. TM’s routine became very efficient. He could see many more patients in a shorter time than with the former, in-person system. Therefore, as staff left his practice, Dr. TM did not replace them and also laid off others. Ultimately, the practice had 1 full-time records/insurance secretary who worked from home and 1 part-time nurse who helped with the in-person day and answered some patient inquiries by email. In part as an effort to add new patients, Dr. TM built an engaging website through which his current patients could receive medical information and new patients could sign up.
In late 2022, Dr. TM offered a $100 credit to any current patient who referred a friend or family member who then became a patient. This promotion was surprisingly effective and resulted in an influx of new patients. For example, Patient Z (a long-time patient) received 3 credits for referring her 3 sisters who lived out of state and became telepatients: Patient D, who lived 200 hundred miles away; Patient E, who lived 50 miles away in the adjoining state; and Patient F, who lived 150 miles away. Patient D contacted Dr. TM because she thought she was pregnant and wanted prenatal care, Patient E thought she might have a sexually transmitted infection (STI) and wanted treatment, and Patient F wanted general care and was inquiring about a medical abortion. Dr. TM agreed to treat Patient D but required 1 in-person visit. After 1 brief telemedicine session each with Patients E and F, Dr. TM wrote prescriptions for them.
By 2023, Dr. TM was enthusiastic about telemedicine as a professional practice. However, problems would ensue.
Dos and don’ts of telemedicine2
- Do take the initiative and inform patients of the availability of telemedicine/telehealth services
- Do use the services of medical malpractice insurance companies with regard to telemedicine
- Do integrate telemedicine into practice protocols and account for their limitations
- Don’t assume there are blanket exemptions or waivers in the states where your patients are located
Medical considerations
Telemedicine is endorsed by the American College of Obstetricians and Gynecologists (ACOG) as a vehicle for delivering prenatal and postpartum care.5 This represents an effort to reduce maternal and neonatal morbidity and mortality,5 as well as expandaccess to care and address the deficit in primary care providers and services, especially in rural and underserved populations.5,6 For obstetrics, prenatal care is designed to optimize pregnancy, childbirth, and postpartum care, with a focus on nutrition and genetic consultation and patient education on pregnancy, childbearing, breastfeeding, and newborn care.7
Benefits of telemedicine include its convenience for patients and providers, its efficiency and lower costs for providers (and hopefully patients, as well), and the potential improved access to care for patients.8 It is estimated that if a woman inititates obstetric care at 6 weeks, over the course of the 40-week gestation period, 15 prenatal visits will occur.9 Ultimately, the number of visits is determined based on the specifics of the pregnancy. With telemedicine, clinicians can provide those consultations, and information related to: ultrasonography, fetal echocardiography, and postpartum care services remotely.10 Using telemedicine may reduce missed visits, and remote monitoring may improve the quality of care.11
Barriers to telemedicine care include technical limitations, time constraints, and patient concerns of telehealth (visits). Technical limitations include the lack of a high speed internet connection and/or a smart device and the initial technical set-up–related problems,12 which affect providers as well as patients. Time constraints primarly refer to the ObGyn practice’s lack of time to establish telehealth services.13 Other challenges include integrating translation services, billing-related problems,10 and reimbursement and licensing barriers.14
Before the COVID-19 pandemic, obstetrics led the way in telemedicine with the development of the OB Nest model. Designed to replace in-person obstetrics care visits with telehealth,15 it includes home management tools such as blood pressure cuffs, cardiotocography, scales for weight checks, and Doppler ultrasounds.10 Patients can be instructed to measure fundal height and receive medications by mail. Anesthesia consultation can occur via this venue by having the patient complete a questionnaire prior to arriving at the labor and delivery unit.16
Legal considerations
With the COVID-19 pandemic, temporary changes were made to encourage the rapid adoption of telemedicine, including changes to licensing laws, certain prescription requirements, Health Insurance Portability and Accountability Act (HIPAA) privacy-security regulations, and reimbursement rules that required in-person visits. Thus, many ObGyns started using telemedicine during this rarified period, in which the rules appeared to be few and far between, with limited enforcement of the law and professional obligations.17 However, now that many of the legal rules that were suspended or ignored have been (or are being) reimposed and enforced, it is important for providers to become familiar with the legal issues involved in practicing telemedicine.
First, where is the patient? When discussing the legal issues of telemedicine, it is important to remember that many legal rules for medical care (ie, liability, informed consent, and licensing) vary from state to state. If the patient resides in a different state (“foreign” state) from the physician’s practice location (the physician’s “home” state), the care is considered delivered in the state where the patient is located. Thus, the patient’s location generally establishes the law covering the telemedicine transaction. In the following discussion, the rules refer to the law and professional obligations, with commentary on some key legal issues that are relevant to ObGyn telemedicine.
Continue to: Reinforcing the rules...
Reinforcing the rules
Licensing
During the height of the COVID-19 pandemic, the federal government and almost all states temporarily modified the licensing requirement to allow telemedicine based on an existing medical license in any state—disregarding the “where is the patient” rule. As those rules begin to lapse or change with the official end of the pandemic declared by President Biden as May 2023,17 the rules under which a physician began telemedicine interstate practice in 2020 also may be changing.
Simply put, “The same standards for licensure apply to health care providers regardless of whether care is delivered in-person or virtually through telehealth services.”18 When a physician is engaged in telemedicine treatment of a patient in the physician’s home state, there is generally no licensing issue. Telemedicine generally does not require a separate specific license.19 However, when the patient is in another state (a “foreign” state), there can be a substantial licensing issue.20 Ordinarily, to provide that treatment, the physician must, in some manner, be approved to practice in the patient’s state. That may occur, for example, in the following ways: (1) the physician may hold an additional regular license in the patient’s state, which allows practice there, or (2) the physician may have received permission for “temporary practice” in another state.
Many states (often adjoining states) have formal agreements with other states that allow telemedicine practice by providers in each other’s states. There also are “compacts”, or agreements that enable providers in any of the participating states to practice in the other associated states without a separate license.18 Although several websites provide information about compact licensing and the like, clinicians should not rely on simple lists or maps. Individual states may have special provisions about applying their laws to out-of-state “compact” physicians. In addition, under the Interstate Medical Licensure Compact, “physicians have to pay licensing fees and satisfy the requirements of each medical board in the states where they wish to practice.”21
Consequences. Practicing telemedicine with a patient in a state where the physician does not have a license is generally a crime. Furthermore, it may be the basis for license discipline in the physician’s home state and result in a report to the National Practi-tioner Databank.22 In addition, reimbursement often depends on the practitioner being licensed, and the absence of a license may be a basis for denying payment for services.23 Finally, malpractice insurance generally is limited to licensed practice. Thus, the insurer may decline to defend the unlicensed clinician against a malpractice claim or pay any damages.
Prescribing privileges
Prescribing privileges usually are connected to licensing, so as the rules for licensing change postpandemic, so do the rules for prescribing. In most cases, the physician must have a license in the state where care is given to prescribe medication—which in telemedicine, as noted, typically means the state where the patient is located. Exceptions vary by state, but in general, if a physician does not have a license to provide care, the physician is unlikely to be authorized to prescribe medication.24 Failure to abide by the applicable state rules may result in civil and even criminal liability for illegal prescribing activity.
In addition, the US Food and Drug Administration (FDA) and Drug Enforcement Administration (DEA, which enforces laws concerning controlled substances) also regulate the prescription and sale of pharmaceuticals.25 There are state and federal limits on the ability of clinicians to order controlled substances without an in-person visit. The Ryan Haight Online Pharmacy Consumer Protection Act, for example, sets limits on controlled substance prescriptions without an in-person examination.26 Federal law was modified due to COVID-19 to permit prescribing of many controlled substances by telemedicine if there is synchronous audio and visual examination of the patient. Physicians who write such prescriptions also are required to have a DEA registration in the patient’s state. This is an essential consideration for physicians considering interstate telemedicine practice.27
HIPAA and privacy
Governments waived some of the legal requirements related to health information during the pandemic, but those waivers either have expired or will do so soon. Federal and state laws regarding privacy and security—notably including HIPAA—apply to telemedicine and are of particular concern given the considerable amount of communication of protected health information with telemedicine.
HIPAA security rules essentially require making sure health information cannot be hacked or intercepted. Audio-only telemedicine by landline (not cell) is acceptable under the security rules, but almost all other remote communication requires secure communications.28
Clinicians also need to adhere to the more usual HIPAA privacy rules when practicingtelehealth. State laws protecting patient privacy vary and may be more stringent than HIPAA, so clinicians also must know the requirements in any state where they practice—whether in office or telemedicine.29
Making sure telemedicine practices are consistent with these security and privacy rules often requires particular technical expertise that is outside the realm of most practicing clinicians. However, without modification, the pre-telemedicine technology of many medical offices likely is insufficient for the full range of telemedicine services.30
Reimbursement and fraud
Before COVID-19, Medicare and Medicaid reimbursement for telemedicine was limited. Government decisions to substantially broaden those reimbursement rules (at least temporarily) provided a substantial boost to telemedicine early in the pandemic.23 Federal regulations and statutes also expanded telemedicine reimbursement for various services. Some will end shortly after the health emergency, and others will be permanent. Parts of that will not be sorted out for several years, so it will likely be a changing landscape for reimbursement.
Continue to: Rules that are evolving...
Rules that are evolving
Informed consent
The ethical and legal obligations to obtain informed consent are present in telemedicineas well as in-person care, with the same basic requirements regarding risks, benefits, alternative care, etc.32 However, with telemedicine, information related to remote care should be included and is outlined in TABLE 1.
Certain states may have somewhat unique informed consent requirements—especially for reproductive care, including abortion.34 Therefore, it is important for clinicians to ensure their consent process and forms comply with any legal jurisdiction in which a patient is located.
Medical malpractice
The basics of medical malpractice (or negligence) are the same in telemedicine as in in-person care: duty, breach of duty, and injury caused by the breach. That is, there may be liability when a medical professional breaches the duty of care, causing the patient’s injury. The physician’s duty is defined by the quality of care that the profession (specialty) accepts as reasonably good. This is defined by the opinions of physicians within the specialty and formal statements from professional organizations, including ACOG.3
Maintaining the standard of care and quality. The use of telemedicine is not an excuse to lower the quality of health care. There are some circumstances for which it is medically better to have an in-person visit. In these instances, the provider should recommend the appropriate care, even if telemedicine would be more convenient for the provider and staff.35
If the patient insists and telemedicine might result in less than optimal care, the reasons for using a remote visit should be clearly documented contemporaneously with the decision. Furthermore, when the limitations of being unable to physically examine the patient result in less information than is needed for the patient’s care, the provider must find alternatives to make up for the information gap.11,36 It also may be necessary to inform patients about how to maximize telemedicine care.37 At the beginning of telemedicine care the provider should include information about the nature and limits of telehealth, and the patient’s responsibilities. (See TABLE 1) Throughout treatment of the patient, that information should be updated by the provider. That, of course, is particularly important for patients who have not previously used telemedice services.
Malpractice rules vary by state. Many states have special rules regarding malpractice cases. These differences in malpractice standards and regulations “can be problematic for physicians who use telemedicine services to provide care outside the state in which they practice.”38 Caps on noneconomic damages are an example. Those state rules would apply to telemedicine in the patient’s state.
Malpractice insurance
Malpractice insurance now commonly includes telemedicine legally practiced within the physician’s home state. Practitioners who treat patients in foreign states should carefully examine their malpractice insurance policies to confirm that the coverage extends to practice in those states.39 Malpractice carriers may require notification by a covered physician who routinely provides services to patients in another state.3
Keep in mind, malpractice insurance generally does not cover the practice of medicine that is illegal. Practicing telemedicine in a foreign state, where the physician or other provider does not have a license and where that state does not otherwise permit the practice, is illegal. Most likely, the physician’s malpractice insurance will not cover claims that arise from this illegal practice in a foreign state or provide defense for malpractice claims, including frivolous lawsuits. Thus, the physician will pay out of pocket for the costs of a defense attorney.
Telemedicine treatment of minors
Children and adolescents present special legal issues for ObGyn care, which may become more complicated with telemedicine. Historically, parents are responsible for minors (those aged <18 years): they consent to medical treatment, are responsible for paying for it, and have the right to receive information about treatment.
Over the years, though, many states have made exceptions to these principles, especially with regard to contraception and treatment of sexually transmitted diseases.40 For abortion, in particular, there is considerable variation among the states in parental consent and notification.41 The Supreme Court’s decision in Dobbs v Jackson Women’s Health42 may (depending on the state) be followed with more stringent limitations on adolescent consent to abortions, including medical abortions.43
Use of telehealth does not change any obligations regarding adolescent consent or parental notification. Because those differ considerably among states, it is important for all practitioners to know their states’ requirements and keep reasonably complete records demonstrating their compliance with state law.
