Annual Skin Check: Examining the Dermatology Headlines of 2019

Article Type
Changed
Tue, 02/18/2020 - 09:09
Display Headline
Annual Skin Check: Examining the Dermatology Headlines of 2019

From chemical sunscreen to the measles outbreak and drug approvals to product recalls, dermatology experienced its share of firsts and controversies in 2019.

Chemical Sunscreen Controversies

Controversial concerns about the effects of chemical sunscreen on coral reefs took an unprecedented turn in the United States this last year. On February 5, 2019, an ordinance was passed in Key West, Florida, prohibiting the sale of sunscreen containing the organic UV filters oxybenzone and/or octinoxate within city limits.1 On June 25, 2019, a similar law that also included octocrylene was passed in the US Virgin Islands.2 In so doing, these areas joined Hawaii, the Republic of Palau, and parts of Mexico in restricting chemical sunscreen sales.1 Although the Key West ordinance is set to take effect in January 2021, opponents, including dermatologists who believe it will discourage sunscreen use, currently are trying to overturn the ban.3 In the US Virgin Islands, part of the ban went into effect in September 2019, with the rest of the ban set to start in March 2020.2 Companies have started to follow suit. On August 1, 2019, CVS Pharmacy announced that, by the end of 2020, it will remove oxybenzone and octinoxate from some of its store-brand chemical sunscreens.4

On February 26, 2019, the US Food and Drug Administration (FDA) proposed that there are insufficient data to determine if 12 organic UV filters—including the aforementioned oxybenzone, octinoxate, and octocrylene—are generally recognized as safe and effective (GRASE).5 Although these ingredients were listed as GRASE by the FDA in 2011, the rise in sunscreen use since then, as well as changes in sunscreen formulations, prompted the FDA to ask manufacturers to perform additional studies on safety parameters such as systemic absorption.5,6 One study conducted by the FDA itself was published in May 2019 and showed that maximal use of 4 sunscreens resulted in systemic absorption of 4 organic UV filters above 0.5 ng/mL, the FDA’s threshold for requiring nonclinical toxicology assessment. The study authors concluded that “further studies [are needed] to determine the clinical significance of these findings. [But] These results do not indicate that individuals should refrain from the use of sunscreen.”7 Some in the industry have suggested it may take at least 5 years to generate all the data the FDA has requested.6

End of the New York City Measles Outbreak

On September 3, 2019, New York City’s largest measles outbreak in nearly 30 years was declared over. This announcement reflected the fact that 2 incubation periods for measles—42 days—had passed since the last measles patient was considered contagious. In total, there were 654 cases of measles and 52 associated hospitalizations, including 16 admissions to the intensive care unit. Most patients were younger than 18 years and unvaccinated.8

The outbreak began in October 2018 after Orthodox Jewish children from Brooklyn became infected while visiting Israel and imported the measles virus upon their return home.8,9 All 5 boroughs in New York City were ultimately affected, although 4 zip codes in Williamsburg, a neighborhood in Brooklyn with an undervaccinated Orthodox Jewish community, accounted for 72% of cases.8,10 As part of a $6 million effort to stop the outbreak, an emergency order was placed on these 4 zip codes, posing potential fines on people living or working there if they were unvaccinated.8 In addition, a bill was passed and signed into law in New York State that eliminated religious exemptions for immunizations.11 In collaboration with Jewish leaders, these efforts increased the administration of measles-mumps-rubella vaccines by 41% compared with the year before in Williamsburg and Borough Park, another heavily Orthodox Jewish neighborhood in Brooklyn.8

Drug Approvals for Pediatric Dermatology

On March 11, 2019, the IL-4/IL-13 inhibitor dupilumab became the third biologic with a pediatric dermatology indication when the FDA extended its approval to adolescents for the treatment of atopic dermatitis.12 The FDA approval was based on a randomized, double-blind, placebo-controlled trial in which 42% (34/82) of adolescents treated with dupilumab monotherapy every other week achieved 75% or more improvement in the Eczema Area and Severity Index at week 16 compared with 8% (7/85) in the placebo group (P<.001).13

