U.S. Health Chief Kennedy Targets Vaccine Injury Compensation Program

Article Type
Changed
Tue, 08/05/2025 - 17:34

WASHINGTON (Reuters) - U.S. Health Secretary Robert F. Kennedy Jr. said on July 28 that he will work to “fix” the program that compensates victims of vaccine injuries, the National Vaccine Injury Compensation Program.

Kennedy, a long-time vaccine skeptic and former vaccine injury plaintiff lawyer, accused the program and its so-called “Vaccine Court” of corruption and inefficiency in a post on X. He has long been an outspoken critic of the program.

“I will not allow the VICP to continue to ignore its mandate and fail its mission of quickly and fairly compensating vaccine-injured individuals,” he wrote, adding he was working with Attorney General Pam Bondi. “Together, we will steer the Vaccine Court back to its original congressional intent.”

He said the structure disadvantaged claimants because the Department of Health & Human Services – which he now leads – is the defendant, as opposed to vaccine makers.

Changing the VICP would be the latest in a series of far-reaching actions by Kennedy to reshape U.S. regulation of vaccines, food and medicine.

In June, he fired all 17 members of the Centers for Disease Control and Prevention’s Advisory Committee on Immunization Practices, a panel of vaccine experts, replacing them with 7 handpicked members, including known vaccine skeptics.

One of them earned thousands of dollars as an expert witness in litigation against Merck’s, Gardasil vaccine, court records show. Kennedy himself played an instrumental role in organizing mass litigation over the vaccine.

He also is planning to remove all the members of another advisory panel that determines what preventive health measures insurers must cover, the Wall Street Journal reported on July 25. An HHS spokesperson said Kennedy had not yet made a decision regarding the 16-member U.S. Preventive Services Task Force.

Kennedy has for years sown doubt about the safety and efficacy of vaccines. He has a history of clashing with the medical establishment and spreading misinformation about vaccines, including promoting a debunked link between vaccines and autism despite scientific evidence to the contrary.

He has also said the measles vaccine contains cells from aborted fetuses and that the mumps vaccination does not work, comments he made as the U.S. battles one of its worst outbreaks of measles in 25 years.

Kennedy made millions over the years from advocating against vaccines through case referrals, book sales, and consulting fees paid by a nonprofit he founded, according to ethics disclosures.

(Reporting by Ahmed Aboulenein; Additional reporting by Ryan Patrick Jones in Toronto; Editing by Doina Chiacu and Nia Williams)

A version of this article appeared on Medscape.com.

Publications
Topics
Sections

WASHINGTON (Reuters) - U.S. Health Secretary Robert F. Kennedy Jr. said on July 28 that he will work to “fix” the program that compensates victims of vaccine injuries, the National Vaccine Injury Compensation Program.

Kennedy, a long-time vaccine skeptic and former vaccine injury plaintiff lawyer, accused the program and its so-called “Vaccine Court” of corruption and inefficiency in a post on X. He has long been an outspoken critic of the program.

“I will not allow the VICP to continue to ignore its mandate and fail its mission of quickly and fairly compensating vaccine-injured individuals,” he wrote, adding he was working with Attorney General Pam Bondi. “Together, we will steer the Vaccine Court back to its original congressional intent.”

He said the structure disadvantaged claimants because the Department of Health & Human Services – which he now leads – is the defendant, as opposed to vaccine makers.

Changing the VICP would be the latest in a series of far-reaching actions by Kennedy to reshape U.S. regulation of vaccines, food and medicine.

In June, he fired all 17 members of the Centers for Disease Control and Prevention’s Advisory Committee on Immunization Practices, a panel of vaccine experts, replacing them with 7 handpicked members, including known vaccine skeptics.

One of them earned thousands of dollars as an expert witness in litigation against Merck’s, Gardasil vaccine, court records show. Kennedy himself played an instrumental role in organizing mass litigation over the vaccine.

He also is planning to remove all the members of another advisory panel that determines what preventive health measures insurers must cover, the Wall Street Journal reported on July 25. An HHS spokesperson said Kennedy had not yet made a decision regarding the 16-member U.S. Preventive Services Task Force.

Kennedy has for years sown doubt about the safety and efficacy of vaccines. He has a history of clashing with the medical establishment and spreading misinformation about vaccines, including promoting a debunked link between vaccines and autism despite scientific evidence to the contrary.

He has also said the measles vaccine contains cells from aborted fetuses and that the mumps vaccination does not work, comments he made as the U.S. battles one of its worst outbreaks of measles in 25 years.

Kennedy made millions over the years from advocating against vaccines through case referrals, book sales, and consulting fees paid by a nonprofit he founded, according to ethics disclosures.

(Reporting by Ahmed Aboulenein; Additional reporting by Ryan Patrick Jones in Toronto; Editing by Doina Chiacu and Nia Williams)

A version of this article appeared on Medscape.com.

WASHINGTON (Reuters) - U.S. Health Secretary Robert F. Kennedy Jr. said on July 28 that he will work to “fix” the program that compensates victims of vaccine injuries, the National Vaccine Injury Compensation Program.

Kennedy, a long-time vaccine skeptic and former vaccine injury plaintiff lawyer, accused the program and its so-called “Vaccine Court” of corruption and inefficiency in a post on X. He has long been an outspoken critic of the program.

“I will not allow the VICP to continue to ignore its mandate and fail its mission of quickly and fairly compensating vaccine-injured individuals,” he wrote, adding he was working with Attorney General Pam Bondi. “Together, we will steer the Vaccine Court back to its original congressional intent.”

He said the structure disadvantaged claimants because the Department of Health & Human Services – which he now leads – is the defendant, as opposed to vaccine makers.

Changing the VICP would be the latest in a series of far-reaching actions by Kennedy to reshape U.S. regulation of vaccines, food and medicine.

In June, he fired all 17 members of the Centers for Disease Control and Prevention’s Advisory Committee on Immunization Practices, a panel of vaccine experts, replacing them with 7 handpicked members, including known vaccine skeptics.

One of them earned thousands of dollars as an expert witness in litigation against Merck’s, Gardasil vaccine, court records show. Kennedy himself played an instrumental role in organizing mass litigation over the vaccine.

He also is planning to remove all the members of another advisory panel that determines what preventive health measures insurers must cover, the Wall Street Journal reported on July 25. An HHS spokesperson said Kennedy had not yet made a decision regarding the 16-member U.S. Preventive Services Task Force.

Kennedy has for years sown doubt about the safety and efficacy of vaccines. He has a history of clashing with the medical establishment and spreading misinformation about vaccines, including promoting a debunked link between vaccines and autism despite scientific evidence to the contrary.

He has also said the measles vaccine contains cells from aborted fetuses and that the mumps vaccination does not work, comments he made as the U.S. battles one of its worst outbreaks of measles in 25 years.

Kennedy made millions over the years from advocating against vaccines through case referrals, book sales, and consulting fees paid by a nonprofit he founded, according to ethics disclosures.

(Reporting by Ahmed Aboulenein; Additional reporting by Ryan Patrick Jones in Toronto; Editing by Doina Chiacu and Nia Williams)

A version of this article appeared on Medscape.com.

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Mon, 08/04/2025 - 12:06
Un-Gate On Date
Mon, 08/04/2025 - 12:06
Use ProPublica
CFC Schedule Remove Status
Mon, 08/04/2025 - 12:06
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
survey writer start date
Mon, 08/04/2025 - 12:06

Rurality and Age May Shape Phone-Only Mental Health Care Access Among Veterans

Article Type
Changed
Thu, 06/26/2025 - 12:37

TOPLINE:

Patients living in rural areas and those aged ≥ 65 y had increased odds of receiving mental health care exclusively by phone.

METHODOLOGY:

  • Researchers explored factors linked to receiving phone-only mental health care among patients within the Department of Veterans Affairs.
  • They included data for 1,156,146 veteran patients with at least one mental health-specific outpatient encounter between October 2021 and September 2022 and at least one between October 2022 and September 2023.
  • Patients were categorized as those who received care through phone only (n = 49,125) and those who received care through other methods (n = 1,107,021. Care was received exclusively through video (6.39%), in-person (6.63%), or a combination of in-person, video, and/or phone (86.98%).
  • Demographic and clinical predictors, including rurality, age, sex, race, ethnicity, and the number of mental health diagnoses (< 3 vs ≥ 3), were evaluated.

TAKEAWAY:

  • The phone-only group had a mean of 6.27 phone visits, whereas those who received care through other methods had a mean of 4.79 phone visits.
  • Highly rural patients had 1.50 times higher odds of receiving phone-only mental health care than their urban counterparts (adjusted odds ratio [aOR], 1.50; P < .0001).
  • Patients aged 65 years or older were more than twice as likely to receive phone-only care than those younger than 30 years (aOR, ≥ 2.17; P < .0001).
  • Having fewer than three mental health diagnoses and more than 50% of mental health visits conducted by medical providers was associated with higher odds of receiving mental health care exclusively by phone (aORs, 2.03 and 1.87, respectively; P < .0001).

IN PRACTICE:

“The results of this work help to characterize the phone-only patient population and can serve to inform future implementation efforts to ensure that patients are receiving care via the modality that best meets their needs,” the authors wrote.

SOURCE:

This study was led by Samantha L. Connolly, PhD, at the VA Boston Healthcare System in Boston. It was published online in The Journal of Rural Health.

LIMITATIONS:

This study focused on a veteran population which may limit the generalizability of the findings to other groups. Additionally, its cross-sectional design restricted the ability to determine cause-and-effect relationships between factors and phone-only care.

DISCLOSURES:

This study was supported by the US Department of Veterans Affairs. The authors declared having no conflicts of interest.

This article was created using several editorial tools, including AI, as part of the process. Human editors reviewed this content before publication.

A version of this article first appeared on Medscape.com.

Publications
Topics
Sections

TOPLINE:

Patients living in rural areas and those aged ≥ 65 y had increased odds of receiving mental health care exclusively by phone.

METHODOLOGY:

  • Researchers explored factors linked to receiving phone-only mental health care among patients within the Department of Veterans Affairs.
  • They included data for 1,156,146 veteran patients with at least one mental health-specific outpatient encounter between October 2021 and September 2022 and at least one between October 2022 and September 2023.
  • Patients were categorized as those who received care through phone only (n = 49,125) and those who received care through other methods (n = 1,107,021. Care was received exclusively through video (6.39%), in-person (6.63%), or a combination of in-person, video, and/or phone (86.98%).
  • Demographic and clinical predictors, including rurality, age, sex, race, ethnicity, and the number of mental health diagnoses (< 3 vs ≥ 3), were evaluated.

TAKEAWAY:

  • The phone-only group had a mean of 6.27 phone visits, whereas those who received care through other methods had a mean of 4.79 phone visits.
  • Highly rural patients had 1.50 times higher odds of receiving phone-only mental health care than their urban counterparts (adjusted odds ratio [aOR], 1.50; P < .0001).
  • Patients aged 65 years or older were more than twice as likely to receive phone-only care than those younger than 30 years (aOR, ≥ 2.17; P < .0001).
  • Having fewer than three mental health diagnoses and more than 50% of mental health visits conducted by medical providers was associated with higher odds of receiving mental health care exclusively by phone (aORs, 2.03 and 1.87, respectively; P < .0001).

IN PRACTICE:

“The results of this work help to characterize the phone-only patient population and can serve to inform future implementation efforts to ensure that patients are receiving care via the modality that best meets their needs,” the authors wrote.

SOURCE:

This study was led by Samantha L. Connolly, PhD, at the VA Boston Healthcare System in Boston. It was published online in The Journal of Rural Health.

LIMITATIONS:

This study focused on a veteran population which may limit the generalizability of the findings to other groups. Additionally, its cross-sectional design restricted the ability to determine cause-and-effect relationships between factors and phone-only care.

DISCLOSURES:

This study was supported by the US Department of Veterans Affairs. The authors declared having no conflicts of interest.

This article was created using several editorial tools, including AI, as part of the process. Human editors reviewed this content before publication.

A version of this article first appeared on Medscape.com.

TOPLINE:

Patients living in rural areas and those aged ≥ 65 y had increased odds of receiving mental health care exclusively by phone.

METHODOLOGY:

  • Researchers explored factors linked to receiving phone-only mental health care among patients within the Department of Veterans Affairs.
  • They included data for 1,156,146 veteran patients with at least one mental health-specific outpatient encounter between October 2021 and September 2022 and at least one between October 2022 and September 2023.
  • Patients were categorized as those who received care through phone only (n = 49,125) and those who received care through other methods (n = 1,107,021. Care was received exclusively through video (6.39%), in-person (6.63%), or a combination of in-person, video, and/or phone (86.98%).
  • Demographic and clinical predictors, including rurality, age, sex, race, ethnicity, and the number of mental health diagnoses (< 3 vs ≥ 3), were evaluated.

TAKEAWAY:

  • The phone-only group had a mean of 6.27 phone visits, whereas those who received care through other methods had a mean of 4.79 phone visits.
  • Highly rural patients had 1.50 times higher odds of receiving phone-only mental health care than their urban counterparts (adjusted odds ratio [aOR], 1.50; P < .0001).
  • Patients aged 65 years or older were more than twice as likely to receive phone-only care than those younger than 30 years (aOR, ≥ 2.17; P < .0001).
  • Having fewer than three mental health diagnoses and more than 50% of mental health visits conducted by medical providers was associated with higher odds of receiving mental health care exclusively by phone (aORs, 2.03 and 1.87, respectively; P < .0001).

IN PRACTICE:

“The results of this work help to characterize the phone-only patient population and can serve to inform future implementation efforts to ensure that patients are receiving care via the modality that best meets their needs,” the authors wrote.

SOURCE:

This study was led by Samantha L. Connolly, PhD, at the VA Boston Healthcare System in Boston. It was published online in The Journal of Rural Health.

LIMITATIONS:

This study focused on a veteran population which may limit the generalizability of the findings to other groups. Additionally, its cross-sectional design restricted the ability to determine cause-and-effect relationships between factors and phone-only care.

DISCLOSURES:

This study was supported by the US Department of Veterans Affairs. The authors declared having no conflicts of interest.

This article was created using several editorial tools, including AI, as part of the process. Human editors reviewed this content before publication.

A version of this article first appeared on Medscape.com.

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Wed, 06/25/2025 - 11:01
Un-Gate On Date
Wed, 06/25/2025 - 11:01
Use ProPublica
CFC Schedule Remove Status
Wed, 06/25/2025 - 11:01
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
survey writer start date
Wed, 06/25/2025 - 11:01

Searching for the Optimal CRC Surveillance Test

Article Type
Changed
Mon, 12/09/2024 - 15:19

About a third of the US population are eligible for colorectal cancer screening but aren’t up to date on screening.

Many patients are reluctant to test for colon cancer for a variety of reasons, said Jeffrey K. Lee, MD, MPH, a research scientist at the Kaiser Permanente Northern California Division of Research and an attending gastroenterologist at Kaiser Permanente San Francisco Medical Center.

“As a gastroenterologist, I strongly believe we should emphasize the importance of colorectal cancer screening. And there’s many tests available, not just a colonoscopy, to help reduce your chances of developing colorectal cancer and even dying from colorectal cancer,” said Dr. Lee. 

Many patients prefer a test that’s more convenient, that doesn’t require them to take time out of their busy schedules. “We must educate our patients that there are some noninvasive screening options that are helpful, and to be able to share with them some of the benefits, but also some of the drawbacks compared to colonoscopy and allow them to have a choice,” he advised.

Kaiser Permanente Medical Center
Dr. Jeffrey K. Lee



Dr. Lee has devoted his research to colorectal cancer screening, as well as the causes and prevention of CRC. He is a recipient of the AGA Research Scholar Award, and has in turn supported other researchers by contributing to the AGA Research Foundation. In 2012, Dr. Lee received a grant from the Sylvia Allison Kaplan Clinical Research Fund to fund a study on long-term colorectal cancer risk in patients with normal colonoscopy results.

The findings, published in JAMA Internal Medicine, determined that 10 years after a negative colonoscopy, Kaiser Permanente members had a 46% lower risk of being diagnosed with CRC and were 88% less likely to die from disease compared with patients who didn’t undergo screening.

“Furthermore, the reduced risk of developing colorectal cancer, even dying from it, persisted for more than 12 years after the examination compared with an unscreened population,” said Dr. Lee. “I firmly believe our study really supports the ten-year screening interval after a normal colonoscopy, as currently recommended by our guidelines.”

In an interview, he discussed his research efforts to find the best detection regimens for CRC, and the mentors who guided his career path as a GI scientist. 
 

Q: Why did you choose GI?

During medical school I was fortunate to work in the lab of Dr. John M. Carethers at UC San Diego. He introduced me to GI and inspired me to choose GI as a career. His mentorship was invaluable because he not only solidified my interest in GI, but also inspired me to become a physician scientist, focusing on colorectal cancer prevention and control. His amazing mentorship drew me to this field. 

Q: One of your clinical focus areas is hereditary gastrointestinal cancer syndromes. How did you become interested in this area of GI medicine? 

My interest in hereditary GI cancer syndromes stemmed from my work as a medical student in Dr. Carethers’ lab. One of my research projects was looking at certain gene mutations among patients with hereditary GI cancer syndromes, specifically, familial hamartomatous polyposis syndrome. It was through these research projects and seeing how these genetic mutations impacted their risk of developing colorectal cancer, inspired me to care for patients with hereditary GI cancer syndromes. 

 

 

Q: Have you been doing any research on the reasons why more young people are getting colon cancer? 

We recently published work looking at the potential factors that may be driving the rising rates of early onset colorectal cancer. One hypothesis that’s been floating around is antibiotic exposure in early adulthood or childhood because of its effect on the microbiome. Using our large database at Kaiser Permanente Northern California, we did not find an association between oral antibiotic use during early adulthood and the risk of early-onset colorectal cancer.

You have the usual suspects like obesity and diabetes, but it’s not explaining all that risk. While familial colorectal cancer syndromes contribute to a small proportion of early-onset colorectal, these syndromes are not increasing across generations. I really do feel it’s something in the diet or how foods are processed and environmental factors that’s driving some of the risk of early onset colorectal cancer and this should be explored further. 
 

Q: In 2018, you issued a landmark study which found an association between a 10-year follow-up after negative colonoscopy and reduced risk of disease and mortality. Has there been any updates to these findings over the last 6 years? 

We recently saw a study in JAMA Oncology of a Swedish cohort that showed a negative colonoscopy result was associated with a reduced risk of developing and even dying from colorectal cancer 15 years from that examination, compared to the general population of Sweden. I think there’s some things that we need to be cautious about regarding that study. We have to think about the comparison group that they used and the lack of information regarding the indication of the colonoscopy and the quality of the examination. So, it remains uncertain whether future guidelines are going to stretch out that 10-year interval to 15 years.

Q: What other CRC studies are you working on now? 

We have several studies that we are working on right now. One is called the PREVENT CRC study, which is looking at whether a polygenic risk score can improve risk stratification following adenoma removal for colorectal cancer prevention and tailoring post-polypectomy surveillance. This is a large observational cohort study that we have teamed up with the Fred Hutchinson Cancer Center, Erasmus University, and Kaiser Permanente Northwest to answer this important question that may have implications for personalized medicine. 

Then there’s the COOP study, funded by the Patient-Centered Outcomes Research Institute. This is looking at the best surveillance test to use among older adults 65 years and older with a history of polyps. The trial is randomizing them to either getting a colonoscopy for surveillance or annual fecal immunochemical test (FIT) for surveillance. This is to see which test is best for detecting colorectal cancer among older adults with a history of polyps.  
 

Q: Do you think FIT tests could eventually replace colonoscopy, given that it’s less invasive? 

Although FIT and other stool-based tests are less invasive and have been shown to have high accuracy for detecting colorectal cancer, I personally do not think they are going to replace colonoscopy as the most popular screening modality in the United States. Colonoscopy remains the gold standard for detecting and removing precancerous polyps and has the highest accuracy for detecting colorectal cancer. 

 

 

Q: Besides Dr. Carethers, what teacher or mentor had the greatest impact on you? 

Clinically it’s been Dr. Jonathan Terdiman from UCSF, who taught me everything I know about clinical GI, and the art of colonoscopy. In addition, Douglas A. Corley, MD, PhD, the Permanente Medical Group’s chief research officer, has made the greatest impact on my research career. He’s really taught me how to rigorously design a research study to answer important clinically relevant questions, and has given me the skill set to write NIH grants. I would not be here without these mentors who are truly giants in the field of GI.

Q: When you’re not being a GI, how do you spend your free weekend afternoons? Are you still a “Cal Bears” fan at your alma mater, UC Berkeley? 

I spend a lot of time taking my kids to their activities on the weekends. I just took my son to a Cal Bears Game Day, which was hosted by ESPN at Berkeley.

Dr. Lee
Dr. Jeffrey K. Lee, a graduate of the University of California, Berkeley, is pictured here with his son at a 2024 Cal football game.

It was an incredible experience hearing sports analyst Pat McAfee lead all the Cal chants, seeing Nick Saban from the University of Alabama take off his red tie and replace it with a Cal Bears tie, and watching a Cal student win a hundred thousand dollars by kicking a football through the goal posts wearing checkered vans. 

Lightning Round

Texting or talking?

Text

Favorite breakfast?

Taiwanese breakfast



Place you most want to travel to?

Japan



Favorite junk food?

Trader Joe’s chili lime chips



Favorite season?

Springtime, baseball season



Favorite ice cream flavor?

Mint chocolate chip



How many cups of coffee do you drink per day?

2-3



Last movie you watched?

Oppenheimer 



Best place you ever went on vacation?

Hawaii



If you weren’t a gastroenterologist, what would you be?

Barber



Best Halloween costume you ever wore?

SpongeBob SquarePants



Favorite sport?

Tennis

What song do you have to sing along with when you hear it?

Any classic 80s song



Introvert or extrovert?

Introvert

Publications
Topics
Sections

About a third of the US population are eligible for colorectal cancer screening but aren’t up to date on screening.

Many patients are reluctant to test for colon cancer for a variety of reasons, said Jeffrey K. Lee, MD, MPH, a research scientist at the Kaiser Permanente Northern California Division of Research and an attending gastroenterologist at Kaiser Permanente San Francisco Medical Center.

“As a gastroenterologist, I strongly believe we should emphasize the importance of colorectal cancer screening. And there’s many tests available, not just a colonoscopy, to help reduce your chances of developing colorectal cancer and even dying from colorectal cancer,” said Dr. Lee. 

Many patients prefer a test that’s more convenient, that doesn’t require them to take time out of their busy schedules. “We must educate our patients that there are some noninvasive screening options that are helpful, and to be able to share with them some of the benefits, but also some of the drawbacks compared to colonoscopy and allow them to have a choice,” he advised.

Kaiser Permanente Medical Center
Dr. Jeffrey K. Lee



Dr. Lee has devoted his research to colorectal cancer screening, as well as the causes and prevention of CRC. He is a recipient of the AGA Research Scholar Award, and has in turn supported other researchers by contributing to the AGA Research Foundation. In 2012, Dr. Lee received a grant from the Sylvia Allison Kaplan Clinical Research Fund to fund a study on long-term colorectal cancer risk in patients with normal colonoscopy results.

The findings, published in JAMA Internal Medicine, determined that 10 years after a negative colonoscopy, Kaiser Permanente members had a 46% lower risk of being diagnosed with CRC and were 88% less likely to die from disease compared with patients who didn’t undergo screening.

“Furthermore, the reduced risk of developing colorectal cancer, even dying from it, persisted for more than 12 years after the examination compared with an unscreened population,” said Dr. Lee. “I firmly believe our study really supports the ten-year screening interval after a normal colonoscopy, as currently recommended by our guidelines.”

In an interview, he discussed his research efforts to find the best detection regimens for CRC, and the mentors who guided his career path as a GI scientist. 
 

Q: Why did you choose GI?

During medical school I was fortunate to work in the lab of Dr. John M. Carethers at UC San Diego. He introduced me to GI and inspired me to choose GI as a career. His mentorship was invaluable because he not only solidified my interest in GI, but also inspired me to become a physician scientist, focusing on colorectal cancer prevention and control. His amazing mentorship drew me to this field. 

Q: One of your clinical focus areas is hereditary gastrointestinal cancer syndromes. How did you become interested in this area of GI medicine? 

My interest in hereditary GI cancer syndromes stemmed from my work as a medical student in Dr. Carethers’ lab. One of my research projects was looking at certain gene mutations among patients with hereditary GI cancer syndromes, specifically, familial hamartomatous polyposis syndrome. It was through these research projects and seeing how these genetic mutations impacted their risk of developing colorectal cancer, inspired me to care for patients with hereditary GI cancer syndromes. 

 

 

Q: Have you been doing any research on the reasons why more young people are getting colon cancer? 

We recently published work looking at the potential factors that may be driving the rising rates of early onset colorectal cancer. One hypothesis that’s been floating around is antibiotic exposure in early adulthood or childhood because of its effect on the microbiome. Using our large database at Kaiser Permanente Northern California, we did not find an association between oral antibiotic use during early adulthood and the risk of early-onset colorectal cancer.

You have the usual suspects like obesity and diabetes, but it’s not explaining all that risk. While familial colorectal cancer syndromes contribute to a small proportion of early-onset colorectal, these syndromes are not increasing across generations. I really do feel it’s something in the diet or how foods are processed and environmental factors that’s driving some of the risk of early onset colorectal cancer and this should be explored further. 
 

Q: In 2018, you issued a landmark study which found an association between a 10-year follow-up after negative colonoscopy and reduced risk of disease and mortality. Has there been any updates to these findings over the last 6 years? 

We recently saw a study in JAMA Oncology of a Swedish cohort that showed a negative colonoscopy result was associated with a reduced risk of developing and even dying from colorectal cancer 15 years from that examination, compared to the general population of Sweden. I think there’s some things that we need to be cautious about regarding that study. We have to think about the comparison group that they used and the lack of information regarding the indication of the colonoscopy and the quality of the examination. So, it remains uncertain whether future guidelines are going to stretch out that 10-year interval to 15 years.

Q: What other CRC studies are you working on now? 

We have several studies that we are working on right now. One is called the PREVENT CRC study, which is looking at whether a polygenic risk score can improve risk stratification following adenoma removal for colorectal cancer prevention and tailoring post-polypectomy surveillance. This is a large observational cohort study that we have teamed up with the Fred Hutchinson Cancer Center, Erasmus University, and Kaiser Permanente Northwest to answer this important question that may have implications for personalized medicine. 

Then there’s the COOP study, funded by the Patient-Centered Outcomes Research Institute. This is looking at the best surveillance test to use among older adults 65 years and older with a history of polyps. The trial is randomizing them to either getting a colonoscopy for surveillance or annual fecal immunochemical test (FIT) for surveillance. This is to see which test is best for detecting colorectal cancer among older adults with a history of polyps.  
 

Q: Do you think FIT tests could eventually replace colonoscopy, given that it’s less invasive? 

Although FIT and other stool-based tests are less invasive and have been shown to have high accuracy for detecting colorectal cancer, I personally do not think they are going to replace colonoscopy as the most popular screening modality in the United States. Colonoscopy remains the gold standard for detecting and removing precancerous polyps and has the highest accuracy for detecting colorectal cancer. 

 

 

Q: Besides Dr. Carethers, what teacher or mentor had the greatest impact on you? 

Clinically it’s been Dr. Jonathan Terdiman from UCSF, who taught me everything I know about clinical GI, and the art of colonoscopy. In addition, Douglas A. Corley, MD, PhD, the Permanente Medical Group’s chief research officer, has made the greatest impact on my research career. He’s really taught me how to rigorously design a research study to answer important clinically relevant questions, and has given me the skill set to write NIH grants. I would not be here without these mentors who are truly giants in the field of GI.

Q: When you’re not being a GI, how do you spend your free weekend afternoons? Are you still a “Cal Bears” fan at your alma mater, UC Berkeley? 

I spend a lot of time taking my kids to their activities on the weekends. I just took my son to a Cal Bears Game Day, which was hosted by ESPN at Berkeley.

Dr. Lee
Dr. Jeffrey K. Lee, a graduate of the University of California, Berkeley, is pictured here with his son at a 2024 Cal football game.

It was an incredible experience hearing sports analyst Pat McAfee lead all the Cal chants, seeing Nick Saban from the University of Alabama take off his red tie and replace it with a Cal Bears tie, and watching a Cal student win a hundred thousand dollars by kicking a football through the goal posts wearing checkered vans. 

Lightning Round

Texting or talking?

Text

Favorite breakfast?

Taiwanese breakfast



Place you most want to travel to?

Japan



Favorite junk food?

Trader Joe’s chili lime chips



Favorite season?

Springtime, baseball season



Favorite ice cream flavor?

Mint chocolate chip



How many cups of coffee do you drink per day?

2-3



Last movie you watched?

Oppenheimer 



Best place you ever went on vacation?

Hawaii



If you weren’t a gastroenterologist, what would you be?

Barber



Best Halloween costume you ever wore?

SpongeBob SquarePants



Favorite sport?

Tennis

What song do you have to sing along with when you hear it?

Any classic 80s song



Introvert or extrovert?

Introvert

About a third of the US population are eligible for colorectal cancer screening but aren’t up to date on screening.

Many patients are reluctant to test for colon cancer for a variety of reasons, said Jeffrey K. Lee, MD, MPH, a research scientist at the Kaiser Permanente Northern California Division of Research and an attending gastroenterologist at Kaiser Permanente San Francisco Medical Center.

“As a gastroenterologist, I strongly believe we should emphasize the importance of colorectal cancer screening. And there’s many tests available, not just a colonoscopy, to help reduce your chances of developing colorectal cancer and even dying from colorectal cancer,” said Dr. Lee. 

Many patients prefer a test that’s more convenient, that doesn’t require them to take time out of their busy schedules. “We must educate our patients that there are some noninvasive screening options that are helpful, and to be able to share with them some of the benefits, but also some of the drawbacks compared to colonoscopy and allow them to have a choice,” he advised.

Kaiser Permanente Medical Center
Dr. Jeffrey K. Lee



Dr. Lee has devoted his research to colorectal cancer screening, as well as the causes and prevention of CRC. He is a recipient of the AGA Research Scholar Award, and has in turn supported other researchers by contributing to the AGA Research Foundation. In 2012, Dr. Lee received a grant from the Sylvia Allison Kaplan Clinical Research Fund to fund a study on long-term colorectal cancer risk in patients with normal colonoscopy results.

The findings, published in JAMA Internal Medicine, determined that 10 years after a negative colonoscopy, Kaiser Permanente members had a 46% lower risk of being diagnosed with CRC and were 88% less likely to die from disease compared with patients who didn’t undergo screening.

“Furthermore, the reduced risk of developing colorectal cancer, even dying from it, persisted for more than 12 years after the examination compared with an unscreened population,” said Dr. Lee. “I firmly believe our study really supports the ten-year screening interval after a normal colonoscopy, as currently recommended by our guidelines.”

In an interview, he discussed his research efforts to find the best detection regimens for CRC, and the mentors who guided his career path as a GI scientist. 
 

Q: Why did you choose GI?

During medical school I was fortunate to work in the lab of Dr. John M. Carethers at UC San Diego. He introduced me to GI and inspired me to choose GI as a career. His mentorship was invaluable because he not only solidified my interest in GI, but also inspired me to become a physician scientist, focusing on colorectal cancer prevention and control. His amazing mentorship drew me to this field. 

Q: One of your clinical focus areas is hereditary gastrointestinal cancer syndromes. How did you become interested in this area of GI medicine? 

My interest in hereditary GI cancer syndromes stemmed from my work as a medical student in Dr. Carethers’ lab. One of my research projects was looking at certain gene mutations among patients with hereditary GI cancer syndromes, specifically, familial hamartomatous polyposis syndrome. It was through these research projects and seeing how these genetic mutations impacted their risk of developing colorectal cancer, inspired me to care for patients with hereditary GI cancer syndromes. 

 

 

Q: Have you been doing any research on the reasons why more young people are getting colon cancer? 

We recently published work looking at the potential factors that may be driving the rising rates of early onset colorectal cancer. One hypothesis that’s been floating around is antibiotic exposure in early adulthood or childhood because of its effect on the microbiome. Using our large database at Kaiser Permanente Northern California, we did not find an association between oral antibiotic use during early adulthood and the risk of early-onset colorectal cancer.

You have the usual suspects like obesity and diabetes, but it’s not explaining all that risk. While familial colorectal cancer syndromes contribute to a small proportion of early-onset colorectal, these syndromes are not increasing across generations. I really do feel it’s something in the diet or how foods are processed and environmental factors that’s driving some of the risk of early onset colorectal cancer and this should be explored further. 
 

Q: In 2018, you issued a landmark study which found an association between a 10-year follow-up after negative colonoscopy and reduced risk of disease and mortality. Has there been any updates to these findings over the last 6 years? 

We recently saw a study in JAMA Oncology of a Swedish cohort that showed a negative colonoscopy result was associated with a reduced risk of developing and even dying from colorectal cancer 15 years from that examination, compared to the general population of Sweden. I think there’s some things that we need to be cautious about regarding that study. We have to think about the comparison group that they used and the lack of information regarding the indication of the colonoscopy and the quality of the examination. So, it remains uncertain whether future guidelines are going to stretch out that 10-year interval to 15 years.

Q: What other CRC studies are you working on now? 

We have several studies that we are working on right now. One is called the PREVENT CRC study, which is looking at whether a polygenic risk score can improve risk stratification following adenoma removal for colorectal cancer prevention and tailoring post-polypectomy surveillance. This is a large observational cohort study that we have teamed up with the Fred Hutchinson Cancer Center, Erasmus University, and Kaiser Permanente Northwest to answer this important question that may have implications for personalized medicine. 

Then there’s the COOP study, funded by the Patient-Centered Outcomes Research Institute. This is looking at the best surveillance test to use among older adults 65 years and older with a history of polyps. The trial is randomizing them to either getting a colonoscopy for surveillance or annual fecal immunochemical test (FIT) for surveillance. This is to see which test is best for detecting colorectal cancer among older adults with a history of polyps.  
 

Q: Do you think FIT tests could eventually replace colonoscopy, given that it’s less invasive? 

Although FIT and other stool-based tests are less invasive and have been shown to have high accuracy for detecting colorectal cancer, I personally do not think they are going to replace colonoscopy as the most popular screening modality in the United States. Colonoscopy remains the gold standard for detecting and removing precancerous polyps and has the highest accuracy for detecting colorectal cancer. 

 

 

Q: Besides Dr. Carethers, what teacher or mentor had the greatest impact on you? 

Clinically it’s been Dr. Jonathan Terdiman from UCSF, who taught me everything I know about clinical GI, and the art of colonoscopy. In addition, Douglas A. Corley, MD, PhD, the Permanente Medical Group’s chief research officer, has made the greatest impact on my research career. He’s really taught me how to rigorously design a research study to answer important clinically relevant questions, and has given me the skill set to write NIH grants. I would not be here without these mentors who are truly giants in the field of GI.

Q: When you’re not being a GI, how do you spend your free weekend afternoons? Are you still a “Cal Bears” fan at your alma mater, UC Berkeley? 

I spend a lot of time taking my kids to their activities on the weekends. I just took my son to a Cal Bears Game Day, which was hosted by ESPN at Berkeley.

Dr. Lee
Dr. Jeffrey K. Lee, a graduate of the University of California, Berkeley, is pictured here with his son at a 2024 Cal football game.

It was an incredible experience hearing sports analyst Pat McAfee lead all the Cal chants, seeing Nick Saban from the University of Alabama take off his red tie and replace it with a Cal Bears tie, and watching a Cal student win a hundred thousand dollars by kicking a football through the goal posts wearing checkered vans. 

Lightning Round

Texting or talking?

Text

Favorite breakfast?

Taiwanese breakfast



Place you most want to travel to?

Japan



Favorite junk food?

Trader Joe’s chili lime chips



Favorite season?

Springtime, baseball season



Favorite ice cream flavor?

Mint chocolate chip



How many cups of coffee do you drink per day?

2-3



Last movie you watched?

Oppenheimer 



Best place you ever went on vacation?

Hawaii



If you weren’t a gastroenterologist, what would you be?

Barber



Best Halloween costume you ever wore?

SpongeBob SquarePants



Favorite sport?

Tennis

What song do you have to sing along with when you hear it?

Any classic 80s song



Introvert or extrovert?

Introvert

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Mon, 12/02/2024 - 09:02
Un-Gate On Date
Mon, 12/02/2024 - 09:02
Use ProPublica
CFC Schedule Remove Status
Mon, 12/02/2024 - 09:02
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
survey writer start date
Mon, 12/02/2024 - 09:02

Hyperpigmented Macules Caused by Burrowing Bugs (Cydnidae) May Mimic More Serious Conditions

Article Type
Changed
Fri, 09/05/2025 - 12:20
Display Headline

Hyperpigmented Macules Caused by Burrowing Bugs (Cydnidae) May Mimic More Serious Conditions

Cydnidae is a family of small to medium-sized shield bugs with spiny legs that commonly are known as burrowing bugs (or burrower bugs). The family Cydnidae includes more than 100 genera and approximately 600 species worldwide.1 These insects are arthropods of the order Hemiptera (suborder: Heteroptera; superfamily: Pentatomoidae) and largely are concentrated in tropical and temperate regions. Approximately 145 species have been recorded in the Neotropical Region and have been included in the subfamilies Amnestinae, Cephalocteinae, and Sehirinae, in addition to Cydnidae.2 Burrowing bugs are ovoid in shape and 2 to 20 mm in length and morphologically are well adapted for burrowing. Their life span is 100 to 300 days. Being phytophagous, they burrow to feed on plants and roots. Adult burrowing bugs have wings and can fly. They have specialized glands located in either the abdomen (nymph) or thorax (adult) that secrete odorous chemicals for self-protection.3 The secretions contain hydrocarbonates that function as repellents and danger signals, can cause paralysis in prey, and act as a chemoattractant for mates.4-6 They also cause hyperpigmentation upon contact with the skin.

In this article, we present a series of cases from the same community to demonstrate the characteristic features of hyperpigmented macules caused by exposure to burrowing bugs. Dermatologists should be aware of this entity to prevent misdiagnosis and unnecessary investigations and treatment.

Case Series

A 36-year-old woman and 6 children (age range, 6-12 years) presented with a widespread, acute, brown-pigmented, macular eruption with lesions that increased in number over a 1-week period. All 7 patients resided in the same locality and were otherwise systemically healthy. Initially, the index case, a 7-year-old girl, was referred to our tertiary care center by a dermatologist with a provisional diagnosis of idiopathic macular eruptive pigmentation. The patient’s mother recalled noticing a tiny black insect on the child's scalp that left pigment on the skin when she crushed it between her fingers. The rest of the patients presented over the next few days: 3 of the children belonged to the same household as the index case, and there was history of all 6 children playing in the neighborhood park during late evening hours. The adult patient was the parent of one of the affected children. The lesions were associated with mild itching and tingling in 3 children but were asymptomatic in the other patients.