Abortion
The most heated current controversy about telemedicine involves abortion—specifically medical abortion, which is the combination of mifepristone and misoprostol.44,45 The FDA approved the combination in 2000. Almost immediately, many states required in-person visits with a certified clinician to receive a prescription for mifepristone and misoprostol, and eventually, the FDA adopted similar requirements.46 However, during the pandemic from 2021 to 2022, the FDA permitted telemedicine prescriptions. Several states still require in-person physician visits, although the constitutionality of those requirements has not been established.47
With the Supreme Court’s decision in Dobbs v Jackson Women’s Health in 2022,42 disagreements have ensued about the degree to which states may regulate the prescription of FDA-approved medical abortion drugs. Thorny constitutional issues exist in the plans of both abortion opponents and proponents in the battle over medical abortion in antiabortion states. It may be that federal drug law preempts state laws limiting access to FDA-approved drugs. On the other hand, it may be that states can make it a crime within the state to possess or provide abortion-inducing drugs. Courts will probably take years to resolve the many tangled legal questions.48
Thus, while the pandemic telemedicine rules may have advanced access to abortion,34 there may be some pending downsides.49 States that prohibit abortion will likely include prohibitions on medical abortions. In addition, they may prohibit anyone in the state (including pharmacies) from selling, possessing, or obtaining any drug used for causing or inducing an abortion.50 If, for constitutional reasons, they cannot press criminal charges or undertake licensing discipline for prescribing abortion, some states will likely withdraw from telehealth licensing compacts to avoid out-of-state prescriptions. This area of telemedicine has considerable uncertainty.
Continue to: CASE Conclusion...
CASE Conclusion
Patient concerns come to the fore
By 2023, Dr. TM started receiving bad news. Patient D called complaining that after following the advice on the website, she suffered a severe reaction and had to be rushed to an emergency department. Patient E (who had only 1 in-office visit early in her pregnancy) notified the office that she developed very high blood pressure that resulted in severe placental abruption, requiring emergency care and resulting in the loss of the fetus. Patient F complained that someone hacked the TikTok direct message communication with Dr. TM and tried to “blackmail” or harass her.
Discussion. Patients D, E, and F represent potential problems of telemedicine practice. Patient D was injured because she relied on her doctor’s website (to which Dr. TM directed patients). It contained an error that caused an injury. A doctor-patient relationship existed, and bad medical advice likely caused the injury. Physicians providing advice online must ensure the advice is correct and kept current.
Patient E demonstrates the importance of monitoring patients remotely (blood pressure transmitted to the office) or with periodic in-office visits. It is not clear whether she was a no-show for office visits (and whether the office followed up on any missed appointments) or if such visits were never scheduled. Liability for failure to monitor adequately is a possibility.
Patient F’s seemingly minor complaint could be a potential problem. Dr. TM used an insecure mode of communication. Although some HIPAA security regulations were modified or suspended during the pandemic, using such an unsecure platform is problematic, especially if temporary HIPAA rules expired. The outcome of the complaint is in doubt.
(See TABLE 2 for additional comments on patients D, E, and F.)
Out-of-state practice
Dr. TM treated 3 out-of-state residents (D, E, and F) via telemedicine. Recently Dr. TM received a complaint from the State Medical Licensure Board for practicing medicine without a license (Patient D), followed by similar charges from Patient E’s and Patient F’s state licensing boards. He has received a licensing inquiry from his home state board about those claims of illegal practice in other states and incompetent treatment.
Patient D’s pregnancy did not go well. The 1 in-person visit did not occur and she has filed a malpractice suit against Dr. TM. Patient E is threatening a malpractice case because the STI was not appropriately diagnosed and had advanced before another physician treated it.
In addition, a private citizen in Patient F’s state has filed suit against Dr. TM for abetting an illegal abortion (for Patient F).
Discussion. Patients D, E, and F illustrate the risk of even incidental out-of-state practice. The medical board inquiries arose from anonymous tips to all 4 states reporting Dr. TM was “practicing medicine without a license.” Patient E’s home state did have a licensing compact with the adjoining state (ie, Dr. TM’s home state). However, it required physicians to register and file an annual report, which Dr. TM had not done. The other 2 states did not have compacts with Dr. TM’s home state. Thus, he was illegally practicing medicine and would be subject to penalties. His home state also might impose license discipline based on his illegal practice in other states.
Continue to: What’s the verdict?...
What’s the verdict?
Dr. TM’s malpractice carrier is refusing to defend the claims of medical malpractice threatened by Patients D, E, and F. The company first notes that the terms of the malpractice policy specifically exclude the illegal practice of medicine. Furthermore, when a physician legally practices in another state, the policy requires a written notice to the insurance carrier of such practice. Dr. TM will likely have to engage and pay for a malpractice attorney for these cases. Because the claims are filed in 3 different states, more than a home-state attorney will likely be involved in the defense of these cases. Dr. TM will need to pay the attorneys and any damages from a settlement or trial.
Malpractice claims. Patient D claims that the doctor essentially abandoned her by never reaching out to her or arranging an in-person visit. Dr. TM claims the patient was responsible for scheduling the in-person visit. Patient E claims it was malpractice not to determine the specific nature of the STI and to do follow-up testing to determine that it was cured. All patients claim there was no genuine informed consent to the telemedicine. An attorney has warned Dr. TM that it is “not going to look good to the jury” that he was practicing without a license in the state and suggests he settle the cases quickly by paying damages.
Abortion-related claims. Patient F presents a different set of problems. Dr. TM’s home state is “proabortion.” Patient F’s home state is strongly “antiabortion,” making it a felony to participate in, assist, or facilitate an abortion (including medical abortion). Criminal charges have been filed against Dr. TM for the illegal practice of medicine, for aiding and facilitating an abortion, and for failure to notify a parent that a minor is seeking an abortion. For now, Dr. TM’s state is refusing to extradite on the abortion charge. Still, the patient’s state insists that it do so on the illegal practice of medicine charges and new charges of insurance fraud and failure to report suspected sexual abuse of a child. (Under the patient’s state law, anyone having sex with Patient F would have engaged in sexual abuse or “statutory rape,” so the state insists that the fact she was pregnant proves someone had sex with her.)
Patient F’s state also has a statute that allows private citizens to file civil claims against anyone procuring or assisting with an abortion (a successful private citizen can receive a minimum of $10,000 from the defendant). Several citizens from the patient’s state have already filed claims against Dr. TM in his state courts. Only one of them, probably the first to file, could succeed. Courts in the state have issued subpoenas and ordered Dr. TM to appear and reply to the civil suits. If he does not respond, there will be a default judgment.
Dr. TM’s attorney tells him that these lawsuits will not settle and will take a long time to defend and resolve. That will be expensive.
Billing and fraud. Dr. TM’s office recently received a series of notices from private health insurers stating they are investigating previously made payments as being fraudulent (unlicensed). They will not pay any new claims pending the investigation. On behalf of Medicare-Medicaid and other federal programs, the US Attorney’s office has notified Dr. TM that it has opened an investigation into fraudulent federal payments. F’s home state also is filing a (criminal) insurance fraud case, although the basis for it is unclear. (Dr. TM’s attorney believes it might be to increase pressure on the physician’s state to extradite Dr. TM for Patient F’s case.)
In addition, a disgruntled former employee of Dr. TM has filed a federal FCA case against him for filing inflated claims with various federally funded programs. The employee also made whistleblower calls to insurance companies and some state-funded medical programs. A forensic accounting investigation by Dr. TM’s accountant confirmed a pattern of very sloppy records and recurring billing for televisits that did not occur. Dr. TM believes that this was the act of one of the temporary assistants he hired in a pinch, who did not understand the system and just guessed when filing some insurance claims.
During the investigation, the federal and state attorneys are looking into a possible violation of state and federal Anti-Kickback Statutes. This is based on the original offer of a $100 credit for referrals to Dr. TM’s telemedicine practice.
The attorneys are concerned that other legal problems may present themselves. They are thoroughly reviewing Dr. TM’s practice and making several critical but somewhat modest changes to his practice. They also have insisted that Dr. TM have appropriate staff to handle the details of the practice and billing.
Conclusions
Telemedicine presents notable legal challenges to medical practice. As the pandemic status ends, ObGyn physicians practicing telemedicine need to be aware of the rules and how they are changing. For those physicians who want to continue or start a telemedicine practice, securing legal and technical support to ensure your operations are inline with the legal requirements can minimize any risk of legal troubles in the future. ●
A physician in State A, where abortion is legal, has a telemedicine patient in State B, where it is illegal to assist, provide, or procure an abortion. If the physician prescribes a medical abortion, he would violate the law of State B by using telemedicine to help the patient (located in State B) obtain an abortion. This could result in criminal charges against the prescribing physician.
- Board on Health Care Services; Institute of Medicine. The Role of Telehealth in an Evolving Health Care Environment: Workshop Summary. National Academies Press: 2012. https://www.ncbi.nlm.nih.gov/books/NBK207145/. Accessed March 30, 2023.
- Bruhn HK. Telemedicine: dos and don’ts to mitigate liability risk. J APPOS. 2020;24:195-196. doi:10.1016/j.jaapos. 2020.07.002
- Implementing telehealth in practice: ACOG Committee Opinion Summary, number 798. Obstet Gynecol. 2020; 2135:493-494. doi:10.1097/AOG.0000000000003672
- Bestsennyy O, Gilbert G, Harris A, et al. Telehealth: a quarter-trillion-dollar post-COVID-19 reality? McKinsey & Company. July 9, 2021. Accessed March 2, 2023. https://www.mckinsey.com/industries/healthcare/our-insights /telehealth-a-quarter-trillion-dollar-post-covid-19-reality
- Stanley AY, Wallace JB. Telehealth to improve perinatal care access. MCN Am J Matern Child Nurs. 2022;47:281-287. doi: 10.1097/NMC.0000000000000841
- Warshaw R. Health disparities affect millions in rural US communities. Association of American Medical Colleges. Published October 31, 2017. Accessed March 31, 2023. https://www.aamc.org/news-insights/health-disparities -affect-millions-rural-us-communities
- Almuslin H, AlDossary S. Models of incorporating telehealth into obstetric care during the COVID-19 pandemic, its benefits and barriers: a scoping review. Telemed J E Health. 2022;28:24-38. doi:10.1089/tmj.2020.0553
- Gold AE, Gilbert A, McMichael BJ. Socially distant health care. Tul L Rev. 2021;96:423-468. https://scholarship .law.ua.edu/cgi/viewcontent.cgi?article=1713&context =fac_articles. Accessed March 4, 2023.
- Zolotor AJ, Carlough MC. Update on prenatal care. Am Fam Physician. 2014;89:199-208.
- Odibo IN, Wendel PJ, Magann EF. Telemedicine in obstetrics. Clin Obstet Gynecol. 2013;56:422-433. doi:10.1097/ GRF.0b013e318290fef0
- Shmerling A, Hoss M, Malam N, et al. Prenatal care via telehealth. Prim Care. 2022;49:609-619. doi:10.1016/j. pop.2022.05.002
- Madden N, Emeruwa UN, Friedman AM, et al. Telehealth uptake into prenatal care and provider attitudes during COVID-19 pandemic in New York City: a quantitative and qualitative analysis. Am J Perinatol. 2020;37:1005-1014. doi:10.1055/s-0040-1712939
- Dosaj A, Thiyagarajan D, Ter Haar C, et al. Rapid implementation of telehealth services during the COVID-19 pandemic. Telemed J E Health. 2020;27:116-120. doi:10.1089/ tmj.2020.0219
- Lurie N, Carr B. The role of telehealth in the medical response to disasters. JAMA Intern Med. 2018;187:745-746. doi: 10.1001/jamainternmed.2018.1314
- Tobah YSB, LeBlanc A, Branda E, et al. Randomized comparison of a reduced-visit prenatal care model enhanced with remote monitoring. Am J Obstet Gynecol. 2019;221:638-e1-638.e8. doi:10.1016/j.ajog.2019.06.034
- Vivanti AJ, Deruelle P, Piccone O, et al. Follow-up for pregnant women during the COVID-19 pandemic: French national authority for health recommendations. J Gynecol Obstet Hum Reprod. 2020;49:101804. doi:10.1016/j. jogoh.2020.101804
- Ellimoottil C. Takeaways from 2 key studies on interstate telehealth use among Medicare fee-for-service beneficiaries. JAMA Health Forum. 2022;3:e223020-E223020. doi:10.1001/ jamahealthforum.2022.3020
- Harris J, Hartnett T, Hoagland GW, et al. What eliminating barriers to interstate telehealth taught us during the pandemic. Bipartisan Policy Center. Published November 2021. Accessed March 9, 2023. https://bipartisanpolicy .org/download/?file=/wp-content/uploads/2021/11/BPC -Health-Licensure-Brief_WEB.pdf.