In October 2019, trifarotene cream and minocycline foam were approved by the FDA for the treatment of acne in patients 9 years and older.14,15 As such, both became the first acne therapies to include patients as young as 9 years in their studies and indication—a milestone, considering the fact that children have historically been excluded from clinical trials.16 The 2 topical treatments also are noteworthy for being first in class: trifarotene cream is the only topical retinoid to selectively target the retinoic acid receptor γ and to have been studied specifically for both facial and truncal acne,14,17 and minocycline foam is the first topical tetracycline.15

 

 

Drug Approvals for Rare Dermatologic Diseases

On July 19, 2019, apremilast, a phosphodiesterase 4 inhibitor, became the first medication approved by the FDA for the treatment of adults with oral ulcers due to Behçet disease, a rare multisystem inflammatory disease.18 The FDA approval was based on a double-blind, randomized, placebo-controlled trial in which 53% (55/104) of patients receiving apremilast monotherapy were ulcer free at week 12 compared to 22% (23/103) receiving placebo (P<.0001)(ClinicalTrials.gov Identifier NCT02307513).19

On October 8, 2019, afamelanotide was approved by the FDA to increase pain-free light exposure in adults with erythropoietic protoporphyria, a rare metabolic disorder associated with photosensitivity.20 A melanocortin receptor agonist, afamelanotide is believed to confer photoprotection by increasing the production of eumelanin in the epidermis. The FDA approval was based on 2 randomized, double-blind, placebo-controlled trials, both of which found that patients given afamelanotide spent significantly more time in direct sunlight without pain compared to patients in the placebo group (P=.005 and P=.04).21

Recalls of Popular Skin Products

On July 5, 2019, Neutrogena recalled its cult-favorite Light Therapy Acne Mask. The recall was driven by rare reports of transient visual side effects due to insufficient eye protection from the mask’s light-emitting diodes.22,23 Reported in association with 0.02% of masks sold at the time of the recall, these side effects included eye pain, irritation, tearing, blurry vision, seeing spots, and changes in color vision.24 In addition, a risk for potentially irreversible eye injury from the mask was cited in people taking photosensitizing medications, such as doxycycline, and people with certain underlying eye conditions, such as retinitis pigmentosa and ocular albinism.22,24,25

Following decades of asbestos-related controversy, 1 lot of the iconic Johnson’s Baby Powder was recalled for the first time on October 18, 2019, after the FDA found subtrace levels of asbestos in 1 of the lot’s bottles.26 After the recall, Johnson & Johnson reported that 2 third-party laboratories did not ultimately find asbestos when they tested the bottle of interest as well as other bottles from the recalled lot. Three of 5 samples prepared in 1 room by the third-party laboratories initially did test positive for asbestos, but this result was attributed to the room’s air conditioner, which was found to be contaminated with asbestos. When the same samples were prepared in another room, no asbestos was detected.27 The FDA maintained there was “no indication of cross-contamination” when they originally tested the implicated bottle.28