Clinical examination of the patients revealed multiple dark- to light-brown, discrete, irregularly shaped macules over the trunk, arms, and soles (eFigure 1). Dermoscopic examination of a pigmented macule showed an irregularly shaped, brownish, structureless area with accentuation of the pigment at skin creases and perieccrine pigmentation (eFigure 2). The pigmentation was unaffected by rubbing with alcohol or water. Clinicoepidemiologic parameters of the patients are summarized in the eTable.

CT116003094-Fig-1_ABC
eFIGURE 1. A-C, Discrete, irregularly shaped, brown to dark brown macules on the trunk, elbow, and soles.
CT116003094-Fig2_AB
eFIGURE 2. A and B, Dermoscopy showed irregularly shaped, homogenous, brownish, structureless areas with accentuation along skin creases and around eccrine A B openings (original magnification ×10 and ×10).

CT116003094-Table

One of the children’s parents conducted a geological examination of the ground in the neighborhood park during evening hours and found tiny burrowing bugs (eFigure 3). When crushed between the fingers, these insects left a similar brownish hyperpigmentation on the skin. The parents were counseled on the nature of the eruption, and the patients were kept under observation for 2 weeks. On follow-up after 5 days, the lesions showed markedly decreased intensity of hyperpigmentation, and no new lesions were observed in any of the 7 patients.

Baskaran-3
eFIGURE 3. A burrowing bug (Cydnidae) found at the neighborhood park visited by all patients.

Comment

Pentatomoidae insects generally are benign and harmless to humans. There have been isolated reports of erythematous plaques caused by Antiteuchus mixtus and Edessa maculate.7 Malhotra et al8 reported the first known series of cases with Cydnidae insect–induced hyperpigmented macules. The reported patients presented with asymptomatic, brown, hyperpigmented macules over exposed sites such as the feet, neck, and chest. All the cases occurred during the monsoon season in tropical and temperate regions of the world, and the patients were characteristically clustered in similar geographic areas. The causative insect was identified as Chilocoris assmuthi Breddin, 1904, belonging to the family Cydnidae. When it was crushed between the fingers, the skin became hyperpigmented, confirming the role of the secretions from the insect in the etiology.8

A second case was described by Sonthalia,9 who also described the dermoscopic features of hyperpigmented macules caused by burrowing bugs. The lesions showed a stuck-on, clustered appearance of ovoid and bizarre pigmented clods, globules, and granules.9 Although the lesions occur mainly over exposed sites, pigmented macules occurring over unusual sites such as the abdomen and back also have been reported in association with burrowing bugs.10 Characteristically, the lesions initially are faint and darken with time and usually fade within a week. They can be rubbed off with acetone but persist when washed with soap and water. The fleeting nature of the pigmentation also has led to the term transient pseudo-lentigines sign to describe hyperpigmentation caused by burrowing bugs.11

Soil and plants are burrowing bugs’ natural habitats, and the insects typically are seen in vegetation-rich, moist areas adjoining human dwellings (eg, parks, gardens), where clusters of cases can occur. These insects proliferate during the monsoon season in tropical and temperate areas, leading to more cases occurring during these months. 

Compared to prior reports,8,9 a few of our patients had predominant trunk and neck involvement with an occasional tingling sensation or pruritus while the rest were asymptomatic. Dermoscopic features from our patients shared similar reported features of Cydnidae pigmentation.4,5 The accentuation of pigment over skin creases seen on dermoscopy was due to accumulation of Cydnidae secretion at these sites. 

The differential diagnosis commonly includes idiopathic macular eruptive pigmentation, which is characterized by an asymptomatic progressive eruption of hyperpigmented macules over the trunk that persists from a few months up to 3 years. Other conditions in the differential include benign conditions such as acral benign melanocytic nevi, lentigines, pigmented purpuric dermatosis, and postinflammatory hyperpigmentation, as well as malignant conditions such as acral melanoma. Dermoscopy is a helpful, easy-to-use tool in differentiating these pigmentation disorders, obviating the need for an invasive investigation such as histopathologic analysis. Simultaneous involvement in a group of people living together or visiting the same place, abrupt onset, predominant involvement of the exposed sites, characteristic clinical and dermoscopic features, self-limiting course, and timing with the monsoon season should suggest a possibility of Cydnidae dermatitis/pigmentation, which can be confirmed by finding the causative bug in the affected locality.

Management

No specific treatment is required for the pigmentation caused by Cydnidae, as it is self-resolving. The macules can, however, be removed with acetone. Patients must be counseled regarding the benign and fleeting nature of this condition, as the abrupt onset may alarm them of a systemic disease. Affected patients should be advised against walking barefoot in areas where the insects can be found. Spraying insecticides in the affected locality also helps to reduce the presence of burrowing bugs.

References
  1. Hosokawa T, Kikuchi Y, Nikoh N, et al. Polyphyly of gut symbionts in stinkbugs of the family Cydnidae. Appl Environ Microbiol. 2012; 78:4758-4761.
  2. Schwertner CF, Nardi C. Burrower bugs (Cydnidae). In: Panizzi A, ­Grazia J, eds. True Bugs (Heteroptera) of the Neotropics. Entomology in Focus, vol 2. Springer; 2015.
  3. Lis JA. Burrower bugs of the Old World: a catalogue (Hemiptera: Heteroptera: Cydnidae). Genus (Wroclaw). 1999;10:165-249.
  4. Hayashi N, Yamamura Y, Ôhama S, et al. Defensive substances from stink bugs of Cydnidae. Experientia. 1976;32:418-419.
  5. Smith RM. The defensive secretion of the bugs Lampropharadifasciata, Adrisanumeensis, and Tectocorisdiophthalmus from Fiji. NZ J Zool. 1978;5:821-822.
  6. Krall BS, Zilkowski BW, Kight SL, et al. Chemistry and defensive efficacy of secretion of burrowing bugs. J Chem Ecol. 1997;23:1951-1962.
  7. Haddad V Jr, Cardoso J, Moraes R. Skin lesions caused by stink bugs (Insecta: Heteroptera: Pentatomidae): first report of dermatological injuries in humans. Wilderness Environ Med. 2002;13:48-50.
  8. Malhotra AK, Lis JA, Ramam M. Cydnidae (burrowing bug) pigmentation: a novel arthropod dermatosis. JAMA Dermatol. 2015;151:232-233.
  9. Sonthalia S. Dermoscopy of Cydnidae pigmentation: a novel disorder of pigmentation. Dermatol Pract Concept. 2019;9:228-229.
  10. Poojary S, Baddireddy K. Demystifying the stinking reddish brown stains through the dermoscope: Cydnidae pigmentation. Indian ­Dermatol Online J. 2019;10:757-758.
  11. Amrani A, Das A. Cydnidae pigmentation: unusual location on the abdomen and back. Br J Dermatol. 2021;184:E125.
Article PDF
Author and Disclosure Information

From the Department of Dermatology, Venereology and Leprology, Postgraduate Institute of Medical Education and Research, Chandigarh, India.

The authors have no relevant financial disclosures to report.

Correspondence: Muthu Sendhil Kumaran, MD, DNB, MNAMS (drsen_2000@yahoo.com).

Cutis. 2025 September;116(3):94-95, E11-E12. doi:10.12788/cutis.1261

Issue
Cutis - 116(3)
Publications
Topics
Page Number
94-95
Sections
Author and Disclosure Information

From the Department of Dermatology, Venereology and Leprology, Postgraduate Institute of Medical Education and Research, Chandigarh, India.

The authors have no relevant financial disclosures to report.

Correspondence: Muthu Sendhil Kumaran, MD, DNB, MNAMS (drsen_2000@yahoo.com).

Cutis. 2025 September;116(3):94-95, E11-E12. doi:10.12788/cutis.1261

Author and Disclosure Information

From the Department of Dermatology, Venereology and Leprology, Postgraduate Institute of Medical Education and Research, Chandigarh, India.

The authors have no relevant financial disclosures to report.

Correspondence: Muthu Sendhil Kumaran, MD, DNB, MNAMS (drsen_2000@yahoo.com).

Cutis. 2025 September;116(3):94-95, E11-E12. doi:10.12788/cutis.1261

Article PDF
Article PDF

Cydnidae is a family of small to medium-sized shield bugs with spiny legs that commonly are known as burrowing bugs (or burrower bugs). The family Cydnidae includes more than 100 genera and approximately 600 species worldwide.1 These insects are arthropods of the order Hemiptera (suborder: Heteroptera; superfamily: Pentatomoidae) and largely are concentrated in tropical and temperate regions. Approximately 145 species have been recorded in the Neotropical Region and have been included in the subfamilies Amnestinae, Cephalocteinae, and Sehirinae, in addition to Cydnidae.2 Burrowing bugs are ovoid in shape and 2 to 20 mm in length and morphologically are well adapted for burrowing. Their life span is 100 to 300 days. Being phytophagous, they burrow to feed on plants and roots. Adult burrowing bugs have wings and can fly. They have specialized glands located in either the abdomen (nymph) or thorax (adult) that secrete odorous chemicals for self-protection.3 The secretions contain hydrocarbonates that function as repellents and danger signals, can cause paralysis in prey, and act as a chemoattractant for mates.4-6 They also cause hyperpigmentation upon contact with the skin.

In this article, we present a series of cases from the same community to demonstrate the characteristic features of hyperpigmented macules caused by exposure to burrowing bugs. Dermatologists should be aware of this entity to prevent misdiagnosis and unnecessary investigations and treatment.

Case Series

A 36-year-old woman and 6 children (age range, 6-12 years) presented with a widespread, acute, brown-pigmented, macular eruption with lesions that increased in number over a 1-week period. All 7 patients resided in the same locality and were otherwise systemically healthy. Initially, the index case, a 7-year-old girl, was referred to our tertiary care center by a dermatologist with a provisional diagnosis of idiopathic macular eruptive pigmentation. The patient’s mother recalled noticing a tiny black insect on the child's scalp that left pigment on the skin when she crushed it between her fingers. The rest of the patients presented over the next few days: 3 of the children belonged to the same household as the index case, and there was history of all 6 children playing in the neighborhood park during late evening hours. The adult patient was the parent of one of the affected children. The lesions were associated with mild itching and tingling in 3 children but were asymptomatic in the other patients.

Clinical examination of the patients revealed multiple dark- to light-brown, discrete, irregularly shaped macules over the trunk, arms, and soles (eFigure 1). Dermoscopic examination of a pigmented macule showed an irregularly shaped, brownish, structureless area with accentuation of the pigment at skin creases and perieccrine pigmentation (eFigure 2). The pigmentation was unaffected by rubbing with alcohol or water. Clinicoepidemiologic parameters of the patients are summarized in the eTable.

CT116003094-Fig-1_ABC
eFIGURE 1. A-C, Discrete, irregularly shaped, brown to dark brown macules on the trunk, elbow, and soles.
CT116003094-Fig2_AB
eFIGURE 2. A and B, Dermoscopy showed irregularly shaped, homogenous, brownish, structureless areas with accentuation along skin creases and around eccrine A B openings (original magnification ×10 and ×10).

CT116003094-Table

One of the children’s parents conducted a geological examination of the ground in the neighborhood park during evening hours and found tiny burrowing bugs (eFigure 3). When crushed between the fingers, these insects left a similar brownish hyperpigmentation on the skin. The parents were counseled on the nature of the eruption, and the patients were kept under observation for 2 weeks. On follow-up after 5 days, the lesions showed markedly decreased intensity of hyperpigmentation, and no new lesions were observed in any of the 7 patients.

Baskaran-3
eFIGURE 3. A burrowing bug (Cydnidae) found at the neighborhood park visited by all patients.

Comment

Pentatomoidae insects generally are benign and harmless to humans. There have been isolated reports of erythematous plaques caused by Antiteuchus mixtus and Edessa maculate.7 Malhotra et al8 reported the first known series of cases with Cydnidae insect–induced hyperpigmented macules. The reported patients presented with asymptomatic, brown, hyperpigmented macules over exposed sites such as the feet, neck, and chest. All the cases occurred during the monsoon season in tropical and temperate regions of the world, and the patients were characteristically clustered in similar geographic areas. The causative insect was identified as Chilocoris assmuthi Breddin, 1904, belonging to the family Cydnidae. When it was crushed between the fingers, the skin became hyperpigmented, confirming the role of the secretions from the insect in the etiology.8

A second case was described by Sonthalia,9 who also described the dermoscopic features of hyperpigmented macules caused by burrowing bugs. The lesions showed a stuck-on, clustered appearance of ovoid and bizarre pigmented clods, globules, and granules.9 Although the lesions occur mainly over exposed sites, pigmented macules occurring over unusual sites such as the abdomen and back also have been reported in association with burrowing bugs.10 Characteristically, the lesions initially are faint and darken with time and usually fade within a week. They can be rubbed off with acetone but persist when washed with soap and water. The fleeting nature of the pigmentation also has led to the term transient pseudo-lentigines sign to describe hyperpigmentation caused by burrowing bugs.11

Soil and plants are burrowing bugs’ natural habitats, and the insects typically are seen in vegetation-rich, moist areas adjoining human dwellings (eg, parks, gardens), where clusters of cases can occur. These insects proliferate during the monsoon season in tropical and temperate areas, leading to more cases occurring during these months. 

Compared to prior reports,8,9 a few of our patients had predominant trunk and neck involvement with an occasional tingling sensation or pruritus while the rest were asymptomatic. Dermoscopic features from our patients shared similar reported features of Cydnidae pigmentation.4,5 The accentuation of pigment over skin creases seen on dermoscopy was due to accumulation of Cydnidae secretion at these sites. 

The differential diagnosis commonly includes idiopathic macular eruptive pigmentation, which is characterized by an asymptomatic progressive eruption of hyperpigmented macules over the trunk that persists from a few months up to 3 years. Other conditions in the differential include benign conditions such as acral benign melanocytic nevi, lentigines, pigmented purpuric dermatosis, and postinflammatory hyperpigmentation, as well as malignant conditions such as acral melanoma. Dermoscopy is a helpful, easy-to-use tool in differentiating these pigmentation disorders, obviating the need for an invasive investigation such as histopathologic analysis. Simultaneous involvement in a group of people living together or visiting the same place, abrupt onset, predominant involvement of the exposed sites, characteristic clinical and dermoscopic features, self-limiting course, and timing with the monsoon season should suggest a possibility of Cydnidae dermatitis/pigmentation, which can be confirmed by finding the causative bug in the affected locality.

Management

No specific treatment is required for the pigmentation caused by Cydnidae, as it is self-resolving. The macules can, however, be removed with acetone. Patients must be counseled regarding the benign and fleeting nature of this condition, as the abrupt onset may alarm them of a systemic disease. Affected patients should be advised against walking barefoot in areas where the insects can be found. Spraying insecticides in the affected locality also helps to reduce the presence of burrowing bugs.

Cydnidae is a family of small to medium-sized shield bugs with spiny legs that commonly are known as burrowing bugs (or burrower bugs). The family Cydnidae includes more than 100 genera and approximately 600 species worldwide.1 These insects are arthropods of the order Hemiptera (suborder: Heteroptera; superfamily: Pentatomoidae) and largely are concentrated in tropical and temperate regions. Approximately 145 species have been recorded in the Neotropical Region and have been included in the subfamilies Amnestinae, Cephalocteinae, and Sehirinae, in addition to Cydnidae.2 Burrowing bugs are ovoid in shape and 2 to 20 mm in length and morphologically are well adapted for burrowing. Their life span is 100 to 300 days. Being phytophagous, they burrow to feed on plants and roots. Adult burrowing bugs have wings and can fly. They have specialized glands located in either the abdomen (nymph) or thorax (adult) that secrete odorous chemicals for self-protection.3 The secretions contain hydrocarbonates that function as repellents and danger signals, can cause paralysis in prey, and act as a chemoattractant for mates.4-6 They also cause hyperpigmentation upon contact with the skin.

In this article, we present a series of cases from the same community to demonstrate the characteristic features of hyperpigmented macules caused by exposure to burrowing bugs. Dermatologists should be aware of this entity to prevent misdiagnosis and unnecessary investigations and treatment.

Case Series

A 36-year-old woman and 6 children (age range, 6-12 years) presented with a widespread, acute, brown-pigmented, macular eruption with lesions that increased in number over a 1-week period. All 7 patients resided in the same locality and were otherwise systemically healthy. Initially, the index case, a 7-year-old girl, was referred to our tertiary care center by a dermatologist with a provisional diagnosis of idiopathic macular eruptive pigmentation. The patient’s mother recalled noticing a tiny black insect on the child's scalp that left pigment on the skin when she crushed it between her fingers. The rest of the patients presented over the next few days: 3 of the children belonged to the same household as the index case, and there was history of all 6 children playing in the neighborhood park during late evening hours. The adult patient was the parent of one of the affected children. The lesions were associated with mild itching and tingling in 3 children but were asymptomatic in the other patients.

Clinical examination of the patients revealed multiple dark- to light-brown, discrete, irregularly shaped macules over the trunk, arms, and soles (eFigure 1). Dermoscopic examination of a pigmented macule showed an irregularly shaped, brownish, structureless area with accentuation of the pigment at skin creases and perieccrine pigmentation (eFigure 2). The pigmentation was unaffected by rubbing with alcohol or water. Clinicoepidemiologic parameters of the patients are summarized in the eTable.

CT116003094-Fig-1_ABC
eFIGURE 1. A-C, Discrete, irregularly shaped, brown to dark brown macules on the trunk, elbow, and soles.
CT116003094-Fig2_AB
eFIGURE 2. A and B, Dermoscopy showed irregularly shaped, homogenous, brownish, structureless areas with accentuation along skin creases and around eccrine A B openings (original magnification ×10 and ×10).

CT116003094-Table

One of the children’s parents conducted a geological examination of the ground in the neighborhood park during evening hours and found tiny burrowing bugs (eFigure 3). When crushed between the fingers, these insects left a similar brownish hyperpigmentation on the skin. The parents were counseled on the nature of the eruption, and the patients were kept under observation for 2 weeks. On follow-up after 5 days, the lesions showed markedly decreased intensity of hyperpigmentation, and no new lesions were observed in any of the 7 patients.

Baskaran-3
eFIGURE 3. A burrowing bug (Cydnidae) found at the neighborhood park visited by all patients.

Comment

Pentatomoidae insects generally are benign and harmless to humans. There have been isolated reports of erythematous plaques caused by Antiteuchus mixtus and Edessa maculate.7 Malhotra et al8 reported the first known series of cases with Cydnidae insect–induced hyperpigmented macules. The reported patients presented with asymptomatic, brown, hyperpigmented macules over exposed sites such as the feet, neck, and chest. All the cases occurred during the monsoon season in tropical and temperate regions of the world, and the patients were characteristically clustered in similar geographic areas. The causative insect was identified as Chilocoris assmuthi Breddin, 1904, belonging to the family Cydnidae. When it was crushed between the fingers, the skin became hyperpigmented, confirming the role of the secretions from the insect in the etiology.8

A second case was described by Sonthalia,9 who also described the dermoscopic features of hyperpigmented macules caused by burrowing bugs. The lesions showed a stuck-on, clustered appearance of ovoid and bizarre pigmented clods, globules, and granules.9 Although the lesions occur mainly over exposed sites, pigmented macules occurring over unusual sites such as the abdomen and back also have been reported in association with burrowing bugs.10 Characteristically, the lesions initially are faint and darken with time and usually fade within a week. They can be rubbed off with acetone but persist when washed with soap and water. The fleeting nature of the pigmentation also has led to the term transient pseudo-lentigines sign to describe hyperpigmentation caused by burrowing bugs.11

Soil and plants are burrowing bugs’ natural habitats, and the insects typically are seen in vegetation-rich, moist areas adjoining human dwellings (eg, parks, gardens), where clusters of cases can occur. These insects proliferate during the monsoon season in tropical and temperate areas, leading to more cases occurring during these months. 

Compared to prior reports,8,9 a few of our patients had predominant trunk and neck involvement with an occasional tingling sensation or pruritus while the rest were asymptomatic. Dermoscopic features from our patients shared similar reported features of Cydnidae pigmentation.4,5 The accentuation of pigment over skin creases seen on dermoscopy was due to accumulation of Cydnidae secretion at these sites. 

The differential diagnosis commonly includes idiopathic macular eruptive pigmentation, which is characterized by an asymptomatic progressive eruption of hyperpigmented macules over the trunk that persists from a few months up to 3 years. Other conditions in the differential include benign conditions such as acral benign melanocytic nevi, lentigines, pigmented purpuric dermatosis, and postinflammatory hyperpigmentation, as well as malignant conditions such as acral melanoma. Dermoscopy is a helpful, easy-to-use tool in differentiating these pigmentation disorders, obviating the need for an invasive investigation such as histopathologic analysis. Simultaneous involvement in a group of people living together or visiting the same place, abrupt onset, predominant involvement of the exposed sites, characteristic clinical and dermoscopic features, self-limiting course, and timing with the monsoon season should suggest a possibility of Cydnidae dermatitis/pigmentation, which can be confirmed by finding the causative bug in the affected locality.

Management

No specific treatment is required for the pigmentation caused by Cydnidae, as it is self-resolving. The macules can, however, be removed with acetone. Patients must be counseled regarding the benign and fleeting nature of this condition, as the abrupt onset may alarm them of a systemic disease. Affected patients should be advised against walking barefoot in areas where the insects can be found. Spraying insecticides in the affected locality also helps to reduce the presence of burrowing bugs.

References
  1. Hosokawa T, Kikuchi Y, Nikoh N, et al. Polyphyly of gut symbionts in stinkbugs of the family Cydnidae. Appl Environ Microbiol. 2012; 78:4758-4761.
  2. Schwertner CF, Nardi C. Burrower bugs (Cydnidae). In: Panizzi A, ­Grazia J, eds. True Bugs (Heteroptera) of the Neotropics. Entomology in Focus, vol 2. Springer; 2015.
  3. Lis JA. Burrower bugs of the Old World: a catalogue (Hemiptera: Heteroptera: Cydnidae). Genus (Wroclaw). 1999;10:165-249.
  4. Hayashi N, Yamamura Y, Ôhama S, et al. Defensive substances from stink bugs of Cydnidae. Experientia. 1976;32:418-419.
  5. Smith RM. The defensive secretion of the bugs Lampropharadifasciata, Adrisanumeensis, and Tectocorisdiophthalmus from Fiji. NZ J Zool. 1978;5:821-822.
  6. Krall BS, Zilkowski BW, Kight SL, et al. Chemistry and defensive efficacy of secretion of burrowing bugs. J Chem Ecol. 1997;23:1951-1962.
  7. Haddad V Jr, Cardoso J, Moraes R. Skin lesions caused by stink bugs (Insecta: Heteroptera: Pentatomidae): first report of dermatological injuries in humans. Wilderness Environ Med. 2002;13:48-50.
  8. Malhotra AK, Lis JA, Ramam M. Cydnidae (burrowing bug) pigmentation: a novel arthropod dermatosis. JAMA Dermatol. 2015;151:232-233.
  9. Sonthalia S. Dermoscopy of Cydnidae pigmentation: a novel disorder of pigmentation. Dermatol Pract Concept. 2019;9:228-229.
  10. Poojary S, Baddireddy K. Demystifying the stinking reddish brown stains through the dermoscope: Cydnidae pigmentation. Indian ­Dermatol Online J. 2019;10:757-758.
  11. Amrani A, Das A. Cydnidae pigmentation: unusual location on the abdomen and back. Br J Dermatol. 2021;184:E125.
References
  1. Hosokawa T, Kikuchi Y, Nikoh N, et al. Polyphyly of gut symbionts in stinkbugs of the family Cydnidae. Appl Environ Microbiol. 2012; 78:4758-4761.
  2. Schwertner CF, Nardi C. Burrower bugs (Cydnidae). In: Panizzi A, ­Grazia J, eds. True Bugs (Heteroptera) of the Neotropics. Entomology in Focus, vol 2. Springer; 2015.
  3. Lis JA. Burrower bugs of the Old World: a catalogue (Hemiptera: Heteroptera: Cydnidae). Genus (Wroclaw). 1999;10:165-249.
  4. Hayashi N, Yamamura Y, Ôhama S, et al. Defensive substances from stink bugs of Cydnidae. Experientia. 1976;32:418-419.
  5. Smith RM. The defensive secretion of the bugs Lampropharadifasciata, Adrisanumeensis, and Tectocorisdiophthalmus from Fiji. NZ J Zool. 1978;5:821-822.
  6. Krall BS, Zilkowski BW, Kight SL, et al. Chemistry and defensive efficacy of secretion of burrowing bugs. J Chem Ecol. 1997;23:1951-1962.
  7. Haddad V Jr, Cardoso J, Moraes R. Skin lesions caused by stink bugs (Insecta: Heteroptera: Pentatomidae): first report of dermatological injuries in humans. Wilderness Environ Med. 2002;13:48-50.
  8. Malhotra AK, Lis JA, Ramam M. Cydnidae (burrowing bug) pigmentation: a novel arthropod dermatosis. JAMA Dermatol. 2015;151:232-233.
  9. Sonthalia S. Dermoscopy of Cydnidae pigmentation: a novel disorder of pigmentation. Dermatol Pract Concept. 2019;9:228-229.
  10. Poojary S, Baddireddy K. Demystifying the stinking reddish brown stains through the dermoscope: Cydnidae pigmentation. Indian ­Dermatol Online J. 2019;10:757-758.
  11. Amrani A, Das A. Cydnidae pigmentation: unusual location on the abdomen and back. Br J Dermatol. 2021;184:E125.
Issue
Cutis - 116(3)
Issue
Cutis - 116(3)
Page Number
94-95
Page Number
94-95
Publications
Publications
Topics
Article Type
Display Headline

Hyperpigmented Macules Caused by Burrowing Bugs (Cydnidae) May Mimic More Serious Conditions

Display Headline

Hyperpigmented Macules Caused by Burrowing Bugs (Cydnidae) May Mimic More Serious Conditions

Sections
Inside the Article

Practice Points

  • Burrowing bugs (Cydnidae) are phytophagous and burrow to feed on plants and roots. They are more numerous during the monsoon season in tropical and temperate regions.
  • Secretions from burrowing bugs cause asymptomatic, hyperpigmented, irregularly shaped macules suggestive of an exogenous cause that commonly affect clusters of patients from the same geographic locality.
  • The lesions are self-limiting and must be differentiated from close mimickers to ensure adequate and appropriate patient counseling.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Fri, 09/05/2025 - 11:36
Un-Gate On Date
Fri, 09/05/2025 - 11:36
Use ProPublica
CFC Schedule Remove Status
Fri, 09/05/2025 - 11:36
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
survey writer start date
Fri, 09/05/2025 - 11:36

Tapping Into Relief: A Distraction Technique to Reduce Pain During Dermatologic Procedures

Article Type
Changed
Fri, 09/05/2025 - 10:55
Display Headline

Tapping Into Relief: A Distraction Technique to Reduce Pain During Dermatologic Procedures

Practice Gap

Pain during minimally invasive dermatologic procedures such as lidocaine injections, cryotherapy, nail unit injections, and cosmetic procedures including neurotoxin injections can cause patient discomfort leading to procedural anxiety, poor compliance with treatment regimens, and avoidance of necessary care. Current solutions to manage pain during dermatologic procedures present several limitations; for example, topical anesthetics seldom alleviate procedural pain,1 particularly in sensitive areas (eg, nail unit, face) or for patients with a needle phobia. Additionally, topical anesthetics often require up to 2 hours to take effect, making them impractical for quick outpatient procedures. Other pain reduction strategies including vibration devices or cold sprays2,3 can be effective but are an added expense to the physician or clinic, which may preclude their use in resource-limited settings. Psychological distraction techniques such as deep breathing require active patient participation and might reinforce pain expectations and increase patient anxiety.4 Given these challenges, there is a need for effective, affordable, nonpharmacologic pain reduction strategies that can be integrated seamlessly into clinical practice to enhance the patient experience.

The Technique

Tapping is a simple noninvasive distraction technique that may alleviate procedural pain by exploiting the gate control theory of pain.5 According to this theory, tactile stimuli activate mechanoreceptors that send inhibitory signals to the spinal cord, effectively closing the gate to pain transmission. Unlike the Helfer skin tap technique,6 which involves 15 preinjection taps and 3 postinjection taps directly on the injection site, our approach targets distant bony prominences. This modification allows for immediate needle insertion without interfering with the sterile field or increasing the risk for needlestick injuries from tapping near the injection site. Bony sites such as the shoulder or knee are ideal for this technique due to their high density and rigidity that efficiently transmit tactile stimuli––similar to how sound travels faster through solids than through liquids or gases.7

To implement this technique in practice, we first stabilize the injection site to reduce movement from tapping. This can be done by stabilizing the injection site (eg, resting the hand on an instrument stand during a nail unit injection). A second person—such as a medical assistant, medical student, resident, or even the patient’s family member—taps at a distant site at least an arm’s length away from the injection site (Figure). The tapping pressure should be firm enough for the patient to feel the vibration but not forceful enough that it becomes unpleasant or disrupts the injection area. Tapping starts just before needle insertion and continues through the injection. No warning is given to the patient, as the surprise element may help distract them from pain. Varying the rhythm, intensity, or location of the tapping can enhance its distracting effect. 

Ong-Pearls-0925
FIGURE. Demonstration of a medical student tapping a patient’s shoulder during nail unit injections.

This tapping technique can be effectively combined with other pain reduction strategies in a multimodal approach; for example, when used concurrently with topical anesthetics, both the central (tapping) and peripheral (anesthetic) pain pathways are addressed, potentially yielding additive effects. For patients with a needle ­phobia, pairing tapping with cognitive distraction (eg, talkesthesia) may further reduce anxiety. In our nail specialty clinic at Weill Cornell Medicine (New York, New York), we often combine tapping with cold sprays and talkesthesia, which improves patient comfort without prolonging the visit. Importantly, the technique enables seamless integration with most pharmacologic and nonpharmacologic methods, eliminating the need for additional patient education or procedure time.

Practice Implications

The tapping technique described here is free, easy to implement, and requires no additional resources aside from another person to tap the patient during the procedure. It can be used for a wide range of dermatologic procedures, including biopsies, intralesional injections, and cosmetic treatments, including neurotoxin injections. The minimal learning curve and ease of integration into procedural workflows make this technique a valuable tool for dermatologists aiming to improve patient comfort without disrupting workflow. In our practice, we have observed that tapping reduces self-reported pain and helps ease anxiety, particularly in patients with a needle phobia. Its simplicity and accessibility make it a valuable addition to a wide range of dermatologic procedures. Prospective studies investigating patient-reported outcomes could help establish this technique’s role in clinical practice.

References
  1. Navarro-Rodriguez JM, Suarez-Serrano C, Martin-Valero R, et al. Effectiveness of topical anesthetics in pain management for dermal injuries: a systematic review. J Clin Med. 2021;10:2522. doi:10.3390/jcm10112522
  2. Lipner SR. Pain-minimizing strategies for nail surgery. Cutis. 2018;101:76-77.
  3. Ricardo JW, Lipner SR. Air cooling for improved analgesia during local anesthetic infiltration for nail surgery. J Am Acad Dermatol. 2021;84:e231-e232. doi:10.1016/j.jaad.2019.11.032
  4. Hill RC, Chernoff KA, Lipner SR. A breath of fresh air: use of deep breathing technique to minimize pain with nail injections. J Am Acad Dermatol. 2024;90:e163. doi:10.1016/j.jaad.2023.10.043
  5. Mendell LM. Constructing and deconstructing the gate theory of pain. Pain. 2014;155:210-216. doi:10.1016/j.pain.2013.12.010
  6. Jyoti G, Arora S, Sharma B. Helfer Skin Tap Tech Technique for the IM injection pain among adult patients. Nursing & Midwifery Research Journal. 2018;14:18-30. doi:10.1177/0974150X20180304
  7. Iowa State University. Nondestructive Evaluation Physics: Sound. Published 2021. Accessed July 31, 2025. https://www.nde-ed.org/Physics/Sound/speedinmaterials.xhtml
Article PDF
Author and Disclosure Information

Michael M. Ong and Dr. Lipner are from Weill Cornell Medicine, New York, New York. Dr. Lipner is from the Department of Dermatology. Zachary Neubauer is from the Thomas Jefferson University, Philadelphia, Pennsylvania. Naeha Pathak is from Icahn School of Medicine, Mount Sinai, New York. Amit Singal is from Rutgers New Jersey Medical School, Newark.

Michael M. Ong, Zachary Neubauer, Naeha Pathak, and Amit Singal have no relevant financial disclosures to report. Dr. Lipner has served as a consultant for BelleTorus Corporation and Moberg Pharmaceuticals.

Correspondence: Shari R. Lipner, MD, PhD, 1305 York Ave, 9th Floor, New York, NY 10021 (shl9032@med.cornell.edu).

Cutis. 2025 September;116(3):96-97. doi:10.12788/cutis.1257

Issue
Cutis - 116(3)
Publications
Topics
Page Number
96-97
Sections
Author and Disclosure Information

Michael M. Ong and Dr. Lipner are from Weill Cornell Medicine, New York, New York. Dr. Lipner is from the Department of Dermatology. Zachary Neubauer is from the Thomas Jefferson University, Philadelphia, Pennsylvania. Naeha Pathak is from Icahn School of Medicine, Mount Sinai, New York. Amit Singal is from Rutgers New Jersey Medical School, Newark.

Michael M. Ong, Zachary Neubauer, Naeha Pathak, and Amit Singal have no relevant financial disclosures to report. Dr. Lipner has served as a consultant for BelleTorus Corporation and Moberg Pharmaceuticals.

Correspondence: Shari R. Lipner, MD, PhD, 1305 York Ave, 9th Floor, New York, NY 10021 (shl9032@med.cornell.edu).

Cutis. 2025 September;116(3):96-97. doi:10.12788/cutis.1257

Author and Disclosure Information

Michael M. Ong and Dr. Lipner are from Weill Cornell Medicine, New York, New York. Dr. Lipner is from the Department of Dermatology. Zachary Neubauer is from the Thomas Jefferson University, Philadelphia, Pennsylvania. Naeha Pathak is from Icahn School of Medicine, Mount Sinai, New York. Amit Singal is from Rutgers New Jersey Medical School, Newark.

Michael M. Ong, Zachary Neubauer, Naeha Pathak, and Amit Singal have no relevant financial disclosures to report. Dr. Lipner has served as a consultant for BelleTorus Corporation and Moberg Pharmaceuticals.

Correspondence: Shari R. Lipner, MD, PhD, 1305 York Ave, 9th Floor, New York, NY 10021 (shl9032@med.cornell.edu).

Cutis. 2025 September;116(3):96-97. doi:10.12788/cutis.1257

Article PDF
Article PDF

Practice Gap

Pain during minimally invasive dermatologic procedures such as lidocaine injections, cryotherapy, nail unit injections, and cosmetic procedures including neurotoxin injections can cause patient discomfort leading to procedural anxiety, poor compliance with treatment regimens, and avoidance of necessary care. Current solutions to manage pain during dermatologic procedures present several limitations; for example, topical anesthetics seldom alleviate procedural pain,1 particularly in sensitive areas (eg, nail unit, face) or for patients with a needle phobia. Additionally, topical anesthetics often require up to 2 hours to take effect, making them impractical for quick outpatient procedures. Other pain reduction strategies including vibration devices or cold sprays2,3 can be effective but are an added expense to the physician or clinic, which may preclude their use in resource-limited settings. Psychological distraction techniques such as deep breathing require active patient participation and might reinforce pain expectations and increase patient anxiety.4 Given these challenges, there is a need for effective, affordable, nonpharmacologic pain reduction strategies that can be integrated seamlessly into clinical practice to enhance the patient experience.

The Technique

Tapping is a simple noninvasive distraction technique that may alleviate procedural pain by exploiting the gate control theory of pain.5 According to this theory, tactile stimuli activate mechanoreceptors that send inhibitory signals to the spinal cord, effectively closing the gate to pain transmission. Unlike the Helfer skin tap technique,6 which involves 15 preinjection taps and 3 postinjection taps directly on the injection site, our approach targets distant bony prominences. This modification allows for immediate needle insertion without interfering with the sterile field or increasing the risk for needlestick injuries from tapping near the injection site. Bony sites such as the shoulder or knee are ideal for this technique due to their high density and rigidity that efficiently transmit tactile stimuli––similar to how sound travels faster through solids than through liquids or gases.7

To implement this technique in practice, we first stabilize the injection site to reduce movement from tapping. This can be done by stabilizing the injection site (eg, resting the hand on an instrument stand during a nail unit injection). A second person—such as a medical assistant, medical student, resident, or even the patient’s family member—taps at a distant site at least an arm’s length away from the injection site (Figure). The tapping pressure should be firm enough for the patient to feel the vibration but not forceful enough that it becomes unpleasant or disrupts the injection area. Tapping starts just before needle insertion and continues through the injection. No warning is given to the patient, as the surprise element may help distract them from pain. Varying the rhythm, intensity, or location of the tapping can enhance its distracting effect. 

Ong-Pearls-0925
FIGURE. Demonstration of a medical student tapping a patient’s shoulder during nail unit injections.

This tapping technique can be effectively combined with other pain reduction strategies in a multimodal approach; for example, when used concurrently with topical anesthetics, both the central (tapping) and peripheral (anesthetic) pain pathways are addressed, potentially yielding additive effects. For patients with a needle ­phobia, pairing tapping with cognitive distraction (eg, talkesthesia) may further reduce anxiety. In our nail specialty clinic at Weill Cornell Medicine (New York, New York), we often combine tapping with cold sprays and talkesthesia, which improves patient comfort without prolonging the visit. Importantly, the technique enables seamless integration with most pharmacologic and nonpharmacologic methods, eliminating the need for additional patient education or procedure time.

Practice Implications

The tapping technique described here is free, easy to implement, and requires no additional resources aside from another person to tap the patient during the procedure. It can be used for a wide range of dermatologic procedures, including biopsies, intralesional injections, and cosmetic treatments, including neurotoxin injections. The minimal learning curve and ease of integration into procedural workflows make this technique a valuable tool for dermatologists aiming to improve patient comfort without disrupting workflow. In our practice, we have observed that tapping reduces self-reported pain and helps ease anxiety, particularly in patients with a needle phobia. Its simplicity and accessibility make it a valuable addition to a wide range of dermatologic procedures. Prospective studies investigating patient-reported outcomes could help establish this technique’s role in clinical practice.