- Center for Connected Health Policy. Cross-state licensing. Accessed February 21, 2023. https://www.cchpca.org/topic /cross-state-licensing-professional-requirements.
- US Department of Health & Human Services. Telehealth. Getting started with licensure. Published February 3, 2023. Accessed February 27, 2023. https://telehealth.hhs.gov /licensure/getting-started-licensure/
- US Department of Health & Human Services. Telehealth. Licensure. Accessed February 27, 2023. https://telehealth .hhs.gov/licensure
- US Department of Health & Human Services. National Practitioner Data Bank (NPDB) code lists. Published December 2022. Accessed March 9, 2023. https://www.npdb .hrsa.gov/software/CodeLists.pdf
- American College of Obstetricians and Gynecologists. COVID-19 FAQs for obstetrician-gynecologists, telehealth. 2020. Accessed March 5, 2023. https://www.acog.org /clinical-information/physician-faqs/covid-19-faqs-for -ob-gyns-telehealth
- Gorman RK. Prescribing medication through the practice of telemedicine: a comparative analysis of federal and state online prescribing policies, and policy considerations for the future. S Cal Interdisc Law J. 2020;30:739-769. https://gould .usc.edu/why/students/orgs/ilj/assets/docs/30-3-Gorman. pdf. Accessed March 10, 2023.
- Farringer DR. A telehealth explosion: using lessons from the pandemic to shape the future of telehealth regulation. Tex A&M Law Rev. 2021;9:1-47. https://scholarship.law.tamu. edu/cgi/viewcontent.cgi?article=1232&context=lawreview. Accessed February 28, 2023.
- Sterba KR, Johnson EE, Douglas E, et al. Implementation of a women’s reproductive behavioral health telemedicine program: a qualitative study of barriers and facilitators in obstetric and pediatric clinics. BMC Pregnancy Childbirth. 2023;23:167, 1-10. doi:10.1186/s12884-023-05463-2.
- US Department of Justice. COVID-19 FAQ (telemedicine). https://www.deadiversion.usdoj.gov/faq/coronavirus_faq .htm#TELE_FAQ2. Accessed March 13, 2023.
- US Department of Health & Human Services. Guidance on how the HIPAA rules permit covered health care providers and health plans to use remote communication technologies for audio-only telehealth. Published June 13, 2022. Accessed February 22, 2023. https://www.hhs.gov/hipaa/for-professionals/privacy /guidance/hipaa-audio-telehealth/index.html.
- Gray JME. HIPAA, telehealth, and the treatment of mental illness in a post-COVID world. Okla City Uni Law Rev. 2021;46:1-26. https://law.okcu.edu/wp-content /uploads/2022/04/J-Michael-E-Gray-HIPAA-Telehealth -and-Treament.pdf. Accessed March 9, 2023.
- Kurzweil C. Telemental health care and data privacy: current HIPAA privacy pitfalls and a proposed solution. Ann Health L Adv Dir. 2022;31:165.
- US Department of Health & Human Services and US Department of Justice. Health care fraud and abuse control program FY 2020: annual report. July 2021. Accessed March 9, 2023. https://oig.hhs.gov/publications/docs/hcfac /FY2020-hcfac.pdf
- Copeland KB. Telemedicine scams. Iowa Law Rev. 2022: 108:69-126. https://ilr.law.uiowa.edu/sites/ilr.law.uiowa.edu /files/2023-01/A2_Copeland.pdf. Accessed March 10, 2023.
- Solimini R, Busardò FP, Gibelli F, et al. Ethical and legal challenges of telemedicine in the era of the COVID-19 pandemic. Medicina (Kaunas). 2021;57:13141324. doi:10.3390/medicina57121314
- Reed A. COVID: a silver linings playbook. mobilizing pandemic era success stories to advance reproductive justice. Berkeley J Gender Law Justice. 2022;37:221-266. https://lawcat.berkeley.edu/record/1237158/files/16%20 Reed_final.pdf. Accessed March 11, 2023.
- Women’s Preventive Services Initiative and The American College of Obstetricians and Gynecologists. FAQ for telehealth services. Accessed March 2, 2023. https://www .womenspreventivehealth.org/wp-content/uploads/WPSI -Telehealth-FAQ.pdf
- Warren L, Chen KT. Telehealth apps in ObGyn practice. OBG Manag. 2022;34:46-47. doi:10.12788/obgm.0178
- American College of Obstetricians and Gynecologists. 10 telehealth tips for an Ob-Gyn visit. 2020. Accessed March 2, 2023. https://www.acog.org/womens-health /infographics/10-telehealth-tips-for-an-ob-gyn-visit
- Wolf TD. Telemedicine and malpractice: creating uniformity at the national level. Wm Mary Law Rev. 2019;61:15051536. https://scholarship.law.wm.edu/cgi/viewcontent.cgi ?article=3862&context=wmlr. Accessed March 11, 2023.
- Cahan E. Lawsuits, reimbursement, and liability insurance— facing the realities of a post-Roe era. JAMA. 2022;328:515517. doi:10.1001/jama.2022.9193
- Heinrich L, Hernandez AK, Laurie AR. Telehealth considerations for the adolescent patient. Prim Care. 2022;49:597-607. doi:10.1016/j.pop.2022.04.006
- Guttmacher Institute. An overview of consent to reproductive health services by young people. Published March 1, 2023. Accessed April 1, 2023. https://www.guttmacher.org /state-policy/explore/overview-minors-consent-law.
- Dobbs v. Jackson Women’s Health. No. 19–1392. June 24, 2022. Accessed April 1, 2023. https://www.supremecourt .gov/opinions/21pdf/19-1392_6j37.pdf
- Lindgren Y. Dobbs v. Jackson Women’s Health and the post-Roe landscape. J Am Acad Matrimonial Law. 2022;35:235283. https://www.aaml.org/wp-content/uploads/MAT110-1 .pdf. Accessed March 11, 2023.
- Mohiuddin H. The use of telemedicine during a pandemic to provide access to medication abortion. Hous J Health Law Policy. 2021;21:483-525. https://houstonhealthlaw. scholasticahq.com/article/34611.pdf. Accessed March 10, 2023.
- Rebouché R. The public health turn in reproductive rights. Wash & Lee Law Rev. 2021;78:1355-1432. https:// scholarlycommons.law.wlu.edu/cgi/viewcontent .cgi?article=4743&context=wlulr. Accessed March 10, 2023.
- Fliegel R. Access to medication abortion: now more important than ever. Am J Law Med. 2022;48:286-304. doi:10.1017/amj.2022.24
- Guttmacher Institute. Medication abortion. March 1, 2023. Accessed April 1, 2023 https://www.guttmacher.org /state-policy/explore/medication-abortion#:~:text=In%20 January%202023%2C%20the%20FDA,order%20to%20 dispense%20the%20pills
- Cohen DS, Donley G, Rebouché R. The new abortion battleground. Columbia Law Rev. 2023;123:1-100. https:// columbialawreview.org/content/the-new-abortion -battleground/. Accessed March 1, 2023.
- Hunt SA. Call me, beep me, if you want to reach me: utilizing telemedicine to expand abortion access. Vanderbilt Law Rev. 2023;76:323-359. Accessed March 10, 2023. https:// vanderbiltlawreview.org/lawreview/wp-content/uploads /sites/278/2023/01/Call-Me-Beep-Me-If-You-Want-toReach-Me-Utilizing-Telemedicine-to-Expand-AbortionAccess.pdf
- Gleckel JA, Wulkan SL. Abortion and telemedicine: looking beyond COVID-19 and the shadow docket. UC Davis Law Rev Online. 2020;54:105-121. https://lawreview.law.ucdavis. edu/online/54/files/54-online-Gleckel_Wulkan.pdf. Accessed April 1, 2023.
Telemedicine (or telehealth) originated in the early 1900s, when radios were used to communicate medical advice to clinics aboard ships.1 According to the American Telemedicine Association, telemedicine is namely “the use of medical information exchanged from one site to another via electronic communications to improve a patient’s clinical health status.”2 These communications use 2-way video, email, smartphones, wireless tools, and other forms of telecommunications technology.
During the COVID-19 pandemic, many ObGyns—encouraged and advised by professional organizations—began providing telemedicine services.3 The first reported case of COVID-19 was in late 2019; the use of telemedicine was 38 times higher in February 2021 than in February 2020,4 illustrating how many physicians quickly moved to telemedicine practices.
CASE Dr. TM’s telemedicine dream
Before COVID-19, Dr. TM (an ObGyn practi-tioner) practiced in-person medicine in his home state. With the onset of the pandemic, Dr. TM struggled to switch to primarily seeing patients online (generally using Zoom or Facebook Live), with 1 day per week in the office for essential in-person visits.
After several months, however, Dr. TM’s routine became very efficient. He could see many more patients in a shorter time than with the former, in-person system. Therefore, as staff left his practice, Dr. TM did not replace them and also laid off others. Ultimately, the practice had 1 full-time records/insurance secretary who worked from home and 1 part-time nurse who helped with the in-person day and answered some patient inquiries by email. In part as an effort to add new patients, Dr. TM built an engaging website through which his current patients could receive medical information and new patients could sign up.
In late 2022, Dr. TM offered a $100 credit to any current patient who referred a friend or family member who then became a patient. This promotion was surprisingly effective and resulted in an influx of new patients. For example, Patient Z (a long-time patient) received 3 credits for referring her 3 sisters who lived out of state and became telepatients: Patient D, who lived 200 hundred miles away; Patient E, who lived 50 miles away in the adjoining state; and Patient F, who lived 150 miles away. Patient D contacted Dr. TM because she thought she was pregnant and wanted prenatal care, Patient E thought she might have a sexually transmitted infection (STI) and wanted treatment, and Patient F wanted general care and was inquiring about a medical abortion. Dr. TM agreed to treat Patient D but required 1 in-person visit. After 1 brief telemedicine session each with Patients E and F, Dr. TM wrote prescriptions for them.
By 2023, Dr. TM was enthusiastic about telemedicine as a professional practice. However, problems would ensue.
Dos and don’ts of telemedicine2
- Do take the initiative and inform patients of the availability of telemedicine/telehealth services
- Do use the services of medical malpractice insurance companies with regard to telemedicine
- Do integrate telemedicine into practice protocols and account for their limitations
- Don’t assume there are blanket exemptions or waivers in the states where your patients are located
Medical considerations
Telemedicine is endorsed by the American College of Obstetricians and Gynecologists (ACOG) as a vehicle for delivering prenatal and postpartum care.5 This represents an effort to reduce maternal and neonatal morbidity and mortality,5 as well as expandaccess to care and address the deficit in primary care providers and services, especially in rural and underserved populations.5,6 For obstetrics, prenatal care is designed to optimize pregnancy, childbirth, and postpartum care, with a focus on nutrition and genetic consultation and patient education on pregnancy, childbearing, breastfeeding, and newborn care.7
Benefits of telemedicine include its convenience for patients and providers, its efficiency and lower costs for providers (and hopefully patients, as well), and the potential improved access to care for patients.8 It is estimated that if a woman inititates obstetric care at 6 weeks, over the course of the 40-week gestation period, 15 prenatal visits will occur.9 Ultimately, the number of visits is determined based on the specifics of the pregnancy. With telemedicine, clinicians can provide those consultations, and information related to: ultrasonography, fetal echocardiography, and postpartum care services remotely.10 Using telemedicine may reduce missed visits, and remote monitoring may improve the quality of care.11
Barriers to telemedicine care include technical limitations, time constraints, and patient concerns of telehealth (visits). Technical limitations include the lack of a high speed internet connection and/or a smart device and the initial technical set-up–related problems,12 which affect providers as well as patients. Time constraints primarly refer to the ObGyn practice’s lack of time to establish telehealth services.13 Other challenges include integrating translation services, billing-related problems,10 and reimbursement and licensing barriers.14
Before the COVID-19 pandemic, obstetrics led the way in telemedicine with the development of the OB Nest model. Designed to replace in-person obstetrics care visits with telehealth,15 it includes home management tools such as blood pressure cuffs, cardiotocography, scales for weight checks, and Doppler ultrasounds.10 Patients can be instructed to measure fundal height and receive medications by mail. Anesthesia consultation can occur via this venue by having the patient complete a questionnaire prior to arriving at the labor and delivery unit.16
Legal considerations
With the COVID-19 pandemic, temporary changes were made to encourage the rapid adoption of telemedicine, including changes to licensing laws, certain prescription requirements, Health Insurance Portability and Accountability Act (HIPAA) privacy-security regulations, and reimbursement rules that required in-person visits. Thus, many ObGyns started using telemedicine during this rarified period, in which the rules appeared to be few and far between, with limited enforcement of the law and professional obligations.17 However, now that many of the legal rules that were suspended or ignored have been (or are being) reimposed and enforced, it is important for providers to become familiar with the legal issues involved in practicing telemedicine.