References
  1. Zraick K. Key West bans sunscreen containing chemicals believed to harm coral reefs. New York Times. February 7, 2019. https://www.nytimes.com/2019/02/07/us/sunscreen-coral-reef-key-west.html. Accessed December 23, 2019.
  2. Gies H. The U.S. Virigin Islands becomes the first American jurisdiction to ban common chemical sunscreens. Pacific Standard. July 18, 2019. https://psmag.com/environment/sunscreen-is-corals-biggest-anemone. Accessed December 23, 2019.
  3. Luscombe R. Republicans seek to overturn Key West ban on coral-damaging sunscreens. The Guardian. November 9, 2019. https://www.theguardian.com/us-news/2019/nov/09/key-west-sunscreen-coral-reef-backlash-skin-cancer. Accessed December 23, 2019.
  4. Salazar D. CVS to remove 2 chemicals from 60 store-brand sunscreens. Drug Store News. August 2, 2019. https://drugstorenews.com/retail-news/cvs-to-remove-2-chemicals-from-60-store-brand-sunscreens. Accessed December 23, 2019.
  5. Sunscreen drug products for over-the-counter human use. Fed Registr. 2019;84(38):6204-6275. To be codified at 21 CFR §201, 310, 347, and 352.
  6. DeLeo VA. Sunscreen regulations and advice for your patients. Cutis. 2019;103:251-253.
  7. Matta MK, Zusterzeel R, Pilli NR, et al. Effect of sunscreen application under maximal use conditions on plasma concentration of sunscreen active ingredients: a randomized clinical trial. JAMA. 2019;321:2082-2091.
  8. Mayor de Blasio, health officials declare end of measles outbreak in New York City [news release]. New York, NY: City of New York; September 3, 2019. https://www1.nyc.gov/office-of-the-mayor/news/409-19/mayor-de-blasio-health-officials-declare-end-measles-outbreak-new-york-city. Accessed December 23, 2019.
  9. Health department reports eleven new cases of measles in Brooklyn’s Orthodox Jewish community, urges on time vaccination for all children, especially before traveling to Israel and other countries experiencing measles outbreaks [news release]. New York, NY: City of New York; November 2, 2018. https://www1.nyc.gov/site/doh/about/press/pr2018/pr091-18.page. Accessed December 23, 2019.
  10. Centers for Disease Control and Prevention. Measles elimination. https://www.cdc.gov/measles/elimination.html. Updated October 4, 2019. Accessed December 23, 2019.
  11. McKinley J. Measles outbreak: N.Y. eliminates religious exemptions for vaccinations. New York Times. June 13, 2019. https://www.nytimes.com/2019/06/13/nyregion/measles-vaccinations-new-york.html. Accessed December 23, 2019.
  12. FDA approves Dupixent® (dupilumab) for moderate-to-severe atopic dermatitis in adolescents [news release]. Cambridge, MA: Sanofi; March 11, 2019. http://www.news.sanofi.us/2019-03-11-FDA-approves-Dupixent-R-dupilumab-for-moderate-to-severe-atopic-dermatitis-in-adolescents. Accessed December 23, 2019.
  13. Simpson EL, Paller AS, Siegfried EC, et al. Efficacy and safety of dupilumab in adolescents with uncontrolled moderate to severe atopic dermatitis: a phase 3 randomized clinical trial [published online ahead of print November 6, 2019]. JAMA Dermatol. doi:10.1001/jamadermatol.2019.3336.
  14. Galderma receives FDA approval for AKLIEF® (trifarotene) cream, 0.005%, the first new retinoid molecule for the treatment of acne in over 20 years [news release]. Fort Worth, TX: Galderma Laboratories, LP; October 4, 2019. https://www.multivu.com/players/English/8613051-galderma-aklief-retinoid-molecule-acne-treatment/. Accessed December 23, 2019.
  15. Update—Foamix receives FDA approval of AMZEEQ™ topical minocycline treatment for millions of moderate to severe acne sufferers [news release]. Bridgewater, NJ: Foamix Pharmaceuticals Ltd; October 18, 2019. http://www.foamix.com/news-releases/news-release-details/update-foamix-receives-fda-approval-amzeeqtm-topical-minocycline. Accessed December 23, 2019.