Practice Gap

Pain during minimally invasive dermatologic procedures such as lidocaine injections, cryotherapy, nail unit injections, and cosmetic procedures including neurotoxin injections can cause patient discomfort leading to procedural anxiety, poor compliance with treatment regimens, and avoidance of necessary care. Current solutions to manage pain during dermatologic procedures present several limitations; for example, topical anesthetics seldom alleviate procedural pain,1 particularly in sensitive areas (eg, nail unit, face) or for patients with a needle phobia. Additionally, topical anesthetics often require up to 2 hours to take effect, making them impractical for quick outpatient procedures. Other pain reduction strategies including vibration devices or cold sprays2,3 can be effective but are an added expense to the physician or clinic, which may preclude their use in resource-limited settings. Psychological distraction techniques such as deep breathing require active patient participation and might reinforce pain expectations and increase patient anxiety.4 Given these challenges, there is a need for effective, affordable, nonpharmacologic pain reduction strategies that can be integrated seamlessly into clinical practice to enhance the patient experience.

The Technique

Tapping is a simple noninvasive distraction technique that may alleviate procedural pain by exploiting the gate control theory of pain.5 According to this theory, tactile stimuli activate mechanoreceptors that send inhibitory signals to the spinal cord, effectively closing the gate to pain transmission. Unlike the Helfer skin tap technique,6 which involves 15 preinjection taps and 3 postinjection taps directly on the injection site, our approach targets distant bony prominences. This modification allows for immediate needle insertion without interfering with the sterile field or increasing the risk for needlestick injuries from tapping near the injection site. Bony sites such as the shoulder or knee are ideal for this technique due to their high density and rigidity that efficiently transmit tactile stimuli––similar to how sound travels faster through solids than through liquids or gases.7

To implement this technique in practice, we first stabilize the injection site to reduce movement from tapping. This can be done by stabilizing the injection site (eg, resting the hand on an instrument stand during a nail unit injection). A second person—such as a medical assistant, medical student, resident, or even the patient’s family member—taps at a distant site at least an arm’s length away from the injection site (Figure). The tapping pressure should be firm enough for the patient to feel the vibration but not forceful enough that it becomes unpleasant or disrupts the injection area. Tapping starts just before needle insertion and continues through the injection. No warning is given to the patient, as the surprise element may help distract them from pain. Varying the rhythm, intensity, or location of the tapping can enhance its distracting effect. 

Ong-Pearls-0925
FIGURE. Demonstration of a medical student tapping a patient’s shoulder during nail unit injections.

This tapping technique can be effectively combined with other pain reduction strategies in a multimodal approach; for example, when used concurrently with topical anesthetics, both the central (tapping) and peripheral (anesthetic) pain pathways are addressed, potentially yielding additive effects. For patients with a needle ­phobia, pairing tapping with cognitive distraction (eg, talkesthesia) may further reduce anxiety. In our nail specialty clinic at Weill Cornell Medicine (New York, New York), we often combine tapping with cold sprays and talkesthesia, which improves patient comfort without prolonging the visit. Importantly, the technique enables seamless integration with most pharmacologic and nonpharmacologic methods, eliminating the need for additional patient education or procedure time.

Practice Implications

The tapping technique described here is free, easy to implement, and requires no additional resources aside from another person to tap the patient during the procedure. It can be used for a wide range of dermatologic procedures, including biopsies, intralesional injections, and cosmetic treatments, including neurotoxin injections. The minimal learning curve and ease of integration into procedural workflows make this technique a valuable tool for dermatologists aiming to improve patient comfort without disrupting workflow. In our practice, we have observed that tapping reduces self-reported pain and helps ease anxiety, particularly in patients with a needle phobia. Its simplicity and accessibility make it a valuable addition to a wide range of dermatologic procedures. Prospective studies investigating patient-reported outcomes could help establish this technique’s role in clinical practice.

References
  1. Navarro-Rodriguez JM, Suarez-Serrano C, Martin-Valero R, et al. Effectiveness of topical anesthetics in pain management for dermal injuries: a systematic review. J Clin Med. 2021;10:2522. doi:10.3390/jcm10112522
  2. Lipner SR. Pain-minimizing strategies for nail surgery. Cutis. 2018;101:76-77.
  3. Ricardo JW, Lipner SR. Air cooling for improved analgesia during local anesthetic infiltration for nail surgery. J Am Acad Dermatol. 2021;84:e231-e232. doi:10.1016/j.jaad.2019.11.032
  4. Hill RC, Chernoff KA, Lipner SR. A breath of fresh air: use of deep breathing technique to minimize pain with nail injections. J Am Acad Dermatol. 2024;90:e163. doi:10.1016/j.jaad.2023.10.043
  5. Mendell LM. Constructing and deconstructing the gate theory of pain. Pain. 2014;155:210-216. doi:10.1016/j.pain.2013.12.010
  6. Jyoti G, Arora S, Sharma B. Helfer Skin Tap Tech Technique for the IM injection pain among adult patients. Nursing & Midwifery Research Journal. 2018;14:18-30. doi:10.1177/0974150X20180304
  7. Iowa State University. Nondestructive Evaluation Physics: Sound. Published 2021. Accessed July 31, 2025. https://www.nde-ed.org/Physics/Sound/speedinmaterials.xhtml
References
  1. Navarro-Rodriguez JM, Suarez-Serrano C, Martin-Valero R, et al. Effectiveness of topical anesthetics in pain management for dermal injuries: a systematic review. J Clin Med. 2021;10:2522. doi:10.3390/jcm10112522
  2. Lipner SR. Pain-minimizing strategies for nail surgery. Cutis. 2018;101:76-77.
  3. Ricardo JW, Lipner SR. Air cooling for improved analgesia during local anesthetic infiltration for nail surgery. J Am Acad Dermatol. 2021;84:e231-e232. doi:10.1016/j.jaad.2019.11.032
  4. Hill RC, Chernoff KA, Lipner SR. A breath of fresh air: use of deep breathing technique to minimize pain with nail injections. J Am Acad Dermatol. 2024;90:e163. doi:10.1016/j.jaad.2023.10.043
  5. Mendell LM. Constructing and deconstructing the gate theory of pain. Pain. 2014;155:210-216. doi:10.1016/j.pain.2013.12.010
  6. Jyoti G, Arora S, Sharma B. Helfer Skin Tap Tech Technique for the IM injection pain among adult patients. Nursing & Midwifery Research Journal. 2018;14:18-30. doi:10.1177/0974150X20180304
  7. Iowa State University. Nondestructive Evaluation Physics: Sound. Published 2021. Accessed July 31, 2025. https://www.nde-ed.org/Physics/Sound/speedinmaterials.xhtml
Issue
Cutis - 116(3)
Issue
Cutis - 116(3)
Page Number
96-97
Page Number
96-97
Publications
Publications
Topics
Article Type
Display Headline

Tapping Into Relief: A Distraction Technique to Reduce Pain During Dermatologic Procedures

Display Headline

Tapping Into Relief: A Distraction Technique to Reduce Pain During Dermatologic Procedures

Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Fri, 09/05/2025 - 10:37
Un-Gate On Date
Fri, 09/05/2025 - 10:37
Use ProPublica
CFC Schedule Remove Status
Fri, 09/05/2025 - 10:37
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
survey writer start date
Fri, 09/05/2025 - 10:37

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

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

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

To the Editor:

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

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

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

CT116003107-Table

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

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

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

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

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

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

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

Correspondence: Shari R. Lipner, MD, PhD, 1305 York Avenue, New York, NY 10021 (shl9032@med.cornell.edu).

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

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

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

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

Correspondence: Shari R. Lipner, MD, PhD, 1305 York Avenue, New York, NY 10021 (shl9032@med.cornell.edu).

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

Author and Disclosure Information

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

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

Correspondence: Shari R. Lipner, MD, PhD, 1305 York Avenue, New York, NY 10021 (shl9032@med.cornell.edu).

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

Article PDF
Article PDF

To the Editor:

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

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

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

CT116003107-Table

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

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

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

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

To the Editor:

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

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

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

CT116003107-Table

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

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

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

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

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

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

Display Headline

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

Sections
Inside the Article

PRACTICE POINTS

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

From Refractory to Responsive: The Expanding Therapeutic Landscape of Prurigo Nodularis

Article Type
Changed
Fri, 09/05/2025 - 10:26
Display Headline

From Refractory to Responsive: The Expanding Therapeutic Landscape of Prurigo Nodularis

Prurigo nodularis (PN) is a chronic, severely pruritic neuroimmunologic skin disorder characterized by multiple firm hyperkeratotic nodules and intense pruritus, often leading to considerable impairment in quality of life and increased rates of depression and anxiety.1 It is considered difficult to treat due to its complex pathogenesis, the severity and chronicity of pruritus, and the limited efficacy of conventional therapies.2,3 The disease is driven by a self-perpetuating itch-scratch cycle, underpinned by dysregulation of both immune and neural pathways including type 2 (interleukin [IL] 4, IL-13, IL-31), Th17, and Th22 cytokines as well as neuropeptides and altered cutaneous nerve architecture.1,3 This results in persistent severe pruritus and nodular lesions that are highly refractory to standard treatments.1 Conventional therapies (eg, locally acting agents, phototherapy, and systemic immunomodulators and neuromodulators) have varied efficacy and notable adverse effect profiles.3 While the approval of targeted biologics has transformed the therapeutic landscape, several other treatment options also are being explored in clinical trials. Herein, we review all recently approved therapies as well as emerging treatments currently under investigation.

Dupilumab

Dupilumab, the first therapy for PN approved by the US Food and Drug Administration (FDA) in 2022—is a monoclonal antibody that inhibits signaling of IL-4 and IL-13, key drivers of type 2 inflammation implicated in PN pathogenesis.4,5 In 2 pivotal phase 3 randomized controlled trials (LIBERTY-PN PRIME and PRIME2),5 dupilumab demonstrated notable efficacy in adults with moderate to severe PN. A reduction of 4 points or more on the Worst Itch Numeric Rating Scale (WI-NRS) was achieved by 60.0% (45/75) of patients treated with dupilumab at week 24 compared with 18.4% (14/76) receiving placebo in the PRIME trial. In PRIME2, the same outcome was achieved by 37.2% (29/78) of patients receiving dupilumab at week 12 compared with 22.0% (18/82) of patients receiving placebo.5 Dupilumab also led to a greater proportion of patients achieving a substantial reduction in nodule count (5 nodules) and improved quality of life compared with placebo.5,6 The safety profile of dupilumab for treatment of PN was favorable and consistent with prior experience in atopic dermatitis; conjunctivitis was the most common adverse event.5,6

Nemolizumab

Nemolizumab, an IL-31 receptor A antagonist, is the most recent agent approved by the FDA for PN in 2024.7 In the OLYMPIA 1 and OLYMPIA 2 phase 3 trials,8 nemolizumab produced a clinically meaningful reduction in itch (defined as a 4-point improvement in the Peak Pruritus Numerical Rating Scale score) in 56.3% (103/183) of patients at week 16 compared with 20.9% (19/91) receiving placebo. Additionally, 37.7% (69/183) of patients receiving nemolizumab achieved clear or almost clear skin (Investigator’s Global Assessment score of 0 or 1 with a 2-point reduction) vs 11.0% with placebo (both P<.001). Benefits were observed as early as week 4, including rapid improvements in itch, sleep disturbance, and nodule count.8 Nemolizumab also improved quality of life and reduced symptoms of anxiety and depression. The safety profile was favorable, with headache and atopic dermatitis the most common adverse events; serious adverse events were infrequent and similar between groups.8

Abrocitinib

Abrocitinib, an oral selective Janus kinase 1 inhibitor, is an investigational therapy for PN and currently has not been approved by the FDA for this indication. In a phase 2 open-label trial, abrocitinib 200 mg daily for 12 weeks led to a 78.3% reduction in weekly Peak Pruritus Numerical Rating Scale scores in PN, with 80.0% (8/10) of patients achieving a clinically meaningful improvement of 4 points or higher. Nodule counts and quality of life also improved, with an onset of itch relief as early as week 2. The safety profile was favorable, with acneform eruptions the most common adverse event and no serious adverse events reported9; however, these results are based on small, nonrandomized studies and require confirmation in larger randomized controlled trials before abrocitinib can be considered a standard therapy for PN.

Cryosim-1

Transient receptor potential melastatin 8 (TRPM8) is a cold-sensing ion channel found in unmyelinated sensory neurons within the dorsal root and trigeminal ­ganglia.10 It is activated by cool temperatures (15-28 °C) and compounds such as menthol, leading to calcium influx and a cooling sensation. In a randomized, double-blind, vehicle-controlled trial, researchers investigated the efficacy of cryosim-1 (a synthetic TRPM8 agonist) in treating PN.10 Thirty patients were enrolled, with 18 (60.0%) receiving cryosim-1 and 12 (40.0%) receiving placebo over 8 weeks. By week 8, cryosim-1 significantly reduced itch severity (mean numerical rating scale score postapplication, 2.8 vs 4.3; P=.031) and improved sleep disturbances (2.2 vs 4.2; P=.031) compared to placebo. Patients reported higher satisfaction with itch relief, and no adverse effects were observed. The study concluded that cryosim-1 is a safe, effective topical therapy for PN, likely working by interrupting the itch-scratch cycle and potentially modulating inflammatory pathways involved in chronic itch.10

Nalbuphine

Nalbuphine is a κ opioid receptor agonist and μ opioid receptor antagonist that has been investigated for the treatment of PN.11 In a phase 2 randomized controlled trial, oral nalbuphine extended release (NAL-ER) 162 mg twice daily provided measurable antipruritic efficacy, with 44.4% (8/18) of patients achieving at least a 30% reduction in 7-day WI-NRS at week 10 compared with 36.4% (8/22) in the placebo group. Among those who completed the study, 66.7% (8/12) of patients receiving NAL-ER 162 mg achieved significant itch reduction vs 40% (8/20) receiving placebo (P=.03). At least a 50% reduction in WI-NRS was achieved by 33.3% (6/18) of patients receiving NAL-ER 162 mg twice daily. Extended open-label treatment was associated with further improvements in itch and lesion activity. Adverse events were mostly mild to moderate (eg, nausea, dizziness, headache, and fatigue) and occurred during dose titration. Physiologic opioid withdrawal symptoms were limited and resolved within a few days of discontinuing the medication.11

Final Thoughts

In conclusion, PN remains one of the most challenging chronic dermatologic conditions to manage and is driven by a complex interplay of neuroimmune mechanisms and resistance to many conventional therapies. The approval of dupilumab and nemolizumab has marked a pivotal shift in the therapeutic landscape, offering hope to patients who previously had limited options5,8; however, the burden of PN remains substantial, and many patients continue to experience relentless itch, poor sleep, and reduced quality of life.1 Emerging therapies such as TRPM8 agonists, Janus kinase inhibitors, and opioid modulators represent promising additions to the treatment options, targeting novel pathways beyond traditional immunosuppression.9-11

References
  1. Williams KA, Huang AH, Belzberg M, et al. Prurigo nodularis: pathogenesis and management. J Am Acad Dermatol. 2020;83:1567-1575. doi:10.1016/j.jaad.2020.04.182
  2. Gründel S, Pereira MP, Storck M, et al. Analysis of 325 patients with chronic nodular prurigo: clinics, burden of disease and course of treatment. Acta Derm Venereol. 2020;100:adv00269. doi:10.2340/00015555-3571
  3. Liao V, Cornman HL, Ma E, et al. Prurigo nodularis: new insights into pathogenesis and novel therapeutics. Br J Dermatol. 2024;190:798-810. doi:10.1093/bjd/ljae052
  4. Elmariah SB, Tao L, Valdes-Rodriguez R, et al. Individual article: management of prurigo nodularis. J Drugs Dermatol. 2023;22:SF365502s15-SF365502s22. doi:10.36849/JDD.SF365502
  5. Yosipovitch G, Mollanazar N, Ständer S, et al. Dupilumab in patients with prurigo nodularis: two randomized, double-blind, placebo-controlled phase 3 trials. Nat Med. 2023;29:1180-1190. doi:10.1038/s41591-023-02320-9
  6. Cao P, Xu W, Jiang S, et al. Dupilumab for the treatment of prurigo nodularis: a systematic review. Front Immunol. 2023;14:1092685. doi:10.3389/fimmu.2023.1092685
  7. Dagenet CB, Saadi C, Phillips MA, et al. Landscape of prurigo nodularis clinical trials. JAAD Rev. 2024;2:127-136. doi:10.1016/j.jdrv.2024.09.006
  8. Kwatra SG, Yosipovitch G, Legat FJ, et al. Phase 3 trial of nemolizumab in patients with prurigo nodularis. N Engl J Med. 2023;389:1579-1589. doi:10.1056/NEJMoa2301333
  9. Kwatra SG, Bordeaux ZA, Parthasarathy V, et al. Efficacy and safety of abrocitinib in prurigo nodularis and chronic pruritus of unknown origin: a nonrandomized controlled trial. JAMA Dermatol. 2024;160:717-724. doi:10.1001/jamadermatol.2024.1464
  10. Choi ME, Lee JH, Jung CJ, et al. A randomized, double-blinded, vehicle-controlled clinical trial of topical cryosim-1, a synthetic TRPM8 agonist, in prurigo nodularis. J Cosmet Dermatol. 2024;23:931-937. doi:10.1111/jocd.16079
  11. Weisshaar E, Szepietowski JC, Bernhard JD, et al. Efficacy and safety of oral nalbuphine extended release in prurigo nodularis: results of a phase 2 randomized controlled trial with an open-label extension phase. J Eur Acad Dermatol Venereol. 2022;36:453-461. doi:10.1111/jdv.17816
Article PDF
Author and Disclosure Information

Dr. Wan is from Inspira Medical Center Mullica Hill, New Jersey. Dr. Khachemoune (ORCID: 0000-0003-1622-1097) is from Premier Dermatology, Ashburn, Virginia, and the Department of Dermatology, Istanbul Medipol University, International School of Medicine, Istanbul, Türkiye.

The authors have no relevant financial disclosures to report.

Correspondence: Amor Khachemoune, MD 44121 Harry Byrd Hwy, Ste 210, Ashburn, VA 20147 (amorkh@gmail.com).

Cutis. 2025 September;116(3):80-81. doi:10.12788/cutis.1260

Issue
Cutis - 116(3)
Publications
Topics
Page Number
80-81
Sections
Author and Disclosure Information

Dr. Wan is from Inspira Medical Center Mullica Hill, New Jersey. Dr. Khachemoune (ORCID: 0000-0003-1622-1097) is from Premier Dermatology, Ashburn, Virginia, and the Department of Dermatology, Istanbul Medipol University, International School of Medicine, Istanbul, Türkiye.

The authors have no relevant financial disclosures to report.

Correspondence: Amor Khachemoune, MD 44121 Harry Byrd Hwy, Ste 210, Ashburn, VA 20147 (amorkh@gmail.com).

Cutis. 2025 September;116(3):80-81. doi:10.12788/cutis.1260

Author and Disclosure Information

Dr. Wan is from Inspira Medical Center Mullica Hill, New Jersey. Dr. Khachemoune (ORCID: 0000-0003-1622-1097) is from Premier Dermatology, Ashburn, Virginia, and the Department of Dermatology, Istanbul Medipol University, International School of Medicine, Istanbul, Türkiye.

The authors have no relevant financial disclosures to report.

Correspondence: Amor Khachemoune, MD 44121 Harry Byrd Hwy, Ste 210, Ashburn, VA 20147 (amorkh@gmail.com).

Cutis. 2025 September;116(3):80-81. doi:10.12788/cutis.1260

Article PDF
Article PDF

Prurigo nodularis (PN) is a chronic, severely pruritic neuroimmunologic skin disorder characterized by multiple firm hyperkeratotic nodules and intense pruritus, often leading to considerable impairment in quality of life and increased rates of depression and anxiety.1 It is considered difficult to treat due to its complex pathogenesis, the severity and chronicity of pruritus, and the limited efficacy of conventional therapies.2,3 The disease is driven by a self-perpetuating itch-scratch cycle, underpinned by dysregulation of both immune and neural pathways including type 2 (interleukin [IL] 4, IL-13, IL-31), Th17, and Th22 cytokines as well as neuropeptides and altered cutaneous nerve architecture.1,3 This results in persistent severe pruritus and nodular lesions that are highly refractory to standard treatments.1 Conventional therapies (eg, locally acting agents, phototherapy, and systemic immunomodulators and neuromodulators) have varied efficacy and notable adverse effect profiles.3 While the approval of targeted biologics has transformed the therapeutic landscape, several other treatment options also are being explored in clinical trials. Herein, we review all recently approved therapies as well as emerging treatments currently under investigation.

Dupilumab

Dupilumab, the first therapy for PN approved by the US Food and Drug Administration (FDA) in 2022—is a monoclonal antibody that inhibits signaling of IL-4 and IL-13, key drivers of type 2 inflammation implicated in PN pathogenesis.4,5 In 2 pivotal phase 3 randomized controlled trials (LIBERTY-PN PRIME and PRIME2),5 dupilumab demonstrated notable efficacy in adults with moderate to severe PN. A reduction of 4 points or more on the Worst Itch Numeric Rating Scale (WI-NRS) was achieved by 60.0% (45/75) of patients treated with dupilumab at week 24 compared with 18.4% (14/76) receiving placebo in the PRIME trial. In PRIME2, the same outcome was achieved by 37.2% (29/78) of patients receiving dupilumab at week 12 compared with 22.0% (18/82) of patients receiving placebo.5 Dupilumab also led to a greater proportion of patients achieving a substantial reduction in nodule count (5 nodules) and improved quality of life compared with placebo.5,6 The safety profile of dupilumab for treatment of PN was favorable and consistent with prior experience in atopic dermatitis; conjunctivitis was the most common adverse event.5,6

Nemolizumab

Nemolizumab, an IL-31 receptor A antagonist, is the most recent agent approved by the FDA for PN in 2024.7 In the OLYMPIA 1 and OLYMPIA 2 phase 3 trials,8 nemolizumab produced a clinically meaningful reduction in itch (defined as a 4-point improvement in the Peak Pruritus Numerical Rating Scale score) in 56.3% (103/183) of patients at week 16 compared with 20.9% (19/91) receiving placebo. Additionally, 37.7% (69/183) of patients receiving nemolizumab achieved clear or almost clear skin (Investigator’s Global Assessment score of 0 or 1 with a 2-point reduction) vs 11.0% with placebo (both P<.001). Benefits were observed as early as week 4, including rapid improvements in itch, sleep disturbance, and nodule count.8 Nemolizumab also improved quality of life and reduced symptoms of anxiety and depression. The safety profile was favorable, with headache and atopic dermatitis the most common adverse events; serious adverse events were infrequent and similar between groups.8

Abrocitinib

Abrocitinib, an oral selective Janus kinase 1 inhibitor, is an investigational therapy for PN and currently has not been approved by the FDA for this indication. In a phase 2 open-label trial, abrocitinib 200 mg daily for 12 weeks led to a 78.3% reduction in weekly Peak Pruritus Numerical Rating Scale scores in PN, with 80.0% (8/10) of patients achieving a clinically meaningful improvement of 4 points or higher. Nodule counts and quality of life also improved, with an onset of itch relief as early as week 2. The safety profile was favorable, with acneform eruptions the most common adverse event and no serious adverse events reported9; however, these results are based on small, nonrandomized studies and require confirmation in larger randomized controlled trials before abrocitinib can be considered a standard therapy for PN.

Cryosim-1

Transient receptor potential melastatin 8 (TRPM8) is a cold-sensing ion channel found in unmyelinated sensory neurons within the dorsal root and trigeminal ­ganglia.10 It is activated by cool temperatures (15-28 °C) and compounds such as menthol, leading to calcium influx and a cooling sensation. In a randomized, double-blind, vehicle-controlled trial, researchers investigated the efficacy of cryosim-1 (a synthetic TRPM8 agonist) in treating PN.10 Thirty patients were enrolled, with 18 (60.0%) receiving cryosim-1 and 12 (40.0%) receiving placebo over 8 weeks. By week 8, cryosim-1 significantly reduced itch severity (mean numerical rating scale score postapplication, 2.8 vs 4.3; P=.031) and improved sleep disturbances (2.2 vs 4.2; P=.031) compared to placebo. Patients reported higher satisfaction with itch relief, and no adverse effects were observed. The study concluded that cryosim-1 is a safe, effective topical therapy for PN, likely working by interrupting the itch-scratch cycle and potentially modulating inflammatory pathways involved in chronic itch.10

Nalbuphine

Nalbuphine is a κ opioid receptor agonist and μ opioid receptor antagonist that has been investigated for the treatment of PN.11 In a phase 2 randomized controlled trial, oral nalbuphine extended release (NAL-ER) 162 mg twice daily provided measurable antipruritic efficacy, with 44.4% (8/18) of patients achieving at least a 30% reduction in 7-day WI-NRS at week 10 compared with 36.4% (8/22) in the placebo group. Among those who completed the study, 66.7% (8/12) of patients receiving NAL-ER 162 mg achieved significant itch reduction vs 40% (8/20) receiving placebo (P=.03). At least a 50% reduction in WI-NRS was achieved by 33.3% (6/18) of patients receiving NAL-ER 162 mg twice daily. Extended open-label treatment was associated with further improvements in itch and lesion activity. Adverse events were mostly mild to moderate (eg, nausea, dizziness, headache, and fatigue) and occurred during dose titration. Physiologic opioid withdrawal symptoms were limited and resolved within a few days of discontinuing the medication.11

Final Thoughts

In conclusion, PN remains one of the most challenging chronic dermatologic conditions to manage and is driven by a complex interplay of neuroimmune mechanisms and resistance to many conventional therapies. The approval of dupilumab and nemolizumab has marked a pivotal shift in the therapeutic landscape, offering hope to patients who previously had limited options5,8; however, the burden of PN remains substantial, and many patients continue to experience relentless itch, poor sleep, and reduced quality of life.1 Emerging therapies such as TRPM8 agonists, Janus kinase inhibitors, and opioid modulators represent promising additions to the treatment options, targeting novel pathways beyond traditional immunosuppression.9-11

Prurigo nodularis (PN) is a chronic, severely pruritic neuroimmunologic skin disorder characterized by multiple firm hyperkeratotic nodules and intense pruritus, often leading to considerable impairment in quality of life and increased rates of depression and anxiety.1 It is considered difficult to treat due to its complex pathogenesis, the severity and chronicity of pruritus, and the limited efficacy of conventional therapies.2,3 The disease is driven by a self-perpetuating itch-scratch cycle, underpinned by dysregulation of both immune and neural pathways including type 2 (interleukin [IL] 4, IL-13, IL-31), Th17, and Th22 cytokines as well as neuropeptides and altered cutaneous nerve architecture.1,3 This results in persistent severe pruritus and nodular lesions that are highly refractory to standard treatments.1 Conventional therapies (eg, locally acting agents, phototherapy, and systemic immunomodulators and neuromodulators) have varied efficacy and notable adverse effect profiles.3 While the approval of targeted biologics has transformed the therapeutic landscape, several other treatment options also are being explored in clinical trials. Herein, we review all recently approved therapies as well as emerging treatments currently under investigation.

Dupilumab

Dupilumab, the first therapy for PN approved by the US Food and Drug Administration (FDA) in 2022—is a monoclonal antibody that inhibits signaling of IL-4 and IL-13, key drivers of type 2 inflammation implicated in PN pathogenesis.4,5 In 2 pivotal phase 3 randomized controlled trials (LIBERTY-PN PRIME and PRIME2),5 dupilumab demonstrated notable efficacy in adults with moderate to severe PN. A reduction of 4 points or more on the Worst Itch Numeric Rating Scale (WI-NRS) was achieved by 60.0% (45/75) of patients treated with dupilumab at week 24 compared with 18.4% (14/76) receiving placebo in the PRIME trial. In PRIME2, the same outcome was achieved by 37.2% (29/78) of patients receiving dupilumab at week 12 compared with 22.0% (18/82) of patients receiving placebo.5 Dupilumab also led to a greater proportion of patients achieving a substantial reduction in nodule count (5 nodules) and improved quality of life compared with placebo.5,6 The safety profile of dupilumab for treatment of PN was favorable and consistent with prior experience in atopic dermatitis; conjunctivitis was the most common adverse event.5,6

Nemolizumab

Nemolizumab, an IL-31 receptor A antagonist, is the most recent agent approved by the FDA for PN in 2024.7 In the OLYMPIA 1 and OLYMPIA 2 phase 3 trials,8 nemolizumab produced a clinically meaningful reduction in itch (defined as a 4-point improvement in the Peak Pruritus Numerical Rating Scale score) in 56.3% (103/183) of patients at week 16 compared with 20.9% (19/91) receiving placebo. Additionally, 37.7% (69/183) of patients receiving nemolizumab achieved clear or almost clear skin (Investigator’s Global Assessment score of 0 or 1 with a 2-point reduction) vs 11.0% with placebo (both P<.001). Benefits were observed as early as week 4, including rapid improvements in itch, sleep disturbance, and nodule count.8 Nemolizumab also improved quality of life and reduced symptoms of anxiety and depression. The safety profile was favorable, with headache and atopic dermatitis the most common adverse events; serious adverse events were infrequent and similar between groups.8

Abrocitinib

Abrocitinib, an oral selective Janus kinase 1 inhibitor, is an investigational therapy for PN and currently has not been approved by the FDA for this indication. In a phase 2 open-label trial, abrocitinib 200 mg daily for 12 weeks led to a 78.3% reduction in weekly Peak Pruritus Numerical Rating Scale scores in PN, with 80.0% (8/10) of patients achieving a clinically meaningful improvement of 4 points or higher. Nodule counts and quality of life also improved, with an onset of itch relief as early as week 2. The safety profile was favorable, with acneform eruptions the most common adverse event and no serious adverse events reported9; however, these results are based on small, nonrandomized studies and require confirmation in larger randomized controlled trials before abrocitinib can be considered a standard therapy for PN.

Cryosim-1

Transient receptor potential melastatin 8 (TRPM8) is a cold-sensing ion channel found in unmyelinated sensory neurons within the dorsal root and trigeminal ­ganglia.10 It is activated by cool temperatures (15-28 °C) and compounds such as menthol, leading to calcium influx and a cooling sensation. In a randomized, double-blind, vehicle-controlled trial, researchers investigated the efficacy of cryosim-1 (a synthetic TRPM8 agonist) in treating PN.10 Thirty patients were enrolled, with 18 (60.0%) receiving cryosim-1 and 12 (40.0%) receiving placebo over 8 weeks. By week 8, cryosim-1 significantly reduced itch severity (mean numerical rating scale score postapplication, 2.8 vs 4.3; P=.031) and improved sleep disturbances (2.2 vs 4.2; P=.031) compared to placebo. Patients reported higher satisfaction with itch relief, and no adverse effects were observed. The study concluded that cryosim-1 is a safe, effective topical therapy for PN, likely working by interrupting the itch-scratch cycle and potentially modulating inflammatory pathways involved in chronic itch.10

Nalbuphine

Nalbuphine is a κ opioid receptor agonist and μ opioid receptor antagonist that has been investigated for the treatment of PN.11 In a phase 2 randomized controlled trial, oral nalbuphine extended release (NAL-ER) 162 mg twice daily provided measurable antipruritic efficacy, with 44.4% (8/18) of patients achieving at least a 30% reduction in 7-day WI-NRS at week 10 compared with 36.4% (8/22) in the placebo group. Among those who completed the study, 66.7% (8/12) of patients receiving NAL-ER 162 mg achieved significant itch reduction vs 40% (8/20) receiving placebo (P=.03). At least a 50% reduction in WI-NRS was achieved by 33.3% (6/18) of patients receiving NAL-ER 162 mg twice daily. Extended open-label treatment was associated with further improvements in itch and lesion activity. Adverse events were mostly mild to moderate (eg, nausea, dizziness, headache, and fatigue) and occurred during dose titration. Physiologic opioid withdrawal symptoms were limited and resolved within a few days of discontinuing the medication.11

Final Thoughts

In conclusion, PN remains one of the most challenging chronic dermatologic conditions to manage and is driven by a complex interplay of neuroimmune mechanisms and resistance to many conventional therapies. The approval of dupilumab and nemolizumab has marked a pivotal shift in the therapeutic landscape, offering hope to patients who previously had limited options5,8; however, the burden of PN remains substantial, and many patients continue to experience relentless itch, poor sleep, and reduced quality of life.1 Emerging therapies such as TRPM8 agonists, Janus kinase inhibitors, and opioid modulators represent promising additions to the treatment options, targeting novel pathways beyond traditional immunosuppression.9-11

References
  1. Williams KA, Huang AH, Belzberg M, et al. Prurigo nodularis: pathogenesis and management. J Am Acad Dermatol. 2020;83:1567-1575. doi:10.1016/j.jaad.2020.04.182
  2. Gründel S, Pereira MP, Storck M, et al. Analysis of 325 patients with chronic nodular prurigo: clinics, burden of disease and course of treatment. Acta Derm Venereol. 2020;100:adv00269. doi:10.2340/00015555-3571
  3. Liao V, Cornman HL, Ma E, et al. Prurigo nodularis: new insights into pathogenesis and novel therapeutics. Br J Dermatol. 2024;190:798-810. doi:10.1093/bjd/ljae052
  4. Elmariah SB, Tao L, Valdes-Rodriguez R, et al. Individual article: management of prurigo nodularis. J Drugs Dermatol. 2023;22:SF365502s15-SF365502s22. doi:10.36849/JDD.SF365502
  5. Yosipovitch G, Mollanazar N, Ständer S, et al. Dupilumab in patients with prurigo nodularis: two randomized, double-blind, placebo-controlled phase 3 trials. Nat Med. 2023;29:1180-1190. doi:10.1038/s41591-023-02320-9
  6. Cao P, Xu W, Jiang S, et al. Dupilumab for the treatment of prurigo nodularis: a systematic review. Front Immunol. 2023;14:1092685. doi:10.3389/fimmu.2023.1092685
  7. Dagenet CB, Saadi C, Phillips MA, et al. Landscape of prurigo nodularis clinical trials. JAAD Rev. 2024;2:127-136. doi:10.1016/j.jdrv.2024.09.006
  8. Kwatra SG, Yosipovitch G, Legat FJ, et al. Phase 3 trial of nemolizumab in patients with prurigo nodularis. N Engl J Med. 2023;389:1579-1589. doi:10.1056/NEJMoa2301333
  9. Kwatra SG, Bordeaux ZA, Parthasarathy V, et al. Efficacy and safety of abrocitinib in prurigo nodularis and chronic pruritus of unknown origin: a nonrandomized controlled trial. JAMA Dermatol. 2024;160:717-724. doi:10.1001/jamadermatol.2024.1464
  10. Choi ME, Lee JH, Jung CJ, et al. A randomized, double-blinded, vehicle-controlled clinical trial of topical cryosim-1, a synthetic TRPM8 agonist, in prurigo nodularis. J Cosmet Dermatol. 2024;23:931-937. doi:10.1111/jocd.16079
  11. Weisshaar E, Szepietowski JC, Bernhard JD, et al. Efficacy and safety of oral nalbuphine extended release in prurigo nodularis: results of a phase 2 randomized controlled trial with an open-label extension phase. J Eur Acad Dermatol Venereol. 2022;36:453-461. doi:10.1111/jdv.17816
References
  1. Williams KA, Huang AH, Belzberg M, et al. Prurigo nodularis: pathogenesis and management. J Am Acad Dermatol. 2020;83:1567-1575. doi:10.1016/j.jaad.2020.04.182
  2. Gründel S, Pereira MP, Storck M, et al. Analysis of 325 patients with chronic nodular prurigo: clinics, burden of disease and course of treatment. Acta Derm Venereol. 2020;100:adv00269. doi:10.2340/00015555-3571
  3. Liao V, Cornman HL, Ma E, et al. Prurigo nodularis: new insights into pathogenesis and novel therapeutics. Br J Dermatol. 2024;190:798-810. doi:10.1093/bjd/ljae052
  4. Elmariah SB, Tao L, Valdes-Rodriguez R, et al. Individual article: management of prurigo nodularis. J Drugs Dermatol. 2023;22:SF365502s15-SF365502s22. doi:10.36849/JDD.SF365502
  5. Yosipovitch G, Mollanazar N, Ständer S, et al. Dupilumab in patients with prurigo nodularis: two randomized, double-blind, placebo-controlled phase 3 trials. Nat Med. 2023;29:1180-1190. doi:10.1038/s41591-023-02320-9
  6. Cao P, Xu W, Jiang S, et al. Dupilumab for the treatment of prurigo nodularis: a systematic review. Front Immunol. 2023;14:1092685. doi:10.3389/fimmu.2023.1092685
  7. Dagenet CB, Saadi C, Phillips MA, et al. Landscape of prurigo nodularis clinical trials. JAAD Rev. 2024;2:127-136. doi:10.1016/j.jdrv.2024.09.006
  8. Kwatra SG, Yosipovitch G, Legat FJ, et al. Phase 3 trial of nemolizumab in patients with prurigo nodularis. N Engl J Med. 2023;389:1579-1589. doi:10.1056/NEJMoa2301333
  9. Kwatra SG, Bordeaux ZA, Parthasarathy V, et al. Efficacy and safety of abrocitinib in prurigo nodularis and chronic pruritus of unknown origin: a nonrandomized controlled trial. JAMA Dermatol. 2024;160:717-724. doi:10.1001/jamadermatol.2024.1464
  10. Choi ME, Lee JH, Jung CJ, et al. A randomized, double-blinded, vehicle-controlled clinical trial of topical cryosim-1, a synthetic TRPM8 agonist, in prurigo nodularis. J Cosmet Dermatol. 2024;23:931-937. doi:10.1111/jocd.16079
  11. Weisshaar E, Szepietowski JC, Bernhard JD, et al. Efficacy and safety of oral nalbuphine extended release in prurigo nodularis: results of a phase 2 randomized controlled trial with an open-label extension phase. J Eur Acad Dermatol Venereol. 2022;36:453-461. doi:10.1111/jdv.17816
Issue
Cutis - 116(3)
Issue
Cutis - 116(3)
Page Number
80-81
Page Number
80-81
Publications
Publications
Topics
Article Type
Display Headline

From Refractory to Responsive: The Expanding Therapeutic Landscape of Prurigo Nodularis

Display Headline

From Refractory to Responsive: The Expanding Therapeutic Landscape of Prurigo Nodularis

Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Thu, 09/04/2025 - 17:58
Un-Gate On Date
Thu, 09/04/2025 - 17:58
Use ProPublica
CFC Schedule Remove Status
Thu, 09/04/2025 - 17:58
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
survey writer start date
Thu, 09/04/2025 - 17:58

Type VII Collagen Disorders Simplified

Article Type
Changed
Thu, 09/04/2025 - 16:47
Display Headline

Type VII Collagen Disorders Simplified

There are 3 uncommon types of mechanobullous skin diseases caused by relative reduction or complete loss of functional type VII collagen, which is the main component of anchoring fibrils in the lamina densa of the basement membrane zone (BMZ) of the skin and mucous membrane epithelium.1 The function of the anchoring fibrils is to maintain adherence of the basement membrane of the epithelium to the connective tissue of the papillary dermis and submucosa.1 The mechanism of action of the loss of type VII collagen function is via autoimmunity in epidermolysis bullosa acquisita (EBA)2 and bullous systemic lupus erythematosus (BSLE).3 In the heritable family of 4 epidermolysis bullosa (EB) variants, only one of the subtypes—dystrophic EB (DEB)—is caused by various recessive and dominant mutations of the type VII collagen gene (COL7A1).4 The other 3 diseases in the family—EB simplex, junctional EB, and Kindler syndrome—are caused by diverse mutations that corrupt the integrity of keratinocytes and the BMZ.5,6 This article provides an overview of these 3 subtypes to help differentiate them from DEB.