First, where is the patient? When discussing the legal issues of telemedicine, it is important to remember that many legal rules for medical care (ie, liability, informed consent, and licensing) vary from state to state. If the patient resides in a different state (“foreign” state) from the physician’s practice location (the physician’s “home” state), the care is considered delivered in the state where the patient is located. Thus, the patient’s location generally establishes the law covering the telemedicine transaction. In the following discussion, the rules refer to the law and professional obligations, with commentary on some key legal issues that are relevant to ObGyn telemedicine.
Continue to: Reinforcing the rules...
Reinforcing the rules
Licensing
During the height of the COVID-19 pandemic, the federal government and almost all states temporarily modified the licensing requirement to allow telemedicine based on an existing medical license in any state—disregarding the “where is the patient” rule. As those rules begin to lapse or change with the official end of the pandemic declared by President Biden as May 2023,17 the rules under which a physician began telemedicine interstate practice in 2020 also may be changing.
Simply put, “The same standards for licensure apply to health care providers regardless of whether care is delivered in-person or virtually through telehealth services.”18 When a physician is engaged in telemedicine treatment of a patient in the physician’s home state, there is generally no licensing issue. Telemedicine generally does not require a separate specific license.19 However, when the patient is in another state (a “foreign” state), there can be a substantial licensing issue.20 Ordinarily, to provide that treatment, the physician must, in some manner, be approved to practice in the patient’s state. That may occur, for example, in the following ways: (1) the physician may hold an additional regular license in the patient’s state, which allows practice there, or (2) the physician may have received permission for “temporary practice” in another state.
Many states (often adjoining states) have formal agreements with other states that allow telemedicine practice by providers in each other’s states. There also are “compacts”, or agreements that enable providers in any of the participating states to practice in the other associated states without a separate license.18 Although several websites provide information about compact licensing and the like, clinicians should not rely on simple lists or maps. Individual states may have special provisions about applying their laws to out-of-state “compact” physicians. In addition, under the Interstate Medical Licensure Compact, “physicians have to pay licensing fees and satisfy the requirements of each medical board in the states where they wish to practice.”21
Consequences. Practicing telemedicine with a patient in a state where the physician does not have a license is generally a crime. Furthermore, it may be the basis for license discipline in the physician’s home state and result in a report to the National Practi-tioner Databank.22 In addition, reimbursement often depends on the practitioner being licensed, and the absence of a license may be a basis for denying payment for services.23 Finally, malpractice insurance generally is limited to licensed practice. Thus, the insurer may decline to defend the unlicensed clinician against a malpractice claim or pay any damages.
Prescribing privileges
Prescribing privileges usually are connected to licensing, so as the rules for licensing change postpandemic, so do the rules for prescribing. In most cases, the physician must have a license in the state where care is given to prescribe medication—which in telemedicine, as noted, typically means the state where the patient is located. Exceptions vary by state, but in general, if a physician does not have a license to provide care, the physician is unlikely to be authorized to prescribe medication.24 Failure to abide by the applicable state rules may result in civil and even criminal liability for illegal prescribing activity.
In addition, the US Food and Drug Administration (FDA) and Drug Enforcement Administration (DEA, which enforces laws concerning controlled substances) also regulate the prescription and sale of pharmaceuticals.25 There are state and federal limits on the ability of clinicians to order controlled substances without an in-person visit. The Ryan Haight Online Pharmacy Consumer Protection Act, for example, sets limits on controlled substance prescriptions without an in-person examination.26 Federal law was modified due to COVID-19 to permit prescribing of many controlled substances by telemedicine if there is synchronous audio and visual examination of the patient. Physicians who write such prescriptions also are required to have a DEA registration in the patient’s state. This is an essential consideration for physicians considering interstate telemedicine practice.27
HIPAA and privacy
Governments waived some of the legal requirements related to health information during the pandemic, but those waivers either have expired or will do so soon. Federal and state laws regarding privacy and security—notably including HIPAA—apply to telemedicine and are of particular concern given the considerable amount of communication of protected health information with telemedicine.
HIPAA security rules essentially require making sure health information cannot be hacked or intercepted. Audio-only telemedicine by landline (not cell) is acceptable under the security rules, but almost all other remote communication requires secure communications.28
Clinicians also need to adhere to the more usual HIPAA privacy rules when practicingtelehealth. State laws protecting patient privacy vary and may be more stringent than HIPAA, so clinicians also must know the requirements in any state where they practice—whether in office or telemedicine.29
Making sure telemedicine practices are consistent with these security and privacy rules often requires particular technical expertise that is outside the realm of most practicing clinicians. However, without modification, the pre-telemedicine technology of many medical offices likely is insufficient for the full range of telemedicine services.30
Reimbursement and fraud
Before COVID-19, Medicare and Medicaid reimbursement for telemedicine was limited. Government decisions to substantially broaden those reimbursement rules (at least temporarily) provided a substantial boost to telemedicine early in the pandemic.23 Federal regulations and statutes also expanded telemedicine reimbursement for various services. Some will end shortly after the health emergency, and others will be permanent. Parts of that will not be sorted out for several years, so it will likely be a changing landscape for reimbursement.
Continue to: Rules that are evolving...
Rules that are evolving
Informed consent
The ethical and legal obligations to obtain informed consent are present in telemedicineas well as in-person care, with the same basic requirements regarding risks, benefits, alternative care, etc.32 However, with telemedicine, information related to remote care should be included and is outlined in TABLE 1.
Certain states may have somewhat unique informed consent requirements—especially for reproductive care, including abortion.34 Therefore, it is important for clinicians to ensure their consent process and forms comply with any legal jurisdiction in which a patient is located.
Medical malpractice
The basics of medical malpractice (or negligence) are the same in telemedicine as in in-person care: duty, breach of duty, and injury caused by the breach. That is, there may be liability when a medical professional breaches the duty of care, causing the patient’s injury. The physician’s duty is defined by the quality of care that the profession (specialty) accepts as reasonably good. This is defined by the opinions of physicians within the specialty and formal statements from professional organizations, including ACOG.3
Maintaining the standard of care and quality. The use of telemedicine is not an excuse to lower the quality of health care. There are some circumstances for which it is medically better to have an in-person visit. In these instances, the provider should recommend the appropriate care, even if telemedicine would be more convenient for the provider and staff.35
If the patient insists and telemedicine might result in less than optimal care, the reasons for using a remote visit should be clearly documented contemporaneously with the decision. Furthermore, when the limitations of being unable to physically examine the patient result in less information than is needed for the patient’s care, the provider must find alternatives to make up for the information gap.11,36 It also may be necessary to inform patients about how to maximize telemedicine care.37 At the beginning of telemedicine care the provider should include information about the nature and limits of telehealth, and the patient’s responsibilities. (See TABLE 1) Throughout treatment of the patient, that information should be updated by the provider. That, of course, is particularly important for patients who have not previously used telemedice services.
Malpractice rules vary by state. Many states have special rules regarding malpractice cases. These differences in malpractice standards and regulations “can be problematic for physicians who use telemedicine services to provide care outside the state in which they practice.”38 Caps on noneconomic damages are an example. Those state rules would apply to telemedicine in the patient’s state.
Malpractice insurance
Malpractice insurance now commonly includes telemedicine legally practiced within the physician’s home state. Practitioners who treat patients in foreign states should carefully examine their malpractice insurance policies to confirm that the coverage extends to practice in those states.39 Malpractice carriers may require notification by a covered physician who routinely provides services to patients in another state.3
Keep in mind, malpractice insurance generally does not cover the practice of medicine that is illegal. Practicing telemedicine in a foreign state, where the physician or other provider does not have a license and where that state does not otherwise permit the practice, is illegal. Most likely, the physician’s malpractice insurance will not cover claims that arise from this illegal practice in a foreign state or provide defense for malpractice claims, including frivolous lawsuits. Thus, the physician will pay out of pocket for the costs of a defense attorney.
Telemedicine treatment of minors
Children and adolescents present special legal issues for ObGyn care, which may become more complicated with telemedicine. Historically, parents are responsible for minors (those aged <18 years): they consent to medical treatment, are responsible for paying for it, and have the right to receive information about treatment.
Over the years, though, many states have made exceptions to these principles, especially with regard to contraception and treatment of sexually transmitted diseases.40 For abortion, in particular, there is considerable variation among the states in parental consent and notification.41 The Supreme Court’s decision in Dobbs v Jackson Women’s Health42 may (depending on the state) be followed with more stringent limitations on adolescent consent to abortions, including medical abortions.43
Use of telehealth does not change any obligations regarding adolescent consent or parental notification. Because those differ considerably among states, it is important for all practitioners to know their states’ requirements and keep reasonably complete records demonstrating their compliance with state law.
Abortion
The most heated current controversy about telemedicine involves abortion—specifically medical abortion, which is the combination of mifepristone and misoprostol.44,45 The FDA approved the combination in 2000. Almost immediately, many states required in-person visits with a certified clinician to receive a prescription for mifepristone and misoprostol, and eventually, the FDA adopted similar requirements.46 However, during the pandemic from 2021 to 2022, the FDA permitted telemedicine prescriptions. Several states still require in-person physician visits, although the constitutionality of those requirements has not been established.47
With the Supreme Court’s decision in Dobbs v Jackson Women’s Health in 2022,42 disagreements have ensued about the degree to which states may regulate the prescription of FDA-approved medical abortion drugs. Thorny constitutional issues exist in the plans of both abortion opponents and proponents in the battle over medical abortion in antiabortion states. It may be that federal drug law preempts state laws limiting access to FDA-approved drugs. On the other hand, it may be that states can make it a crime within the state to possess or provide abortion-inducing drugs. Courts will probably take years to resolve the many tangled legal questions.48
Thus, while the pandemic telemedicine rules may have advanced access to abortion,34 there may be some pending downsides.49 States that prohibit abortion will likely include prohibitions on medical abortions. In addition, they may prohibit anyone in the state (including pharmacies) from selling, possessing, or obtaining any drug used for causing or inducing an abortion.50 If, for constitutional reasons, they cannot press criminal charges or undertake licensing discipline for prescribing abortion, some states will likely withdraw from telehealth licensing compacts to avoid out-of-state prescriptions. This area of telemedicine has considerable uncertainty.
Continue to: CASE Conclusion...
CASE Conclusion
Patient concerns come to the fore
By 2023, Dr. TM started receiving bad news. Patient D called complaining that after following the advice on the website, she suffered a severe reaction and had to be rushed to an emergency department. Patient E (who had only 1 in-office visit early in her pregnancy) notified the office that she developed very high blood pressure that resulted in severe placental abruption, requiring emergency care and resulting in the loss of the fetus. Patient F complained that someone hacked the TikTok direct message communication with Dr. TM and tried to “blackmail” or harass her.
Discussion. Patients D, E, and F represent potential problems of telemedicine practice. Patient D was injured because she relied on her doctor’s website (to which Dr. TM directed patients). It contained an error that caused an injury. A doctor-patient relationship existed, and bad medical advice likely caused the injury. Physicians providing advice online must ensure the advice is correct and kept current.
Patient E demonstrates the importance of monitoring patients remotely (blood pressure transmitted to the office) or with periodic in-office visits. It is not clear whether she was a no-show for office visits (and whether the office followed up on any missed appointments) or if such visits were never scheduled. Liability for failure to monitor adequately is a possibility.
Patient F’s seemingly minor complaint could be a potential problem. Dr. TM used an insecure mode of communication. Although some HIPAA security regulations were modified or suspended during the pandemic, using such an unsecure platform is problematic, especially if temporary HIPAA rules expired. The outcome of the complaint is in doubt.
(See TABLE 2 for additional comments on patients D, E, and F.)
Out-of-state practice
Dr. TM treated 3 out-of-state residents (D, E, and F) via telemedicine. Recently Dr. TM received a complaint from the State Medical Licensure Board for practicing medicine without a license (Patient D), followed by similar charges from Patient E’s and Patient F’s state licensing boards. He has received a licensing inquiry from his home state board about those claims of illegal practice in other states and incompetent treatment.
Patient D’s pregnancy did not go well. The 1 in-person visit did not occur and she has filed a malpractice suit against Dr. TM. Patient E is threatening a malpractice case because the STI was not appropriately diagnosed and had advanced before another physician treated it.
In addition, a private citizen in Patient F’s state has filed suit against Dr. TM for abetting an illegal abortion (for Patient F).