  16. Redfearn S. Clinical trial patient inclusion and exclusion criteria need an overhaul, say experts. CenterWatch website. April 23, 2018. https://www.centerwatch.com/cwweekly/2018/04/23/clinical-trial-patient-inclusion-and-exclusion-criteria-need-an-overhaul-say-experts. Accessed December 23, 2019.
  17. Tan J, Thiboutot D, Popp G, et al. Randomized phase 3 evaluation of trifarotene 50 mug/g cream treatment of moderate facial and truncal acne. J Am Acad Dermatol. 2019;80:1691-1699.
  18. FDA approves OTEZLA® (apremilast) for the treatment of oral ulcers associated with Behçet’s disease [news release]. Summit, NJ: Celgene; July 19, 2019. https://ir.celgene.com/press-releases/press-release-details/2019/FDA-Approves-OTEZLA-apremilast-for-the-Treatment-of-Oral-Ulcers-Associated-with-Behets-Disease/default.aspx. Accessed December 23, 2019.
  19. Apremilast [package insert]. Summit, NJ: Celgene Corporation; 2019.
  20. FDA approves first treatment to increase pain-free light exposure in patients with a rare disorder [news release]. Silver Spring, MD: US Food and Drug Administration; October 8, 2019. https://www.fda.gov/news-events/press-announcements/fda-approves-first-treatment-increase-pain-free-light-exposure-patients-rare-disorder. Accessed December 23, 2019.
  21. Langendonk JG, Balwani M, Anderson KE, et al. Afamelanotide for erythropoietic protoporphyria. N Engl J Med. 2015;373:48-59.
  22. Light Therapy Mask recall statement. Neutrogena website. https://www.neutrogena.com/light-therapy-statement.html. Accessed December 23, 2019.
  23. Bromwich JE. Neutrogena recalls Light Therapy Masks, citing risk of eye injury. New York Times. July 18, 2019. https://www.nytimes.com/2019/07/18/style/neutrogena-light-therapy-mask-recall.html. Accessed December 23, 2019, 2019.
  24. Nguyen T. Neutrogena recalls acne mask over concerns about blue light. Chemical & Engineering News. August 6, 2019. https://cen.acs.org/safety/lab-safety/Neutrogena-recalls-acne-mask-over-concerns-about-blue-light/97/web/2019/08. Accessed November 16, 2019.
  25. Australian Government Department of Health, Therapeutic Goods Administration. Neutrogena Visibly Clear Light Therapy Acne Mask and Activator: Recall - potential for eye damage. https://www.tga.gov.au/alert/neutrogena-visibly-clear-light-therapy-acne-mask-and-activator. Published July 17, 2019. Accessed December 23, 2019.
  26. Johnson & Johnson Consumer Inc. to voluntarily recall a single lot of Johnson’s Baby Powder in the United States [press release]. New Brunswick, NJ: Johnson & Johnson Consumer Inc; October 18, 2019. https://www.factsabouttalc.com/_document/15-new-tests-from-the-same-bottle-of-johnsons-baby-powder-previously-tested-by-fda-find-no-asbestos?id=0000016e-1915-dc68-af7e-df3f147c0000. Accessed December 23, 2019.
  27. 15 new tests from the same bottle of Johnson’s Baby Powder previously tested by FDA find no asbestos [press release]. New Brunswick, NJ: Johnson & Johnson Consumer Inc; October 29, 2019. https://www.factsabouttalc.com/_document/johnson-johnson-consumer-inc-to-voluntarily-recall-a-single-lot-of-johnsons-baby-powder-in-the-united-states?id=0000016d-debf-d71d-a77d-dfbfebeb0000. Accessed December 23, 2019.
  28. Hsu T. Johnson & Johnson says recalled baby powder doesn’t have asbestos. New York Times. October 29, 2019. https://www.nytimes.com/2019/10/29/business/johnson-baby-powder-asbestos.html. Accessed December 23, 2019.
Article PDF
Author and Disclosure Information