Epidermolysis Bullosa

Epidermolysis bullosa consists of a heterogeneous family of 4 major genetic mechanobullous diseases that affect the skin and mucous membranes with more than 30 subtypes.1 Dystrophic EB is caused by mutations in the COL7A1 gene, which encodes for the α-1 chain of collagen type VII. Classically, EB is divided into 4 main variants based on the location of the cleavage plane or split occurring in the epithelium, which in turn helps to predict the severity of the illness.

Epidermolysis bullosa may be inherited in an ­autosomal-dominant or autosomal-recessive fashion, or it may occur as a spontaneous mutation. All sexes and races are affected equally. Patients present at birth or in early childhood with fragile skin and mucous membranes that may develop blisters, erosions, and ulcerations after minor trauma.7 These lesions are marked by slow healing and scar formation and often are associated with itching and pain.

Dystrophic Epidermolysis Bullosa

Dystrophic EB accounts for approximately 25%6 of all EB cases in the United States and may be inherited as either a dominant or recessive trait. Hundreds of different pathogenic mutations have been discovered in the COL7A1 gene in the subtypes of DEB.4,8 Dominant DEB tends to cause milder disease because the patients retain one normal COL7A1 allele and produce some type VII collagen (Figure 1), whereas patients with recessive DEB lack type VII collagen completely.9 The cleavage plane is between the lamina densa and the superficial dermis or submucosa. Severity is variable and ranges from ­localization to the hands and feet to severe generalized blistering and painful ulcerations depending on which of the many possible gene mutations have been inherited. Sequelae include mitten deformities, malalignment and tooth decay, and the development of early aggressive squamous cell carcinomas, which may be fatal. The most severe cases of recessive DEB also may have internal organ involvement.

Camisa-1
FIGURE 1. Bulla on the ankle of an infant and scarring on the hands and severe nail dystrophy with nail loss in a parent with autosomal-dominant dystrophic epidermolysis bullosa.

Epidermolysis Bullosa Simplex

Epidermolysis bullosa simplex is the most common variant, comprising approximately 70%of EB cases in the United States.6 Epidermolysis bullosa simplex usually is inherited as autosomal-dominant mutations in the keratin 5 or keratin 14 genes,10 not COL7A1. Skin blistering results from cleavage within the basal cell layer where the keratin genes are primarily expressed. Blisters tend to occur in acral areas such as hands and feet and may heal without scarring in the localized form of epidermolysis bullosa simplex (Figure 2).

Camisa-2
FIGURE 2. Epidermolysis bullosa simplex in a child with healing blisters localized to the hands and wrist.

Junctional Epidermolysis Bullosa and Kindler Syndrome

Junctional epidermolysis bullosa (JEB) and Kindler syndrome11 are the rarest of the autosomal-recessive EB ­variants.6 The plane of cleavage in JEB is through the lamina lucida of the BMZ. Junctional epidermolysis bullosa is caused by mutations of the genes that encode for the 3 chains of laminin 332 protein and type XVII collagen,5,12 not to be confused with type VII collagen. As with DEB, there is a wide range of severity in JEB, from localized effects on the eyes, oral cavity, and tooth enamel to widespread blistering and skin cancers. In JEB cases involving newborns, nonhealing wounds on the face, buttocks, fingers, and toes may be seen, with devastating complications in the oral cavity, esophagus, and larynx. Life expectancy is limited to 2 years or less.6 There have only been approximately 40,013 cases of Kindler syndrome reported worldwide6 and there is clinical overlap with DEB. Patients also may demonstrate poikiloderma and photosensitivity. Kindler syndrome is caused by mutations in the FERMT1 gene which encodes for kindlin-1. This protein mediates anchorage between the actin cytoskeleton and the extracellular matrix.5,11 Loss of function produces variable cleavage planes around the dermoepidermal junction.

Clinical management of all EB variants, especially the severe recessive types, traditionally has been limited to the prevention of trauma to the skin and mucous membranes and supportive care, including dressing changes to erosions and ulcerations, antibiotic ointments as needed, and amelioration of pain and pruritus. Bone marrow and pluripotential stem cell transplants have been attempted.12 Complications of EB, such as deformities of the hands and feet caused by excessive scarring, esophageal strictures, poor dentition, and squamous cell carcinomas, must be addressed by a multidisciplinary team of specialists, including plastic surgery, gastroenterology, dentistry/oral surgery, ophthalmology, and dermatology/Mohs surgery. 

Until recently, there were no medications approved by the US Food and Drug Administration (FDA) specifically indicated for EB. In 2023, topical gene therapy was approved by the FDA for both recessive and dominant forms of DEB. Normal COL7A1 sequences are delivered by an attenuated herpes simplex virus 1 vector (beremagene geperpavec) in a gel applied directly to the wounds of patients with DEB. In a clinical trial, matching wounds on 31 patients (62 wounds total) were treated with the active agent or placebo gel. After 6 months, complete wound closure was observed in 67% (21/31) of those treated with the active agent and 22% (7/31) of those treated with placebo (P=.002).14 In a single case report, a patient with recessive DEB and cicatrizing conjunctivitis (Figure 3) was given ophthalmic beremagene geperpavec after surgery and had improved visual acuity.15 A topical gel consisting of birch triterpenes to promote healing of partial-thickness wounds also was approved for patients with DEB and JEB by the FDA and the European Commission. In a study of 223 patients, 41% of those using active gel and 29% of those using placebo gel achieved the primary end point of percentage of target wounds that had first complete closure at 45 days.16 

Camisa-3
FIGURE 3. Recessive dystrophic epidermolysis bullosa complicated by cicatrizing conjunctivitis in a teen who underwent successful surgery and ophthalmic gene therapy to restore his sight. Photograph courtesy of Alfonso L. Sabater, MD (Miami, Florida).


The most recent FDA approval for DEB involves transferring the functional COL7A1 gene to the patient’s skin cells, then expanding the gene-corrected cells into sheets of keratinocytes that can be surgically applied to the chronic wound sites. In a phase 3 trial of prademagene zamikeracel (pz-cel), 11 patients with 86 matched wounds were randomized to receive pz-cel (50%) or standard wound care (50%). After 24 weeks, 35 wounds treated with pz-cel were at least 50% healed compared to 7 control wounds.17 The results for healing and reduction of pain were statistically significant (P<.0001 and P<.0002, respectively).17 Recombinant collagen VII as replacement therapy also is under study to be given by intravenous infusion to increase tissue collagen VII where it is lacking. This treatment has shown early biologic and therapeutic effects.9,18 Larger long-term follow-up studies are necessary to confirm persistence of the gene-corrected skin cells, the functionality of the replacement collagen VII, and the potential risk for the development of autoantibodies to type VII collagen.

Epidermolysis Bullosa Acquisita

Epidermolysis bullosa acquisita is a rare autoimmune subepithelial bullous disease that primarily affects middle-aged adults but also has been reported in children.19 Epidermolysis bullosa acquisita is caused by circulating pathogenic IgG autoantibodies that target and bind to type VII collagen in the anchoring fibrils,20-22 thereby disrupting the attachment of the epithelium to its underlying connective tissue.

The 2 major clinical manifestations of EBA include a mechanobullous disease resembling inherited forms of DEB (Figure 4) and an inflammatory bullous pemphigoid (BP)–like disease,23 as well as a combination of both types of skin lesions (Figure 5). The skin and mucous membranes of the oral cavity, esophagus, eyes, and urogenital areas are affected in both types; scarring may cause functional disabilities. In the mechanobullous type of EBA, it is common for blisters and erosions to develop in trauma-prone areas such as the hands, feet, elbows, and knees. The blisters tend to heal with scarring and milia formation as might be seen in porphyria cutanea tarda or cicatricial pemphigoid, which are in the differential diagnosis. Dystrophy of the fingernails or complete nail loss may be observed, resembling DEB. In the BP-like presentation, tense blisters arise upon inflamed or urticarial skin and mucous membranes, which may then become generalized. 

Camisa-4
FIGURE 4. Epidermolysis bullosa acquisita (mechanobullous type) of the hands shows small blisters, scarring, and erosions of the lateral nail folds.

 

Camisa-5
FIGURE 5. Epidermolysis bullosa acquisita with severe ulcerations on the neck and back secondary to blisters with scarring.


Histopathology in both forms of EBA demonstrates subepithelial separation as clefts or blisters. The mechanobullous type shows a sparse inflammatory infiltrate compared to large collections of neutrophils and eosinophils in the blister cavity and in the superficial dermis in the BP-like cases. The final diagnosis rests on the results of immunopathology testing.24 Direct immunofluorescence of perilesional skin and mucosa shows a linear-granular band of IgG and C3 and other conjugates along the BMZ. Deposits of IgA alone in EBA occur in only about 2.4% of cases and are observed more often when there is mucous membrane involvement.2 Indirect immunofluorescence of sera against salt-split skin substrates detects immunoreactants in the floor of the blister rather than in the roof, as would be seen in BP. Highly specific and sensitive enzyme-linked immunosorbent assay (ELISA) kits now are commercially available and can detect autoantibodies against the N-terminal domain of type VII collagen in more than 90% of cases of EBA.25 

Inflammatory bowel disease (IBD), particularly Crohn disease (CD), precedes the onset of EBA in approximately 25% of cases.26,27 Ulcerative colitis is much less common. Type VII collagen is normally present in the basement membrane of intestinal epithelium. In a survey of patients with IBD, 68% of those with CD and 13% of those with ulcerative colitis had circulating anti–type VII collagen antibodies detected by ELISA without having symptoms of EBA.28 A case report of a patient with both well-proven EBA and CD highlighted the clinical difficulty of controlling EBA: treatment with prednisolone and sulfasalazine improved the CD but had little effect on the skin blisters.29 A variety of malignancies have been reported in association with EBA, including cancers of the uterine cervix,30 thyroid, and pancreas,31 lymphoma, and chronic lymphatic leukemia. Some of these cases have met the criteria for classification as paraneoplastic, whereas others may have been coincidental. 

Treatment for chronic EBA generally has been limited.2,24 Putative antineutrophil drugs such as dapsone and colchicine combined with systemic corticosteroids may be useful in milder or juvenile cases, which tend to have a better prognosis than adult cases.19 In more severe EBA, systemic corticosteroids and/or immunosuppressive drugs such as azathioprine,23 cyclophosphamide,23 mycophenolate mofetil,31 methotrexate,23 cyclosporine,33 and infliximab23 have been used. More recently, rituximab infusion monotherapy33 and rituximab combined with intravenous immunoglobulin or adjuvant immunoadsorption of the pathogenic autoantibodies have induced remission of refractory EBA.32 Adjuvant immunoadsorption therapy is not widely available. Multispecialty care often is required, especially ophthalmology for conjunctival involvement and gastroenterology for potential esophageal stenosis and the early detection and treatment of IBD.

Bullous Systemic Lupus Erythematosus

Bullous systemic lupus erythematosus is a rare and specific autoimmune skin complication that mostly is seen in patients with an established diagnosis of systemic lupus erythematosus (SLE) who are experiencing a disease flare. Although more common in women, it has been reported in all sexes and races as well as in children. Vesicles and bullae may arise on sun-exposed (Figure 6) and sun-protected areas of skin.

Camisa-6
FIGURE 6. Bullous systemic lupus erythematosus demonstrates active vesicles and bullae on a sun-exposed area of the wrist.

Histopathology shows subepidermal separation with collections of neutrophils and nuclear fragments in the blister cavity. The differential diagnosis of BSLE includes EBA, BP, dermatitis herpetiformis, and linear IgA bullous dermatosis. Direct immunofluorescence testing shows linear-granular deposits of IgG and/or IgM and IgA along the BMZ.34 When utilizing the indirect immunofluorescence split-skin assay, the autoantibody to type VII collagen would be detected in the floor of the blister if the serum titer was sufficiently high.3 Proposed criteria for the diagnosis of BSLE have been published: 1) diagnosis of SLE now based on the 2019 European League Against Rheumatism/American College of Rheumatology classification35; 2) vesicles and bullae arising upon but not limited to sun-exposed skin; 3) histopathology featuring neutrophil-rich subepithelial bullae; 4) positive indirect immunofluorescence for circulating BMZ antibodies using separated human skin as substrate; 5) and direct immunofluorescence showing IgG and/or IgM and often IgA at the BMZ.36 Using ELISA to detect circulating antibodies against type VII collagen24 should now be added to the criteria. The new criteria for SLE34 do not include BSLE, perhaps because it occurs in less than 1% of patients with SLE.37 

Further investigation by Gammon et al3 confirmed that the autoantibodies in BSLE are identical to those found in EBA (ie, directed against type VII collagen in the lamina densa). Bullous systemic lupus erythematosus is not considered to be the coexistence of EBA with SLE but rather a specific entity wherein type VII collagen autoantibodies are expressed in the autoimmune spectrum of SLE. It is especially important to make the diagnosis of BSLE because it is predictive of more serious systemic complications of SLE (eg, hematologic and renal disease is found in up to 90% of cases).38 

The natural course of BSLE is variable. Treatments include systemic corticosteroids, dapsone, and immunosuppressive drugs such as azathioprine, methotrexate, mycophenolate mofetil, and cyclophosphamide, especially in cases with nephritis.37 There may be spontaneous resolution of the rash as the inflammatory activity of SLE subsides. Rituximab has been used effectively in several refractory cases of BSLE that failed to respond to all other conventional treatments.39

Conclusion

Anchoring fibrils are composed primarily of type VII collagen. Their role is to maintain the attachment of epithelium to the upper dermis and submucosa. The reduction or complete loss of type VII collagen caused by mutations of the COL7A1 gene results in dominant DEB or recessive DEB, respectively. Two distinct non-heritable immunobullous diseases, EBA and BSLE, are caused by autoantibodies that target type VII collagen. A comparison of the 4 type VII collagen disorders can be found in the Table.

CT116002012_e-Table_part1CT116002012_e-Table_part2
References
  1. Bardhan A, Bruckner-Tuderman L, Chapple ILC, et al. Epidermolysis bullosa. Nat Rev Dis Primers. 2020;6:78. doi:10.1038/s41572-020-0210-0
  2. Miyamoto D, Gordilho JO, Santi CG, et al. Epidermolysis bullosa acquisita. An Bras Dermatol. 2022;97:409-423. doi:10.1016/j.abd.2021.09.010.
  3. Gammon WR, Woodley DT, Dole KC, et al. Evidence that anti-basement membrane zone antibodies in bullous eruption of systemic lupus erythematosus recognize epidermolysis bullosa acquisita autoantigen. J Invest Dermatol. 1985;84:472-476. doi:10.1111/1523-1747.ep12272402.
  4. Yadav RS, Jaswal A, Shrestha S, et al. Dystrophic epidermolysis bullosa. J Nepal Med Assoc. 2018;56:879-882. doi:10.31729/jnma.3791
  5. Mariath LM, Santin JT, Schuler-Faccini L, et al. Inherited epidermolysis bullosa: update on the clinical and genetic aspects. An Bras Dermatol. 2020;95:551-569. doi:10.1016/j.abd.2020.05.001
  6. Understanding epidermolysis bullosa (EB). DEBRA website. Accessed August 17, 2025. https://www.debra.org/about-eb/understanding-epidermolysis-bullosa-eb
  7. Hon KL, Chu S, Leung AKC. Epidermolysis bullosa: pediatric perspectives. Curr Pediatr Rev. 2022;18:182-190. doi:10.2174/1573396317666210525161252
  8. Dang N, Klingberg S, Marr P, et al. Review of collagen VII sequence variants found in Australasian patients with dystrophic epidermolysis bullosa reveals nine COL7A1 variants. J Dermatol Sci. 2007;46:169-178. doi:10.1016/j.jdermsci.2007.02.006
  9. Payne AS. Topical gene therapy for epidermolysis bullosa. N Engl J Med. 2022;387:2281-2284. doi:10.1056/NEJMe2213203
  10. Khani P, Ghazi F, Zekri A, et al. Keratins and epidermolysis bullosa simplex. J Cell Physiol. 2018;234:289-297. doi:10.1002/jcp.26898
  11. Lai-Cheong JE, Tanaka A, Hawche G, et al. Kindler syndrome: a focal adhesion genodermatosis. Br J Dermatol. 2009;160:233-242. doi:10.1111/j.1365-2133.2008.08976.x
  12. Hou P-C, Wang H-T, Abhee S, et al. Investigational treatments for epidermolysis bullosa. Am J Clin Dermatol. 2021;22:801-817. doi:10.1007/s40257-021-00626-3
  13. Youseffian L, Vahidnezhad H, Uitto J. Kindler Syndrome. GeneReviews [Internet]. Updated January 6, 2022. Accessed August 21, 2025.
  14. Guide SV, Gonzalez ME, Bagci S, et al. Trial of beremagene geperpavec (B-VEC) for dystrophic epidermolysis bullosa. N Engl J Med. 2022;387:2211-2219. doi:10.1056/NEJMoa2206663
  15. Vetencourt AT, Sayed-Ahmed I, Gomez J, et al. Ocular gene therapy in a patient with dystrophic epidermolysis bullosa. N Engl J Med. 2024;390:530-535. doi:10.1056/NEJMoa2301244
  16. Kern JS, Sprecher E, Fernandez MF, et al. Efficacy and safety of Oleogel-S10 (birch triterpenes for epidermolysis bullosa: results from the phase III randomized double-blind phase of the EASE study. Br J Dermatol. 2023;188:12-21. doi:10.1093/bjd/ljac001
  17. Tang JY, Marinkovich MP, Wiss K, et al. Prademagene zamikeracel for recessive dystrophic epidermolysis bullosa wounds (VIITAL): a two-centre, randomized, open-label, intrapatient-controlled phase 3 trial. Lancet. 2025;406:163-173. doi:10.1016/S0140-6736(25)00778-0
  18. Gretzmeier C, Pin D, Kern JS, et al. Systemic collagen VII replacement therapy for advanced recessive dystrophic epidermolysis bullosa. J Invest Dermatol. 2022;142:1094-1102. doi:10.1016/j.jid.2021.09.008
  19. Hignett E, Sami N. Pediatric epidermolysis bullosa acquisita. A review. Pediatr Dermatol. 2021;38:1047-1050. doi:10.1111/pde.14722
  20. Chen M, Kim GH, Prakash L, et al. Autoimmunity to anchoring fibril collagen. Autoimmunity. 2012;45:91-101. doi:10.1007/s12016-007-0027-6.
  21. Kridin K, Kneiber D, Kowalski EH, et al. Epidermolysis bullosa acquisita: a comprehensive review. Autoimmun Rev. 2019;18:786-795. doi:10.1016/j.autrev.2019.06.007
  22. Hofmann SC, Weidinger A. Epidermolysis bullosa acquisita. Hautarzt. 2019;70:265-270. doi:10.1007/s00105-019-4387-7
  23. Ishi N, Hamada T, Dainichi T, et al. Epidermolysis bullosa acquisita: what’s new? J Dermatol. 2010;37:220-230. doi:10.1111/j.1346-8138.2009.00799.x
  24. Iwata H, Vorobyev A, Koga H, et al. Meta-analysis of the clinical and immunopathological characteristics and treatment outcomes in epidermolysis bullosa acquisita patients. Orphanet J Rare Dis. 2018;13:153. doi:10.1186/s13023-018-0896-1
  25. Komorowski L, Muller R, Vorobyev A, et al. Sensitive and specific assays for routine serological diagnosis of epidermolysis bullosa acquisita. J Am Acad Dermatol. 2013;68:e89-95. doi:10.1016/j.jaad.2011.12.032
  26. Antonelli E, Bassotti G, Tramontana M, et al. Dermatological manifestations in inflammatory bowel diseases. J Clin Med. 2021;10:364-390. doi:10.3390/jcm10020364
  27. Bezzio C, Della Corte C, Vernero M, et al. Inflammatory bowel disease and immune-mediated inflammatory diseases: looking at less frequent associations. Therap Adv Gastroenterol. 2022;15:17562848221115312. doi:10.1177/17562848221115312
  28. Chen M, O’Toole EA, Sanghavi J, et al. The epidermolysis acquisita antigen (type VII collagen) is present in human colon and patients with Crohn’s disease have antibodies to type VII collagen. J Invest Dermatol. 2002;118:1059-1064. doi:10.1046/j.1523-1747.2002.01772.x
  29. Labeille B, Gineston JL, Denoeux JP, et al. Epidermolysis bullosa acquisita and Crohn’s disease. A case report with immunological and electron microscopic studies. Arch Intern Med. 1988;148:1457-1459.
  30. Etienne A, Ruffieux P, Didierjean L, et al. Epidermolysis bullosa acquisita and metastatic cancer of the uterine cervix. Ann Dermatol Venereol. 1998;125:321-323.
  31. Busch J-O, Sticherling M. Epidermolysis bullosa acquisita and neuroendocrine pancreatic cancer-Coincidence or patho-genetic relationship? J Dtsch Dermatol Ges. 2007;5:916-918. doi:10.111/j.1610-0387.2007.06338.x
  32. Bevans SL, Sami N. The use of rituximab in treatment of epidermolysis bullosa acquisita: three new cases and a review of the literature. Dermatol Ther. 2018;31:e12726. doi:10.1111/j.1610-0387.2007.06338.x
  33. Yang A, Kim M, Craig P, et al. A case report of the use of rituximab and the epidermolysis bullosa disease activity scoring index (EBDASI) in a patient with epidermolysis bullosa acquisita with extensive esophageal involvement. Arch Dermatovenerol Croat. 2018;26:325-328.
  34. Burrows NP, Bhogal BS, Black MM, et al. Bullous eruption of systemic lupus erythematosus: a clinicopathological study of four cases. Br J Dermatol. 1993;128:332-338. doi:10.1111/j.1365-2133.1993.tb00180.x
  35. Aringer M, Leuchten N, Johnson SR. New criteria for lupus. Curr Rheum Rep. 2020;22:18. doi:10.1007/s11926-020-00896-6
  36. Camisa C. Vesiculobullous systemic lupus erythematosus. A report of four cases. J Am Acad Dermatol. 1988;18:93-100. doi:10.1016/s0190-9622(88)70014-6
  37. Duan L, Chen L, Zhong S, et al. Treatment of bullous systemic lupus erythematosus. J Immunol Res. 2015;2015:167064. doi:10.1155/2015/167064
  38. Sprow G, Afarideh M, Dan J, et al. Bullous systemic lupus erythematosus in females. Int J Womens Dermatol. 2022;8:e034. doi:10.1097/JW9.0000000000000034
  39. Contestable JJ, Edhegard KD, Meyerle JH. Bullous systemic lupus erythematosus: a review and update to diagnosis and treatment. Am J Clin Dermatol. 2014;15:517-524. doi:10.1007/s40257-014-0098-0
  40. Fine JD, Mellerio JE. Epidermolysis bullosa. In: Bolognia JL, Jorizzo JL, Schaffer JV (eds), Dermatology (ed 3), Elsevier Saunders; 2012: 501-513.
Article PDF
Author and Disclosure Information

From OnSpot Dermatology, Naples, Florida, and Riverchase Dermatology, Fort Myers, Florida.

Dr. Camisa has no relevant financial disclosures to report.

Correspondence: Charles Camisa, MD (drcamisa@gmail.com).

Cutis. 2025 August;116(2):E12-E19. doi:10.12788/cutis.1267

Issue
Cutis - 116(2)
Publications
Topics
Page Number
E12-E19
Sections
Author and Disclosure Information

From OnSpot Dermatology, Naples, Florida, and Riverchase Dermatology, Fort Myers, Florida.

Dr. Camisa has no relevant financial disclosures to report.

Correspondence: Charles Camisa, MD (drcamisa@gmail.com).

Cutis. 2025 August;116(2):E12-E19. doi:10.12788/cutis.1267

Author and Disclosure Information

From OnSpot Dermatology, Naples, Florida, and Riverchase Dermatology, Fort Myers, Florida.

Dr. Camisa has no relevant financial disclosures to report.

Correspondence: Charles Camisa, MD (drcamisa@gmail.com).

Cutis. 2025 August;116(2):E12-E19. doi:10.12788/cutis.1267

Article PDF
Article PDF

There are 3 uncommon types of mechanobullous skin diseases caused by relative reduction or complete loss of functional type VII collagen, which is the main component of anchoring fibrils in the lamina densa of the basement membrane zone (BMZ) of the skin and mucous membrane epithelium.1 The function of the anchoring fibrils is to maintain adherence of the basement membrane of the epithelium to the connective tissue of the papillary dermis and submucosa.1 The mechanism of action of the loss of type VII collagen function is via autoimmunity in epidermolysis bullosa acquisita (EBA)2 and bullous systemic lupus erythematosus (BSLE).3 In the heritable family of 4 epidermolysis bullosa (EB) variants, only one of the subtypes—dystrophic EB (DEB)—is caused by various recessive and dominant mutations of the type VII collagen gene (COL7A1).4 The other 3 diseases in the family—EB simplex, junctional EB, and Kindler syndrome—are caused by diverse mutations that corrupt the integrity of keratinocytes and the BMZ.5,6 This article provides an overview of these 3 subtypes to help differentiate them from DEB.

Epidermolysis Bullosa

Epidermolysis bullosa consists of a heterogeneous family of 4 major genetic mechanobullous diseases that affect the skin and mucous membranes with more than 30 subtypes.1 Dystrophic EB is caused by mutations in the COL7A1 gene, which encodes for the α-1 chain of collagen type VII. Classically, EB is divided into 4 main variants based on the location of the cleavage plane or split occurring in the epithelium, which in turn helps to predict the severity of the illness.

Epidermolysis bullosa may be inherited in an ­autosomal-dominant or autosomal-recessive fashion, or it may occur as a spontaneous mutation. All sexes and races are affected equally. Patients present at birth or in early childhood with fragile skin and mucous membranes that may develop blisters, erosions, and ulcerations after minor trauma.7 These lesions are marked by slow healing and scar formation and often are associated with itching and pain.

Dystrophic Epidermolysis Bullosa

Dystrophic EB accounts for approximately 25%6 of all EB cases in the United States and may be inherited as either a dominant or recessive trait. Hundreds of different pathogenic mutations have been discovered in the COL7A1 gene in the subtypes of DEB.4,8 Dominant DEB tends to cause milder disease because the patients retain one normal COL7A1 allele and produce some type VII collagen (Figure 1), whereas patients with recessive DEB lack type VII collagen completely.9 The cleavage plane is between the lamina densa and the superficial dermis or submucosa. Severity is variable and ranges from ­localization to the hands and feet to severe generalized blistering and painful ulcerations depending on which of the many possible gene mutations have been inherited. Sequelae include mitten deformities, malalignment and tooth decay, and the development of early aggressive squamous cell carcinomas, which may be fatal. The most severe cases of recessive DEB also may have internal organ involvement.

Camisa-1
FIGURE 1. Bulla on the ankle of an infant and scarring on the hands and severe nail dystrophy with nail loss in a parent with autosomal-dominant dystrophic epidermolysis bullosa.

Epidermolysis Bullosa Simplex

Epidermolysis bullosa simplex is the most common variant, comprising approximately 70%of EB cases in the United States.6 Epidermolysis bullosa simplex usually is inherited as autosomal-dominant mutations in the keratin 5 or keratin 14 genes,10 not COL7A1. Skin blistering results from cleavage within the basal cell layer where the keratin genes are primarily expressed. Blisters tend to occur in acral areas such as hands and feet and may heal without scarring in the localized form of epidermolysis bullosa simplex (Figure 2).

Camisa-2
FIGURE 2. Epidermolysis bullosa simplex in a child with healing blisters localized to the hands and wrist.

Junctional Epidermolysis Bullosa and Kindler Syndrome

Junctional epidermolysis bullosa (JEB) and Kindler syndrome11 are the rarest of the autosomal-recessive EB ­variants.6 The plane of cleavage in JEB is through the lamina lucida of the BMZ. Junctional epidermolysis bullosa is caused by mutations of the genes that encode for the 3 chains of laminin 332 protein and type XVII collagen,5,12 not to be confused with type VII collagen. As with DEB, there is a wide range of severity in JEB, from localized effects on the eyes, oral cavity, and tooth enamel to widespread blistering and skin cancers. In JEB cases involving newborns, nonhealing wounds on the face, buttocks, fingers, and toes may be seen, with devastating complications in the oral cavity, esophagus, and larynx. Life expectancy is limited to 2 years or less.6 There have only been approximately 40,013 cases of Kindler syndrome reported worldwide6 and there is clinical overlap with DEB. Patients also may demonstrate poikiloderma and photosensitivity. Kindler syndrome is caused by mutations in the FERMT1 gene which encodes for kindlin-1. This protein mediates anchorage between the actin cytoskeleton and the extracellular matrix.5,11 Loss of function produces variable cleavage planes around the dermoepidermal junction.

Clinical management of all EB variants, especially the severe recessive types, traditionally has been limited to the prevention of trauma to the skin and mucous membranes and supportive care, including dressing changes to erosions and ulcerations, antibiotic ointments as needed, and amelioration of pain and pruritus. Bone marrow and pluripotential stem cell transplants have been attempted.12 Complications of EB, such as deformities of the hands and feet caused by excessive scarring, esophageal strictures, poor dentition, and squamous cell carcinomas, must be addressed by a multidisciplinary team of specialists, including plastic surgery, gastroenterology, dentistry/oral surgery, ophthalmology, and dermatology/Mohs surgery. 

Until recently, there were no medications approved by the US Food and Drug Administration (FDA) specifically indicated for EB. In 2023, topical gene therapy was approved by the FDA for both recessive and dominant forms of DEB. Normal COL7A1 sequences are delivered by an attenuated herpes simplex virus 1 vector (beremagene geperpavec) in a gel applied directly to the wounds of patients with DEB. In a clinical trial, matching wounds on 31 patients (62 wounds total) were treated with the active agent or placebo gel. After 6 months, complete wound closure was observed in 67% (21/31) of those treated with the active agent and 22% (7/31) of those treated with placebo (P=.002).14 In a single case report, a patient with recessive DEB and cicatrizing conjunctivitis (Figure 3) was given ophthalmic beremagene geperpavec after surgery and had improved visual acuity.15 A topical gel consisting of birch triterpenes to promote healing of partial-thickness wounds also was approved for patients with DEB and JEB by the FDA and the European Commission. In a study of 223 patients, 41% of those using active gel and 29% of those using placebo gel achieved the primary end point of percentage of target wounds that had first complete closure at 45 days.16 

Camisa-3
FIGURE 3. Recessive dystrophic epidermolysis bullosa complicated by cicatrizing conjunctivitis in a teen who underwent successful surgery and ophthalmic gene therapy to restore his sight. Photograph courtesy of Alfonso L. Sabater, MD (Miami, Florida).


The most recent FDA approval for DEB involves transferring the functional COL7A1 gene to the patient’s skin cells, then expanding the gene-corrected cells into sheets of keratinocytes that can be surgically applied to the chronic wound sites. In a phase 3 trial of prademagene zamikeracel (pz-cel), 11 patients with 86 matched wounds were randomized to receive pz-cel (50%) or standard wound care (50%). After 24 weeks, 35 wounds treated with pz-cel were at least 50% healed compared to 7 control wounds.17 The results for healing and reduction of pain were statistically significant (P<.0001 and P<.0002, respectively).17 Recombinant collagen VII as replacement therapy also is under study to be given by intravenous infusion to increase tissue collagen VII where it is lacking. This treatment has shown early biologic and therapeutic effects.9,18 Larger long-term follow-up studies are necessary to confirm persistence of the gene-corrected skin cells, the functionality of the replacement collagen VII, and the potential risk for the development of autoantibodies to type VII collagen.

Epidermolysis Bullosa Acquisita

Epidermolysis bullosa acquisita is a rare autoimmune subepithelial bullous disease that primarily affects middle-aged adults but also has been reported in children.19 Epidermolysis bullosa acquisita is caused by circulating pathogenic IgG autoantibodies that target and bind to type VII collagen in the anchoring fibrils,20-22 thereby disrupting the attachment of the epithelium to its underlying connective tissue.

The 2 major clinical manifestations of EBA include a mechanobullous disease resembling inherited forms of DEB (Figure 4) and an inflammatory bullous pemphigoid (BP)–like disease,23 as well as a combination of both types of skin lesions (Figure 5). The skin and mucous membranes of the oral cavity, esophagus, eyes, and urogenital areas are affected in both types; scarring may cause functional disabilities. In the mechanobullous type of EBA, it is common for blisters and erosions to develop in trauma-prone areas such as the hands, feet, elbows, and knees. The blisters tend to heal with scarring and milia formation as might be seen in porphyria cutanea tarda or cicatricial pemphigoid, which are in the differential diagnosis. Dystrophy of the fingernails or complete nail loss may be observed, resembling DEB. In the BP-like presentation, tense blisters arise upon inflamed or urticarial skin and mucous membranes, which may then become generalized. 

Camisa-4
FIGURE 4. Epidermolysis bullosa acquisita (mechanobullous type) of the hands shows small blisters, scarring, and erosions of the lateral nail folds.

 

Camisa-5
FIGURE 5. Epidermolysis bullosa acquisita with severe ulcerations on the neck and back secondary to blisters with scarring.


Histopathology in both forms of EBA demonstrates subepithelial separation as clefts or blisters. The mechanobullous type shows a sparse inflammatory infiltrate compared to large collections of neutrophils and eosinophils in the blister cavity and in the superficial dermis in the BP-like cases. The final diagnosis rests on the results of immunopathology testing.24 Direct immunofluorescence of perilesional skin and mucosa shows a linear-granular band of IgG and C3 and other conjugates along the BMZ. Deposits of IgA alone in EBA occur in only about 2.4% of cases and are observed more often when there is mucous membrane involvement.2 Indirect immunofluorescence of sera against salt-split skin substrates detects immunoreactants in the floor of the blister rather than in the roof, as would be seen in BP. Highly specific and sensitive enzyme-linked immunosorbent assay (ELISA) kits now are commercially available and can detect autoantibodies against the N-terminal domain of type VII collagen in more than 90% of cases of EBA.25 

Inflammatory bowel disease (IBD), particularly Crohn disease (CD), precedes the onset of EBA in approximately 25% of cases.26,27 Ulcerative colitis is much less common. Type VII collagen is normally present in the basement membrane of intestinal epithelium. In a survey of patients with IBD, 68% of those with CD and 13% of those with ulcerative colitis had circulating anti–type VII collagen antibodies detected by ELISA without having symptoms of EBA.28 A case report of a patient with both well-proven EBA and CD highlighted the clinical difficulty of controlling EBA: treatment with prednisolone and sulfasalazine improved the CD but had little effect on the skin blisters.29 A variety of malignancies have been reported in association with EBA, including cancers of the uterine cervix,30 thyroid, and pancreas,31 lymphoma, and chronic lymphatic leukemia. Some of these cases have met the criteria for classification as paraneoplastic, whereas others may have been coincidental. 

Treatment for chronic EBA generally has been limited.2,24 Putative antineutrophil drugs such as dapsone and colchicine combined with systemic corticosteroids may be useful in milder or juvenile cases, which tend to have a better prognosis than adult cases.19 In more severe EBA, systemic corticosteroids and/or immunosuppressive drugs such as azathioprine,23 cyclophosphamide,23 mycophenolate mofetil,31 methotrexate,23 cyclosporine,33 and infliximab23 have been used. More recently, rituximab infusion monotherapy33 and rituximab combined with intravenous immunoglobulin or adjuvant immunoadsorption of the pathogenic autoantibodies have induced remission of refractory EBA.32 Adjuvant immunoadsorption therapy is not widely available. Multispecialty care often is required, especially ophthalmology for conjunctival involvement and gastroenterology for potential esophageal stenosis and the early detection and treatment of IBD.

Bullous Systemic Lupus Erythematosus

Bullous systemic lupus erythematosus is a rare and specific autoimmune skin complication that mostly is seen in patients with an established diagnosis of systemic lupus erythematosus (SLE) who are experiencing a disease flare. Although more common in women, it has been reported in all sexes and races as well as in children. Vesicles and bullae may arise on sun-exposed (Figure 6) and sun-protected areas of skin.

Camisa-6
FIGURE 6. Bullous systemic lupus erythematosus demonstrates active vesicles and bullae on a sun-exposed area of the wrist.

Histopathology shows subepidermal separation with collections of neutrophils and nuclear fragments in the blister cavity. The differential diagnosis of BSLE includes EBA, BP, dermatitis herpetiformis, and linear IgA bullous dermatosis. Direct immunofluorescence testing shows linear-granular deposits of IgG and/or IgM and IgA along the BMZ.34 When utilizing the indirect immunofluorescence split-skin assay, the autoantibody to type VII collagen would be detected in the floor of the blister if the serum titer was sufficiently high.3 Proposed criteria for the diagnosis of BSLE have been published: 1) diagnosis of SLE now based on the 2019 European League Against Rheumatism/American College of Rheumatology classification35; 2) vesicles and bullae arising upon but not limited to sun-exposed skin; 3) histopathology featuring neutrophil-rich subepithelial bullae; 4) positive indirect immunofluorescence for circulating BMZ antibodies using separated human skin as substrate; 5) and direct immunofluorescence showing IgG and/or IgM and often IgA at the BMZ.36 Using ELISA to detect circulating antibodies against type VII collagen24 should now be added to the criteria. The new criteria for SLE34 do not include BSLE, perhaps because it occurs in less than 1% of patients with SLE.37 

Further investigation by Gammon et al3 confirmed that the autoantibodies in BSLE are identical to those found in EBA (ie, directed against type VII collagen in the lamina densa). Bullous systemic lupus erythematosus is not considered to be the coexistence of EBA with SLE but rather a specific entity wherein type VII collagen autoantibodies are expressed in the autoimmune spectrum of SLE. It is especially important to make the diagnosis of BSLE because it is predictive of more serious systemic complications of SLE (eg, hematologic and renal disease is found in up to 90% of cases).38 

The natural course of BSLE is variable. Treatments include systemic corticosteroids, dapsone, and immunosuppressive drugs such as azathioprine, methotrexate, mycophenolate mofetil, and cyclophosphamide, especially in cases with nephritis.37 There may be spontaneous resolution of the rash as the inflammatory activity of SLE subsides. Rituximab has been used effectively in several refractory cases of BSLE that failed to respond to all other conventional treatments.39

Conclusion

Anchoring fibrils are composed primarily of type VII collagen. Their role is to maintain the attachment of epithelium to the upper dermis and submucosa. The reduction or complete loss of type VII collagen caused by mutations of the COL7A1 gene results in dominant DEB or recessive DEB, respectively. Two distinct non-heritable immunobullous diseases, EBA and BSLE, are caused by autoantibodies that target type VII collagen. A comparison of the 4 type VII collagen disorders can be found in the Table.