Discussion. Patients D, E, and F illustrate the risk of even incidental out-of-state practice. The medical board inquiries arose from anonymous tips to all 4 states reporting Dr. TM was “practicing medicine without a license.” Patient E’s home state did have a licensing compact with the adjoining state (ie, Dr. TM’s home state). However, it required physicians to register and file an annual report, which Dr. TM had not done. The other 2 states did not have compacts with Dr. TM’s home state. Thus, he was illegally practicing medicine and would be subject to penalties. His home state also might impose license discipline based on his illegal practice in other states.
Continue to: What’s the verdict?...
What’s the verdict?
Dr. TM’s malpractice carrier is refusing to defend the claims of medical malpractice threatened by Patients D, E, and F. The company first notes that the terms of the malpractice policy specifically exclude the illegal practice of medicine. Furthermore, when a physician legally practices in another state, the policy requires a written notice to the insurance carrier of such practice. Dr. TM will likely have to engage and pay for a malpractice attorney for these cases. Because the claims are filed in 3 different states, more than a home-state attorney will likely be involved in the defense of these cases. Dr. TM will need to pay the attorneys and any damages from a settlement or trial.
Malpractice claims. Patient D claims that the doctor essentially abandoned her by never reaching out to her or arranging an in-person visit. Dr. TM claims the patient was responsible for scheduling the in-person visit. Patient E claims it was malpractice not to determine the specific nature of the STI and to do follow-up testing to determine that it was cured. All patients claim there was no genuine informed consent to the telemedicine. An attorney has warned Dr. TM that it is “not going to look good to the jury” that he was practicing without a license in the state and suggests he settle the cases quickly by paying damages.
Abortion-related claims. Patient F presents a different set of problems. Dr. TM’s home state is “proabortion.” Patient F’s home state is strongly “antiabortion,” making it a felony to participate in, assist, or facilitate an abortion (including medical abortion). Criminal charges have been filed against Dr. TM for the illegal practice of medicine, for aiding and facilitating an abortion, and for failure to notify a parent that a minor is seeking an abortion. For now, Dr. TM’s state is refusing to extradite on the abortion charge. Still, the patient’s state insists that it do so on the illegal practice of medicine charges and new charges of insurance fraud and failure to report suspected sexual abuse of a child. (Under the patient’s state law, anyone having sex with Patient F would have engaged in sexual abuse or “statutory rape,” so the state insists that the fact she was pregnant proves someone had sex with her.)
Patient F’s state also has a statute that allows private citizens to file civil claims against anyone procuring or assisting with an abortion (a successful private citizen can receive a minimum of $10,000 from the defendant). Several citizens from the patient’s state have already filed claims against Dr. TM in his state courts. Only one of them, probably the first to file, could succeed. Courts in the state have issued subpoenas and ordered Dr. TM to appear and reply to the civil suits. If he does not respond, there will be a default judgment.
Dr. TM’s attorney tells him that these lawsuits will not settle and will take a long time to defend and resolve. That will be expensive.
Billing and fraud. Dr. TM’s office recently received a series of notices from private health insurers stating they are investigating previously made payments as being fraudulent (unlicensed). They will not pay any new claims pending the investigation. On behalf of Medicare-Medicaid and other federal programs, the US Attorney’s office has notified Dr. TM that it has opened an investigation into fraudulent federal payments. F’s home state also is filing a (criminal) insurance fraud case, although the basis for it is unclear. (Dr. TM’s attorney believes it might be to increase pressure on the physician’s state to extradite Dr. TM for Patient F’s case.)
In addition, a disgruntled former employee of Dr. TM has filed a federal FCA case against him for filing inflated claims with various federally funded programs. The employee also made whistleblower calls to insurance companies and some state-funded medical programs. A forensic accounting investigation by Dr. TM’s accountant confirmed a pattern of very sloppy records and recurring billing for televisits that did not occur. Dr. TM believes that this was the act of one of the temporary assistants he hired in a pinch, who did not understand the system and just guessed when filing some insurance claims.
During the investigation, the federal and state attorneys are looking into a possible violation of state and federal Anti-Kickback Statutes. This is based on the original offer of a $100 credit for referrals to Dr. TM’s telemedicine practice.
The attorneys are concerned that other legal problems may present themselves. They are thoroughly reviewing Dr. TM’s practice and making several critical but somewhat modest changes to his practice. They also have insisted that Dr. TM have appropriate staff to handle the details of the practice and billing.
Conclusions
Telemedicine presents notable legal challenges to medical practice. As the pandemic status ends, ObGyn physicians practicing telemedicine need to be aware of the rules and how they are changing. For those physicians who want to continue or start a telemedicine practice, securing legal and technical support to ensure your operations are inline with the legal requirements can minimize any risk of legal troubles in the future. ●
A physician in State A, where abortion is legal, has a telemedicine patient in State B, where it is illegal to assist, provide, or procure an abortion. If the physician prescribes a medical abortion, he would violate the law of State B by using telemedicine to help the patient (located in State B) obtain an abortion. This could result in criminal charges against the prescribing physician.
Telemedicine (or telehealth) originated in the early 1900s, when radios were used to communicate medical advice to clinics aboard ships.1 According to the American Telemedicine Association, telemedicine is namely “the use of medical information exchanged from one site to another via electronic communications to improve a patient’s clinical health status.”2 These communications use 2-way video, email, smartphones, wireless tools, and other forms of telecommunications technology.
During the COVID-19 pandemic, many ObGyns—encouraged and advised by professional organizations—began providing telemedicine services.3 The first reported case of COVID-19 was in late 2019; the use of telemedicine was 38 times higher in February 2021 than in February 2020,4 illustrating how many physicians quickly moved to telemedicine practices.
CASE Dr. TM’s telemedicine dream
Before COVID-19, Dr. TM (an ObGyn practi-tioner) practiced in-person medicine in his home state. With the onset of the pandemic, Dr. TM struggled to switch to primarily seeing patients online (generally using Zoom or Facebook Live), with 1 day per week in the office for essential in-person visits.
After several months, however, Dr. TM’s routine became very efficient. He could see many more patients in a shorter time than with the former, in-person system. Therefore, as staff left his practice, Dr. TM did not replace them and also laid off others. Ultimately, the practice had 1 full-time records/insurance secretary who worked from home and 1 part-time nurse who helped with the in-person day and answered some patient inquiries by email. In part as an effort to add new patients, Dr. TM built an engaging website through which his current patients could receive medical information and new patients could sign up.
In late 2022, Dr. TM offered a $100 credit to any current patient who referred a friend or family member who then became a patient. This promotion was surprisingly effective and resulted in an influx of new patients. For example, Patient Z (a long-time patient) received 3 credits for referring her 3 sisters who lived out of state and became telepatients: Patient D, who lived 200 hundred miles away; Patient E, who lived 50 miles away in the adjoining state; and Patient F, who lived 150 miles away. Patient D contacted Dr. TM because she thought she was pregnant and wanted prenatal care, Patient E thought she might have a sexually transmitted infection (STI) and wanted treatment, and Patient F wanted general care and was inquiring about a medical abortion. Dr. TM agreed to treat Patient D but required 1 in-person visit. After 1 brief telemedicine session each with Patients E and F, Dr. TM wrote prescriptions for them.
By 2023, Dr. TM was enthusiastic about telemedicine as a professional practice. However, problems would ensue.
Dos and don’ts of telemedicine2
- Do take the initiative and inform patients of the availability of telemedicine/telehealth services
- Do use the services of medical malpractice insurance companies with regard to telemedicine
- Do integrate telemedicine into practice protocols and account for their limitations
- Don’t assume there are blanket exemptions or waivers in the states where your patients are located
Medical considerations
Telemedicine is endorsed by the American College of Obstetricians and Gynecologists (ACOG) as a vehicle for delivering prenatal and postpartum care.5 This represents an effort to reduce maternal and neonatal morbidity and mortality,5 as well as expandaccess to care and address the deficit in primary care providers and services, especially in rural and underserved populations.5,6 For obstetrics, prenatal care is designed to optimize pregnancy, childbirth, and postpartum care, with a focus on nutrition and genetic consultation and patient education on pregnancy, childbearing, breastfeeding, and newborn care.7
Benefits of telemedicine include its convenience for patients and providers, its efficiency and lower costs for providers (and hopefully patients, as well), and the potential improved access to care for patients.8 It is estimated that if a woman inititates obstetric care at 6 weeks, over the course of the 40-week gestation period, 15 prenatal visits will occur.9 Ultimately, the number of visits is determined based on the specifics of the pregnancy. With telemedicine, clinicians can provide those consultations, and information related to: ultrasonography, fetal echocardiography, and postpartum care services remotely.10 Using telemedicine may reduce missed visits, and remote monitoring may improve the quality of care.11
Barriers to telemedicine care include technical limitations, time constraints, and patient concerns of telehealth (visits). Technical limitations include the lack of a high speed internet connection and/or a smart device and the initial technical set-up–related problems,12 which affect providers as well as patients. Time constraints primarly refer to the ObGyn practice’s lack of time to establish telehealth services.13 Other challenges include integrating translation services, billing-related problems,10 and reimbursement and licensing barriers.14
Before the COVID-19 pandemic, obstetrics led the way in telemedicine with the development of the OB Nest model. Designed to replace in-person obstetrics care visits with telehealth,15 it includes home management tools such as blood pressure cuffs, cardiotocography, scales for weight checks, and Doppler ultrasounds.10 Patients can be instructed to measure fundal height and receive medications by mail. Anesthesia consultation can occur via this venue by having the patient complete a questionnaire prior to arriving at the labor and delivery unit.16
Legal considerations
With the COVID-19 pandemic, temporary changes were made to encourage the rapid adoption of telemedicine, including changes to licensing laws, certain prescription requirements, Health Insurance Portability and Accountability Act (HIPAA) privacy-security regulations, and reimbursement rules that required in-person visits. Thus, many ObGyns started using telemedicine during this rarified period, in which the rules appeared to be few and far between, with limited enforcement of the law and professional obligations.17 However, now that many of the legal rules that were suspended or ignored have been (or are being) reimposed and enforced, it is important for providers to become familiar with the legal issues involved in practicing telemedicine.
First, where is the patient? When discussing the legal issues of telemedicine, it is important to remember that many legal rules for medical care (ie, liability, informed consent, and licensing) vary from state to state. If the patient resides in a different state (“foreign” state) from the physician’s practice location (the physician’s “home” state), the care is considered delivered in the state where the patient is located. Thus, the patient’s location generally establishes the law covering the telemedicine transaction. In the following discussion, the rules refer to the law and professional obligations, with commentary on some key legal issues that are relevant to ObGyn telemedicine.
Continue to: Reinforcing the rules...
Reinforcing the rules
Licensing
During the height of the COVID-19 pandemic, the federal government and almost all states temporarily modified the licensing requirement to allow telemedicine based on an existing medical license in any state—disregarding the “where is the patient” rule. As those rules begin to lapse or change with the official end of the pandemic declared by President Biden as May 2023,17 the rules under which a physician began telemedicine interstate practice in 2020 also may be changing.
Simply put, “The same standards for licensure apply to health care providers regardless of whether care is delivered in-person or virtually through telehealth services.”18 When a physician is engaged in telemedicine treatment of a patient in the physician’s home state, there is generally no licensing issue. Telemedicine generally does not require a separate specific license.19 However, when the patient is in another state (a “foreign” state), there can be a substantial licensing issue.20 Ordinarily, to provide that treatment, the physician must, in some manner, be approved to practice in the patient’s state. That may occur, for example, in the following ways: (1) the physician may hold an additional regular license in the patient’s state, which allows practice there, or (2) the physician may have received permission for “temporary practice” in another state.
Many states (often adjoining states) have formal agreements with other states that allow telemedicine practice by providers in each other’s states. There also are “compacts”, or agreements that enable providers in any of the participating states to practice in the other associated states without a separate license.18 Although several websites provide information about compact licensing and the like, clinicians should not rely on simple lists or maps. Individual states may have special provisions about applying their laws to out-of-state “compact” physicians. In addition, under the Interstate Medical Licensure Compact, “physicians have to pay licensing fees and satisfy the requirements of each medical board in the states where they wish to practice.”21
Consequences. Practicing telemedicine with a patient in a state where the physician does not have a license is generally a crime. Furthermore, it may be the basis for license discipline in the physician’s home state and result in a report to the National Practi-tioner Databank.22 In addition, reimbursement often depends on the practitioner being licensed, and the absence of a license may be a basis for denying payment for services.23 Finally, malpractice insurance generally is limited to licensed practice. Thus, the insurer may decline to defend the unlicensed clinician against a malpractice claim or pay any damages.