From the Department of Dermatology, State University of New York Downstate Health Sciences University, Brooklyn.

The author reports no conflict of interest.

Correspondence: Daniel R. Mazori, MD, Department of Dermatology, State University of New York Downstate Health Sciences University, 450 Clarkson Ave, Box 46, Brooklyn, NY 11203 (daniel.mazori@downstate.edu).

Issue
Cutis - 104(6)
Publications
Topics
Page Number
E28-E30
Sections
Author and Disclosure Information

From the Department of Dermatology, State University of New York Downstate Health Sciences University, Brooklyn.

The author reports no conflict of interest.

Correspondence: Daniel R. Mazori, MD, Department of Dermatology, State University of New York Downstate Health Sciences University, 450 Clarkson Ave, Box 46, Brooklyn, NY 11203 (daniel.mazori@downstate.edu).

Author and Disclosure Information

From the Department of Dermatology, State University of New York Downstate Health Sciences University, Brooklyn.

The author reports no conflict of interest.

Correspondence: Daniel R. Mazori, MD, Department of Dermatology, State University of New York Downstate Health Sciences University, 450 Clarkson Ave, Box 46, Brooklyn, NY 11203 (daniel.mazori@downstate.edu).

Article PDF
Article PDF

From chemical sunscreen to the measles outbreak and drug approvals to product recalls, dermatology experienced its share of firsts and controversies in 2019.

Chemical Sunscreen Controversies

Controversial concerns about the effects of chemical sunscreen on coral reefs took an unprecedented turn in the United States this last year. On February 5, 2019, an ordinance was passed in Key West, Florida, prohibiting the sale of sunscreen containing the organic UV filters oxybenzone and/or octinoxate within city limits.1 On June 25, 2019, a similar law that also included octocrylene was passed in the US Virgin Islands.2 In so doing, these areas joined Hawaii, the Republic of Palau, and parts of Mexico in restricting chemical sunscreen sales.1 Although the Key West ordinance is set to take effect in January 2021, opponents, including dermatologists who believe it will discourage sunscreen use, currently are trying to overturn the ban.3 In the US Virgin Islands, part of the ban went into effect in September 2019, with the rest of the ban set to start in March 2020.2 Companies have started to follow suit. On August 1, 2019, CVS Pharmacy announced that, by the end of 2020, it will remove oxybenzone and octinoxate from some of its store-brand chemical sunscreens.4

On February 26, 2019, the US Food and Drug Administration (FDA) proposed that there are insufficient data to determine if 12 organic UV filters—including the aforementioned oxybenzone, octinoxate, and octocrylene—are generally recognized as safe and effective (GRASE).5 Although these ingredients were listed as GRASE by the FDA in 2011, the rise in sunscreen use since then, as well as changes in sunscreen formulations, prompted the FDA to ask manufacturers to perform additional studies on safety parameters such as systemic absorption.5,6 One study conducted by the FDA itself was published in May 2019 and showed that maximal use of 4 sunscreens resulted in systemic absorption of 4 organic UV filters above 0.5 ng/mL, the FDA’s threshold for requiring nonclinical toxicology assessment. The study authors concluded that “further studies [are needed] to determine the clinical significance of these findings. [But] These results do not indicate that individuals should refrain from the use of sunscreen.”7 Some in the industry have suggested it may take at least 5 years to generate all the data the FDA has requested.6

End of the New York City Measles Outbreak

On September 3, 2019, New York City’s largest measles outbreak in nearly 30 years was declared over. This announcement reflected the fact that 2 incubation periods for measles—42 days—had passed since the last measles patient was considered contagious. In total, there were 654 cases of measles and 52 associated hospitalizations, including 16 admissions to the intensive care unit. Most patients were younger than 18 years and unvaccinated.8

The outbreak began in October 2018 after Orthodox Jewish children from Brooklyn became infected while visiting Israel and imported the measles virus upon their return home.8,9 All 5 boroughs in New York City were ultimately affected, although 4 zip codes in Williamsburg, a neighborhood in Brooklyn with an undervaccinated Orthodox Jewish community, accounted for 72% of cases.8,10 As part of a $6 million effort to stop the outbreak, an emergency order was placed on these 4 zip codes, posing potential fines on people living or working there if they were unvaccinated.8 In addition, a bill was passed and signed into law in New York State that eliminated religious exemptions for immunizations.11 In collaboration with Jewish leaders, these efforts increased the administration of measles-mumps-rubella vaccines by 41% compared with the year before in Williamsburg and Borough Park, another heavily Orthodox Jewish neighborhood in Brooklyn.8