CT116002012_e-Table_part1CT116002012_e-Table_part2

There are 3 uncommon types of mechanobullous skin diseases caused by relative reduction or complete loss of functional type VII collagen, which is the main component of anchoring fibrils in the lamina densa of the basement membrane zone (BMZ) of the skin and mucous membrane epithelium.1 The function of the anchoring fibrils is to maintain adherence of the basement membrane of the epithelium to the connective tissue of the papillary dermis and submucosa.1 The mechanism of action of the loss of type VII collagen function is via autoimmunity in epidermolysis bullosa acquisita (EBA)2 and bullous systemic lupus erythematosus (BSLE).3 In the heritable family of 4 epidermolysis bullosa (EB) variants, only one of the subtypes—dystrophic EB (DEB)—is caused by various recessive and dominant mutations of the type VII collagen gene (COL7A1).4 The other 3 diseases in the family—EB simplex, junctional EB, and Kindler syndrome—are caused by diverse mutations that corrupt the integrity of keratinocytes and the BMZ.5,6 This article provides an overview of these 3 subtypes to help differentiate them from DEB.

Epidermolysis Bullosa

Epidermolysis bullosa consists of a heterogeneous family of 4 major genetic mechanobullous diseases that affect the skin and mucous membranes with more than 30 subtypes.1 Dystrophic EB is caused by mutations in the COL7A1 gene, which encodes for the α-1 chain of collagen type VII. Classically, EB is divided into 4 main variants based on the location of the cleavage plane or split occurring in the epithelium, which in turn helps to predict the severity of the illness.

Epidermolysis bullosa may be inherited in an ­autosomal-dominant or autosomal-recessive fashion, or it may occur as a spontaneous mutation. All sexes and races are affected equally. Patients present at birth or in early childhood with fragile skin and mucous membranes that may develop blisters, erosions, and ulcerations after minor trauma.7 These lesions are marked by slow healing and scar formation and often are associated with itching and pain.

Dystrophic Epidermolysis Bullosa

Dystrophic EB accounts for approximately 25%6 of all EB cases in the United States and may be inherited as either a dominant or recessive trait. Hundreds of different pathogenic mutations have been discovered in the COL7A1 gene in the subtypes of DEB.4,8 Dominant DEB tends to cause milder disease because the patients retain one normal COL7A1 allele and produce some type VII collagen (Figure 1), whereas patients with recessive DEB lack type VII collagen completely.9 The cleavage plane is between the lamina densa and the superficial dermis or submucosa. Severity is variable and ranges from ­localization to the hands and feet to severe generalized blistering and painful ulcerations depending on which of the many possible gene mutations have been inherited. Sequelae include mitten deformities, malalignment and tooth decay, and the development of early aggressive squamous cell carcinomas, which may be fatal. The most severe cases of recessive DEB also may have internal organ involvement.

Camisa-1
FIGURE 1. Bulla on the ankle of an infant and scarring on the hands and severe nail dystrophy with nail loss in a parent with autosomal-dominant dystrophic epidermolysis bullosa.

Epidermolysis Bullosa Simplex

Epidermolysis bullosa simplex is the most common variant, comprising approximately 70%of EB cases in the United States.6 Epidermolysis bullosa simplex usually is inherited as autosomal-dominant mutations in the keratin 5 or keratin 14 genes,10 not COL7A1. Skin blistering results from cleavage within the basal cell layer where the keratin genes are primarily expressed. Blisters tend to occur in acral areas such as hands and feet and may heal without scarring in the localized form of epidermolysis bullosa simplex (Figure 2).

Camisa-2
FIGURE 2. Epidermolysis bullosa simplex in a child with healing blisters localized to the hands and wrist.

Junctional Epidermolysis Bullosa and Kindler Syndrome

Junctional epidermolysis bullosa (JEB) and Kindler syndrome11 are the rarest of the autosomal-recessive EB ­variants.6 The plane of cleavage in JEB is through the lamina lucida of the BMZ. Junctional epidermolysis bullosa is caused by mutations of the genes that encode for the 3 chains of laminin 332 protein and type XVII collagen,5,12 not to be confused with type VII collagen. As with DEB, there is a wide range of severity in JEB, from localized effects on the eyes, oral cavity, and tooth enamel to widespread blistering and skin cancers. In JEB cases involving newborns, nonhealing wounds on the face, buttocks, fingers, and toes may be seen, with devastating complications in the oral cavity, esophagus, and larynx. Life expectancy is limited to 2 years or less.6 There have only been approximately 40,013 cases of Kindler syndrome reported worldwide6 and there is clinical overlap with DEB. Patients also may demonstrate poikiloderma and photosensitivity. Kindler syndrome is caused by mutations in the FERMT1 gene which encodes for kindlin-1. This protein mediates anchorage between the actin cytoskeleton and the extracellular matrix.5,11 Loss of function produces variable cleavage planes around the dermoepidermal junction.

Clinical management of all EB variants, especially the severe recessive types, traditionally has been limited to the prevention of trauma to the skin and mucous membranes and supportive care, including dressing changes to erosions and ulcerations, antibiotic ointments as needed, and amelioration of pain and pruritus. Bone marrow and pluripotential stem cell transplants have been attempted.12 Complications of EB, such as deformities of the hands and feet caused by excessive scarring, esophageal strictures, poor dentition, and squamous cell carcinomas, must be addressed by a multidisciplinary team of specialists, including plastic surgery, gastroenterology, dentistry/oral surgery, ophthalmology, and dermatology/Mohs surgery. 

Until recently, there were no medications approved by the US Food and Drug Administration (FDA) specifically indicated for EB. In 2023, topical gene therapy was approved by the FDA for both recessive and dominant forms of DEB. Normal COL7A1 sequences are delivered by an attenuated herpes simplex virus 1 vector (beremagene geperpavec) in a gel applied directly to the wounds of patients with DEB. In a clinical trial, matching wounds on 31 patients (62 wounds total) were treated with the active agent or placebo gel. After 6 months, complete wound closure was observed in 67% (21/31) of those treated with the active agent and 22% (7/31) of those treated with placebo (P=.002).14 In a single case report, a patient with recessive DEB and cicatrizing conjunctivitis (Figure 3) was given ophthalmic beremagene geperpavec after surgery and had improved visual acuity.15 A topical gel consisting of birch triterpenes to promote healing of partial-thickness wounds also was approved for patients with DEB and JEB by the FDA and the European Commission. In a study of 223 patients, 41% of those using active gel and 29% of those using placebo gel achieved the primary end point of percentage of target wounds that had first complete closure at 45 days.16 

Camisa-3
FIGURE 3. Recessive dystrophic epidermolysis bullosa complicated by cicatrizing conjunctivitis in a teen who underwent successful surgery and ophthalmic gene therapy to restore his sight. Photograph courtesy of Alfonso L. Sabater, MD (Miami, Florida).


The most recent FDA approval for DEB involves transferring the functional COL7A1 gene to the patient’s skin cells, then expanding the gene-corrected cells into sheets of keratinocytes that can be surgically applied to the chronic wound sites. In a phase 3 trial of prademagene zamikeracel (pz-cel), 11 patients with 86 matched wounds were randomized to receive pz-cel (50%) or standard wound care (50%). After 24 weeks, 35 wounds treated with pz-cel were at least 50% healed compared to 7 control wounds.17 The results for healing and reduction of pain were statistically significant (P<.0001 and P<.0002, respectively).17 Recombinant collagen VII as replacement therapy also is under study to be given by intravenous infusion to increase tissue collagen VII where it is lacking. This treatment has shown early biologic and therapeutic effects.9,18 Larger long-term follow-up studies are necessary to confirm persistence of the gene-corrected skin cells, the functionality of the replacement collagen VII, and the potential risk for the development of autoantibodies to type VII collagen.

Epidermolysis Bullosa Acquisita

Epidermolysis bullosa acquisita is a rare autoimmune subepithelial bullous disease that primarily affects middle-aged adults but also has been reported in children.19 Epidermolysis bullosa acquisita is caused by circulating pathogenic IgG autoantibodies that target and bind to type VII collagen in the anchoring fibrils,20-22 thereby disrupting the attachment of the epithelium to its underlying connective tissue.

The 2 major clinical manifestations of EBA include a mechanobullous disease resembling inherited forms of DEB (Figure 4) and an inflammatory bullous pemphigoid (BP)–like disease,23 as well as a combination of both types of skin lesions (Figure 5). The skin and mucous membranes of the oral cavity, esophagus, eyes, and urogenital areas are affected in both types; scarring may cause functional disabilities. In the mechanobullous type of EBA, it is common for blisters and erosions to develop in trauma-prone areas such as the hands, feet, elbows, and knees. The blisters tend to heal with scarring and milia formation as might be seen in porphyria cutanea tarda or cicatricial pemphigoid, which are in the differential diagnosis. Dystrophy of the fingernails or complete nail loss may be observed, resembling DEB. In the BP-like presentation, tense blisters arise upon inflamed or urticarial skin and mucous membranes, which may then become generalized. 

Camisa-4
FIGURE 4. Epidermolysis bullosa acquisita (mechanobullous type) of the hands shows small blisters, scarring, and erosions of the lateral nail folds.

 

Camisa-5
FIGURE 5. Epidermolysis bullosa acquisita with severe ulcerations on the neck and back secondary to blisters with scarring.


Histopathology in both forms of EBA demonstrates subepithelial separation as clefts or blisters. The mechanobullous type shows a sparse inflammatory infiltrate compared to large collections of neutrophils and eosinophils in the blister cavity and in the superficial dermis in the BP-like cases. The final diagnosis rests on the results of immunopathology testing.24 Direct immunofluorescence of perilesional skin and mucosa shows a linear-granular band of IgG and C3 and other conjugates along the BMZ. Deposits of IgA alone in EBA occur in only about 2.4% of cases and are observed more often when there is mucous membrane involvement.2 Indirect immunofluorescence of sera against salt-split skin substrates detects immunoreactants in the floor of the blister rather than in the roof, as would be seen in BP. Highly specific and sensitive enzyme-linked immunosorbent assay (ELISA) kits now are commercially available and can detect autoantibodies against the N-terminal domain of type VII collagen in more than 90% of cases of EBA.25 

Inflammatory bowel disease (IBD), particularly Crohn disease (CD), precedes the onset of EBA in approximately 25% of cases.26,27 Ulcerative colitis is much less common. Type VII collagen is normally present in the basement membrane of intestinal epithelium. In a survey of patients with IBD, 68% of those with CD and 13% of those with ulcerative colitis had circulating anti–type VII collagen antibodies detected by ELISA without having symptoms of EBA.28 A case report of a patient with both well-proven EBA and CD highlighted the clinical difficulty of controlling EBA: treatment with prednisolone and sulfasalazine improved the CD but had little effect on the skin blisters.29 A variety of malignancies have been reported in association with EBA, including cancers of the uterine cervix,30 thyroid, and pancreas,31 lymphoma, and chronic lymphatic leukemia. Some of these cases have met the criteria for classification as paraneoplastic, whereas others may have been coincidental. 

Treatment for chronic EBA generally has been limited.2,24 Putative antineutrophil drugs such as dapsone and colchicine combined with systemic corticosteroids may be useful in milder or juvenile cases, which tend to have a better prognosis than adult cases.19 In more severe EBA, systemic corticosteroids and/or immunosuppressive drugs such as azathioprine,23 cyclophosphamide,23 mycophenolate mofetil,31 methotrexate,23 cyclosporine,33 and infliximab23 have been used. More recently, rituximab infusion monotherapy33 and rituximab combined with intravenous immunoglobulin or adjuvant immunoadsorption of the pathogenic autoantibodies have induced remission of refractory EBA.32 Adjuvant immunoadsorption therapy is not widely available. Multispecialty care often is required, especially ophthalmology for conjunctival involvement and gastroenterology for potential esophageal stenosis and the early detection and treatment of IBD.

Bullous Systemic Lupus Erythematosus

Bullous systemic lupus erythematosus is a rare and specific autoimmune skin complication that mostly is seen in patients with an established diagnosis of systemic lupus erythematosus (SLE) who are experiencing a disease flare. Although more common in women, it has been reported in all sexes and races as well as in children. Vesicles and bullae may arise on sun-exposed (Figure 6) and sun-protected areas of skin.

Camisa-6
FIGURE 6. Bullous systemic lupus erythematosus demonstrates active vesicles and bullae on a sun-exposed area of the wrist.

Histopathology shows subepidermal separation with collections of neutrophils and nuclear fragments in the blister cavity. The differential diagnosis of BSLE includes EBA, BP, dermatitis herpetiformis, and linear IgA bullous dermatosis. Direct immunofluorescence testing shows linear-granular deposits of IgG and/or IgM and IgA along the BMZ.34 When utilizing the indirect immunofluorescence split-skin assay, the autoantibody to type VII collagen would be detected in the floor of the blister if the serum titer was sufficiently high.3 Proposed criteria for the diagnosis of BSLE have been published: 1) diagnosis of SLE now based on the 2019 European League Against Rheumatism/American College of Rheumatology classification35; 2) vesicles and bullae arising upon but not limited to sun-exposed skin; 3) histopathology featuring neutrophil-rich subepithelial bullae; 4) positive indirect immunofluorescence for circulating BMZ antibodies using separated human skin as substrate; 5) and direct immunofluorescence showing IgG and/or IgM and often IgA at the BMZ.36 Using ELISA to detect circulating antibodies against type VII collagen24 should now be added to the criteria. The new criteria for SLE34 do not include BSLE, perhaps because it occurs in less than 1% of patients with SLE.37 

Further investigation by Gammon et al3 confirmed that the autoantibodies in BSLE are identical to those found in EBA (ie, directed against type VII collagen in the lamina densa). Bullous systemic lupus erythematosus is not considered to be the coexistence of EBA with SLE but rather a specific entity wherein type VII collagen autoantibodies are expressed in the autoimmune spectrum of SLE. It is especially important to make the diagnosis of BSLE because it is predictive of more serious systemic complications of SLE (eg, hematologic and renal disease is found in up to 90% of cases).38 

The natural course of BSLE is variable. Treatments include systemic corticosteroids, dapsone, and immunosuppressive drugs such as azathioprine, methotrexate, mycophenolate mofetil, and cyclophosphamide, especially in cases with nephritis.37 There may be spontaneous resolution of the rash as the inflammatory activity of SLE subsides. Rituximab has been used effectively in several refractory cases of BSLE that failed to respond to all other conventional treatments.39

Conclusion

Anchoring fibrils are composed primarily of type VII collagen. Their role is to maintain the attachment of epithelium to the upper dermis and submucosa. The reduction or complete loss of type VII collagen caused by mutations of the COL7A1 gene results in dominant DEB or recessive DEB, respectively. Two distinct non-heritable immunobullous diseases, EBA and BSLE, are caused by autoantibodies that target type VII collagen. A comparison of the 4 type VII collagen disorders can be found in the Table.

CT116002012_e-Table_part1CT116002012_e-Table_part2
References
  1. Bardhan A, Bruckner-Tuderman L, Chapple ILC, et al. Epidermolysis bullosa. Nat Rev Dis Primers. 2020;6:78. doi:10.1038/s41572-020-0210-0
  2. Miyamoto D, Gordilho JO, Santi CG, et al. Epidermolysis bullosa acquisita. An Bras Dermatol. 2022;97:409-423. doi:10.1016/j.abd.2021.09.010.
  3. Gammon WR, Woodley DT, Dole KC, et al. Evidence that anti-basement membrane zone antibodies in bullous eruption of systemic lupus erythematosus recognize epidermolysis bullosa acquisita autoantigen. J Invest Dermatol. 1985;84:472-476. doi:10.1111/1523-1747.ep12272402.
  4. Yadav RS, Jaswal A, Shrestha S, et al. Dystrophic epidermolysis bullosa. J Nepal Med Assoc. 2018;56:879-882. doi:10.31729/jnma.3791
  5. Mariath LM, Santin JT, Schuler-Faccini L, et al. Inherited epidermolysis bullosa: update on the clinical and genetic aspects. An Bras Dermatol. 2020;95:551-569. doi:10.1016/j.abd.2020.05.001
  6. Understanding epidermolysis bullosa (EB). DEBRA website. Accessed August 17, 2025. https://www.debra.org/about-eb/understanding-epidermolysis-bullosa-eb
  7. Hon KL, Chu S, Leung AKC. Epidermolysis bullosa: pediatric perspectives. Curr Pediatr Rev. 2022;18:182-190. doi:10.2174/1573396317666210525161252
  8. Dang N, Klingberg S, Marr P, et al. Review of collagen VII sequence variants found in Australasian patients with dystrophic epidermolysis bullosa reveals nine COL7A1 variants. J Dermatol Sci. 2007;46:169-178. doi:10.1016/j.jdermsci.2007.02.006
  9. Payne AS. Topical gene therapy for epidermolysis bullosa. N Engl J Med. 2022;387:2281-2284. doi:10.1056/NEJMe2213203
  10. Khani P, Ghazi F, Zekri A, et al. Keratins and epidermolysis bullosa simplex. J Cell Physiol. 2018;234:289-297. doi:10.1002/jcp.26898
  11. Lai-Cheong JE, Tanaka A, Hawche G, et al. Kindler syndrome: a focal adhesion genodermatosis. Br J Dermatol. 2009;160:233-242. doi:10.1111/j.1365-2133.2008.08976.x
  12. Hou P-C, Wang H-T, Abhee S, et al. Investigational treatments for epidermolysis bullosa. Am J Clin Dermatol. 2021;22:801-817. doi:10.1007/s40257-021-00626-3
  13. Youseffian L, Vahidnezhad H, Uitto J. Kindler Syndrome. GeneReviews [Internet]. Updated January 6, 2022. Accessed August 21, 2025.
  14. Guide SV, Gonzalez ME, Bagci S, et al. Trial of beremagene geperpavec (B-VEC) for dystrophic epidermolysis bullosa. N Engl J Med. 2022;387:2211-2219. doi:10.1056/NEJMoa2206663
  15. Vetencourt AT, Sayed-Ahmed I, Gomez J, et al. Ocular gene therapy in a patient with dystrophic epidermolysis bullosa. N Engl J Med. 2024;390:530-535. doi:10.1056/NEJMoa2301244
  16. Kern JS, Sprecher E, Fernandez MF, et al. Efficacy and safety of Oleogel-S10 (birch triterpenes for epidermolysis bullosa: results from the phase III randomized double-blind phase of the EASE study. Br J Dermatol. 2023;188:12-21. doi:10.1093/bjd/ljac001
  17. Tang JY, Marinkovich MP, Wiss K, et al. Prademagene zamikeracel for recessive dystrophic epidermolysis bullosa wounds (VIITAL): a two-centre, randomized, open-label, intrapatient-controlled phase 3 trial. Lancet. 2025;406:163-173. doi:10.1016/S0140-6736(25)00778-0
  18. Gretzmeier C, Pin D, Kern JS, et al. Systemic collagen VII replacement therapy for advanced recessive dystrophic epidermolysis bullosa. J Invest Dermatol. 2022;142:1094-1102. doi:10.1016/j.jid.2021.09.008
  19. Hignett E, Sami N. Pediatric epidermolysis bullosa acquisita. A review. Pediatr Dermatol. 2021;38:1047-1050. doi:10.1111/pde.14722
  20. Chen M, Kim GH, Prakash L, et al. Autoimmunity to anchoring fibril collagen. Autoimmunity. 2012;45:91-101. doi:10.1007/s12016-007-0027-6.
  21. Kridin K, Kneiber D, Kowalski EH, et al. Epidermolysis bullosa acquisita: a comprehensive review. Autoimmun Rev. 2019;18:786-795. doi:10.1016/j.autrev.2019.06.007
  22. Hofmann SC, Weidinger A. Epidermolysis bullosa acquisita. Hautarzt. 2019;70:265-270. doi:10.1007/s00105-019-4387-7
  23. Ishi N, Hamada T, Dainichi T, et al. Epidermolysis bullosa acquisita: what’s new? J Dermatol. 2010;37:220-230. doi:10.1111/j.1346-8138.2009.00799.x
  24. Iwata H, Vorobyev A, Koga H, et al. Meta-analysis of the clinical and immunopathological characteristics and treatment outcomes in epidermolysis bullosa acquisita patients. Orphanet J Rare Dis. 2018;13:153. doi:10.1186/s13023-018-0896-1
  25. Komorowski L, Muller R, Vorobyev A, et al. Sensitive and specific assays for routine serological diagnosis of epidermolysis bullosa acquisita. J Am Acad Dermatol. 2013;68:e89-95. doi:10.1016/j.jaad.2011.12.032
  26. Antonelli E, Bassotti G, Tramontana M, et al. Dermatological manifestations in inflammatory bowel diseases. J Clin Med. 2021;10:364-390. doi:10.3390/jcm10020364
  27. Bezzio C, Della Corte C, Vernero M, et al. Inflammatory bowel disease and immune-mediated inflammatory diseases: looking at less frequent associations. Therap Adv Gastroenterol. 2022;15:17562848221115312. doi:10.1177/17562848221115312
  28. Chen M, O’Toole EA, Sanghavi J, et al. The epidermolysis acquisita antigen (type VII collagen) is present in human colon and patients with Crohn’s disease have antibodies to type VII collagen. J Invest Dermatol. 2002;118:1059-1064. doi:10.1046/j.1523-1747.2002.01772.x
  29. Labeille B, Gineston JL, Denoeux JP, et al. Epidermolysis bullosa acquisita and Crohn’s disease. A case report with immunological and electron microscopic studies. Arch Intern Med. 1988;148:1457-1459.
  30. Etienne A, Ruffieux P, Didierjean L, et al. Epidermolysis bullosa acquisita and metastatic cancer of the uterine cervix. Ann Dermatol Venereol. 1998;125:321-323.
  31. Busch J-O, Sticherling M. Epidermolysis bullosa acquisita and neuroendocrine pancreatic cancer-Coincidence or patho-genetic relationship? J Dtsch Dermatol Ges. 2007;5:916-918. doi:10.111/j.1610-0387.2007.06338.x
  32. Bevans SL, Sami N. The use of rituximab in treatment of epidermolysis bullosa acquisita: three new cases and a review of the literature. Dermatol Ther. 2018;31:e12726. doi:10.1111/j.1610-0387.2007.06338.x
  33. Yang A, Kim M, Craig P, et al. A case report of the use of rituximab and the epidermolysis bullosa disease activity scoring index (EBDASI) in a patient with epidermolysis bullosa acquisita with extensive esophageal involvement. Arch Dermatovenerol Croat. 2018;26:325-328.
  34. Burrows NP, Bhogal BS, Black MM, et al. Bullous eruption of systemic lupus erythematosus: a clinicopathological study of four cases. Br J Dermatol. 1993;128:332-338. doi:10.1111/j.1365-2133.1993.tb00180.x
  35. Aringer M, Leuchten N, Johnson SR. New criteria for lupus. Curr Rheum Rep. 2020;22:18. doi:10.1007/s11926-020-00896-6
  36. Camisa C. Vesiculobullous systemic lupus erythematosus. A report of four cases. J Am Acad Dermatol. 1988;18:93-100. doi:10.1016/s0190-9622(88)70014-6
  37. Duan L, Chen L, Zhong S, et al. Treatment of bullous systemic lupus erythematosus. J Immunol Res. 2015;2015:167064. doi:10.1155/2015/167064
  38. Sprow G, Afarideh M, Dan J, et al. Bullous systemic lupus erythematosus in females. Int J Womens Dermatol. 2022;8:e034. doi:10.1097/JW9.0000000000000034
  39. Contestable JJ, Edhegard KD, Meyerle JH. Bullous systemic lupus erythematosus: a review and update to diagnosis and treatment. Am J Clin Dermatol. 2014;15:517-524. doi:10.1007/s40257-014-0098-0
  40. Fine JD, Mellerio JE. Epidermolysis bullosa. In: Bolognia JL, Jorizzo JL, Schaffer JV (eds), Dermatology (ed 3), Elsevier Saunders; 2012: 501-513.
References
  1. Bardhan A, Bruckner-Tuderman L, Chapple ILC, et al. Epidermolysis bullosa. Nat Rev Dis Primers. 2020;6:78. doi:10.1038/s41572-020-0210-0
  2. Miyamoto D, Gordilho JO, Santi CG, et al. Epidermolysis bullosa acquisita. An Bras Dermatol. 2022;97:409-423. doi:10.1016/j.abd.2021.09.010.
  3. Gammon WR, Woodley DT, Dole KC, et al. Evidence that anti-basement membrane zone antibodies in bullous eruption of systemic lupus erythematosus recognize epidermolysis bullosa acquisita autoantigen. J Invest Dermatol. 1985;84:472-476. doi:10.1111/1523-1747.ep12272402.
  4. Yadav RS, Jaswal A, Shrestha S, et al. Dystrophic epidermolysis bullosa. J Nepal Med Assoc. 2018;56:879-882. doi:10.31729/jnma.3791
  5. Mariath LM, Santin JT, Schuler-Faccini L, et al. Inherited epidermolysis bullosa: update on the clinical and genetic aspects. An Bras Dermatol. 2020;95:551-569. doi:10.1016/j.abd.2020.05.001
  6. Understanding epidermolysis bullosa (EB). DEBRA website. Accessed August 17, 2025. https://www.debra.org/about-eb/understanding-epidermolysis-bullosa-eb
  7. Hon KL, Chu S, Leung AKC. Epidermolysis bullosa: pediatric perspectives. Curr Pediatr Rev. 2022;18:182-190. doi:10.2174/1573396317666210525161252
  8. Dang N, Klingberg S, Marr P, et al. Review of collagen VII sequence variants found in Australasian patients with dystrophic epidermolysis bullosa reveals nine COL7A1 variants. J Dermatol Sci. 2007;46:169-178. doi:10.1016/j.jdermsci.2007.02.006
  9. Payne AS. Topical gene therapy for epidermolysis bullosa. N Engl J Med. 2022;387:2281-2284. doi:10.1056/NEJMe2213203
  10. Khani P, Ghazi F, Zekri A, et al. Keratins and epidermolysis bullosa simplex. J Cell Physiol. 2018;234:289-297. doi:10.1002/jcp.26898
  11. Lai-Cheong JE, Tanaka A, Hawche G, et al. Kindler syndrome: a focal adhesion genodermatosis. Br J Dermatol. 2009;160:233-242. doi:10.1111/j.1365-2133.2008.08976.x
  12. Hou P-C, Wang H-T, Abhee S, et al. Investigational treatments for epidermolysis bullosa. Am J Clin Dermatol. 2021;22:801-817. doi:10.1007/s40257-021-00626-3
  13. Youseffian L, Vahidnezhad H, Uitto J. Kindler Syndrome. GeneReviews [Internet]. Updated January 6, 2022. Accessed August 21, 2025.
  14. Guide SV, Gonzalez ME, Bagci S, et al. Trial of beremagene geperpavec (B-VEC) for dystrophic epidermolysis bullosa. N Engl J Med. 2022;387:2211-2219. doi:10.1056/NEJMoa2206663
  15. Vetencourt AT, Sayed-Ahmed I, Gomez J, et al. Ocular gene therapy in a patient with dystrophic epidermolysis bullosa. N Engl J Med. 2024;390:530-535. doi:10.1056/NEJMoa2301244
  16. Kern JS, Sprecher E, Fernandez MF, et al. Efficacy and safety of Oleogel-S10 (birch triterpenes for epidermolysis bullosa: results from the phase III randomized double-blind phase of the EASE study. Br J Dermatol. 2023;188:12-21. doi:10.1093/bjd/ljac001
  17. Tang JY, Marinkovich MP, Wiss K, et al. Prademagene zamikeracel for recessive dystrophic epidermolysis bullosa wounds (VIITAL): a two-centre, randomized, open-label, intrapatient-controlled phase 3 trial. Lancet. 2025;406:163-173. doi:10.1016/S0140-6736(25)00778-0
  18. Gretzmeier C, Pin D, Kern JS, et al. Systemic collagen VII replacement therapy for advanced recessive dystrophic epidermolysis bullosa. J Invest Dermatol. 2022;142:1094-1102. doi:10.1016/j.jid.2021.09.008
  19. Hignett E, Sami N. Pediatric epidermolysis bullosa acquisita. A review. Pediatr Dermatol. 2021;38:1047-1050. doi:10.1111/pde.14722
  20. Chen M, Kim GH, Prakash L, et al. Autoimmunity to anchoring fibril collagen. Autoimmunity. 2012;45:91-101. doi:10.1007/s12016-007-0027-6.
  21. Kridin K, Kneiber D, Kowalski EH, et al. Epidermolysis bullosa acquisita: a comprehensive review. Autoimmun Rev. 2019;18:786-795. doi:10.1016/j.autrev.2019.06.007
  22. Hofmann SC, Weidinger A. Epidermolysis bullosa acquisita. Hautarzt. 2019;70:265-270. doi:10.1007/s00105-019-4387-7
  23. Ishi N, Hamada T, Dainichi T, et al. Epidermolysis bullosa acquisita: what’s new? J Dermatol. 2010;37:220-230. doi:10.1111/j.1346-8138.2009.00799.x
  24. Iwata H, Vorobyev A, Koga H, et al. Meta-analysis of the clinical and immunopathological characteristics and treatment outcomes in epidermolysis bullosa acquisita patients. Orphanet J Rare Dis. 2018;13:153. doi:10.1186/s13023-018-0896-1
  25. Komorowski L, Muller R, Vorobyev A, et al. Sensitive and specific assays for routine serological diagnosis of epidermolysis bullosa acquisita. J Am Acad Dermatol. 2013;68:e89-95. doi:10.1016/j.jaad.2011.12.032
  26. Antonelli E, Bassotti G, Tramontana M, et al. Dermatological manifestations in inflammatory bowel diseases. J Clin Med. 2021;10:364-390. doi:10.3390/jcm10020364
  27. Bezzio C, Della Corte C, Vernero M, et al. Inflammatory bowel disease and immune-mediated inflammatory diseases: looking at less frequent associations. Therap Adv Gastroenterol. 2022;15:17562848221115312. doi:10.1177/17562848221115312
  28. Chen M, O’Toole EA, Sanghavi J, et al. The epidermolysis acquisita antigen (type VII collagen) is present in human colon and patients with Crohn’s disease have antibodies to type VII collagen. J Invest Dermatol. 2002;118:1059-1064. doi:10.1046/j.1523-1747.2002.01772.x
  29. Labeille B, Gineston JL, Denoeux JP, et al. Epidermolysis bullosa acquisita and Crohn’s disease. A case report with immunological and electron microscopic studies. Arch Intern Med. 1988;148:1457-1459.
  30. Etienne A, Ruffieux P, Didierjean L, et al. Epidermolysis bullosa acquisita and metastatic cancer of the uterine cervix. Ann Dermatol Venereol. 1998;125:321-323.
  31. Busch J-O, Sticherling M. Epidermolysis bullosa acquisita and neuroendocrine pancreatic cancer-Coincidence or patho-genetic relationship? J Dtsch Dermatol Ges. 2007;5:916-918. doi:10.111/j.1610-0387.2007.06338.x
  32. Bevans SL, Sami N. The use of rituximab in treatment of epidermolysis bullosa acquisita: three new cases and a review of the literature. Dermatol Ther. 2018;31:e12726. doi:10.1111/j.1610-0387.2007.06338.x
  33. Yang A, Kim M, Craig P, et al. A case report of the use of rituximab and the epidermolysis bullosa disease activity scoring index (EBDASI) in a patient with epidermolysis bullosa acquisita with extensive esophageal involvement. Arch Dermatovenerol Croat. 2018;26:325-328.
  34. Burrows NP, Bhogal BS, Black MM, et al. Bullous eruption of systemic lupus erythematosus: a clinicopathological study of four cases. Br J Dermatol. 1993;128:332-338. doi:10.1111/j.1365-2133.1993.tb00180.x
  35. Aringer M, Leuchten N, Johnson SR. New criteria for lupus. Curr Rheum Rep. 2020;22:18. doi:10.1007/s11926-020-00896-6
  36. Camisa C. Vesiculobullous systemic lupus erythematosus. A report of four cases. J Am Acad Dermatol. 1988;18:93-100. doi:10.1016/s0190-9622(88)70014-6
  37. Duan L, Chen L, Zhong S, et al. Treatment of bullous systemic lupus erythematosus. J Immunol Res. 2015;2015:167064. doi:10.1155/2015/167064
  38. Sprow G, Afarideh M, Dan J, et al. Bullous systemic lupus erythematosus in females. Int J Womens Dermatol. 2022;8:e034. doi:10.1097/JW9.0000000000000034
  39. Contestable JJ, Edhegard KD, Meyerle JH. Bullous systemic lupus erythematosus: a review and update to diagnosis and treatment. Am J Clin Dermatol. 2014;15:517-524. doi:10.1007/s40257-014-0098-0
  40. Fine JD, Mellerio JE. Epidermolysis bullosa. In: Bolognia JL, Jorizzo JL, Schaffer JV (eds), Dermatology (ed 3), Elsevier Saunders; 2012: 501-513.
Issue
Cutis - 116(2)
Issue
Cutis - 116(2)
Page Number
E12-E19
Page Number
E12-E19
Publications
Publications
Topics
Article Type
Display Headline

Type VII Collagen Disorders Simplified

Display Headline

Type VII Collagen Disorders Simplified

Sections
Inside the Article

PRACTICE POINTS

  • The full complement of type VII collagen is required for the normal assembly of anchoring fibrils, whose function is to adhere the basement membrane to the underlying connective tissue of skin and mucous membranes.
  • In the heritable epidermolysis bullosa (EB) family of diseases, only dominant and recessive dystrophic epidermolysis bullosa are caused by partial or total loss of type VII collagen function.
  • New treatments that have been approved for EB include topical gene therapy with COL7A1, topical birch triterpene gel, and skin cells from patients that are genetically corrected with a functional COL7A1 gene.
  • Epidermolysis bullosa acquisita and bullous systemic lupus erythematosus are rare distinct autoimmune subepithelial bullous diseases caused by IgG antibodies that target type VII collagen in the anchoring fibrils.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Thu, 09/04/2025 - 16:34
Un-Gate On Date
Thu, 09/04/2025 - 16:34
Use ProPublica
CFC Schedule Remove Status
Thu, 09/04/2025 - 16:34
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
survey writer start date
Thu, 09/04/2025 - 16:34

Generalized Erythematous Plaques and Pustules in a Pregnant Patient

Article Type
Changed
Thu, 08/28/2025 - 11:47
Display Headline

Generalized Erythematous Plaques and Pustules in a Pregnant Patient

THE DIAGNOSIS: Impetigo Herpetiformis

Histopathology revealed epidermal acanthosis and spongiosis with overlying parakeratosis associated with subcorneal and intracorneal neutrophils, papillary dermal edema, and dermal mixed inflammation with neutrophils and eosinophils. Both direct immunofluorescence and periodic acid–Schiff studies were negative. Blood and pustule cultures were sterile and the skin flora were normal. Based on these findings, a diagnosis of impetigo herpetiformis (IH) was made. The condition improved with systemic and topical steroids, supportive care, and an intravenous infusion of infliximab 5 mg/kg. At 3 weeks’ follow-up, the patient demonstrated near-complete resolution and later delivered successfully at 40 weeks’ gestation without complications.

Impetigo herpetiformis, also known as pustular psoriasis of pregnancy, is an exceedingly rare gestational dermatosis that typically manifests in the third trimester and can be life-threatening for both the mother and fetus. The term was first used in 1872 to describe 5 pregnant women with extensive acute pustular eruptions, all in unstable condition; 4 (80%)of the cases resulted in maternal death, and all resulted in fetal death.1 Impetigo herpetiformis is characterized by pruritic and painful erythematous patches studded at the periphery with subcorneal pustules. Eruptions usually occur in the flexural areas and spread centrifugally, with extension of the lesions peripherally as the center erodes and crusts. Sparing of the face, palms, and soles is expected, and mucosal involvement is rare. Generalized involvement and exfoliation may occur in extreme cases.2 While IH typically manifests during the third trimester, it may occur any time throughout pregnancy or immediately postpartum.3 A few cases have been reported in the puerperium.2 Common symptoms include fever, chills, malaise, anorexia, nausea, vomiting, diarrhea, and arthralgias. Less common complications include hypoalbuminemia and severe hypocalcemia leading to tetany, seizures, and delirium.2,3 While maternal mortality is uncommon, fetal mortality often is a more pressing risk and is attributed to placental insufficiency.3,4 For this reason, early delivery commonly is considered in severe cases.

Whether IH is a separate entity or a variant of pustular psoriasis remains heavily debated. Although the histopathology of IH is identical to pustular psoriasis, the lack of a personal and family history of psoriasis, symptom resolution with delivery, and possible recurrence during successive pregnancies help differentiate IH from generalized pustular psoriasis.2,5 Earlier onset, diffuse involvement, faster progression, and recurrence in subsequent pregnancies all have been linked to a worse prognosis.4

The differential diagnosis for IH includes acute generalized exanthematous pustulosis, pemphigoid gestationis, dermatitis herpetiformis, and subcorneal pustular dermatosis. Acute generalized exanthematous pustulosis is an uncommon severe cutaneous drug reaction characterized by the sudden onset of numerous sterile pustules on erythematous skin within 48 hours of exposure. The most common offending medications include pristinamycin and beta-lactam antibiotics. A high fever, neutrophilic leukocytosis, and hypocalcemia often accompany acute generalized exanthematous pustulosis.6 Prompt diagnosis and withdrawal of the offending drug as well as supportive care and symptomatic treatment are crucial for disease management, as systemic symptoms and even organ involvement may occur.6

Pemphigoid gestationis, also known as gestational pemphigoid or herpes gestationis, is a rare autoimmune blistering disorder that primarily affects pregnant women. It typically manifests in the second or third trimester or shortly after delivery. Clinically, it manifests as an intensely pruritic polymorphic eruption of urticarial papules and plaques accompanied by vesicles and bullae and often is distributed on the abdomen and extends to other body regions. Although the exact etiology is unknown, pemphigoid gestationis is caused by autoantibodies targeting the BP180 and BP230 hemidesmosomal proteins.7 Treatment usually involves systemic corticosteroids and may require additional immunosuppressive therapy. In most cases, patients see resolution within 6 months of delivery.7

Dermatitis herpetiformis is a chronic autoimmune blistering skin disorder characterized by intensely pruritic, grouped vesicles and papules, often distributed symmetrically on extensor surfaces such as the elbows, knees, buttocks, and back. It is closely associated with celiac disease and is triggered by gluten ingestion in genetically predisposed individuals with human leukocyte antigen DQ2 and DQ8 haplotypes. Dermatitis herpetiformis is caused by deposition of IgA antibodies that target tissue transglutaminase 3 at the dermal papillae, leading to inflammation and blister formation. 8 Treatment typically involves a gluten-free diet and medications such as dapsone to alleviate symptoms and prevent recurrence.