Prescribing privileges
Prescribing privileges usually are connected to licensing, so as the rules for licensing change postpandemic, so do the rules for prescribing. In most cases, the physician must have a license in the state where care is given to prescribe medication—which in telemedicine, as noted, typically means the state where the patient is located. Exceptions vary by state, but in general, if a physician does not have a license to provide care, the physician is unlikely to be authorized to prescribe medication.24 Failure to abide by the applicable state rules may result in civil and even criminal liability for illegal prescribing activity.
In addition, the US Food and Drug Administration (FDA) and Drug Enforcement Administration (DEA, which enforces laws concerning controlled substances) also regulate the prescription and sale of pharmaceuticals.25 There are state and federal limits on the ability of clinicians to order controlled substances without an in-person visit. The Ryan Haight Online Pharmacy Consumer Protection Act, for example, sets limits on controlled substance prescriptions without an in-person examination.26 Federal law was modified due to COVID-19 to permit prescribing of many controlled substances by telemedicine if there is synchronous audio and visual examination of the patient. Physicians who write such prescriptions also are required to have a DEA registration in the patient’s state. This is an essential consideration for physicians considering interstate telemedicine practice.27
HIPAA and privacy
Governments waived some of the legal requirements related to health information during the pandemic, but those waivers either have expired or will do so soon. Federal and state laws regarding privacy and security—notably including HIPAA—apply to telemedicine and are of particular concern given the considerable amount of communication of protected health information with telemedicine.
HIPAA security rules essentially require making sure health information cannot be hacked or intercepted. Audio-only telemedicine by landline (not cell) is acceptable under the security rules, but almost all other remote communication requires secure communications.28
Clinicians also need to adhere to the more usual HIPAA privacy rules when practicingtelehealth. State laws protecting patient privacy vary and may be more stringent than HIPAA, so clinicians also must know the requirements in any state where they practice—whether in office or telemedicine.29
Making sure telemedicine practices are consistent with these security and privacy rules often requires particular technical expertise that is outside the realm of most practicing clinicians. However, without modification, the pre-telemedicine technology of many medical offices likely is insufficient for the full range of telemedicine services.30
Reimbursement and fraud
Before COVID-19, Medicare and Medicaid reimbursement for telemedicine was limited. Government decisions to substantially broaden those reimbursement rules (at least temporarily) provided a substantial boost to telemedicine early in the pandemic.23 Federal regulations and statutes also expanded telemedicine reimbursement for various services. Some will end shortly after the health emergency, and others will be permanent. Parts of that will not be sorted out for several years, so it will likely be a changing landscape for reimbursement.
Continue to: Rules that are evolving...
Rules that are evolving
Informed consent
The ethical and legal obligations to obtain informed consent are present in telemedicineas well as in-person care, with the same basic requirements regarding risks, benefits, alternative care, etc.32 However, with telemedicine, information related to remote care should be included and is outlined in TABLE 1.
Certain states may have somewhat unique informed consent requirements—especially for reproductive care, including abortion.34 Therefore, it is important for clinicians to ensure their consent process and forms comply with any legal jurisdiction in which a patient is located.
Medical malpractice
The basics of medical malpractice (or negligence) are the same in telemedicine as in in-person care: duty, breach of duty, and injury caused by the breach. That is, there may be liability when a medical professional breaches the duty of care, causing the patient’s injury. The physician’s duty is defined by the quality of care that the profession (specialty) accepts as reasonably good. This is defined by the opinions of physicians within the specialty and formal statements from professional organizations, including ACOG.3
Maintaining the standard of care and quality. The use of telemedicine is not an excuse to lower the quality of health care. There are some circumstances for which it is medically better to have an in-person visit. In these instances, the provider should recommend the appropriate care, even if telemedicine would be more convenient for the provider and staff.35
If the patient insists and telemedicine might result in less than optimal care, the reasons for using a remote visit should be clearly documented contemporaneously with the decision. Furthermore, when the limitations of being unable to physically examine the patient result in less information than is needed for the patient’s care, the provider must find alternatives to make up for the information gap.11,36 It also may be necessary to inform patients about how to maximize telemedicine care.37 At the beginning of telemedicine care the provider should include information about the nature and limits of telehealth, and the patient’s responsibilities. (See TABLE 1) Throughout treatment of the patient, that information should be updated by the provider. That, of course, is particularly important for patients who have not previously used telemedice services.
Malpractice rules vary by state. Many states have special rules regarding malpractice cases. These differences in malpractice standards and regulations “can be problematic for physicians who use telemedicine services to provide care outside the state in which they practice.”38 Caps on noneconomic damages are an example. Those state rules would apply to telemedicine in the patient’s state.
Malpractice insurance
Malpractice insurance now commonly includes telemedicine legally practiced within the physician’s home state. Practitioners who treat patients in foreign states should carefully examine their malpractice insurance policies to confirm that the coverage extends to practice in those states.39 Malpractice carriers may require notification by a covered physician who routinely provides services to patients in another state.3
Keep in mind, malpractice insurance generally does not cover the practice of medicine that is illegal. Practicing telemedicine in a foreign state, where the physician or other provider does not have a license and where that state does not otherwise permit the practice, is illegal. Most likely, the physician’s malpractice insurance will not cover claims that arise from this illegal practice in a foreign state or provide defense for malpractice claims, including frivolous lawsuits. Thus, the physician will pay out of pocket for the costs of a defense attorney.
Telemedicine treatment of minors
Children and adolescents present special legal issues for ObGyn care, which may become more complicated with telemedicine. Historically, parents are responsible for minors (those aged <18 years): they consent to medical treatment, are responsible for paying for it, and have the right to receive information about treatment.
Over the years, though, many states have made exceptions to these principles, especially with regard to contraception and treatment of sexually transmitted diseases.40 For abortion, in particular, there is considerable variation among the states in parental consent and notification.41 The Supreme Court’s decision in Dobbs v Jackson Women’s Health42 may (depending on the state) be followed with more stringent limitations on adolescent consent to abortions, including medical abortions.43
Use of telehealth does not change any obligations regarding adolescent consent or parental notification. Because those differ considerably among states, it is important for all practitioners to know their states’ requirements and keep reasonably complete records demonstrating their compliance with state law.
Abortion
The most heated current controversy about telemedicine involves abortion—specifically medical abortion, which is the combination of mifepristone and misoprostol.44,45 The FDA approved the combination in 2000. Almost immediately, many states required in-person visits with a certified clinician to receive a prescription for mifepristone and misoprostol, and eventually, the FDA adopted similar requirements.46 However, during the pandemic from 2021 to 2022, the FDA permitted telemedicine prescriptions. Several states still require in-person physician visits, although the constitutionality of those requirements has not been established.47
With the Supreme Court’s decision in Dobbs v Jackson Women’s Health in 2022,42 disagreements have ensued about the degree to which states may regulate the prescription of FDA-approved medical abortion drugs. Thorny constitutional issues exist in the plans of both abortion opponents and proponents in the battle over medical abortion in antiabortion states. It may be that federal drug law preempts state laws limiting access to FDA-approved drugs. On the other hand, it may be that states can make it a crime within the state to possess or provide abortion-inducing drugs. Courts will probably take years to resolve the many tangled legal questions.48
Thus, while the pandemic telemedicine rules may have advanced access to abortion,34 there may be some pending downsides.49 States that prohibit abortion will likely include prohibitions on medical abortions. In addition, they may prohibit anyone in the state (including pharmacies) from selling, possessing, or obtaining any drug used for causing or inducing an abortion.50 If, for constitutional reasons, they cannot press criminal charges or undertake licensing discipline for prescribing abortion, some states will likely withdraw from telehealth licensing compacts to avoid out-of-state prescriptions. This area of telemedicine has considerable uncertainty.
Continue to: CASE Conclusion...
CASE Conclusion
Patient concerns come to the fore
By 2023, Dr. TM started receiving bad news. Patient D called complaining that after following the advice on the website, she suffered a severe reaction and had to be rushed to an emergency department. Patient E (who had only 1 in-office visit early in her pregnancy) notified the office that she developed very high blood pressure that resulted in severe placental abruption, requiring emergency care and resulting in the loss of the fetus. Patient F complained that someone hacked the TikTok direct message communication with Dr. TM and tried to “blackmail” or harass her.
Discussion. Patients D, E, and F represent potential problems of telemedicine practice. Patient D was injured because she relied on her doctor’s website (to which Dr. TM directed patients). It contained an error that caused an injury. A doctor-patient relationship existed, and bad medical advice likely caused the injury. Physicians providing advice online must ensure the advice is correct and kept current.
Patient E demonstrates the importance of monitoring patients remotely (blood pressure transmitted to the office) or with periodic in-office visits. It is not clear whether she was a no-show for office visits (and whether the office followed up on any missed appointments) or if such visits were never scheduled. Liability for failure to monitor adequately is a possibility.
Patient F’s seemingly minor complaint could be a potential problem. Dr. TM used an insecure mode of communication. Although some HIPAA security regulations were modified or suspended during the pandemic, using such an unsecure platform is problematic, especially if temporary HIPAA rules expired. The outcome of the complaint is in doubt.
(See TABLE 2 for additional comments on patients D, E, and F.)
Out-of-state practice
Dr. TM treated 3 out-of-state residents (D, E, and F) via telemedicine. Recently Dr. TM received a complaint from the State Medical Licensure Board for practicing medicine without a license (Patient D), followed by similar charges from Patient E’s and Patient F’s state licensing boards. He has received a licensing inquiry from his home state board about those claims of illegal practice in other states and incompetent treatment.
Patient D’s pregnancy did not go well. The 1 in-person visit did not occur and she has filed a malpractice suit against Dr. TM. Patient E is threatening a malpractice case because the STI was not appropriately diagnosed and had advanced before another physician treated it.
In addition, a private citizen in Patient F’s state has filed suit against Dr. TM for abetting an illegal abortion (for Patient F).
Discussion. Patients D, E, and F illustrate the risk of even incidental out-of-state practice. The medical board inquiries arose from anonymous tips to all 4 states reporting Dr. TM was “practicing medicine without a license.” Patient E’s home state did have a licensing compact with the adjoining state (ie, Dr. TM’s home state). However, it required physicians to register and file an annual report, which Dr. TM had not done. The other 2 states did not have compacts with Dr. TM’s home state. Thus, he was illegally practicing medicine and would be subject to penalties. His home state also might impose license discipline based on his illegal practice in other states.
Continue to: What’s the verdict?...
What’s the verdict?
Dr. TM’s malpractice carrier is refusing to defend the claims of medical malpractice threatened by Patients D, E, and F. The company first notes that the terms of the malpractice policy specifically exclude the illegal practice of medicine. Furthermore, when a physician legally practices in another state, the policy requires a written notice to the insurance carrier of such practice. Dr. TM will likely have to engage and pay for a malpractice attorney for these cases. Because the claims are filed in 3 different states, more than a home-state attorney will likely be involved in the defense of these cases. Dr. TM will need to pay the attorneys and any damages from a settlement or trial.
Malpractice claims. Patient D claims that the doctor essentially abandoned her by never reaching out to her or arranging an in-person visit. Dr. TM claims the patient was responsible for scheduling the in-person visit. Patient E claims it was malpractice not to determine the specific nature of the STI and to do follow-up testing to determine that it was cured. All patients claim there was no genuine informed consent to the telemedicine. An attorney has warned Dr. TM that it is “not going to look good to the jury” that he was practicing without a license in the state and suggests he settle the cases quickly by paying damages.
Abortion-related claims. Patient F presents a different set of problems. Dr. TM’s home state is “proabortion.” Patient F’s home state is strongly “antiabortion,” making it a felony to participate in, assist, or facilitate an abortion (including medical abortion). Criminal charges have been filed against Dr. TM for the illegal practice of medicine, for aiding and facilitating an abortion, and for failure to notify a parent that a minor is seeking an abortion. For now, Dr. TM’s state is refusing to extradite on the abortion charge. Still, the patient’s state insists that it do so on the illegal practice of medicine charges and new charges of insurance fraud and failure to report suspected sexual abuse of a child. (Under the patient’s state law, anyone having sex with Patient F would have engaged in sexual abuse or “statutory rape,” so the state insists that the fact she was pregnant proves someone had sex with her.)
Patient F’s state also has a statute that allows private citizens to file civil claims against anyone procuring or assisting with an abortion (a successful private citizen can receive a minimum of $10,000 from the defendant). Several citizens from the patient’s state have already filed claims against Dr. TM in his state courts. Only one of them, probably the first to file, could succeed. Courts in the state have issued subpoenas and ordered Dr. TM to appear and reply to the civil suits. If he does not respond, there will be a default judgment.
Dr. TM’s attorney tells him that these lawsuits will not settle and will take a long time to defend and resolve. That will be expensive.