Subcorneal pustular dermatosis, also known as Sneddon-Wilkinson disease, is a rare chronic relapsing pustular dermatosis characterized by sterile superficial pustules arranged in annular or circinate patterns on erythematous plaques. It predominantly affects middleaged women and often is associated with underlying conditions such as IgA gammopathy or monoclonal gammopathy of undetermined significance. The pathogenesis remains unclear, but immune dysregulation is thought to play a role. Some authors still question whether subcorneal pustular dermatosis is a distinct entity from pustular psoriasis.4,5,12 Dapsone is the preferred first-line treatment, with adjunct therapies including topical or systemic corticosteroids, other immunosuppressive agents, tumor necrosis factor inhibitors, and UV light therapy.9

Definitive management of IH is achieved through early delivery; however, systemic corticosteroids often are used in varying doses to control the disease and to extend the pregnancy period closer to term or until delivery is considered viable. Additional therapies that can be considered include infliximab, cyclosporine, and topical corticosteroids, in conjunction with fluid and electrolyte maintenance.2,4,10 If symptoms persist despite supportive care and pharmacologic intervention, induction of labor or termination of pregnancy may be indicated. In nonbreastfeeding postpartum mothers with persistent disease, therapies commonly used in generalized pustular psoriasis may be given.11

References
  1. Hebra F. Ueber einzelne wahrend Schwangerschaft, des wacherbette unde bei uterinal. Krankheiten der Frauen zu beobachtende Hautkrankheiten. Wien Med Wochenschr. 1872;48:1197-1202.
  2. Fouda UM, Fouda RM, Ammar HM, et al. Impetigo herpetiformis during the puerperium triggered by secondary hypoparathyroidism: a case report. Cases J. 2009;2:9338. doi:10.1186/1757-1626-2-9338
  3. Kroumpouzos G, Cohen LM. Dermatoses of pregnancy. J Am Acad Dermatol. 2001;45:1-22. doi:10.1067/mjd.2001.114595
  4. Liu J, Ali K, Lou H, et al. First-trimester impetigo herpetiformis leads to stillbirth: a case report. Dermatol Ther (Heidelb). 2022;12:1271-1279. doi:10.1007/s13555-022-00735-9
  5. Lotem M, Katzenelson V, Rotem A, et al. Impetigo herpetiformis: a variant of pustular psoriasis or a separate entity? J Am Acad Dermatol. 1989;20:338-41. doi:10.1016/s0190-9622(89)70042-6
  6. Stadler PC, Oschmann A, Kerl-French K, et al. Acute generalized exanthematous pustulosis: clinical characteristics, pathogenesis, and management. Dermatology. 2023;239:328-333. doi:10.1159/000529218
  7. Abdelhafez MMA, Ahmed KAM, Daud MNBM, et al. Pemphigoid gestationis and adverse pregnancy outcomes: a literature review. J Gynecol Obstet Hum Reprod. 2022;51:102370. doi:10.1016 /j.jogoh.2022.102370
  8. Reunala T, Hervonen K, Salmi T. Dermatitis herpetiformis: an update on diagnosis and management. Am J Clin Dermatol. 2021;22:329-338. doi:10.1007/s40257-020-00584-2
  9. Watts PJ, Khachemoune A. Subcorneal pustular dermatosis: a review of 30 years of progress. Am J Clin Dermatol. 2016;17:653-671. doi:10.1007 /s40257-016-0202-8
  10. Robinson A, Van Voorhees AS, Hsu S, et al. Treatment of pustular psoriasis: from the Medical Board of the National Psoriasis Foundation. J Am Acad Dermatol. 2012;67:279-288. doi:10.1016/j.jaad.2011.01.032
  11. Bukhari IA. Impetigo herpetiformis in a primigravida: successful treatment with etanercept. J Drugs Dermatol. 2004;3:449-451.
  12. Chang SE, Kim HH, Choi JH, et al. Impetigo herpetiformis followed by generalized pustular psoriasis: more evidence of same disease entity. Int J Dermatol. 2003;42(9):754-755.
Article PDF
Author and Disclosure Information

From the College of Medicine, University of Illinois, Peoria; Dr. Kentosh is from the Department of Dermatology. Dr. Kentosh also is from the Soderstrom Skin Institute, Peoria.

The authors have no relevant financial disclosures to report.

Correspondence: Justin Porter, MD, 1 Illini Dr, Peoria, IL 61605 (jporte9@uic.edu).

Cutis. 2025 September;116(3):87, 93, 104. doi:10.12788/cutis.1258

Issue
Cutis - 116(3)
Publications
Topics
Page Number
87, 93, 104
Sections
Author and Disclosure Information

From the College of Medicine, University of Illinois, Peoria; Dr. Kentosh is from the Department of Dermatology. Dr. Kentosh also is from the Soderstrom Skin Institute, Peoria.

The authors have no relevant financial disclosures to report.

Correspondence: Justin Porter, MD, 1 Illini Dr, Peoria, IL 61605 (jporte9@uic.edu).

Cutis. 2025 September;116(3):87, 93, 104. doi:10.12788/cutis.1258

Author and Disclosure Information

From the College of Medicine, University of Illinois, Peoria; Dr. Kentosh is from the Department of Dermatology. Dr. Kentosh also is from the Soderstrom Skin Institute, Peoria.

The authors have no relevant financial disclosures to report.

Correspondence: Justin Porter, MD, 1 Illini Dr, Peoria, IL 61605 (jporte9@uic.edu).

Cutis. 2025 September;116(3):87, 93, 104. doi:10.12788/cutis.1258

Article PDF
Article PDF

THE DIAGNOSIS: Impetigo Herpetiformis

Histopathology revealed epidermal acanthosis and spongiosis with overlying parakeratosis associated with subcorneal and intracorneal neutrophils, papillary dermal edema, and dermal mixed inflammation with neutrophils and eosinophils. Both direct immunofluorescence and periodic acid–Schiff studies were negative. Blood and pustule cultures were sterile and the skin flora were normal. Based on these findings, a diagnosis of impetigo herpetiformis (IH) was made. The condition improved with systemic and topical steroids, supportive care, and an intravenous infusion of infliximab 5 mg/kg. At 3 weeks’ follow-up, the patient demonstrated near-complete resolution and later delivered successfully at 40 weeks’ gestation without complications.

Impetigo herpetiformis, also known as pustular psoriasis of pregnancy, is an exceedingly rare gestational dermatosis that typically manifests in the third trimester and can be life-threatening for both the mother and fetus. The term was first used in 1872 to describe 5 pregnant women with extensive acute pustular eruptions, all in unstable condition; 4 (80%)of the cases resulted in maternal death, and all resulted in fetal death.1 Impetigo herpetiformis is characterized by pruritic and painful erythematous patches studded at the periphery with subcorneal pustules. Eruptions usually occur in the flexural areas and spread centrifugally, with extension of the lesions peripherally as the center erodes and crusts. Sparing of the face, palms, and soles is expected, and mucosal involvement is rare. Generalized involvement and exfoliation may occur in extreme cases.2 While IH typically manifests during the third trimester, it may occur any time throughout pregnancy or immediately postpartum.3 A few cases have been reported in the puerperium.2 Common symptoms include fever, chills, malaise, anorexia, nausea, vomiting, diarrhea, and arthralgias. Less common complications include hypoalbuminemia and severe hypocalcemia leading to tetany, seizures, and delirium.2,3 While maternal mortality is uncommon, fetal mortality often is a more pressing risk and is attributed to placental insufficiency.3,4 For this reason, early delivery commonly is considered in severe cases.

Whether IH is a separate entity or a variant of pustular psoriasis remains heavily debated. Although the histopathology of IH is identical to pustular psoriasis, the lack of a personal and family history of psoriasis, symptom resolution with delivery, and possible recurrence during successive pregnancies help differentiate IH from generalized pustular psoriasis.2,5 Earlier onset, diffuse involvement, faster progression, and recurrence in subsequent pregnancies all have been linked to a worse prognosis.4

The differential diagnosis for IH includes acute generalized exanthematous pustulosis, pemphigoid gestationis, dermatitis herpetiformis, and subcorneal pustular dermatosis. Acute generalized exanthematous pustulosis is an uncommon severe cutaneous drug reaction characterized by the sudden onset of numerous sterile pustules on erythematous skin within 48 hours of exposure. The most common offending medications include pristinamycin and beta-lactam antibiotics. A high fever, neutrophilic leukocytosis, and hypocalcemia often accompany acute generalized exanthematous pustulosis.6 Prompt diagnosis and withdrawal of the offending drug as well as supportive care and symptomatic treatment are crucial for disease management, as systemic symptoms and even organ involvement may occur.6

Pemphigoid gestationis, also known as gestational pemphigoid or herpes gestationis, is a rare autoimmune blistering disorder that primarily affects pregnant women. It typically manifests in the second or third trimester or shortly after delivery. Clinically, it manifests as an intensely pruritic polymorphic eruption of urticarial papules and plaques accompanied by vesicles and bullae and often is distributed on the abdomen and extends to other body regions. Although the exact etiology is unknown, pemphigoid gestationis is caused by autoantibodies targeting the BP180 and BP230 hemidesmosomal proteins.7 Treatment usually involves systemic corticosteroids and may require additional immunosuppressive therapy. In most cases, patients see resolution within 6 months of delivery.7

Dermatitis herpetiformis is a chronic autoimmune blistering skin disorder characterized by intensely pruritic, grouped vesicles and papules, often distributed symmetrically on extensor surfaces such as the elbows, knees, buttocks, and back. It is closely associated with celiac disease and is triggered by gluten ingestion in genetically predisposed individuals with human leukocyte antigen DQ2 and DQ8 haplotypes. Dermatitis herpetiformis is caused by deposition of IgA antibodies that target tissue transglutaminase 3 at the dermal papillae, leading to inflammation and blister formation. 8 Treatment typically involves a gluten-free diet and medications such as dapsone to alleviate symptoms and prevent recurrence.

Subcorneal pustular dermatosis, also known as Sneddon-Wilkinson disease, is a rare chronic relapsing pustular dermatosis characterized by sterile superficial pustules arranged in annular or circinate patterns on erythematous plaques. It predominantly affects middleaged women and often is associated with underlying conditions such as IgA gammopathy or monoclonal gammopathy of undetermined significance. The pathogenesis remains unclear, but immune dysregulation is thought to play a role. Some authors still question whether subcorneal pustular dermatosis is a distinct entity from pustular psoriasis.4,5,12 Dapsone is the preferred first-line treatment, with adjunct therapies including topical or systemic corticosteroids, other immunosuppressive agents, tumor necrosis factor inhibitors, and UV light therapy.9

Definitive management of IH is achieved through early delivery; however, systemic corticosteroids often are used in varying doses to control the disease and to extend the pregnancy period closer to term or until delivery is considered viable. Additional therapies that can be considered include infliximab, cyclosporine, and topical corticosteroids, in conjunction with fluid and electrolyte maintenance.2,4,10 If symptoms persist despite supportive care and pharmacologic intervention, induction of labor or termination of pregnancy may be indicated. In nonbreastfeeding postpartum mothers with persistent disease, therapies commonly used in generalized pustular psoriasis may be given.11

THE DIAGNOSIS: Impetigo Herpetiformis

Histopathology revealed epidermal acanthosis and spongiosis with overlying parakeratosis associated with subcorneal and intracorneal neutrophils, papillary dermal edema, and dermal mixed inflammation with neutrophils and eosinophils. Both direct immunofluorescence and periodic acid–Schiff studies were negative. Blood and pustule cultures were sterile and the skin flora were normal. Based on these findings, a diagnosis of impetigo herpetiformis (IH) was made. The condition improved with systemic and topical steroids, supportive care, and an intravenous infusion of infliximab 5 mg/kg. At 3 weeks’ follow-up, the patient demonstrated near-complete resolution and later delivered successfully at 40 weeks’ gestation without complications.

Impetigo herpetiformis, also known as pustular psoriasis of pregnancy, is an exceedingly rare gestational dermatosis that typically manifests in the third trimester and can be life-threatening for both the mother and fetus. The term was first used in 1872 to describe 5 pregnant women with extensive acute pustular eruptions, all in unstable condition; 4 (80%)of the cases resulted in maternal death, and all resulted in fetal death.1 Impetigo herpetiformis is characterized by pruritic and painful erythematous patches studded at the periphery with subcorneal pustules. Eruptions usually occur in the flexural areas and spread centrifugally, with extension of the lesions peripherally as the center erodes and crusts. Sparing of the face, palms, and soles is expected, and mucosal involvement is rare. Generalized involvement and exfoliation may occur in extreme cases.2 While IH typically manifests during the third trimester, it may occur any time throughout pregnancy or immediately postpartum.3 A few cases have been reported in the puerperium.2 Common symptoms include fever, chills, malaise, anorexia, nausea, vomiting, diarrhea, and arthralgias. Less common complications include hypoalbuminemia and severe hypocalcemia leading to tetany, seizures, and delirium.2,3 While maternal mortality is uncommon, fetal mortality often is a more pressing risk and is attributed to placental insufficiency.3,4 For this reason, early delivery commonly is considered in severe cases.

Whether IH is a separate entity or a variant of pustular psoriasis remains heavily debated. Although the histopathology of IH is identical to pustular psoriasis, the lack of a personal and family history of psoriasis, symptom resolution with delivery, and possible recurrence during successive pregnancies help differentiate IH from generalized pustular psoriasis.2,5 Earlier onset, diffuse involvement, faster progression, and recurrence in subsequent pregnancies all have been linked to a worse prognosis.4

The differential diagnosis for IH includes acute generalized exanthematous pustulosis, pemphigoid gestationis, dermatitis herpetiformis, and subcorneal pustular dermatosis. Acute generalized exanthematous pustulosis is an uncommon severe cutaneous drug reaction characterized by the sudden onset of numerous sterile pustules on erythematous skin within 48 hours of exposure. The most common offending medications include pristinamycin and beta-lactam antibiotics. A high fever, neutrophilic leukocytosis, and hypocalcemia often accompany acute generalized exanthematous pustulosis.6 Prompt diagnosis and withdrawal of the offending drug as well as supportive care and symptomatic treatment are crucial for disease management, as systemic symptoms and even organ involvement may occur.6

Pemphigoid gestationis, also known as gestational pemphigoid or herpes gestationis, is a rare autoimmune blistering disorder that primarily affects pregnant women. It typically manifests in the second or third trimester or shortly after delivery. Clinically, it manifests as an intensely pruritic polymorphic eruption of urticarial papules and plaques accompanied by vesicles and bullae and often is distributed on the abdomen and extends to other body regions. Although the exact etiology is unknown, pemphigoid gestationis is caused by autoantibodies targeting the BP180 and BP230 hemidesmosomal proteins.7 Treatment usually involves systemic corticosteroids and may require additional immunosuppressive therapy. In most cases, patients see resolution within 6 months of delivery.7

Dermatitis herpetiformis is a chronic autoimmune blistering skin disorder characterized by intensely pruritic, grouped vesicles and papules, often distributed symmetrically on extensor surfaces such as the elbows, knees, buttocks, and back. It is closely associated with celiac disease and is triggered by gluten ingestion in genetically predisposed individuals with human leukocyte antigen DQ2 and DQ8 haplotypes. Dermatitis herpetiformis is caused by deposition of IgA antibodies that target tissue transglutaminase 3 at the dermal papillae, leading to inflammation and blister formation. 8 Treatment typically involves a gluten-free diet and medications such as dapsone to alleviate symptoms and prevent recurrence.

Subcorneal pustular dermatosis, also known as Sneddon-Wilkinson disease, is a rare chronic relapsing pustular dermatosis characterized by sterile superficial pustules arranged in annular or circinate patterns on erythematous plaques. It predominantly affects middleaged women and often is associated with underlying conditions such as IgA gammopathy or monoclonal gammopathy of undetermined significance. The pathogenesis remains unclear, but immune dysregulation is thought to play a role. Some authors still question whether subcorneal pustular dermatosis is a distinct entity from pustular psoriasis.4,5,12 Dapsone is the preferred first-line treatment, with adjunct therapies including topical or systemic corticosteroids, other immunosuppressive agents, tumor necrosis factor inhibitors, and UV light therapy.9

Definitive management of IH is achieved through early delivery; however, systemic corticosteroids often are used in varying doses to control the disease and to extend the pregnancy period closer to term or until delivery is considered viable. Additional therapies that can be considered include infliximab, cyclosporine, and topical corticosteroids, in conjunction with fluid and electrolyte maintenance.2,4,10 If symptoms persist despite supportive care and pharmacologic intervention, induction of labor or termination of pregnancy may be indicated. In nonbreastfeeding postpartum mothers with persistent disease, therapies commonly used in generalized pustular psoriasis may be given.11

References
  1. Hebra F. Ueber einzelne wahrend Schwangerschaft, des wacherbette unde bei uterinal. Krankheiten der Frauen zu beobachtende Hautkrankheiten. Wien Med Wochenschr. 1872;48:1197-1202.
  2. Fouda UM, Fouda RM, Ammar HM, et al. Impetigo herpetiformis during the puerperium triggered by secondary hypoparathyroidism: a case report. Cases J. 2009;2:9338. doi:10.1186/1757-1626-2-9338
  3. Kroumpouzos G, Cohen LM. Dermatoses of pregnancy. J Am Acad Dermatol. 2001;45:1-22. doi:10.1067/mjd.2001.114595
  4. Liu J, Ali K, Lou H, et al. First-trimester impetigo herpetiformis leads to stillbirth: a case report. Dermatol Ther (Heidelb). 2022;12:1271-1279. doi:10.1007/s13555-022-00735-9
  5. Lotem M, Katzenelson V, Rotem A, et al. Impetigo herpetiformis: a variant of pustular psoriasis or a separate entity? J Am Acad Dermatol. 1989;20:338-41. doi:10.1016/s0190-9622(89)70042-6
  6. Stadler PC, Oschmann A, Kerl-French K, et al. Acute generalized exanthematous pustulosis: clinical characteristics, pathogenesis, and management. Dermatology. 2023;239:328-333. doi:10.1159/000529218
  7. Abdelhafez MMA, Ahmed KAM, Daud MNBM, et al. Pemphigoid gestationis and adverse pregnancy outcomes: a literature review. J Gynecol Obstet Hum Reprod. 2022;51:102370. doi:10.1016 /j.jogoh.2022.102370
  8. Reunala T, Hervonen K, Salmi T. Dermatitis herpetiformis: an update on diagnosis and management. Am J Clin Dermatol. 2021;22:329-338. doi:10.1007/s40257-020-00584-2
  9. Watts PJ, Khachemoune A. Subcorneal pustular dermatosis: a review of 30 years of progress. Am J Clin Dermatol. 2016;17:653-671. doi:10.1007 /s40257-016-0202-8
  10. Robinson A, Van Voorhees AS, Hsu S, et al. Treatment of pustular psoriasis: from the Medical Board of the National Psoriasis Foundation. J Am Acad Dermatol. 2012;67:279-288. doi:10.1016/j.jaad.2011.01.032
  11. Bukhari IA. Impetigo herpetiformis in a primigravida: successful treatment with etanercept. J Drugs Dermatol. 2004;3:449-451.
  12. Chang SE, Kim HH, Choi JH, et al. Impetigo herpetiformis followed by generalized pustular psoriasis: more evidence of same disease entity. Int J Dermatol. 2003;42(9):754-755.
References
  1. Hebra F. Ueber einzelne wahrend Schwangerschaft, des wacherbette unde bei uterinal. Krankheiten der Frauen zu beobachtende Hautkrankheiten. Wien Med Wochenschr. 1872;48:1197-1202.
  2. Fouda UM, Fouda RM, Ammar HM, et al. Impetigo herpetiformis during the puerperium triggered by secondary hypoparathyroidism: a case report. Cases J. 2009;2:9338. doi:10.1186/1757-1626-2-9338
  3. Kroumpouzos G, Cohen LM. Dermatoses of pregnancy. J Am Acad Dermatol. 2001;45:1-22. doi:10.1067/mjd.2001.114595
  4. Liu J, Ali K, Lou H, et al. First-trimester impetigo herpetiformis leads to stillbirth: a case report. Dermatol Ther (Heidelb). 2022;12:1271-1279. doi:10.1007/s13555-022-00735-9
  5. Lotem M, Katzenelson V, Rotem A, et al. Impetigo herpetiformis: a variant of pustular psoriasis or a separate entity? J Am Acad Dermatol. 1989;20:338-41. doi:10.1016/s0190-9622(89)70042-6
  6. Stadler PC, Oschmann A, Kerl-French K, et al. Acute generalized exanthematous pustulosis: clinical characteristics, pathogenesis, and management. Dermatology. 2023;239:328-333. doi:10.1159/000529218
  7. Abdelhafez MMA, Ahmed KAM, Daud MNBM, et al. Pemphigoid gestationis and adverse pregnancy outcomes: a literature review. J Gynecol Obstet Hum Reprod. 2022;51:102370. doi:10.1016 /j.jogoh.2022.102370
  8. Reunala T, Hervonen K, Salmi T. Dermatitis herpetiformis: an update on diagnosis and management. Am J Clin Dermatol. 2021;22:329-338. doi:10.1007/s40257-020-00584-2
  9. Watts PJ, Khachemoune A. Subcorneal pustular dermatosis: a review of 30 years of progress. Am J Clin Dermatol. 2016;17:653-671. doi:10.1007 /s40257-016-0202-8
  10. Robinson A, Van Voorhees AS, Hsu S, et al. Treatment of pustular psoriasis: from the Medical Board of the National Psoriasis Foundation. J Am Acad Dermatol. 2012;67:279-288. doi:10.1016/j.jaad.2011.01.032
  11. Bukhari IA. Impetigo herpetiformis in a primigravida: successful treatment with etanercept. J Drugs Dermatol. 2004;3:449-451.
  12. Chang SE, Kim HH, Choi JH, et al. Impetigo herpetiformis followed by generalized pustular psoriasis: more evidence of same disease entity. Int J Dermatol. 2003;42(9):754-755.
Issue
Cutis - 116(3)
Issue
Cutis - 116(3)
Page Number
87, 93, 104
Page Number
87, 93, 104
Publications
Publications
Topics
Article Type
Display Headline

Generalized Erythematous Plaques and Pustules in a Pregnant Patient

Display Headline

Generalized Erythematous Plaques and Pustules in a Pregnant Patient

Sections
Questionnaire Body
Porter-Quiz

A 17-year-old girl was admitted to the hospital at 19 weeks' gestation for a widespread eruption of erythematous plaques with pustules covering more than 60% of the body and signs of sepsis. The rash initially appeared as a few small spots on the upper chest and under the breasts 5 weeks prior to hospital admission with subsequent spread to the abdomen and groin. At admission, the patient had a mild fever and tachycardia. She reported a history of eczema, herpes simplex virus, and intertrigo. Physical examination performed by dermatology revealed generalized erythematous plaques with pustule-studded margins and overlying scale involving the neck, torso, arms, and legs favoring the flexural areas. There was no involvement of the face, eyes, oral mucosa, palms, soles, or nails. Laboratory testing revealed hypoalbuminemia (2.4 g/dL [reference range, 3.5-5.5 g/dL]) and elevated inflammatory markers, including leukocytosis (15.83×103μL [reference range, 4.50- 11.00×103/μL]), absolute neutrophil count (12.87×103/μL [reference range, 1.50-8.00×103/μL]), and erythrocyte sedimentation rate (124 mm/h [reference range, 0-20 mm/h]). A culture from an abdominal pustule grew 1 colony of taphylococcus epidermidis, a suspected contaminant. A biopsy from a lesion on the right chest was performed.

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Wed, 08/27/2025 - 16:42
Un-Gate On Date
Wed, 08/27/2025 - 16:42
Use ProPublica
CFC Schedule Remove Status
Wed, 08/27/2025 - 16:42
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
survey writer start date
Wed, 08/27/2025 - 16:42

Fluoroscopy-Induced Chronic Radiation Dermatitis: A Comprehensive Review and Reappraisal

Article Type
Changed
Wed, 08/27/2025 - 16:18
Display Headline

Fluoroscopy-Induced Chronic Radiation Dermatitis: A Comprehensive Review and Reappraisal

Fluoroscopy is an imaging technique that allows for real-time visualization of internal structures in the body using continuous radiography beams. More than 1 million fluoroscopy-guided procedures are performed annually in the United States.1 Utilization of these procedures continues to increase, and so does the probability of related complications, as prolonged exposure to ionizing radiation can cause skin injuries.2 Fortunately, the incidence of radiation-induced skin injuries compared with the total number of fluoroscopic procedures performed remains small,2 although one study suggested the incidence may be as high as 8.9% in at-risk populations.3

Radiation dermatitis is well recognized in dermatology as a complication of oncologic management; however, radiation dermatitis as a complication of fluoroscopic procedures is underrecognized.4 Fluoroscopy-induced radiation dermatitis can be categorized as acute, subacute, or chronic.5 Common fluoroscopic procedures that have been associated with fluoroscopy-induced radiation dermatitis include interventional cardiac procedures, neurovascular procedures, transjugular intrahepatic portosystemic shunt procedures, and endovascular abdominal aortic aneurysm repairs.6,7

Patients with fluoroscopy-induced radiation dermatitis, particularly fluoroscopy-induced chronic radiation dermatitis (FICRD), can present to dermatology up to several years after the initial fluoroscopy procedure with no awareness of the association between the procedure and their skin findings. This presents a diagnostic challenge, and FICRD often is overlooked.5,8-10

We conducted a literature search of PubMed articles indexed for MEDLINE using the search terms ­fluoroscopy and dermatitis. In this reappraisal, we will provide a comprehensive overview of fluoroscopy-induced ­radiation dermatitis with an emphasis on FICRD, covering its clinical manifestations, pathophysiology, risk factors, ­differential diagnosis, histology, and management. The aim of this review is to highlight the salient features and mimickers of FICRD and inform readers how to approach suspected cases, leading to accurate diagnosis and ­effective management.

Pathophysiology 

Fluoroscopy-induced radiation dermatitis is the result of dose-dependent radiation-induced tissue damage. As the peak skin dosage (PSD) of radiation increases over the course of a procedure or multiple procedures, the severity of skin injury predictably increases. During fluoroscopic procedures, the standard irradiation dosage ranges from 0.02 Gy/min to 0.05 Gy/min.11 Transient skin changes may start to be seen around 2 Gy of cumulative exposure. Fluoroscopic procedures typically range in duration from 60 to 120 minutes; however, complex cases may exceed that. Additionally, multiple procedures performed within shorter intervals can result in greater PSD accumulation. Shorter intervals between procedures do not allow enough time for damage repair from the previous ­procedure and can result in further severe damage when the skin is re-exposed to radiation.2 The American College of Radiology recommends medical follow-up after 10 Gy of cumulative exposure, while cumulative exposure above 15 Gy within a 6- to 12-month period is defined as a sentinel event, according to The Joint Commission.12-14

Depending on the patient’s total radiation dosage during one or more procedures, the result of the tissue damage manifests differently at varying times: early skin changes are categorized as fluoroscopy-induced acute radiation dermatitis, and late skin changes are categorized as FICRD (Table 1).

CT116002005_e-Table1

Clinical Manifestations

Acute radiation dermatitis from fluoroscopic procedures manifests within hours to days up to 90 days following radiation exposure and can be characterized by erythema with blistering, desquamation, epilation, pigmentation changes, and even necrosis if the accumulated dosage exceeds 15 Gy.15 Chronic radiation dermatitis (which as related to fluoroscopic procedures is termed FICRD) has a longer onset of weeks to years and is clinically characterized by telangiectasias, permanent erythema, dermal atrophy, or ulcerations. Clinically, subacute radiation dermatitis shares features of both acute and chronic radiation dermatitis; therefore, it is differentiated based on its histologic features.5,16

Although fluoroscopy-induced acute radiation dermatitis (Table 1) may precede FICRD, acute manifestations of fluoroscopy-related dermatitis can be subtle and often manifest in areas not easily visualized. Because referrals to dermatologists for full-skin examinations after fluoroscopy procedures are not standard, patients may not be aware of the association between these procedures and the development of skin lesions. Nonetheless, some patients may report a history of skin changes such as redness days or weeks after a fluoroscopic procedure with accompanying pain and pruritus limited to the fluoroscopy-exposed region, which tend to self-resolve.17 The risk for FICRD is thought to increase if a history of fluoroscopy-induced acute radiation dermatitis is present.18

The location of the skin findings correlates to the area exposed to prolonged radiation during the procedure(s). The most common areas include the scapular and subscapular regions, the right lateral trunk inferior to the axilla, the mid back, and the right anterolateral chest.16,19,20 These regions are associated with more complex (eg, cardiac) procedures that have been reported to lead to prolonged radiation exposure. The skin findings in FICRD are described as geometric, corresponding to the squarish or rectangular radiography beam that is directed at the patient. Additionally, radiography beams spread outward as they travel in space; therefore, skin injuries are common at the region more distal to the path of origination of the beam.21-23 Subsequently, a geometric, dyspigmented, indurated or atrophic plaque with telangiectasias and erosions or ulcerations with progressive worsening is a common manifestation of FICRD.5,16,23 Patients also commonly present with pruritus or severe pain associated with the lesion.24,25  

Dermatologic Manifestations of FICRD

Skin responses seen weeks to years after a fluoroscopic procedure and typically after cumulative radiation exposure of 10 Gy or greater are categorized as FICRD (Table 2). These changes also can be clinically graded based on the Radiation Therapy Oncology Group classification of radiation dermatitis (Tables 3 and 4).26 Chronic changes in the skin largely result from remodeling of the vasculature and the subcutaneous tissue over time. Unlike acute changes, chronic changes typically persist and continue to worsen.27

CT116002005_e-Table2CT116002005_e-Table3CT116002005_e-Table4

Telangiectasias—Anywhere from months to 1 year after exposure to 10 Gy of radiation, proliferation of atypical superficial vessels in the dermis can be seen, typically manifesting as telangiectasias on physical examination. Telangiectasias can increase with time and can even exhibit a dose-dependent relationship to the radiation exposure.28

Atrophy—Atrophic-appearing skin after radiation exposure is the result of direct injury to both the epidermis and fibroblasts in the dermis. The destruction of keratinocytes leads to a thin epidermis, and destruction of dermal fibroblasts causes insufficient collagen production.29 Clinically, this process manifests as an atrophic plaque that can be seen 12 weeks to 1 year after the procedure. 

Fibrosis—Approximately 1 year after the exposure, the initial damage can lead to disruption of molecular pathways, causing fibrosis. Transforming growth factor (TGF) β1 is the main factor involved.29 Damage to the endothelial cells results in increased TGF-β1 levels, which causes increased stimulation of remaining atypical fibroblasts and thus increased irregular collagen deposition.30 Further adding to this knowledge, Wei et al31 recently proposed that damage to the epidermal keratinocytes leads to disruption of yes-associated protein 1, which is a protective factor released from keratinocytes that regulates the dermal fibroblasts. However, extensive damage to the keratinocytes can lead to lower yes-associated protein 1 levels and its downstream activity, leading to increased levels of TGF-β1 and fibroblast activity.31 Clinically, this fibrotic stage is seen as indurated plaques in patients. 

Necrosis—There are 2 forms of necrosis that can be seen. Ischemic dermal necrosis typically occurs in the acute phase after 10 weeks and approximately 18 Gy of cumulative exposure. It results from substantial skin damage, including microvascular damage and reduction in dermal capillaries, leading to ischemia of the tissue.2 Late dermal necrosis is the process seen in the chronic stage of FICRD and radiation dermatitis not related to ­fluoroscopy. It results from the inability of the fibrotic dermis to vascularly support the epidermis above it.2 It can be seen anywhere from 1 to 4 years after the procedure. This stage clinically manifests as worsening ulcerations with major pain and increased risk for secondary infections.16 

Dyspigmentation—Dyspigmentation at the site of the radiation exposure can be seen acutely and chronically. Dosage above 15 to 18 Gy can lead to destruction of melanocytes, which can cause hypopigmentation in exposed areas. However, melanocytes are relatively resistant to radiation; therefore, dosages below the threshold of destruction of 15 to 18 Gy can cause melanocytic hyperactivity leading to hyperpigmentation.32 Hence, pigmentary changes can vary greatly. Classically, a central area of hypopigmentation with surrounding hyperpigmentation is seen.

Histology 

Histologic appearance of radiation dermatitis varies depending on its stage. Acute radiation dermatitis primarily demonstrates superficial dermal edema, damage to the basal cell layer, small vessel dilation with thrombi, and hemorrhage along with a sparse inflammatory cell infiltrate.33 Histology typically is the only way to characterize subacute radiation dermatitis.5 Lichenoid tissue reaction is its characteristic feature. Mononuclear cells are found adjected to necrotic keratinocytes along with prominent vacuolization of the basal cell layer.33

The key histologic features of chronic radiation dermatitis include epidermal atrophy, hyperkeratosis, telangiectasias, loss of adnexal structures, and dermal fibrosis along with sparse atypical stellate fibroblasts.34 However, clinical context of fluoroscopic exposure is required for the dermatopathologist to differentiate chronic radiation dermatitis from its histologic differential of morphea and lichen sclerosus. In a cross-sectional study, only 1 of 6 cases (16.7%) was correctly diagnosed as chronic radiation dermatitis in the absence of correlating clinical history.35 

Risk Factors for FICRD 

Since the diagnosis of FICRD can be a clinical challenge, understanding the risk factors can be helpful. The general likelihood of developing FICRD is related to the duration, frequency, interval, intensity, and area of radiation exposure. Procedures exceeding the normal duration of 60 to 120 minutes have been well documented as a substantial risk factor for radiation dermatitis and FICRD.36-38 The risk tends to be higher in longer procedures because they result in more radiation exposure and higher accumulated PSD. Obesity (ie, body mass index >26) is the major risk factor that has been associated with longer procedure times, as higher radiation dosages are necessary to penetrate the body of a larger patient and a larger skin surface area is exposed.37-39

Other risk factors associated with FICRD relate to how prone a patient is to radiation-induced DNA damage. Older patients are at higher risk due to lower intrinsic ability of the tissue to repair itself.11 Patients with a history of connective tissue diseases—particularly lupus, scleroderma, and mixed connective tissue disease—are at an increased risk.40 Furthermore, patients with genetic disorders that impair DNA repair are more susceptible to radiation-induced DNA damage; therefore, patients with ataxia-telangiectasia, xeroderma pigmentosum, Fanconi anemia, and hereditary nevoid basal cell carcinoma are at higher risk for FICRD.39 Similarly, medications that can affect DNA repair also have been shown to be risk factors. These medications include chemotherapeutic agents such as actinomycin D, cyclophosphamide, doxorubicin, methotrexate, and 5-fluorouracil.2,39 Diabetes, hyperthyroidism, and tobacco use also have been shown to increase a patient’s risk for FICRD.39 It also is reasonable to believe that patients with defects in fibroblasts or with elastin or collagen disorders (eg, Ehlers-Danlos syndrome) would be at higher risk, but there are no known studies highlighting the association in the literature.

Differential Diagnosis of FICRD 

Acute allergic or irritant contact dermatitis manifests with a localized area of erythematous skin accompanied by pruritus.41 Patients with FICRD can present with a localized area of erythema and hyperpigmentation with minimal atrophy. The lesion may accompany substantial pruritus, which can favor the more common diagnosis of contact dermatitis.35,42,43

Fixed-drug eruption manifests as a well-defined, hyperpigmented plaque in a fixed location that occurs upon ingestion of a drug.44 Fluoroscopy-induced chronic radiation dermatitis lesions are well demarcated and geometrically shaped and therefore can mimic lesions seen in fixed-drug eruptions.45 Additionally, the patient population undergoing fluoroscopic procedures tends to have major comorbidities requiring multiple medications.4

Decubitus ulcers are a result of vascular compromise to an area of skin due to constant pressure and are most commonly seen in the sacral region of patients with ­obesity.46 Ulcerated FICRD lesions can manifest on the lower midback. These lesions can be seen after endovascular repair of abdominal aortic aneurysm or prostatic artery embolization.20,21 The location of these lesions can mimic decubitus ulcers if fluoroscopic history is unknown. As mentioned, obesity also increases the risk for FICRD. 

Morphea can manifest as a localized area of induration and hyperpigmentation of the skin.47 When FICRD has progressed to dermal fibrosis, patients can present with indurated plaques without ulcerations, which can be hard to differentiate from morphea.16,48 However, the presence of ulcerations or hyperkeratosis can differentiate morphea from FICRD.16

Ultimately, it is the location of FICRD lesions that remains the biggest diagnostic clue. Any suspicious lesion present on the scapular or subscapular areas, anterolateral chest, and/or mid back should prompt an investigation into recent or remote history of fluoroscopic procedures. 

Management of FICRD

Diagnosis of FICRD should be made clinically based on the history and physical examination whenever possible, since a biopsy is not recommended.35 Wound healing in FICRD is delayed, and biopsies can lead to ulcerations or secondary infections.17 Therefore, it is important to remain suspicious for FICRD. Management of FICRD should correspond to the clinical findings outlined by a recent Delphi consensus survey.49 Regardless, the core of FICRD management framework should always include good hygiene, maintenance of skin hydration to improve epithelialization, and sufficient photoprotection.49,50

Among the first signs of FICRD are telangiectasias. Although asymptomatic, their appearance can be distressing for patients. Pulsed dye laser therapy is a first-line option that has been studied and has shown clinical efficacy for treatment of telangiectasias and vascular changes in patients with FICRD.49,51

If patients develop fibrotic changes, treatment options are limited. Fibrosis is hard to reverse, and the management approach is limited to symptomatic relief. Mechanical and deep-friction massages have been shown to be effective at reducing skin induration in patients.52 Fractional ablative lasers also may be utilized for skin contractures, especially if range of motion is affected.53,54 Although it comes with its own challenges, autologous fat grafting has shown promise in reducing postradiation fibrosis and inducing angiogenesis in tissue.55 Oral pentoxifylline also has shown mild efficacy, as it may be able to suppress TGF-β1 levels.53 However, prevention of fibrotic changes may be the most important. Wei et al31 suggested that low-dose oral prednisolone at 5 mg twice daily for 3 weeks might be an option to prevent the progression of skin changes and even reverse fibrosis to an extent; however, further evidence regarding its efficacy still is necessary. Additionally, no evidence was identified to support the use of topical corticosteroids for fibrotic changes seen in FICRD.56

Patients with FICRD or even acute radiation dermatitis after fluoroscopy tend to develop superficial ulcerations from minor traumas. Good wound hygiene, antiseptic care, and absorbent dressings, such as hydrogel and hydrocolloid, may be sufficient for treating these wounds, as seen in the Figure.42,48 However, once patients develop refractory ulcerations or necrosis, treatment options are then limited to surgical removal with a flap or graft.5,33,42,45
 

CT116002005_e-Fig_AB
FIGURE. A and B, A 65-year-old woman developed this ulcerated lesion 1 month after undergoing a prolonged cardiac stent placement. The lesion showed improvement after use of topical antiseptic and antibiotic/weak steroid for 2 weeks.

Risk for basal cell carcinomas and squamous cell carcinomas is higher in patients with radiation exposure; however, the exact risk from fluoroscopic procedures is unknown. One study demonstrated an increased risk of 6.9% in development of skin cancer after a median radiation exposure of 15.5 Gy and a mean latency period of 38.3 years,57 and in another retrospective study, the risk was higher in Fitzpatrick skin types I and II.58 Unlike the development of radiodermatitis itself, which shows a dose-dependent response, development of skin cancers follows a stochastic pattern (not dose dependent).59 Therefore, it is important to identify these high-risk patients and establish follow-up.

Conclusion

Fluoroscopy-induced chronic radiation dermatitis can be a diagnostic challenge, as skin changes may not be readily associated with the procedure by patients. Therefore, any lesion with a geometric shape and accompanying chronic radiation dermatitis features located on the scapular or subscapular areas, anterolateral chest, and midback should prompt an investigation into history of fluoroscopic procedures. Treatment of chronic skin changes in FICRD depends on the clinical manifestations. Good hygiene, skin hydration, and sufficient photoprotection are crucial. Finally, long-term monitoring with skin examinations is important to assess for the development of skin cancers in the treated area.

References
  1. Benjamin EJ, Muntner P, Alonso A, et al. Heart Disease and Stroke Statistics-2019 Update: a report from the American Heart Association. Circulation. 2019;139:E56-E528. doi:10.1161/CIR.0000000000000659. Published correction appears in Circulation. 2020;141:E33.
  2. Koenig TR, Wolff D, Mettler FA, et al. Skin injuries from fluoroscopically guided procedures: part 1, characteristics of radiation injury. AJR Am J Roentgenol. 2001;177:3-11. doi:10.2214/ajr.177.1.1770003
  3. Guesnier-Dopagne M, Boyer L, Pereira B, et al. Incidence of chronic radiodermatitis after fluoroscopically guided interventions: a retrospective study. J Vasc Interv Radiol. 2019;30:692-698.e13. doi:10.1016/j.jvir.2019.01.010
  4. Cunha N, Cardoso P, Cabete J. Subacute radiation dermatitis following an interventional cardiology procedure. Cutan Ocul Toxicol. 2017;36:297-299. doi:10.1080/15569527.2016.1254649
  5. Frazier TH, Richardson JB, Fabré VC, et al. Fluoroscopy-induced chronic radiation skin injury: a disease perhaps often overlooked. Arch Dermatol. 2007;143:637-640. doi:10.1001/archderm.143.5.637
  6. Koenig TR, Mettler FA, Wagner LK. Skin injuries from fluoroscopically guided procedures: part 2, review of 73 cases and recommendations for minimizing dose delivered to patient. AJR Am J Roentgenol. 2001;177:13-20. doi:10.2214/ajr.177.1.1770013
  7. Shope TB. Radiation-induced skin injuries from fluoroscopy. Radiographics. 1996;16:1195-1199. doi:10.1148/radiographics.16.5.8888398
  8. Tchanque-Fossuo CN, Isseroff RR, Silverstein MA. Fluoroscopy induced chronic radiation dermatitis should be included in the differential diagnosis of notalgia paresthetica. Dermatol Online J. 2016;22:13030/qt0kh726m9.
  9. Berlin L. Radiation-induced skin injuries and fluoroscopy. AJR Am J Roentgenol. 2001;177:21-25. doi:10.2214/ajr.177.1.1770021
  10. Tchanque-Fossuo CN, Kamangar F, Ho B, et al. Fluoroscopy-induced radionecrosis. Dermatol Online J. 2016;22:13030/qt68w910t2.
  11. Wagner LK, Eifel PJ, Geise RA. Potential biological effects following high X-ray dose interventional procedures. J Vasc Interv Radiol. 1994;5:71-84. doi:10.1016/s1051-0443(94)71456-1
  12. Balter S, Hopewell JW, Miller DL, et al. Fluoroscopically guided interventional procedures: a review of radiation effects on patients’ skin and hair. Radiology. 2010;254:326-341. doi:10.1148/radiol.2542082312
  13. Vance AZ, Weinberg BD, Arbique GM, et al. Fluoroscopic sentinel events in neuroendovascular procedures: how to screen, prevent, and address occurrence. AJNR Am J Neuroradiol. 2013;34:1513-1515. doi:10.3174/ajnr.A3185
  14. Aerts A, Decraene T, van den Oord JJ, et al. Chronic radiodermatitis following percutaneous coronary interventions: a report of two cases. J Eur Acad Dermatol Venereol. 2003;17:340-343. doi:10.1046/j.1468-3083.2003.00687.x
  15. Rosenthal A, Israilevich R, Moy R. Management of acute radiation dermatitis: a review of the literature and proposal for treatment algorithm. J Am Acad Dermatol. 2019;81:558-567. doi:10.1016/j.jaad.2019.02.047
  16. Boncher J, Bergfeld WF. Fluoroscopy-induced chronic radiation dermatitis: a report of two additional cases and a brief review of the literature. J Cutan Pathol. 2012;39:63-67. doi:10.1111/j.1600-0560.2011.01754.x
  17. Spiker A, Zinn Z, Carter WH, et al. Fluoroscopy-induced chronic radiation dermatitis. Am J Cardiol. 2012;110:1861-1863. doi:10.1016/j.amjcard.2012.08.023
  18. Batrani M, Kubba A, Sundharam J. Fluoroscopy-induced chronic radiation dermatitis masquerading as morphea: a diagnostic pitfall. Indian J Pathol Microbiol. 2018;61:393-396. doi:10.4103/IJPM.IJPM_566_17
  19. Jeskowiak A, Hubmer M, Prenner G, et al. Radiation induced cutaneous ulcer on the back in a patient with congenital anomaly of the upper cava system. Interact Cardiovasc Thorac Surg. 2011;12:290-292.
  20. Laborda A, De Assis AM, Ioakeim I, et al. Radiodermitis after prostatic artery embolization: case report and review of the literature. Cardiovasc Intervent Radiol. 2015;38:755-759. doi:10.1007/s00270-015-1083-6
  21. Lyons AB, Harvey VM, Gusev J. Fluoroscopy-induced chronic radiation dermatitis (FICRD) after endovascular abdominal aortic aneurysm endoleak repair. JAAD Case Rep. 2015;1:403-405. doi:10.1016/j.jdcr.2015.09.022
  22. Mossman KL. Analysis of risk in computerized tomography and other diagnostic radiology procedures. Comput Radiol. 1982;6:251-256. doi:10.1016/0730-4862(82)90109-3
  23. Henry MF, Maender JL, Shen Y, et al. Fluoroscopy-induced chronic radiation dermatitis: a report of three cases. Dermatol Online J. 2009;15:3.
  24. Balter S, Miller DL. Patient skin reactions from interventional fluoroscopy procedures. AJR Am J Roentgenol. 2014;202:W335-W342. doi:10.2214/AJR.13.12029
  25. Nishimoto S, Fukuda K, Kawai K, et al. Supplementation of bone marrow aspirate-derived platelet-rich plasma for treating radiation-induced ulcer after cardiac fluoroscopic procedures: a preliminary report. Indian J Plast Surg. 2012;45:109-114. doi:10.4103/0970-0358.96599
  26. Cox JD, Stetz J, Pajak TF. Toxicity criteria of the Radiation Therapy Oncology Group (RTOG) and the European Organization for Research and Treatment of Cancer (EORTC). Int J Radiat Oncol Biol Phys. 1995;31:1341-1346. doi:10.1016/0360-3016(95)00060-C
  27. Wong RK, Bensadoun RJ, Boers-Doets CB, et al. Clinical practice guidelines for the prevention and treatment of acute and late radiation reactions from the MASCC Skin Toxicity Study Group. Support Care Cancer. 2013;21:2933-2948. doi:10.1007/s00520-013-1896-2
  28. Turesson I, Notter G. The predictive value of skin telangiectasia for late radiation effects in different normal tissues. Int J Radiat Oncol Biol Phys. 1986;12:603-609. doi:10.1016/0360-3016(86)90069-6
  29. Hegedus F, Mathew LM, Schwartz RA. Radiation dermatitis: an overview. Int J Dermatol. 2017;56:909-914. doi:10.1111/ijd.13371
  30. Denham JW, Hauer-Jensen M. The radiotherapeutic injury—a complex ‘wound.’ Radiother Oncol. 2002;63:129-145. doi:10.1016/s0167-8140(02)00060-9
  31. Wei KC, Lai SF, Huang WL, et al. An innovative targeted therapy for fluoroscopy-induced chronic radiation dermatitis. J Mol Med (Berl). 2022;100:135-146. doi:10.1007/s00109-021-02146-3
  32. Sitton E. Early and late radiation-induced skin alterations. part I: mechanisms of skin changes. Oncol Nurs Forum. 1992;19:801-807.
  33. Pruitt LG, Rogers W, Byarlay JA, et al. Subacute radiation dermatitis after fluoroscopy. J Cutan Pathol. 2016;43:1091-1095. doi:10.1111/cup.12815
  34. Anderson EB, Draft KS, Lee RA, et al. Update in dermatopathology. Am J Clin Pathol. 2006;125(Suppl):S50-S70. doi:10.1309/GMUFNP6LFMPNR86R
  35. Wei KC, Yang KC, Mar GY, et al. STROBE—radiation ulcer: an overlooked complication of fluoroscopic intervention: a cross-sectional study. Medicine (Baltimore). 2015;94:e2178. doi:10.1097/MD.0000000000002178
  36. Otterburn D, Losken A. Iatrogenic fluoroscopy injury to the skin. Ann Plast Surg. 2010;65:462-465. doi:10.1097/SAP.0b013e3181d6e2d3
  37. Cha MJ, Jo SJ, Cho Y, et al. Patient characteristics and the incidence of radiation-induced dermatitis following radiofrequency catheter ablation. Korean Circ J. 2016;46:646-653. doi:10.4070/kcj.2016.46.5.646
  38. Dehen L, Vilmer C, Humilière C, et al. Chronic radiodermatitis following cardiac catheterisation: a report of two cases and a brief review of the literature. Heart. 1999;81:308-312. doi:10.1136/hrt.81.3.308
  39. Brown KR, Rzucidlo E. Acute and chronic radiation injury. J Vasc Surg. 2011;53(Suppl 1):15S-21S. doi:10.1016/j.jvs.2010.06.175. Published correction appears in J Vasc Surg. 2012;55:627.
  40. Hymes SR, Strom EA, Fife C. Radiation dermatitis: clinical presentation, pathophysiology, and treatment 2006. J Am Acad Dermatol. 2006;54:28-46. doi:10.1016/j.jaad.2005.08.054
  41. Scheinman PL, Vocanson M, Thyssen JP, et al. Contact dermatitis. Nat Rev Dis Primers. 2021;7:38. doi:10.1038/s41572-021-00271-4
  42. Cheng TT, Yang HJ. Chronic radiation dermatitis induced by cardiac catheterization: a case report and literature review. Acta Dermatovenerol Alp Pannonica Adriat. 2022;31:147-149.
  43. Minni JP, Nowak M, Usmani A, et al. A unique case of subacute radiodermatitis. Cutis. 2013;91:230-232.
  44. Flowers H, Brodell R, Brents M, et al. Fixed drug eruptions: presentation, diagnosis, and management. South Med J. 2014;107:724-727. doi:10.14423/SMJ.0000000000000195
  45. Hashimoto I, Sedo H, Inatsugi K, et al. Severe radiation-induced injury after cardiac catheter ablation: a case requiring free anterolateral thigh flap and vastus lateralis muscle flap reconstruction on the upper arm. J Plast Reconstr Aesthet Surg. 2008;61:704-708. doi:10.1016/j.bjps.2007.01.003
  46. Mervis JS, Phillips TJ. Pressure ulcers: pathophysiology, epidemiology, risk factors, and presentation. J Am Acad Dermatol. 2019;81:881-890. doi:10.1016/j.jaad.2018.12.069
  47. Careta MF, Romiti R. Localized scleroderma: clinical spectrum and therapeutic update. An Bras Dermatol. 2015;90:62-73. doi:10.1590/abd1806-4841.20152890
  48. Herz-Ruelas ME, Gómez-Flores M, Moxica-Del Angel J, et al. Ulcerated radiodermatitis induced after fluoroscopically guided stent implantation angioplasty. Case Rep Dermatol Med. 2014;2014:768624. doi:10.1155/2014/768624
  49. Wilson BN, Shah R, Menzer C, et al. Consensus on the clinical management of chronic radiation dermatitis and radiation fibrosis: a Delphi survey. Br J Dermatol. 2022;187:1054-1056. doi:10.1111/bjd.21852
  50. Khanna NR, Kumar DP, Laskar SG, et al. Radiation dermatitis: an overview. Indian J Burns. 2013;21:24-31. doi:10.4103/0971-653x.121877
  51. Spalek M. Chronic radiation-induced dermatitis: challenges and solutions. Clin Cosmet Investig Dermatol. 2016;9:473-482. doi:10.2147/CCID.S94320
  52. Bourgeois JF, Gourgou S, Kramar A, et al. A randomized, prospective study using the LPG technique in treating radiation-induced skin fibrosis: clinical and profilometric analysis. Skin Res Technol. 2008;14:71-76. doi:10.1111/j.1600-0846.2007.00263.x
  53. Borrelli MR, Shen AH, Lee GK, et al. Radiation-induced skinfibrosis: pathogenesis, current treatment options, and emerging therapeutics. Ann Plast Surg. 2019;83(4S Suppl 1):S59-S64. doi:10.1097/SAP.0000000000002098
  54. Wilson B, Shah R, Menzer C, et al. Laser therapy as a treatment for chronic radiation fibrosis. Lasers Surg Med. 2023;55:82-88. doi:10.1002/lsm.23617
  55. Rigotti G, Marchi A, Galiè M, et al. Clinical treatment of radiotherapy tissue damage by lipoaspirate transplant: a healing process mediated by adipose-derived adult stem cells. Plast Reconstr Surg. 2007;119:1409-1422. doi:10.1097/01.prs.0000256047.47909.71
  56. Leventhal J, Young MR. Radiation dermatitis: recognition, prevention, and management. Oncology (Williston Park). 2017;31:885-899.
  57. van Vloten WA, Hermans J, van Daal WA. Radiation-induced skin cancer and radiodermatitis of the head and neck. Cancer. 1987;59:411-414. doi:10.1002/1097-0142(19870201)59:3<411::aid-cncr2820590310>3.0.co;2-z
  58. Davis MM, Hanke CW, Zollinger TW, et al. Skin cancer in patients with chronic radiation dermatitis. J Am Acad Dermatol. 1989;20:608-616. doi:10.1016/s0190-9622(89)70072-4
  59. Miller DL, Balter S, Schueler BA, et al. Clinical radiation management for fluoroscopically guided interventional procedures. Radiology. 2010;257:321-332. doi:10.1148/radiol.10091269
Article PDF
Author and Disclosure Information

Dr. Khan is from the Department of Diagnostic Radiology, University of Oklahoma Health Sciences Center, Oklahoma City. Dr. Khachemoune is from the Department of Dermatology, SUNY Downstate Health Sciences University, Brooklyn, New York, and the Department of Dermatology, Veterans Affairs New York Harbor Healthcare System, Brooklyn.

The authors have no relevant financial disclosures to report.

Correspondence: Amor Khachemoune, MD (ORCID 0000-0003-1622-1097), State University of New York Downstate, Veterans Affairs Medical Center, 800 Poly Place, Brooklyn, NY 11209 (amorkh@gmail.com).

Cutis. 2025 August;116(2):E5-E11. doi:10.12788/cutis.1256

Issue
Cutis - 116(2)
Publications
Topics
Page Number
E5-E11
Sections
Author and Disclosure Information

Dr. Khan is from the Department of Diagnostic Radiology, University of Oklahoma Health Sciences Center, Oklahoma City. Dr. Khachemoune is from the Department of Dermatology, SUNY Downstate Health Sciences University, Brooklyn, New York, and the Department of Dermatology, Veterans Affairs New York Harbor Healthcare System, Brooklyn.

The authors have no relevant financial disclosures to report.

Correspondence: Amor Khachemoune, MD (ORCID 0000-0003-1622-1097), State University of New York Downstate, Veterans Affairs Medical Center, 800 Poly Place, Brooklyn, NY 11209 (amorkh@gmail.com).

Cutis. 2025 August;116(2):E5-E11. doi:10.12788/cutis.1256

Author and Disclosure Information

Dr. Khan is from the Department of Diagnostic Radiology, University of Oklahoma Health Sciences Center, Oklahoma City. Dr. Khachemoune is from the Department of Dermatology, SUNY Downstate Health Sciences University, Brooklyn, New York, and the Department of Dermatology, Veterans Affairs New York Harbor Healthcare System, Brooklyn.

The authors have no relevant financial disclosures to report.

Correspondence: Amor Khachemoune, MD (ORCID 0000-0003-1622-1097), State University of New York Downstate, Veterans Affairs Medical Center, 800 Poly Place, Brooklyn, NY 11209 (amorkh@gmail.com).

Cutis. 2025 August;116(2):E5-E11. doi:10.12788/cutis.1256

Article PDF
Article PDF

Fluoroscopy is an imaging technique that allows for real-time visualization of internal structures in the body using continuous radiography beams. More than 1 million fluoroscopy-guided procedures are performed annually in the United States.1 Utilization of these procedures continues to increase, and so does the probability of related complications, as prolonged exposure to ionizing radiation can cause skin injuries.2 Fortunately, the incidence of radiation-induced skin injuries compared with the total number of fluoroscopic procedures performed remains small,2 although one study suggested the incidence may be as high as 8.9% in at-risk populations.3

Radiation dermatitis is well recognized in dermatology as a complication of oncologic management; however, radiation dermatitis as a complication of fluoroscopic procedures is underrecognized.4 Fluoroscopy-induced radiation dermatitis can be categorized as acute, subacute, or chronic.5 Common fluoroscopic procedures that have been associated with fluoroscopy-induced radiation dermatitis include interventional cardiac procedures, neurovascular procedures, transjugular intrahepatic portosystemic shunt procedures, and endovascular abdominal aortic aneurysm repairs.6,7

Patients with fluoroscopy-induced radiation dermatitis, particularly fluoroscopy-induced chronic radiation dermatitis (FICRD), can present to dermatology up to several years after the initial fluoroscopy procedure with no awareness of the association between the procedure and their skin findings. This presents a diagnostic challenge, and FICRD often is overlooked.5,8-10

We conducted a literature search of PubMed articles indexed for MEDLINE using the search terms ­fluoroscopy and dermatitis. In this reappraisal, we will provide a comprehensive overview of fluoroscopy-induced ­radiation dermatitis with an emphasis on FICRD, covering its clinical manifestations, pathophysiology, risk factors, ­differential diagnosis, histology, and management. The aim of this review is to highlight the salient features and mimickers of FICRD and inform readers how to approach suspected cases, leading to accurate diagnosis and ­effective management.

Pathophysiology 

Fluoroscopy-induced radiation dermatitis is the result of dose-dependent radiation-induced tissue damage. As the peak skin dosage (PSD) of radiation increases over the course of a procedure or multiple procedures, the severity of skin injury predictably increases. During fluoroscopic procedures, the standard irradiation dosage ranges from 0.02 Gy/min to 0.05 Gy/min.11 Transient skin changes may start to be seen around 2 Gy of cumulative exposure. Fluoroscopic procedures typically range in duration from 60 to 120 minutes; however, complex cases may exceed that. Additionally, multiple procedures performed within shorter intervals can result in greater PSD accumulation. Shorter intervals between procedures do not allow enough time for damage repair from the previous ­procedure and can result in further severe damage when the skin is re-exposed to radiation.2 The American College of Radiology recommends medical follow-up after 10 Gy of cumulative exposure, while cumulative exposure above 15 Gy within a 6- to 12-month period is defined as a sentinel event, according to The Joint Commission.12-14

Depending on the patient’s total radiation dosage during one or more procedures, the result of the tissue damage manifests differently at varying times: early skin changes are categorized as fluoroscopy-induced acute radiation dermatitis, and late skin changes are categorized as FICRD (Table 1).

CT116002005_e-Table1

Clinical Manifestations

Acute radiation dermatitis from fluoroscopic procedures manifests within hours to days up to 90 days following radiation exposure and can be characterized by erythema with blistering, desquamation, epilation, pigmentation changes, and even necrosis if the accumulated dosage exceeds 15 Gy.15 Chronic radiation dermatitis (which as related to fluoroscopic procedures is termed FICRD) has a longer onset of weeks to years and is clinically characterized by telangiectasias, permanent erythema, dermal atrophy, or ulcerations. Clinically, subacute radiation dermatitis shares features of both acute and chronic radiation dermatitis; therefore, it is differentiated based on its histologic features.5,16

Although fluoroscopy-induced acute radiation dermatitis (Table 1) may precede FICRD, acute manifestations of fluoroscopy-related dermatitis can be subtle and often manifest in areas not easily visualized. Because referrals to dermatologists for full-skin examinations after fluoroscopy procedures are not standard, patients may not be aware of the association between these procedures and the development of skin lesions. Nonetheless, some patients may report a history of skin changes such as redness days or weeks after a fluoroscopic procedure with accompanying pain and pruritus limited to the fluoroscopy-exposed region, which tend to self-resolve.17 The risk for FICRD is thought to increase if a history of fluoroscopy-induced acute radiation dermatitis is present.18

The location of the skin findings correlates to the area exposed to prolonged radiation during the procedure(s). The most common areas include the scapular and subscapular regions, the right lateral trunk inferior to the axilla, the mid back, and the right anterolateral chest.16,19,20 These regions are associated with more complex (eg, cardiac) procedures that have been reported to lead to prolonged radiation exposure. The skin findings in FICRD are described as geometric, corresponding to the squarish or rectangular radiography beam that is directed at the patient. Additionally, radiography beams spread outward as they travel in space; therefore, skin injuries are common at the region more distal to the path of origination of the beam.21-23 Subsequently, a geometric, dyspigmented, indurated or atrophic plaque with telangiectasias and erosions or ulcerations with progressive worsening is a common manifestation of FICRD.5,16,23 Patients also commonly present with pruritus or severe pain associated with the lesion.24,25  

Dermatologic Manifestations of FICRD

Skin responses seen weeks to years after a fluoroscopic procedure and typically after cumulative radiation exposure of 10 Gy or greater are categorized as FICRD (Table 2). These changes also can be clinically graded based on the Radiation Therapy Oncology Group classification of radiation dermatitis (Tables 3 and 4).26 Chronic changes in the skin largely result from remodeling of the vasculature and the subcutaneous tissue over time. Unlike acute changes, chronic changes typically persist and continue to worsen.27

CT116002005_e-Table2CT116002005_e-Table3CT116002005_e-Table4

Telangiectasias—Anywhere from months to 1 year after exposure to 10 Gy of radiation, proliferation of atypical superficial vessels in the dermis can be seen, typically manifesting as telangiectasias on physical examination. Telangiectasias can increase with time and can even exhibit a dose-dependent relationship to the radiation exposure.28

Atrophy—Atrophic-appearing skin after radiation exposure is the result of direct injury to both the epidermis and fibroblasts in the dermis. The destruction of keratinocytes leads to a thin epidermis, and destruction of dermal fibroblasts causes insufficient collagen production.29 Clinically, this process manifests as an atrophic plaque that can be seen 12 weeks to 1 year after the procedure. 

Fibrosis—Approximately 1 year after the exposure, the initial damage can lead to disruption of molecular pathways, causing fibrosis. Transforming growth factor (TGF) β1 is the main factor involved.29 Damage to the endothelial cells results in increased TGF-β1 levels, which causes increased stimulation of remaining atypical fibroblasts and thus increased irregular collagen deposition.30 Further adding to this knowledge, Wei et al31 recently proposed that damage to the epidermal keratinocytes leads to disruption of yes-associated protein 1, which is a protective factor released from keratinocytes that regulates the dermal fibroblasts. However, extensive damage to the keratinocytes can lead to lower yes-associated protein 1 levels and its downstream activity, leading to increased levels of TGF-β1 and fibroblast activity.31 Clinically, this fibrotic stage is seen as indurated plaques in patients. 

Necrosis—There are 2 forms of necrosis that can be seen. Ischemic dermal necrosis typically occurs in the acute phase after 10 weeks and approximately 18 Gy of cumulative exposure. It results from substantial skin damage, including microvascular damage and reduction in dermal capillaries, leading to ischemia of the tissue.2 Late dermal necrosis is the process seen in the chronic stage of FICRD and radiation dermatitis not related to ­fluoroscopy. It results from the inability of the fibrotic dermis to vascularly support the epidermis above it.2 It can be seen anywhere from 1 to 4 years after the procedure. This stage clinically manifests as worsening ulcerations with major pain and increased risk for secondary infections.16 

Dyspigmentation—Dyspigmentation at the site of the radiation exposure can be seen acutely and chronically. Dosage above 15 to 18 Gy can lead to destruction of melanocytes, which can cause hypopigmentation in exposed areas. However, melanocytes are relatively resistant to radiation; therefore, dosages below the threshold of destruction of 15 to 18 Gy can cause melanocytic hyperactivity leading to hyperpigmentation.32 Hence, pigmentary changes can vary greatly. Classically, a central area of hypopigmentation with surrounding hyperpigmentation is seen.

Histology 

Histologic appearance of radiation dermatitis varies depending on its stage. Acute radiation dermatitis primarily demonstrates superficial dermal edema, damage to the basal cell layer, small vessel dilation with thrombi, and hemorrhage along with a sparse inflammatory cell infiltrate.33 Histology typically is the only way to characterize subacute radiation dermatitis.5 Lichenoid tissue reaction is its characteristic feature. Mononuclear cells are found adjected to necrotic keratinocytes along with prominent vacuolization of the basal cell layer.33

The key histologic features of chronic radiation dermatitis include epidermal atrophy, hyperkeratosis, telangiectasias, loss of adnexal structures, and dermal fibrosis along with sparse atypical stellate fibroblasts.34 However, clinical context of fluoroscopic exposure is required for the dermatopathologist to differentiate chronic radiation dermatitis from its histologic differential of morphea and lichen sclerosus. In a cross-sectional study, only 1 of 6 cases (16.7%) was correctly diagnosed as chronic radiation dermatitis in the absence of correlating clinical history.35 

Risk Factors for FICRD 

Since the diagnosis of FICRD can be a clinical challenge, understanding the risk factors can be helpful. The general likelihood of developing FICRD is related to the duration, frequency, interval, intensity, and area of radiation exposure. Procedures exceeding the normal duration of 60 to 120 minutes have been well documented as a substantial risk factor for radiation dermatitis and FICRD.36-38 The risk tends to be higher in longer procedures because they result in more radiation exposure and higher accumulated PSD. Obesity (ie, body mass index >26) is the major risk factor that has been associated with longer procedure times, as higher radiation dosages are necessary to penetrate the body of a larger patient and a larger skin surface area is exposed.37-39

Other risk factors associated with FICRD relate to how prone a patient is to radiation-induced DNA damage. Older patients are at higher risk due to lower intrinsic ability of the tissue to repair itself.11 Patients with a history of connective tissue diseases—particularly lupus, scleroderma, and mixed connective tissue disease—are at an increased risk.40 Furthermore, patients with genetic disorders that impair DNA repair are more susceptible to radiation-induced DNA damage; therefore, patients with ataxia-telangiectasia, xeroderma pigmentosum, Fanconi anemia, and hereditary nevoid basal cell carcinoma are at higher risk for FICRD.39 Similarly, medications that can affect DNA repair also have been shown to be risk factors. These medications include chemotherapeutic agents such as actinomycin D, cyclophosphamide, doxorubicin, methotrexate, and 5-fluorouracil.2,39 Diabetes, hyperthyroidism, and tobacco use also have been shown to increase a patient’s risk for FICRD.39 It also is reasonable to believe that patients with defects in fibroblasts or with elastin or collagen disorders (eg, Ehlers-Danlos syndrome) would be at higher risk, but there are no known studies highlighting the association in the literature.

Differential Diagnosis of FICRD 

Acute allergic or irritant contact dermatitis manifests with a localized area of erythematous skin accompanied by pruritus.41 Patients with FICRD can present with a localized area of erythema and hyperpigmentation with minimal atrophy. The lesion may accompany substantial pruritus, which can favor the more common diagnosis of contact dermatitis.35,42,43

Fixed-drug eruption manifests as a well-defined, hyperpigmented plaque in a fixed location that occurs upon ingestion of a drug.44 Fluoroscopy-induced chronic radiation dermatitis lesions are well demarcated and geometrically shaped and therefore can mimic lesions seen in fixed-drug eruptions.45 Additionally, the patient population undergoing fluoroscopic procedures tends to have major comorbidities requiring multiple medications.4

Decubitus ulcers are a result of vascular compromise to an area of skin due to constant pressure and are most commonly seen in the sacral region of patients with ­obesity.46 Ulcerated FICRD lesions can manifest on the lower midback. These lesions can be seen after endovascular repair of abdominal aortic aneurysm or prostatic artery embolization.20,21 The location of these lesions can mimic decubitus ulcers if fluoroscopic history is unknown. As mentioned, obesity also increases the risk for FICRD. 

Morphea can manifest as a localized area of induration and hyperpigmentation of the skin.47 When FICRD has progressed to dermal fibrosis, patients can present with indurated plaques without ulcerations, which can be hard to differentiate from morphea.16,48 However, the presence of ulcerations or hyperkeratosis can differentiate morphea from FICRD.16

Ultimately, it is the location of FICRD lesions that remains the biggest diagnostic clue. Any suspicious lesion present on the scapular or subscapular areas, anterolateral chest, and/or mid back should prompt an investigation into recent or remote history of fluoroscopic procedures. 

Management of FICRD

Diagnosis of FICRD should be made clinically based on the history and physical examination whenever possible, since a biopsy is not recommended.35 Wound healing in FICRD is delayed, and biopsies can lead to ulcerations or secondary infections.17 Therefore, it is important to remain suspicious for FICRD. Management of FICRD should correspond to the clinical findings outlined by a recent Delphi consensus survey.49 Regardless, the core of FICRD management framework should always include good hygiene, maintenance of skin hydration to improve epithelialization, and sufficient photoprotection.49,50

Among the first signs of FICRD are telangiectasias. Although asymptomatic, their appearance can be distressing for patients. Pulsed dye laser therapy is a first-line option that has been studied and has shown clinical efficacy for treatment of telangiectasias and vascular changes in patients with FICRD.49,51

If patients develop fibrotic changes, treatment options are limited. Fibrosis is hard to reverse, and the management approach is limited to symptomatic relief. Mechanical and deep-friction massages have been shown to be effective at reducing skin induration in patients.52 Fractional ablative lasers also may be utilized for skin contractures, especially if range of motion is affected.53,54 Although it comes with its own challenges, autologous fat grafting has shown promise in reducing postradiation fibrosis and inducing angiogenesis in tissue.55 Oral pentoxifylline also has shown mild efficacy, as it may be able to suppress TGF-β1 levels.53 However, prevention of fibrotic changes may be the most important. Wei et al31 suggested that low-dose oral prednisolone at 5 mg twice daily for 3 weeks might be an option to prevent the progression of skin changes and even reverse fibrosis to an extent; however, further evidence regarding its efficacy still is necessary. Additionally, no evidence was identified to support the use of topical corticosteroids for fibrotic changes seen in FICRD.56

Patients with FICRD or even acute radiation dermatitis after fluoroscopy tend to develop superficial ulcerations from minor traumas. Good wound hygiene, antiseptic care, and absorbent dressings, such as hydrogel and hydrocolloid, may be sufficient for treating these wounds, as seen in the Figure.42,48 However, once patients develop refractory ulcerations or necrosis, treatment options are then limited to surgical removal with a flap or graft.5,33,42,45
 

CT116002005_e-Fig_AB
FIGURE. A and B, A 65-year-old woman developed this ulcerated lesion 1 month after undergoing a prolonged cardiac stent placement. The lesion showed improvement after use of topical antiseptic and antibiotic/weak steroid for 2 weeks.

Risk for basal cell carcinomas and squamous cell carcinomas is higher in patients with radiation exposure; however, the exact risk from fluoroscopic procedures is unknown. One study demonstrated an increased risk of 6.9% in development of skin cancer after a median radiation exposure of 15.5 Gy and a mean latency period of 38.3 years,57 and in another retrospective study, the risk was higher in Fitzpatrick skin types I and II.58 Unlike the development of radiodermatitis itself, which shows a dose-dependent response, development of skin cancers follows a stochastic pattern (not dose dependent).59 Therefore, it is important to identify these high-risk patients and establish follow-up.

Conclusion

Fluoroscopy-induced chronic radiation dermatitis can be a diagnostic challenge, as skin changes may not be readily associated with the procedure by patients. Therefore, any lesion with a geometric shape and accompanying chronic radiation dermatitis features located on the scapular or subscapular areas, anterolateral chest, and midback should prompt an investigation into history of fluoroscopic procedures. Treatment of chronic skin changes in FICRD depends on the clinical manifestations. Good hygiene, skin hydration, and sufficient photoprotection are crucial. Finally, long-term monitoring with skin examinations is important to assess for the development of skin cancers in the treated area.

Fluoroscopy is an imaging technique that allows for real-time visualization of internal structures in the body using continuous radiography beams. More than 1 million fluoroscopy-guided procedures are performed annually in the United States.1 Utilization of these procedures continues to increase, and so does the probability of related complications, as prolonged exposure to ionizing radiation can cause skin injuries.2 Fortunately, the incidence of radiation-induced skin injuries compared with the total number of fluoroscopic procedures performed remains small,2 although one study suggested the incidence may be as high as 8.9% in at-risk populations.3

Radiation dermatitis is well recognized in dermatology as a complication of oncologic management; however, radiation dermatitis as a complication of fluoroscopic procedures is underrecognized.4 Fluoroscopy-induced radiation dermatitis can be categorized as acute, subacute, or chronic.5 Common fluoroscopic procedures that have been associated with fluoroscopy-induced radiation dermatitis include interventional cardiac procedures, neurovascular procedures, transjugular intrahepatic portosystemic shunt procedures, and endovascular abdominal aortic aneurysm repairs.6,7

Patients with fluoroscopy-induced radiation dermatitis, particularly fluoroscopy-induced chronic radiation dermatitis (FICRD), can present to dermatology up to several years after the initial fluoroscopy procedure with no awareness of the association between the procedure and their skin findings. This presents a diagnostic challenge, and FICRD often is overlooked.5,8-10

We conducted a literature search of PubMed articles indexed for MEDLINE using the search terms ­fluoroscopy and dermatitis. In this reappraisal, we will provide a comprehensive overview of fluoroscopy-induced ­radiation dermatitis with an emphasis on FICRD, covering its clinical manifestations, pathophysiology, risk factors, ­differential diagnosis, histology, and management. The aim of this review is to highlight the salient features and mimickers of FICRD and inform readers how to approach suspected cases, leading to accurate diagnosis and ­effective management.

Pathophysiology 

Fluoroscopy-induced radiation dermatitis is the result of dose-dependent radiation-induced tissue damage. As the peak skin dosage (PSD) of radiation increases over the course of a procedure or multiple procedures, the severity of skin injury predictably increases. During fluoroscopic procedures, the standard irradiation dosage ranges from 0.02 Gy/min to 0.05 Gy/min.11 Transient skin changes may start to be seen around 2 Gy of cumulative exposure. Fluoroscopic procedures typically range in duration from 60 to 120 minutes; however, complex cases may exceed that. Additionally, multiple procedures performed within shorter intervals can result in greater PSD accumulation. Shorter intervals between procedures do not allow enough time for damage repair from the previous ­procedure and can result in further severe damage when the skin is re-exposed to radiation.2 The American College of Radiology recommends medical follow-up after 10 Gy of cumulative exposure, while cumulative exposure above 15 Gy within a 6- to 12-month period is defined as a sentinel event, according to The Joint Commission.12-14

Depending on the patient’s total radiation dosage during one or more procedures, the result of the tissue damage manifests differently at varying times: early skin changes are categorized as fluoroscopy-induced acute radiation dermatitis, and late skin changes are categorized as FICRD (Table 1).

CT116002005_e-Table1

Clinical Manifestations

Acute radiation dermatitis from fluoroscopic procedures manifests within hours to days up to 90 days following radiation exposure and can be characterized by erythema with blistering, desquamation, epilation, pigmentation changes, and even necrosis if the accumulated dosage exceeds 15 Gy.15 Chronic radiation dermatitis (which as related to fluoroscopic procedures is termed FICRD) has a longer onset of weeks to years and is clinically characterized by telangiectasias, permanent erythema, dermal atrophy, or ulcerations. Clinically, subacute radiation dermatitis shares features of both acute and chronic radiation dermatitis; therefore, it is differentiated based on its histologic features.5,16

Although fluoroscopy-induced acute radiation dermatitis (Table 1) may precede FICRD, acute manifestations of fluoroscopy-related dermatitis can be subtle and often manifest in areas not easily visualized. Because referrals to dermatologists for full-skin examinations after fluoroscopy procedures are not standard, patients may not be aware of the association between these procedures and the development of skin lesions. Nonetheless, some patients may report a history of skin changes such as redness days or weeks after a fluoroscopic procedure with accompanying pain and pruritus limited to the fluoroscopy-exposed region, which tend to self-resolve.17 The risk for FICRD is thought to increase if a history of fluoroscopy-induced acute radiation dermatitis is present.18

The location of the skin findings correlates to the area exposed to prolonged radiation during the procedure(s). The most common areas include the scapular and subscapular regions, the right lateral trunk inferior to the axilla, the mid back, and the right anterolateral chest.16,19,20 These regions are associated with more complex (eg, cardiac) procedures that have been reported to lead to prolonged radiation exposure. The skin findings in FICRD are described as geometric, corresponding to the squarish or rectangular radiography beam that is directed at the patient. Additionally, radiography beams spread outward as they travel in space; therefore, skin injuries are common at the region more distal to the path of origination of the beam.21-23 Subsequently, a geometric, dyspigmented, indurated or atrophic plaque with telangiectasias and erosions or ulcerations with progressive worsening is a common manifestation of FICRD.5,16,23 Patients also commonly present with pruritus or severe pain associated with the lesion.24,25  

Dermatologic Manifestations of FICRD

Skin responses seen weeks to years after a fluoroscopic procedure and typically after cumulative radiation exposure of 10 Gy or greater are categorized as FICRD (Table 2). These changes also can be clinically graded based on the Radiation Therapy Oncology Group classification of radiation dermatitis (Tables 3 and 4).26 Chronic changes in the skin largely result from remodeling of the vasculature and the subcutaneous tissue over time. Unlike acute changes, chronic changes typically persist and continue to worsen.27

CT116002005_e-Table2CT116002005_e-Table3CT116002005_e-Table4

Telangiectasias—Anywhere from months to 1 year after exposure to 10 Gy of radiation, proliferation of atypical superficial vessels in the dermis can be seen, typically manifesting as telangiectasias on physical examination. Telangiectasias can increase with time and can even exhibit a dose-dependent relationship to the radiation exposure.28

Atrophy—Atrophic-appearing skin after radiation exposure is the result of direct injury to both the epidermis and fibroblasts in the dermis. The destruction of keratinocytes leads to a thin epidermis, and destruction of dermal fibroblasts causes insufficient collagen production.29 Clinically, this process manifests as an atrophic plaque that can be seen 12 weeks to 1 year after the procedure. 