Billing and fraud. Dr. TM’s office recently received a series of notices from private health insurers stating they are investigating previously made payments as being fraudulent (unlicensed). They will not pay any new claims pending the investigation. On behalf of Medicare-Medicaid and other federal programs, the US Attorney’s office has notified Dr. TM that it has opened an investigation into fraudulent federal payments. F’s home state also is filing a (criminal) insurance fraud case, although the basis for it is unclear. (Dr. TM’s attorney believes it might be to increase pressure on the physician’s state to extradite Dr. TM for Patient F’s case.)
In addition, a disgruntled former employee of Dr. TM has filed a federal FCA case against him for filing inflated claims with various federally funded programs. The employee also made whistleblower calls to insurance companies and some state-funded medical programs. A forensic accounting investigation by Dr. TM’s accountant confirmed a pattern of very sloppy records and recurring billing for televisits that did not occur. Dr. TM believes that this was the act of one of the temporary assistants he hired in a pinch, who did not understand the system and just guessed when filing some insurance claims.
During the investigation, the federal and state attorneys are looking into a possible violation of state and federal Anti-Kickback Statutes. This is based on the original offer of a $100 credit for referrals to Dr. TM’s telemedicine practice.
The attorneys are concerned that other legal problems may present themselves. They are thoroughly reviewing Dr. TM’s practice and making several critical but somewhat modest changes to his practice. They also have insisted that Dr. TM have appropriate staff to handle the details of the practice and billing.
Conclusions
Telemedicine presents notable legal challenges to medical practice. As the pandemic status ends, ObGyn physicians practicing telemedicine need to be aware of the rules and how they are changing. For those physicians who want to continue or start a telemedicine practice, securing legal and technical support to ensure your operations are inline with the legal requirements can minimize any risk of legal troubles in the future. ●
A physician in State A, where abortion is legal, has a telemedicine patient in State B, where it is illegal to assist, provide, or procure an abortion. If the physician prescribes a medical abortion, he would violate the law of State B by using telemedicine to help the patient (located in State B) obtain an abortion. This could result in criminal charges against the prescribing physician.
- Board on Health Care Services; Institute of Medicine. The Role of Telehealth in an Evolving Health Care Environment: Workshop Summary. National Academies Press: 2012. https://www.ncbi.nlm.nih.gov/books/NBK207145/. Accessed March 30, 2023.
- Bruhn HK. Telemedicine: dos and don’ts to mitigate liability risk. J APPOS. 2020;24:195-196. doi:10.1016/j.jaapos. 2020.07.002
- Implementing telehealth in practice: ACOG Committee Opinion Summary, number 798. Obstet Gynecol. 2020; 2135:493-494. doi:10.1097/AOG.0000000000003672
- Bestsennyy O, Gilbert G, Harris A, et al. Telehealth: a quarter-trillion-dollar post-COVID-19 reality? McKinsey & Company. July 9, 2021. Accessed March 2, 2023. https://www.mckinsey.com/industries/healthcare/our-insights /telehealth-a-quarter-trillion-dollar-post-covid-19-reality
- Stanley AY, Wallace JB. Telehealth to improve perinatal care access. MCN Am J Matern Child Nurs. 2022;47:281-287. doi: 10.1097/NMC.0000000000000841
- Warshaw R. Health disparities affect millions in rural US communities. Association of American Medical Colleges. Published October 31, 2017. Accessed March 31, 2023. https://www.aamc.org/news-insights/health-disparities -affect-millions-rural-us-communities
- Almuslin H, AlDossary S. Models of incorporating telehealth into obstetric care during the COVID-19 pandemic, its benefits and barriers: a scoping review. Telemed J E Health. 2022;28:24-38. doi:10.1089/tmj.2020.0553
- Gold AE, Gilbert A, McMichael BJ. Socially distant health care. Tul L Rev. 2021;96:423-468. https://scholarship .law.ua.edu/cgi/viewcontent.cgi?article=1713&context =fac_articles. Accessed March 4, 2023.
- Zolotor AJ, Carlough MC. Update on prenatal care. Am Fam Physician. 2014;89:199-208.
- Odibo IN, Wendel PJ, Magann EF. Telemedicine in obstetrics. Clin Obstet Gynecol. 2013;56:422-433. doi:10.1097/ GRF.0b013e318290fef0
- Shmerling A, Hoss M, Malam N, et al. Prenatal care via telehealth. Prim Care. 2022;49:609-619. doi:10.1016/j. pop.2022.05.002
- Madden N, Emeruwa UN, Friedman AM, et al. Telehealth uptake into prenatal care and provider attitudes during COVID-19 pandemic in New York City: a quantitative and qualitative analysis. Am J Perinatol. 2020;37:1005-1014. doi:10.1055/s-0040-1712939
- Dosaj A, Thiyagarajan D, Ter Haar C, et al. Rapid implementation of telehealth services during the COVID-19 pandemic. Telemed J E Health. 2020;27:116-120. doi:10.1089/ tmj.2020.0219
- Lurie N, Carr B. The role of telehealth in the medical response to disasters. JAMA Intern Med. 2018;187:745-746. doi: 10.1001/jamainternmed.2018.1314
- Tobah YSB, LeBlanc A, Branda E, et al. Randomized comparison of a reduced-visit prenatal care model enhanced with remote monitoring. Am J Obstet Gynecol. 2019;221:638-e1-638.e8. doi:10.1016/j.ajog.2019.06.034
- Vivanti AJ, Deruelle P, Piccone O, et al. Follow-up for pregnant women during the COVID-19 pandemic: French national authority for health recommendations. J Gynecol Obstet Hum Reprod. 2020;49:101804. doi:10.1016/j. jogoh.2020.101804
- Ellimoottil C. Takeaways from 2 key studies on interstate telehealth use among Medicare fee-for-service beneficiaries. JAMA Health Forum. 2022;3:e223020-E223020. doi:10.1001/ jamahealthforum.2022.3020
- Harris J, Hartnett T, Hoagland GW, et al. What eliminating barriers to interstate telehealth taught us during the pandemic. Bipartisan Policy Center. Published November 2021. Accessed March 9, 2023. https://bipartisanpolicy .org/download/?file=/wp-content/uploads/2021/11/BPC -Health-Licensure-Brief_WEB.pdf.
- Center for Connected Health Policy. Cross-state licensing. Accessed February 21, 2023. https://www.cchpca.org/topic /cross-state-licensing-professional-requirements.
- US Department of Health & Human Services. Telehealth. Getting started with licensure. Published February 3, 2023. Accessed February 27, 2023. https://telehealth.hhs.gov /licensure/getting-started-licensure/
- US Department of Health & Human Services. Telehealth. Licensure. Accessed February 27, 2023. https://telehealth .hhs.gov/licensure
- US Department of Health & Human Services. National Practitioner Data Bank (NPDB) code lists. Published December 2022. Accessed March 9, 2023. https://www.npdb .hrsa.gov/software/CodeLists.pdf
- American College of Obstetricians and Gynecologists. COVID-19 FAQs for obstetrician-gynecologists, telehealth. 2020. Accessed March 5, 2023. https://www.acog.org /clinical-information/physician-faqs/covid-19-faqs-for -ob-gyns-telehealth
- Gorman RK. Prescribing medication through the practice of telemedicine: a comparative analysis of federal and state online prescribing policies, and policy considerations for the future. S Cal Interdisc Law J. 2020;30:739-769. https://gould .usc.edu/why/students/orgs/ilj/assets/docs/30-3-Gorman. pdf. Accessed March 10, 2023.
- Farringer DR. A telehealth explosion: using lessons from the pandemic to shape the future of telehealth regulation. Tex A&M Law Rev. 2021;9:1-47. https://scholarship.law.tamu. edu/cgi/viewcontent.cgi?article=1232&context=lawreview. Accessed February 28, 2023.
- Sterba KR, Johnson EE, Douglas E, et al. Implementation of a women’s reproductive behavioral health telemedicine program: a qualitative study of barriers and facilitators in obstetric and pediatric clinics. BMC Pregnancy Childbirth. 2023;23:167, 1-10. doi:10.1186/s12884-023-05463-2.
- US Department of Justice. COVID-19 FAQ (telemedicine). https://www.deadiversion.usdoj.gov/faq/coronavirus_faq .htm#TELE_FAQ2. Accessed March 13, 2023.
- US Department of Health & Human Services. Guidance on how the HIPAA rules permit covered health care providers and health plans to use remote communication technologies for audio-only telehealth. Published June 13, 2022. Accessed February 22, 2023. https://www.hhs.gov/hipaa/for-professionals/privacy /guidance/hipaa-audio-telehealth/index.html.
- Gray JME. HIPAA, telehealth, and the treatment of mental illness in a post-COVID world. Okla City Uni Law Rev. 2021;46:1-26. https://law.okcu.edu/wp-content /uploads/2022/04/J-Michael-E-Gray-HIPAA-Telehealth -and-Treament.pdf. Accessed March 9, 2023.
- Kurzweil C. Telemental health care and data privacy: current HIPAA privacy pitfalls and a proposed solution. Ann Health L Adv Dir. 2022;31:165.
- US Department of Health & Human Services and US Department of Justice. Health care fraud and abuse control program FY 2020: annual report. July 2021. Accessed March 9, 2023. https://oig.hhs.gov/publications/docs/hcfac /FY2020-hcfac.pdf
- Copeland KB. Telemedicine scams. Iowa Law Rev. 2022: 108:69-126. https://ilr.law.uiowa.edu/sites/ilr.law.uiowa.edu /files/2023-01/A2_Copeland.pdf. Accessed March 10, 2023.
- Solimini R, Busardò FP, Gibelli F, et al. Ethical and legal challenges of telemedicine in the era of the COVID-19 pandemic. Medicina (Kaunas). 2021;57:13141324. doi:10.3390/medicina57121314
- Reed A. COVID: a silver linings playbook. mobilizing pandemic era success stories to advance reproductive justice. Berkeley J Gender Law Justice. 2022;37:221-266. https://lawcat.berkeley.edu/record/1237158/files/16%20 Reed_final.pdf. Accessed March 11, 2023.
- Women’s Preventive Services Initiative and The American College of Obstetricians and Gynecologists. FAQ for telehealth services. Accessed March 2, 2023. https://www .womenspreventivehealth.org/wp-content/uploads/WPSI -Telehealth-FAQ.pdf
- Warren L, Chen KT. Telehealth apps in ObGyn practice. OBG Manag. 2022;34:46-47. doi:10.12788/obgm.0178
- American College of Obstetricians and Gynecologists. 10 telehealth tips for an Ob-Gyn visit. 2020. Accessed March 2, 2023. https://www.acog.org/womens-health /infographics/10-telehealth-tips-for-an-ob-gyn-visit
- Wolf TD. Telemedicine and malpractice: creating uniformity at the national level. Wm Mary Law Rev. 2019;61:15051536. https://scholarship.law.wm.edu/cgi/viewcontent.cgi ?article=3862&context=wmlr. Accessed March 11, 2023.
- Cahan E. Lawsuits, reimbursement, and liability insurance— facing the realities of a post-Roe era. JAMA. 2022;328:515517. doi:10.1001/jama.2022.9193
- Heinrich L, Hernandez AK, Laurie AR. Telehealth considerations for the adolescent patient. Prim Care. 2022;49:597-607. doi:10.1016/j.pop.2022.04.006
- Guttmacher Institute. An overview of consent to reproductive health services by young people. Published March 1, 2023. Accessed April 1, 2023. https://www.guttmacher.org /state-policy/explore/overview-minors-consent-law.
- Dobbs v. Jackson Women’s Health. No. 19–1392. June 24, 2022. Accessed April 1, 2023. https://www.supremecourt .gov/opinions/21pdf/19-1392_6j37.pdf
- Lindgren Y. Dobbs v. Jackson Women’s Health and the post-Roe landscape. J Am Acad Matrimonial Law. 2022;35:235283. https://www.aaml.org/wp-content/uploads/MAT110-1 .pdf. Accessed March 11, 2023.
- Mohiuddin H. The use of telemedicine during a pandemic to provide access to medication abortion. Hous J Health Law Policy. 2021;21:483-525. https://houstonhealthlaw. scholasticahq.com/article/34611.pdf. Accessed March 10, 2023.
- Rebouché R. The public health turn in reproductive rights. Wash & Lee Law Rev. 2021;78:1355-1432. https:// scholarlycommons.law.wlu.edu/cgi/viewcontent .cgi?article=4743&context=wlulr. Accessed March 10, 2023.
- Fliegel R. Access to medication abortion: now more important than ever. Am J Law Med. 2022;48:286-304. doi:10.1017/amj.2022.24
- Guttmacher Institute. Medication abortion. March 1, 2023. Accessed April 1, 2023 https://www.guttmacher.org /state-policy/explore/medication-abortion#:~:text=In%20 January%202023%2C%20the%20FDA,order%20to%20 dispense%20the%20pills
- Cohen DS, Donley G, Rebouché R. The new abortion battleground. Columbia Law Rev. 2023;123:1-100. https:// columbialawreview.org/content/the-new-abortion -battleground/. Accessed March 1, 2023.