Fibrosis—Approximately 1 year after the exposure, the initial damage can lead to disruption of molecular pathways, causing fibrosis. Transforming growth factor (TGF) β1 is the main factor involved.29 Damage to the endothelial cells results in increased TGF-β1 levels, which causes increased stimulation of remaining atypical fibroblasts and thus increased irregular collagen deposition.30 Further adding to this knowledge, Wei et al31 recently proposed that damage to the epidermal keratinocytes leads to disruption of yes-associated protein 1, which is a protective factor released from keratinocytes that regulates the dermal fibroblasts. However, extensive damage to the keratinocytes can lead to lower yes-associated protein 1 levels and its downstream activity, leading to increased levels of TGF-β1 and fibroblast activity.31 Clinically, this fibrotic stage is seen as indurated plaques in patients. 

Necrosis—There are 2 forms of necrosis that can be seen. Ischemic dermal necrosis typically occurs in the acute phase after 10 weeks and approximately 18 Gy of cumulative exposure. It results from substantial skin damage, including microvascular damage and reduction in dermal capillaries, leading to ischemia of the tissue.2 Late dermal necrosis is the process seen in the chronic stage of FICRD and radiation dermatitis not related to ­fluoroscopy. It results from the inability of the fibrotic dermis to vascularly support the epidermis above it.2 It can be seen anywhere from 1 to 4 years after the procedure. This stage clinically manifests as worsening ulcerations with major pain and increased risk for secondary infections.16 

Dyspigmentation—Dyspigmentation at the site of the radiation exposure can be seen acutely and chronically. Dosage above 15 to 18 Gy can lead to destruction of melanocytes, which can cause hypopigmentation in exposed areas. However, melanocytes are relatively resistant to radiation; therefore, dosages below the threshold of destruction of 15 to 18 Gy can cause melanocytic hyperactivity leading to hyperpigmentation.32 Hence, pigmentary changes can vary greatly. Classically, a central area of hypopigmentation with surrounding hyperpigmentation is seen.

Histology 

Histologic appearance of radiation dermatitis varies depending on its stage. Acute radiation dermatitis primarily demonstrates superficial dermal edema, damage to the basal cell layer, small vessel dilation with thrombi, and hemorrhage along with a sparse inflammatory cell infiltrate.33 Histology typically is the only way to characterize subacute radiation dermatitis.5 Lichenoid tissue reaction is its characteristic feature. Mononuclear cells are found adjected to necrotic keratinocytes along with prominent vacuolization of the basal cell layer.33

The key histologic features of chronic radiation dermatitis include epidermal atrophy, hyperkeratosis, telangiectasias, loss of adnexal structures, and dermal fibrosis along with sparse atypical stellate fibroblasts.34 However, clinical context of fluoroscopic exposure is required for the dermatopathologist to differentiate chronic radiation dermatitis from its histologic differential of morphea and lichen sclerosus. In a cross-sectional study, only 1 of 6 cases (16.7%) was correctly diagnosed as chronic radiation dermatitis in the absence of correlating clinical history.35 

Risk Factors for FICRD 

Since the diagnosis of FICRD can be a clinical challenge, understanding the risk factors can be helpful. The general likelihood of developing FICRD is related to the duration, frequency, interval, intensity, and area of radiation exposure. Procedures exceeding the normal duration of 60 to 120 minutes have been well documented as a substantial risk factor for radiation dermatitis and FICRD.36-38 The risk tends to be higher in longer procedures because they result in more radiation exposure and higher accumulated PSD. Obesity (ie, body mass index >26) is the major risk factor that has been associated with longer procedure times, as higher radiation dosages are necessary to penetrate the body of a larger patient and a larger skin surface area is exposed.37-39

Other risk factors associated with FICRD relate to how prone a patient is to radiation-induced DNA damage. Older patients are at higher risk due to lower intrinsic ability of the tissue to repair itself.11 Patients with a history of connective tissue diseases—particularly lupus, scleroderma, and mixed connective tissue disease—are at an increased risk.40 Furthermore, patients with genetic disorders that impair DNA repair are more susceptible to radiation-induced DNA damage; therefore, patients with ataxia-telangiectasia, xeroderma pigmentosum, Fanconi anemia, and hereditary nevoid basal cell carcinoma are at higher risk for FICRD.39 Similarly, medications that can affect DNA repair also have been shown to be risk factors. These medications include chemotherapeutic agents such as actinomycin D, cyclophosphamide, doxorubicin, methotrexate, and 5-fluorouracil.2,39 Diabetes, hyperthyroidism, and tobacco use also have been shown to increase a patient’s risk for FICRD.39 It also is reasonable to believe that patients with defects in fibroblasts or with elastin or collagen disorders (eg, Ehlers-Danlos syndrome) would be at higher risk, but there are no known studies highlighting the association in the literature.

Differential Diagnosis of FICRD 

Acute allergic or irritant contact dermatitis manifests with a localized area of erythematous skin accompanied by pruritus.41 Patients with FICRD can present with a localized area of erythema and hyperpigmentation with minimal atrophy. The lesion may accompany substantial pruritus, which can favor the more common diagnosis of contact dermatitis.35,42,43

Fixed-drug eruption manifests as a well-defined, hyperpigmented plaque in a fixed location that occurs upon ingestion of a drug.44 Fluoroscopy-induced chronic radiation dermatitis lesions are well demarcated and geometrically shaped and therefore can mimic lesions seen in fixed-drug eruptions.45 Additionally, the patient population undergoing fluoroscopic procedures tends to have major comorbidities requiring multiple medications.4

Decubitus ulcers are a result of vascular compromise to an area of skin due to constant pressure and are most commonly seen in the sacral region of patients with ­obesity.46 Ulcerated FICRD lesions can manifest on the lower midback. These lesions can be seen after endovascular repair of abdominal aortic aneurysm or prostatic artery embolization.20,21 The location of these lesions can mimic decubitus ulcers if fluoroscopic history is unknown. As mentioned, obesity also increases the risk for FICRD. 

Morphea can manifest as a localized area of induration and hyperpigmentation of the skin.47 When FICRD has progressed to dermal fibrosis, patients can present with indurated plaques without ulcerations, which can be hard to differentiate from morphea.16,48 However, the presence of ulcerations or hyperkeratosis can differentiate morphea from FICRD.16

Ultimately, it is the location of FICRD lesions that remains the biggest diagnostic clue. Any suspicious lesion present on the scapular or subscapular areas, anterolateral chest, and/or mid back should prompt an investigation into recent or remote history of fluoroscopic procedures. 

Management of FICRD

Diagnosis of FICRD should be made clinically based on the history and physical examination whenever possible, since a biopsy is not recommended.35 Wound healing in FICRD is delayed, and biopsies can lead to ulcerations or secondary infections.17 Therefore, it is important to remain suspicious for FICRD. Management of FICRD should correspond to the clinical findings outlined by a recent Delphi consensus survey.49 Regardless, the core of FICRD management framework should always include good hygiene, maintenance of skin hydration to improve epithelialization, and sufficient photoprotection.49,50

Among the first signs of FICRD are telangiectasias. Although asymptomatic, their appearance can be distressing for patients. Pulsed dye laser therapy is a first-line option that has been studied and has shown clinical efficacy for treatment of telangiectasias and vascular changes in patients with FICRD.49,51

If patients develop fibrotic changes, treatment options are limited. Fibrosis is hard to reverse, and the management approach is limited to symptomatic relief. Mechanical and deep-friction massages have been shown to be effective at reducing skin induration in patients.52 Fractional ablative lasers also may be utilized for skin contractures, especially if range of motion is affected.53,54 Although it comes with its own challenges, autologous fat grafting has shown promise in reducing postradiation fibrosis and inducing angiogenesis in tissue.55 Oral pentoxifylline also has shown mild efficacy, as it may be able to suppress TGF-β1 levels.53 However, prevention of fibrotic changes may be the most important. Wei et al31 suggested that low-dose oral prednisolone at 5 mg twice daily for 3 weeks might be an option to prevent the progression of skin changes and even reverse fibrosis to an extent; however, further evidence regarding its efficacy still is necessary. Additionally, no evidence was identified to support the use of topical corticosteroids for fibrotic changes seen in FICRD.56

Patients with FICRD or even acute radiation dermatitis after fluoroscopy tend to develop superficial ulcerations from minor traumas. Good wound hygiene, antiseptic care, and absorbent dressings, such as hydrogel and hydrocolloid, may be sufficient for treating these wounds, as seen in the Figure.42,48 However, once patients develop refractory ulcerations or necrosis, treatment options are then limited to surgical removal with a flap or graft.5,33,42,45
 

CT116002005_e-Fig_AB
FIGURE. A and B, A 65-year-old woman developed this ulcerated lesion 1 month after undergoing a prolonged cardiac stent placement. The lesion showed improvement after use of topical antiseptic and antibiotic/weak steroid for 2 weeks.

Risk for basal cell carcinomas and squamous cell carcinomas is higher in patients with radiation exposure; however, the exact risk from fluoroscopic procedures is unknown. One study demonstrated an increased risk of 6.9% in development of skin cancer after a median radiation exposure of 15.5 Gy and a mean latency period of 38.3 years,57 and in another retrospective study, the risk was higher in Fitzpatrick skin types I and II.58 Unlike the development of radiodermatitis itself, which shows a dose-dependent response, development of skin cancers follows a stochastic pattern (not dose dependent).59 Therefore, it is important to identify these high-risk patients and establish follow-up.

Conclusion

Fluoroscopy-induced chronic radiation dermatitis can be a diagnostic challenge, as skin changes may not be readily associated with the procedure by patients. Therefore, any lesion with a geometric shape and accompanying chronic radiation dermatitis features located on the scapular or subscapular areas, anterolateral chest, and midback should prompt an investigation into history of fluoroscopic procedures. Treatment of chronic skin changes in FICRD depends on the clinical manifestations. Good hygiene, skin hydration, and sufficient photoprotection are crucial. Finally, long-term monitoring with skin examinations is important to assess for the development of skin cancers in the treated area.

References
  1. Benjamin EJ, Muntner P, Alonso A, et al. Heart Disease and Stroke Statistics-2019 Update: a report from the American Heart Association. Circulation. 2019;139:E56-E528. doi:10.1161/CIR.0000000000000659. Published correction appears in Circulation. 2020;141:E33.
  2. Koenig TR, Wolff D, Mettler FA, et al. Skin injuries from fluoroscopically guided procedures: part 1, characteristics of radiation injury. AJR Am J Roentgenol. 2001;177:3-11. doi:10.2214/ajr.177.1.1770003
  3. Guesnier-Dopagne M, Boyer L, Pereira B, et al. Incidence of chronic radiodermatitis after fluoroscopically guided interventions: a retrospective study. J Vasc Interv Radiol. 2019;30:692-698.e13. doi:10.1016/j.jvir.2019.01.010
  4. Cunha N, Cardoso P, Cabete J. Subacute radiation dermatitis following an interventional cardiology procedure. Cutan Ocul Toxicol. 2017;36:297-299. doi:10.1080/15569527.2016.1254649
  5. Frazier TH, Richardson JB, Fabré VC, et al. Fluoroscopy-induced chronic radiation skin injury: a disease perhaps often overlooked. Arch Dermatol. 2007;143:637-640. doi:10.1001/archderm.143.5.637
  6. Koenig TR, Mettler FA, Wagner LK. Skin injuries from fluoroscopically guided procedures: part 2, review of 73 cases and recommendations for minimizing dose delivered to patient. AJR Am J Roentgenol. 2001;177:13-20. doi:10.2214/ajr.177.1.1770013
  7. Shope TB. Radiation-induced skin injuries from fluoroscopy. Radiographics. 1996;16:1195-1199. doi:10.1148/radiographics.16.5.8888398
  8. Tchanque-Fossuo CN, Isseroff RR, Silverstein MA. Fluoroscopy induced chronic radiation dermatitis should be included in the differential diagnosis of notalgia paresthetica. Dermatol Online J. 2016;22:13030/qt0kh726m9.
  9. Berlin L. Radiation-induced skin injuries and fluoroscopy. AJR Am J Roentgenol. 2001;177:21-25. doi:10.2214/ajr.177.1.1770021
  10. Tchanque-Fossuo CN, Kamangar F, Ho B, et al. Fluoroscopy-induced radionecrosis. Dermatol Online J. 2016;22:13030/qt68w910t2.
  11. Wagner LK, Eifel PJ, Geise RA. Potential biological effects following high X-ray dose interventional procedures. J Vasc Interv Radiol. 1994;5:71-84. doi:10.1016/s1051-0443(94)71456-1
  12. Balter S, Hopewell JW, Miller DL, et al. Fluoroscopically guided interventional procedures: a review of radiation effects on patients’ skin and hair. Radiology. 2010;254:326-341. doi:10.1148/radiol.2542082312
  13. Vance AZ, Weinberg BD, Arbique GM, et al. Fluoroscopic sentinel events in neuroendovascular procedures: how to screen, prevent, and address occurrence. AJNR Am J Neuroradiol. 2013;34:1513-1515. doi:10.3174/ajnr.A3185
  14. Aerts A, Decraene T, van den Oord JJ, et al. Chronic radiodermatitis following percutaneous coronary interventions: a report of two cases. J Eur Acad Dermatol Venereol. 2003;17:340-343. doi:10.1046/j.1468-3083.2003.00687.x
  15. Rosenthal A, Israilevich R, Moy R. Management of acute radiation dermatitis: a review of the literature and proposal for treatment algorithm. J Am Acad Dermatol. 2019;81:558-567. doi:10.1016/j.jaad.2019.02.047
  16. Boncher J, Bergfeld WF. Fluoroscopy-induced chronic radiation dermatitis: a report of two additional cases and a brief review of the literature. J Cutan Pathol. 2012;39:63-67. doi:10.1111/j.1600-0560.2011.01754.x
  17. Spiker A, Zinn Z, Carter WH, et al. Fluoroscopy-induced chronic radiation dermatitis. Am J Cardiol. 2012;110:1861-1863. doi:10.1016/j.amjcard.2012.08.023
  18. Batrani M, Kubba A, Sundharam J. Fluoroscopy-induced chronic radiation dermatitis masquerading as morphea: a diagnostic pitfall. Indian J Pathol Microbiol. 2018;61:393-396. doi:10.4103/IJPM.IJPM_566_17
  19. Jeskowiak A, Hubmer M, Prenner G, et al. Radiation induced cutaneous ulcer on the back in a patient with congenital anomaly of the upper cava system. Interact Cardiovasc Thorac Surg. 2011;12:290-292.
  20. Laborda A, De Assis AM, Ioakeim I, et al. Radiodermitis after prostatic artery embolization: case report and review of the literature. Cardiovasc Intervent Radiol. 2015;38:755-759. doi:10.1007/s00270-015-1083-6
  21. Lyons AB, Harvey VM, Gusev J. Fluoroscopy-induced chronic radiation dermatitis (FICRD) after endovascular abdominal aortic aneurysm endoleak repair. JAAD Case Rep. 2015;1:403-405. doi:10.1016/j.jdcr.2015.09.022
  22. Mossman KL. Analysis of risk in computerized tomography and other diagnostic radiology procedures. Comput Radiol. 1982;6:251-256. doi:10.1016/0730-4862(82)90109-3
  23. Henry MF, Maender JL, Shen Y, et al. Fluoroscopy-induced chronic radiation dermatitis: a report of three cases. Dermatol Online J. 2009;15:3.
  24. Balter S, Miller DL. Patient skin reactions from interventional fluoroscopy procedures. AJR Am J Roentgenol. 2014;202:W335-W342. doi:10.2214/AJR.13.12029
  25. Nishimoto S, Fukuda K, Kawai K, et al. Supplementation of bone marrow aspirate-derived platelet-rich plasma for treating radiation-induced ulcer after cardiac fluoroscopic procedures: a preliminary report. Indian J Plast Surg. 2012;45:109-114. doi:10.4103/0970-0358.96599
  26. Cox JD, Stetz J, Pajak TF. Toxicity criteria of the Radiation Therapy Oncology Group (RTOG) and the European Organization for Research and Treatment of Cancer (EORTC). Int J Radiat Oncol Biol Phys. 1995;31:1341-1346. doi:10.1016/0360-3016(95)00060-C
  27. Wong RK, Bensadoun RJ, Boers-Doets CB, et al. Clinical practice guidelines for the prevention and treatment of acute and late radiation reactions from the MASCC Skin Toxicity Study Group. Support Care Cancer. 2013;21:2933-2948. doi:10.1007/s00520-013-1896-2
  28. Turesson I, Notter G. The predictive value of skin telangiectasia for late radiation effects in different normal tissues. Int J Radiat Oncol Biol Phys. 1986;12:603-609. doi:10.1016/0360-3016(86)90069-6
  29. Hegedus F, Mathew LM, Schwartz RA. Radiation dermatitis: an overview. Int J Dermatol. 2017;56:909-914. doi:10.1111/ijd.13371
  30. Denham JW, Hauer-Jensen M. The radiotherapeutic injury—a complex ‘wound.’ Radiother Oncol. 2002;63:129-145. doi:10.1016/s0167-8140(02)00060-9
  31. Wei KC, Lai SF, Huang WL, et al. An innovative targeted therapy for fluoroscopy-induced chronic radiation dermatitis. J Mol Med (Berl). 2022;100:135-146. doi:10.1007/s00109-021-02146-3
  32. Sitton E. Early and late radiation-induced skin alterations. part I: mechanisms of skin changes. Oncol Nurs Forum. 1992;19:801-807.
  33. Pruitt LG, Rogers W, Byarlay JA, et al. Subacute radiation dermatitis after fluoroscopy. J Cutan Pathol. 2016;43:1091-1095. doi:10.1111/cup.12815
  34. Anderson EB, Draft KS, Lee RA, et al. Update in dermatopathology. Am J Clin Pathol. 2006;125(Suppl):S50-S70. doi:10.1309/GMUFNP6LFMPNR86R
  35. Wei KC, Yang KC, Mar GY, et al. STROBE—radiation ulcer: an overlooked complication of fluoroscopic intervention: a cross-sectional study. Medicine (Baltimore). 2015;94:e2178. doi:10.1097/MD.0000000000002178
  36. Otterburn D, Losken A. Iatrogenic fluoroscopy injury to the skin. Ann Plast Surg. 2010;65:462-465. doi:10.1097/SAP.0b013e3181d6e2d3
  37. Cha MJ, Jo SJ, Cho Y, et al. Patient characteristics and the incidence of radiation-induced dermatitis following radiofrequency catheter ablation. Korean Circ J. 2016;46:646-653. doi:10.4070/kcj.2016.46.5.646
  38. Dehen L, Vilmer C, Humilière C, et al. Chronic radiodermatitis following cardiac catheterisation: a report of two cases and a brief review of the literature. Heart. 1999;81:308-312. doi:10.1136/hrt.81.3.308
  39. Brown KR, Rzucidlo E. Acute and chronic radiation injury. J Vasc Surg. 2011;53(Suppl 1):15S-21S. doi:10.1016/j.jvs.2010.06.175. Published correction appears in J Vasc Surg. 2012;55:627.
  40. Hymes SR, Strom EA, Fife C. Radiation dermatitis: clinical presentation, pathophysiology, and treatment 2006. J Am Acad Dermatol. 2006;54:28-46. doi:10.1016/j.jaad.2005.08.054
  41. Scheinman PL, Vocanson M, Thyssen JP, et al. Contact dermatitis. Nat Rev Dis Primers. 2021;7:38. doi:10.1038/s41572-021-00271-4
  42. Cheng TT, Yang HJ. Chronic radiation dermatitis induced by cardiac catheterization: a case report and literature review. Acta Dermatovenerol Alp Pannonica Adriat. 2022;31:147-149.
  43. Minni JP, Nowak M, Usmani A, et al. A unique case of subacute radiodermatitis. Cutis. 2013;91:230-232.
  44. Flowers H, Brodell R, Brents M, et al. Fixed drug eruptions: presentation, diagnosis, and management. South Med J. 2014;107:724-727. doi:10.14423/SMJ.0000000000000195
  45. Hashimoto I, Sedo H, Inatsugi K, et al. Severe radiation-induced injury after cardiac catheter ablation: a case requiring free anterolateral thigh flap and vastus lateralis muscle flap reconstruction on the upper arm. J Plast Reconstr Aesthet Surg. 2008;61:704-708. doi:10.1016/j.bjps.2007.01.003
  46. Mervis JS, Phillips TJ. Pressure ulcers: pathophysiology, epidemiology, risk factors, and presentation. J Am Acad Dermatol. 2019;81:881-890. doi:10.1016/j.jaad.2018.12.069
  47. Careta MF, Romiti R. Localized scleroderma: clinical spectrum and therapeutic update. An Bras Dermatol. 2015;90:62-73. doi:10.1590/abd1806-4841.20152890
  48. Herz-Ruelas ME, Gómez-Flores M, Moxica-Del Angel J, et al. Ulcerated radiodermatitis induced after fluoroscopically guided stent implantation angioplasty. Case Rep Dermatol Med. 2014;2014:768624. doi:10.1155/2014/768624
  49. Wilson BN, Shah R, Menzer C, et al. Consensus on the clinical management of chronic radiation dermatitis and radiation fibrosis: a Delphi survey. Br J Dermatol. 2022;187:1054-1056. doi:10.1111/bjd.21852
  50. Khanna NR, Kumar DP, Laskar SG, et al. Radiation dermatitis: an overview. Indian J Burns. 2013;21:24-31. doi:10.4103/0971-653x.121877
  51. Spalek M. Chronic radiation-induced dermatitis: challenges and solutions. Clin Cosmet Investig Dermatol. 2016;9:473-482. doi:10.2147/CCID.S94320
  52. Bourgeois JF, Gourgou S, Kramar A, et al. A randomized, prospective study using the LPG technique in treating radiation-induced skin fibrosis: clinical and profilometric analysis. Skin Res Technol. 2008;14:71-76. doi:10.1111/j.1600-0846.2007.00263.x
  53. Borrelli MR, Shen AH, Lee GK, et al. Radiation-induced skinfibrosis: pathogenesis, current treatment options, and emerging therapeutics. Ann Plast Surg. 2019;83(4S Suppl 1):S59-S64. doi:10.1097/SAP.0000000000002098
  54. Wilson B, Shah R, Menzer C, et al. Laser therapy as a treatment for chronic radiation fibrosis. Lasers Surg Med. 2023;55:82-88. doi:10.1002/lsm.23617
  55. Rigotti G, Marchi A, Galiè M, et al. Clinical treatment of radiotherapy tissue damage by lipoaspirate transplant: a healing process mediated by adipose-derived adult stem cells. Plast Reconstr Surg. 2007;119:1409-1422. doi:10.1097/01.prs.0000256047.47909.71
  56. Leventhal J, Young MR. Radiation dermatitis: recognition, prevention, and management. Oncology (Williston Park). 2017;31:885-899.
  57. van Vloten WA, Hermans J, van Daal WA. Radiation-induced skin cancer and radiodermatitis of the head and neck. Cancer. 1987;59:411-414. doi:10.1002/1097-0142(19870201)59:3<411::aid-cncr2820590310>3.0.co;2-z
  58. Davis MM, Hanke CW, Zollinger TW, et al. Skin cancer in patients with chronic radiation dermatitis. J Am Acad Dermatol. 1989;20:608-616. doi:10.1016/s0190-9622(89)70072-4
  59. Miller DL, Balter S, Schueler BA, et al. Clinical radiation management for fluoroscopically guided interventional procedures. Radiology. 2010;257:321-332. doi:10.1148/radiol.10091269
References
  1. Benjamin EJ, Muntner P, Alonso A, et al. Heart Disease and Stroke Statistics-2019 Update: a report from the American Heart Association. Circulation. 2019;139:E56-E528. doi:10.1161/CIR.0000000000000659. Published correction appears in Circulation. 2020;141:E33.
  2. Koenig TR, Wolff D, Mettler FA, et al. Skin injuries from fluoroscopically guided procedures: part 1, characteristics of radiation injury. AJR Am J Roentgenol. 2001;177:3-11. doi:10.2214/ajr.177.1.1770003
  3. Guesnier-Dopagne M, Boyer L, Pereira B, et al. Incidence of chronic radiodermatitis after fluoroscopically guided interventions: a retrospective study. J Vasc Interv Radiol. 2019;30:692-698.e13. doi:10.1016/j.jvir.2019.01.010
  4. Cunha N, Cardoso P, Cabete J. Subacute radiation dermatitis following an interventional cardiology procedure. Cutan Ocul Toxicol. 2017;36:297-299. doi:10.1080/15569527.2016.1254649
  5. Frazier TH, Richardson JB, Fabré VC, et al. Fluoroscopy-induced chronic radiation skin injury: a disease perhaps often overlooked. Arch Dermatol. 2007;143:637-640. doi:10.1001/archderm.143.5.637
  6. Koenig TR, Mettler FA, Wagner LK. Skin injuries from fluoroscopically guided procedures: part 2, review of 73 cases and recommendations for minimizing dose delivered to patient. AJR Am J Roentgenol. 2001;177:13-20. doi:10.2214/ajr.177.1.1770013
  7. Shope TB. Radiation-induced skin injuries from fluoroscopy. Radiographics. 1996;16:1195-1199. doi:10.1148/radiographics.16.5.8888398
  8. Tchanque-Fossuo CN, Isseroff RR, Silverstein MA. Fluoroscopy induced chronic radiation dermatitis should be included in the differential diagnosis of notalgia paresthetica. Dermatol Online J. 2016;22:13030/qt0kh726m9.
  9. Berlin L. Radiation-induced skin injuries and fluoroscopy. AJR Am J Roentgenol. 2001;177:21-25. doi:10.2214/ajr.177.1.1770021
  10. Tchanque-Fossuo CN, Kamangar F, Ho B, et al. Fluoroscopy-induced radionecrosis. Dermatol Online J. 2016;22:13030/qt68w910t2.
  11. Wagner LK, Eifel PJ, Geise RA. Potential biological effects following high X-ray dose interventional procedures. J Vasc Interv Radiol. 1994;5:71-84. doi:10.1016/s1051-0443(94)71456-1
  12. Balter S, Hopewell JW, Miller DL, et al. Fluoroscopically guided interventional procedures: a review of radiation effects on patients’ skin and hair. Radiology. 2010;254:326-341. doi:10.1148/radiol.2542082312
  13. Vance AZ, Weinberg BD, Arbique GM, et al. Fluoroscopic sentinel events in neuroendovascular procedures: how to screen, prevent, and address occurrence. AJNR Am J Neuroradiol. 2013;34:1513-1515. doi:10.3174/ajnr.A3185
  14. Aerts A, Decraene T, van den Oord JJ, et al. Chronic radiodermatitis following percutaneous coronary interventions: a report of two cases. J Eur Acad Dermatol Venereol. 2003;17:340-343. doi:10.1046/j.1468-3083.2003.00687.x
  15. Rosenthal A, Israilevich R, Moy R. Management of acute radiation dermatitis: a review of the literature and proposal for treatment algorithm. J Am Acad Dermatol. 2019;81:558-567. doi:10.1016/j.jaad.2019.02.047
  16. Boncher J, Bergfeld WF. Fluoroscopy-induced chronic radiation dermatitis: a report of two additional cases and a brief review of the literature. J Cutan Pathol. 2012;39:63-67. doi:10.1111/j.1600-0560.2011.01754.x
  17. Spiker A, Zinn Z, Carter WH, et al. Fluoroscopy-induced chronic radiation dermatitis. Am J Cardiol. 2012;110:1861-1863. doi:10.1016/j.amjcard.2012.08.023
  18. Batrani M, Kubba A, Sundharam J. Fluoroscopy-induced chronic radiation dermatitis masquerading as morphea: a diagnostic pitfall. Indian J Pathol Microbiol. 2018;61:393-396. doi:10.4103/IJPM.IJPM_566_17
  19. Jeskowiak A, Hubmer M, Prenner G, et al. Radiation induced cutaneous ulcer on the back in a patient with congenital anomaly of the upper cava system. Interact Cardiovasc Thorac Surg. 2011;12:290-292.
  20. Laborda A, De Assis AM, Ioakeim I, et al. Radiodermitis after prostatic artery embolization: case report and review of the literature. Cardiovasc Intervent Radiol. 2015;38:755-759. doi:10.1007/s00270-015-1083-6
  21. Lyons AB, Harvey VM, Gusev J. Fluoroscopy-induced chronic radiation dermatitis (FICRD) after endovascular abdominal aortic aneurysm endoleak repair. JAAD Case Rep. 2015;1:403-405. doi:10.1016/j.jdcr.2015.09.022
  22. Mossman KL. Analysis of risk in computerized tomography and other diagnostic radiology procedures. Comput Radiol. 1982;6:251-256. doi:10.1016/0730-4862(82)90109-3
  23. Henry MF, Maender JL, Shen Y, et al. Fluoroscopy-induced chronic radiation dermatitis: a report of three cases. Dermatol Online J. 2009;15:3.
  24. Balter S, Miller DL. Patient skin reactions from interventional fluoroscopy procedures. AJR Am J Roentgenol. 2014;202:W335-W342. doi:10.2214/AJR.13.12029
  25. Nishimoto S, Fukuda K, Kawai K, et al. Supplementation of bone marrow aspirate-derived platelet-rich plasma for treating radiation-induced ulcer after cardiac fluoroscopic procedures: a preliminary report. Indian J Plast Surg. 2012;45:109-114. doi:10.4103/0970-0358.96599
  26. Cox JD, Stetz J, Pajak TF. Toxicity criteria of the Radiation Therapy Oncology Group (RTOG) and the European Organization for Research and Treatment of Cancer (EORTC). Int J Radiat Oncol Biol Phys. 1995;31:1341-1346. doi:10.1016/0360-3016(95)00060-C
  27. Wong RK, Bensadoun RJ, Boers-Doets CB, et al. Clinical practice guidelines for the prevention and treatment of acute and late radiation reactions from the MASCC Skin Toxicity Study Group. Support Care Cancer. 2013;21:2933-2948. doi:10.1007/s00520-013-1896-2
  28. Turesson I, Notter G. The predictive value of skin telangiectasia for late radiation effects in different normal tissues. Int J Radiat Oncol Biol Phys. 1986;12:603-609. doi:10.1016/0360-3016(86)90069-6
  29. Hegedus F, Mathew LM, Schwartz RA. Radiation dermatitis: an overview. Int J Dermatol. 2017;56:909-914. doi:10.1111/ijd.13371
  30. Denham JW, Hauer-Jensen M. The radiotherapeutic injury—a complex ‘wound.’ Radiother Oncol. 2002;63:129-145. doi:10.1016/s0167-8140(02)00060-9
  31. Wei KC, Lai SF, Huang WL, et al. An innovative targeted therapy for fluoroscopy-induced chronic radiation dermatitis. J Mol Med (Berl). 2022;100:135-146. doi:10.1007/s00109-021-02146-3
  32. Sitton E. Early and late radiation-induced skin alterations. part I: mechanisms of skin changes. Oncol Nurs Forum. 1992;19:801-807.
  33. Pruitt LG, Rogers W, Byarlay JA, et al. Subacute radiation dermatitis after fluoroscopy. J Cutan Pathol. 2016;43:1091-1095. doi:10.1111/cup.12815
  34. Anderson EB, Draft KS, Lee RA, et al. Update in dermatopathology. Am J Clin Pathol. 2006;125(Suppl):S50-S70. doi:10.1309/GMUFNP6LFMPNR86R
  35. Wei KC, Yang KC, Mar GY, et al. STROBE—radiation ulcer: an overlooked complication of fluoroscopic intervention: a cross-sectional study. Medicine (Baltimore). 2015;94:e2178. doi:10.1097/MD.0000000000002178
  36. Otterburn D, Losken A. Iatrogenic fluoroscopy injury to the skin. Ann Plast Surg. 2010;65:462-465. doi:10.1097/SAP.0b013e3181d6e2d3
  37. Cha MJ, Jo SJ, Cho Y, et al. Patient characteristics and the incidence of radiation-induced dermatitis following radiofrequency catheter ablation. Korean Circ J. 2016;46:646-653. doi:10.4070/kcj.2016.46.5.646
  38. Dehen L, Vilmer C, Humilière C, et al. Chronic radiodermatitis following cardiac catheterisation: a report of two cases and a brief review of the literature. Heart. 1999;81:308-312. doi:10.1136/hrt.81.3.308
  39. Brown KR, Rzucidlo E. Acute and chronic radiation injury. J Vasc Surg. 2011;53(Suppl 1):15S-21S. doi:10.1016/j.jvs.2010.06.175. Published correction appears in J Vasc Surg. 2012;55:627.
  40. Hymes SR, Strom EA, Fife C. Radiation dermatitis: clinical presentation, pathophysiology, and treatment 2006. J Am Acad Dermatol. 2006;54:28-46. doi:10.1016/j.jaad.2005.08.054
  41. Scheinman PL, Vocanson M, Thyssen JP, et al. Contact dermatitis. Nat Rev Dis Primers. 2021;7:38. doi:10.1038/s41572-021-00271-4
  42. Cheng TT, Yang HJ. Chronic radiation dermatitis induced by cardiac catheterization: a case report and literature review. Acta Dermatovenerol Alp Pannonica Adriat. 2022;31:147-149.
  43. Minni JP, Nowak M, Usmani A, et al. A unique case of subacute radiodermatitis. Cutis. 2013;91:230-232.
  44. Flowers H, Brodell R, Brents M, et al. Fixed drug eruptions: presentation, diagnosis, and management. South Med J. 2014;107:724-727. doi:10.14423/SMJ.0000000000000195
  45. Hashimoto I, Sedo H, Inatsugi K, et al. Severe radiation-induced injury after cardiac catheter ablation: a case requiring free anterolateral thigh flap and vastus lateralis muscle flap reconstruction on the upper arm. J Plast Reconstr Aesthet Surg. 2008;61:704-708. doi:10.1016/j.bjps.2007.01.003
  46. Mervis JS, Phillips TJ. Pressure ulcers: pathophysiology, epidemiology, risk factors, and presentation. J Am Acad Dermatol. 2019;81:881-890. doi:10.1016/j.jaad.2018.12.069
  47. Careta MF, Romiti R. Localized scleroderma: clinical spectrum and therapeutic update. An Bras Dermatol. 2015;90:62-73. doi:10.1590/abd1806-4841.20152890
  48. Herz-Ruelas ME, Gómez-Flores M, Moxica-Del Angel J, et al. Ulcerated radiodermatitis induced after fluoroscopically guided stent implantation angioplasty. Case Rep Dermatol Med. 2014;2014:768624. doi:10.1155/2014/768624
  49. Wilson BN, Shah R, Menzer C, et al. Consensus on the clinical management of chronic radiation dermatitis and radiation fibrosis: a Delphi survey. Br J Dermatol. 2022;187:1054-1056. doi:10.1111/bjd.21852
  50. Khanna NR, Kumar DP, Laskar SG, et al. Radiation dermatitis: an overview. Indian J Burns. 2013;21:24-31. doi:10.4103/0971-653x.121877
  51. Spalek M. Chronic radiation-induced dermatitis: challenges and solutions. Clin Cosmet Investig Dermatol. 2016;9:473-482. doi:10.2147/CCID.S94320
  52. Bourgeois JF, Gourgou S, Kramar A, et al. A randomized, prospective study using the LPG technique in treating radiation-induced skin fibrosis: clinical and profilometric analysis. Skin Res Technol. 2008;14:71-76. doi:10.1111/j.1600-0846.2007.00263.x
  53. Borrelli MR, Shen AH, Lee GK, et al. Radiation-induced skinfibrosis: pathogenesis, current treatment options, and emerging therapeutics. Ann Plast Surg. 2019;83(4S Suppl 1):S59-S64. doi:10.1097/SAP.0000000000002098
  54. Wilson B, Shah R, Menzer C, et al. Laser therapy as a treatment for chronic radiation fibrosis. Lasers Surg Med. 2023;55:82-88. doi:10.1002/lsm.23617
  55. Rigotti G, Marchi A, Galiè M, et al. Clinical treatment of radiotherapy tissue damage by lipoaspirate transplant: a healing process mediated by adipose-derived adult stem cells. Plast Reconstr Surg. 2007;119:1409-1422. doi:10.1097/01.prs.0000256047.47909.71
  56. Leventhal J, Young MR. Radiation dermatitis: recognition, prevention, and management. Oncology (Williston Park). 2017;31:885-899.
  57. van Vloten WA, Hermans J, van Daal WA. Radiation-induced skin cancer and radiodermatitis of the head and neck. Cancer. 1987;59:411-414. doi:10.1002/1097-0142(19870201)59:3<411::aid-cncr2820590310>3.0.co;2-z
  58. Davis MM, Hanke CW, Zollinger TW, et al. Skin cancer in patients with chronic radiation dermatitis. J Am Acad Dermatol. 1989;20:608-616. doi:10.1016/s0190-9622(89)70072-4
  59. Miller DL, Balter S, Schueler BA, et al. Clinical radiation management for fluoroscopically guided interventional procedures. Radiology. 2010;257:321-332. doi:10.1148/radiol.10091269
Issue
Cutis - 116(2)
Issue
Cutis - 116(2)
Page Number
E5-E11
Page Number
E5-E11
Publications
Publications
Topics
Article Type
Display Headline

Fluoroscopy-Induced Chronic Radiation Dermatitis: A Comprehensive Review and Reappraisal

Display Headline

Fluoroscopy-Induced Chronic Radiation Dermatitis: A Comprehensive Review and Reappraisal

Sections
Inside the Article

PRACTICE POINTS

  • Fluoroscopy-induced chronic radiation dermatitis poses diagnostic challenges, as patients often are unable to associate a history of fluoroscopic procedures with the development of skin lesions.
  • Scapular and subscapular lesions as well as those on the anterolateral chest and mid back should prompt clinicians to inquire about the patient’s history of fluoroscopic procedures.
  • Because lesions can remain refractory to treatment, longterm monitoring is necessary if they are not excised.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Wed, 08/27/2025 - 15:52
Un-Gate On Date
Wed, 08/27/2025 - 15:52
Use ProPublica
CFC Schedule Remove Status
Wed, 08/27/2025 - 15:52
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
survey writer start date
Wed, 08/27/2025 - 15:52