- Hunt SA. Call me, beep me, if you want to reach me: utilizing telemedicine to expand abortion access. Vanderbilt Law Rev. 2023;76:323-359. Accessed March 10, 2023. https:// vanderbiltlawreview.org/lawreview/wp-content/uploads /sites/278/2023/01/Call-Me-Beep-Me-If-You-Want-toReach-Me-Utilizing-Telemedicine-to-Expand-AbortionAccess.pdf
- Gleckel JA, Wulkan SL. Abortion and telemedicine: looking beyond COVID-19 and the shadow docket. UC Davis Law Rev Online. 2020;54:105-121. https://lawreview.law.ucdavis. edu/online/54/files/54-online-Gleckel_Wulkan.pdf. Accessed April 1, 2023.
- Board on Health Care Services; Institute of Medicine. The Role of Telehealth in an Evolving Health Care Environment: Workshop Summary. National Academies Press: 2012. https://www.ncbi.nlm.nih.gov/books/NBK207145/. Accessed March 30, 2023.
- Bruhn HK. Telemedicine: dos and don’ts to mitigate liability risk. J APPOS. 2020;24:195-196. doi:10.1016/j.jaapos. 2020.07.002
- Implementing telehealth in practice: ACOG Committee Opinion Summary, number 798. Obstet Gynecol. 2020; 2135:493-494. doi:10.1097/AOG.0000000000003672
- Bestsennyy O, Gilbert G, Harris A, et al. Telehealth: a quarter-trillion-dollar post-COVID-19 reality? McKinsey & Company. July 9, 2021. Accessed March 2, 2023. https://www.mckinsey.com/industries/healthcare/our-insights /telehealth-a-quarter-trillion-dollar-post-covid-19-reality
- Stanley AY, Wallace JB. Telehealth to improve perinatal care access. MCN Am J Matern Child Nurs. 2022;47:281-287. doi: 10.1097/NMC.0000000000000841
- Warshaw R. Health disparities affect millions in rural US communities. Association of American Medical Colleges. Published October 31, 2017. Accessed March 31, 2023. https://www.aamc.org/news-insights/health-disparities -affect-millions-rural-us-communities
- Almuslin H, AlDossary S. Models of incorporating telehealth into obstetric care during the COVID-19 pandemic, its benefits and barriers: a scoping review. Telemed J E Health. 2022;28:24-38. doi:10.1089/tmj.2020.0553
- Gold AE, Gilbert A, McMichael BJ. Socially distant health care. Tul L Rev. 2021;96:423-468. https://scholarship .law.ua.edu/cgi/viewcontent.cgi?article=1713&context =fac_articles. Accessed March 4, 2023.
- Zolotor AJ, Carlough MC. Update on prenatal care. Am Fam Physician. 2014;89:199-208.
- Odibo IN, Wendel PJ, Magann EF. Telemedicine in obstetrics. Clin Obstet Gynecol. 2013;56:422-433. doi:10.1097/ GRF.0b013e318290fef0
- Shmerling A, Hoss M, Malam N, et al. Prenatal care via telehealth. Prim Care. 2022;49:609-619. doi:10.1016/j. pop.2022.05.002
- Madden N, Emeruwa UN, Friedman AM, et al. Telehealth uptake into prenatal care and provider attitudes during COVID-19 pandemic in New York City: a quantitative and qualitative analysis. Am J Perinatol. 2020;37:1005-1014. doi:10.1055/s-0040-1712939
- Dosaj A, Thiyagarajan D, Ter Haar C, et al. Rapid implementation of telehealth services during the COVID-19 pandemic. Telemed J E Health. 2020;27:116-120. doi:10.1089/ tmj.2020.0219
- Lurie N, Carr B. The role of telehealth in the medical response to disasters. JAMA Intern Med. 2018;187:745-746. doi: 10.1001/jamainternmed.2018.1314
- Tobah YSB, LeBlanc A, Branda E, et al. Randomized comparison of a reduced-visit prenatal care model enhanced with remote monitoring. Am J Obstet Gynecol. 2019;221:638-e1-638.e8. doi:10.1016/j.ajog.2019.06.034
- Vivanti AJ, Deruelle P, Piccone O, et al. Follow-up for pregnant women during the COVID-19 pandemic: French national authority for health recommendations. J Gynecol Obstet Hum Reprod. 2020;49:101804. doi:10.1016/j. jogoh.2020.101804
- Ellimoottil C. Takeaways from 2 key studies on interstate telehealth use among Medicare fee-for-service beneficiaries. JAMA Health Forum. 2022;3:e223020-E223020. doi:10.1001/ jamahealthforum.2022.3020
- Harris J, Hartnett T, Hoagland GW, et al. What eliminating barriers to interstate telehealth taught us during the pandemic. Bipartisan Policy Center. Published November 2021. Accessed March 9, 2023. https://bipartisanpolicy .org/download/?file=/wp-content/uploads/2021/11/BPC -Health-Licensure-Brief_WEB.pdf.
- Center for Connected Health Policy. Cross-state licensing. Accessed February 21, 2023. https://www.cchpca.org/topic /cross-state-licensing-professional-requirements.
- US Department of Health & Human Services. Telehealth. Getting started with licensure. Published February 3, 2023. Accessed February 27, 2023. https://telehealth.hhs.gov /licensure/getting-started-licensure/
- US Department of Health & Human Services. Telehealth. Licensure. Accessed February 27, 2023. https://telehealth .hhs.gov/licensure
- US Department of Health & Human Services. National Practitioner Data Bank (NPDB) code lists. Published December 2022. Accessed March 9, 2023. https://www.npdb .hrsa.gov/software/CodeLists.pdf
- American College of Obstetricians and Gynecologists. COVID-19 FAQs for obstetrician-gynecologists, telehealth. 2020. Accessed March 5, 2023. https://www.acog.org /clinical-information/physician-faqs/covid-19-faqs-for -ob-gyns-telehealth
- Gorman RK. Prescribing medication through the practice of telemedicine: a comparative analysis of federal and state online prescribing policies, and policy considerations for the future. S Cal Interdisc Law J. 2020;30:739-769. https://gould .usc.edu/why/students/orgs/ilj/assets/docs/30-3-Gorman. pdf. Accessed March 10, 2023.
- Farringer DR. A telehealth explosion: using lessons from the pandemic to shape the future of telehealth regulation. Tex A&M Law Rev. 2021;9:1-47. https://scholarship.law.tamu. edu/cgi/viewcontent.cgi?article=1232&context=lawreview. Accessed February 28, 2023.
- Sterba KR, Johnson EE, Douglas E, et al. Implementation of a women’s reproductive behavioral health telemedicine program: a qualitative study of barriers and facilitators in obstetric and pediatric clinics. BMC Pregnancy Childbirth. 2023;23:167, 1-10. doi:10.1186/s12884-023-05463-2.
- US Department of Justice. COVID-19 FAQ (telemedicine). https://www.deadiversion.usdoj.gov/faq/coronavirus_faq .htm#TELE_FAQ2. Accessed March 13, 2023.
- US Department of Health & Human Services. Guidance on how the HIPAA rules permit covered health care providers and health plans to use remote communication technologies for audio-only telehealth. Published June 13, 2022. Accessed February 22, 2023. https://www.hhs.gov/hipaa/for-professionals/privacy /guidance/hipaa-audio-telehealth/index.html.
- Gray JME. HIPAA, telehealth, and the treatment of mental illness in a post-COVID world. Okla City Uni Law Rev. 2021;46:1-26. https://law.okcu.edu/wp-content /uploads/2022/04/J-Michael-E-Gray-HIPAA-Telehealth -and-Treament.pdf. Accessed March 9, 2023.
- Kurzweil C. Telemental health care and data privacy: current HIPAA privacy pitfalls and a proposed solution. Ann Health L Adv Dir. 2022;31:165.
- US Department of Health & Human Services and US Department of Justice. Health care fraud and abuse control program FY 2020: annual report. July 2021. Accessed March 9, 2023. https://oig.hhs.gov/publications/docs/hcfac /FY2020-hcfac.pdf
- Copeland KB. Telemedicine scams. Iowa Law Rev. 2022: 108:69-126. https://ilr.law.uiowa.edu/sites/ilr.law.uiowa.edu /files/2023-01/A2_Copeland.pdf. Accessed March 10, 2023.
- Solimini R, Busardò FP, Gibelli F, et al. Ethical and legal challenges of telemedicine in the era of the COVID-19 pandemic. Medicina (Kaunas). 2021;57:13141324. doi:10.3390/medicina57121314
- Reed A. COVID: a silver linings playbook. mobilizing pandemic era success stories to advance reproductive justice. Berkeley J Gender Law Justice. 2022;37:221-266. https://lawcat.berkeley.edu/record/1237158/files/16%20 Reed_final.pdf. Accessed March 11, 2023.
- Women’s Preventive Services Initiative and The American College of Obstetricians and Gynecologists. FAQ for telehealth services. Accessed March 2, 2023. https://www .womenspreventivehealth.org/wp-content/uploads/WPSI -Telehealth-FAQ.pdf
- Warren L, Chen KT. Telehealth apps in ObGyn practice. OBG Manag. 2022;34:46-47. doi:10.12788/obgm.0178
- American College of Obstetricians and Gynecologists. 10 telehealth tips for an Ob-Gyn visit. 2020. Accessed March 2, 2023. https://www.acog.org/womens-health /infographics/10-telehealth-tips-for-an-ob-gyn-visit
- Wolf TD. Telemedicine and malpractice: creating uniformity at the national level. Wm Mary Law Rev. 2019;61:15051536. https://scholarship.law.wm.edu/cgi/viewcontent.cgi ?article=3862&context=wmlr. Accessed March 11, 2023.
- Cahan E. Lawsuits, reimbursement, and liability insurance— facing the realities of a post-Roe era. JAMA. 2022;328:515517. doi:10.1001/jama.2022.9193
- Heinrich L, Hernandez AK, Laurie AR. Telehealth considerations for the adolescent patient. Prim Care. 2022;49:597-607. doi:10.1016/j.pop.2022.04.006
- Guttmacher Institute. An overview of consent to reproductive health services by young people. Published March 1, 2023. Accessed April 1, 2023. https://www.guttmacher.org /state-policy/explore/overview-minors-consent-law.
- Dobbs v. Jackson Women’s Health. No. 19–1392. June 24, 2022. Accessed April 1, 2023. https://www.supremecourt .gov/opinions/21pdf/19-1392_6j37.pdf
- Lindgren Y. Dobbs v. Jackson Women’s Health and the post-Roe landscape. J Am Acad Matrimonial Law. 2022;35:235283. https://www.aaml.org/wp-content/uploads/MAT110-1 .pdf. Accessed March 11, 2023.
- Mohiuddin H. The use of telemedicine during a pandemic to provide access to medication abortion. Hous J Health Law Policy. 2021;21:483-525. https://houstonhealthlaw. scholasticahq.com/article/34611.pdf. Accessed March 10, 2023.
- Rebouché R. The public health turn in reproductive rights. Wash & Lee Law Rev. 2021;78:1355-1432. https:// scholarlycommons.law.wlu.edu/cgi/viewcontent .cgi?article=4743&context=wlulr. Accessed March 10, 2023.
- Fliegel R. Access to medication abortion: now more important than ever. Am J Law Med. 2022;48:286-304. doi:10.1017/amj.2022.24
- Guttmacher Institute. Medication abortion. March 1, 2023. Accessed April 1, 2023 https://www.guttmacher.org /state-policy/explore/medication-abortion#:~:text=In%20 January%202023%2C%20the%20FDA,order%20to%20 dispense%20the%20pills
- Cohen DS, Donley G, Rebouché R. The new abortion battleground. Columbia Law Rev. 2023;123:1-100. https:// columbialawreview.org/content/the-new-abortion -battleground/. Accessed March 1, 2023.
- Hunt SA. Call me, beep me, if you want to reach me: utilizing telemedicine to expand abortion access. Vanderbilt Law Rev. 2023;76:323-359. Accessed March 10, 2023. https:// vanderbiltlawreview.org/lawreview/wp-content/uploads /sites/278/2023/01/Call-Me-Beep-Me-If-You-Want-toReach-Me-Utilizing-Telemedicine-to-Expand-AbortionAccess.pdf
- Gleckel JA, Wulkan SL. Abortion and telemedicine: looking beyond COVID-19 and the shadow docket. UC Davis Law Rev Online. 2020;54:105-121. https://lawreview.law.ucdavis. edu/online/54/files/54-online-Gleckel_Wulkan.pdf. Accessed April 1, 2023.