Drug Approvals for Pediatric Dermatology

On March 11, 2019, the IL-4/IL-13 inhibitor dupilumab became the third biologic with a pediatric dermatology indication when the FDA extended its approval to adolescents for the treatment of atopic dermatitis.12 The FDA approval was based on a randomized, double-blind, placebo-controlled trial in which 42% (34/82) of adolescents treated with dupilumab monotherapy every other week achieved 75% or more improvement in the Eczema Area and Severity Index at week 16 compared with 8% (7/85) in the placebo group (P<.001).13

In October 2019, trifarotene cream and minocycline foam were approved by the FDA for the treatment of acne in patients 9 years and older.14,15 As such, both became the first acne therapies to include patients as young as 9 years in their studies and indication—a milestone, considering the fact that children have historically been excluded from clinical trials.16 The 2 topical treatments also are noteworthy for being first in class: trifarotene cream is the only topical retinoid to selectively target the retinoic acid receptor γ and to have been studied specifically for both facial and truncal acne,14,17 and minocycline foam is the first topical tetracycline.15

 

 

Drug Approvals for Rare Dermatologic Diseases

On July 19, 2019, apremilast, a phosphodiesterase 4 inhibitor, became the first medication approved by the FDA for the treatment of adults with oral ulcers due to Behçet disease, a rare multisystem inflammatory disease.18 The FDA approval was based on a double-blind, randomized, placebo-controlled trial in which 53% (55/104) of patients receiving apremilast monotherapy were ulcer free at week 12 compared to 22% (23/103) receiving placebo (P<.0001)(ClinicalTrials.gov Identifier NCT02307513).19

On October 8, 2019, afamelanotide was approved by the FDA to increase pain-free light exposure in adults with erythropoietic protoporphyria, a rare metabolic disorder associated with photosensitivity.20 A melanocortin receptor agonist, afamelanotide is believed to confer photoprotection by increasing the production of eumelanin in the epidermis. The FDA approval was based on 2 randomized, double-blind, placebo-controlled trials, both of which found that patients given afamelanotide spent significantly more time in direct sunlight without pain compared to patients in the placebo group (P=.005 and P=.04).21

Recalls of Popular Skin Products

On July 5, 2019, Neutrogena recalled its cult-favorite Light Therapy Acne Mask. The recall was driven by rare reports of transient visual side effects due to insufficient eye protection from the mask’s light-emitting diodes.22,23 Reported in association with 0.02% of masks sold at the time of the recall, these side effects included eye pain, irritation, tearing, blurry vision, seeing spots, and changes in color vision.24 In addition, a risk for potentially irreversible eye injury from the mask was cited in people taking photosensitizing medications, such as doxycycline, and people with certain underlying eye conditions, such as retinitis pigmentosa and ocular albinism.22,24,25

Following decades of asbestos-related controversy, 1 lot of the iconic Johnson’s Baby Powder was recalled for the first time on October 18, 2019, after the FDA found subtrace levels of asbestos in 1 of the lot’s bottles.26 After the recall, Johnson & Johnson reported that 2 third-party laboratories did not ultimately find asbestos when they tested the bottle of interest as well as other bottles from the recalled lot. Three of 5 samples prepared in 1 room by the third-party laboratories initially did test positive for asbestos, but this result was attributed to the room’s air conditioner, which was found to be contaminated with asbestos. When the same samples were prepared in another room, no asbestos was detected.27 The FDA maintained there was “no indication of cross-contamination” when they originally tested the implicated bottle.28

From chemical sunscreen to the measles outbreak and drug approvals to product recalls, dermatology experienced its share of firsts and controversies in 2019.

Chemical Sunscreen Controversies

Controversial concerns about the effects of chemical sunscreen on coral reefs took an unprecedented turn in the United States this last year. On February 5, 2019, an ordinance was passed in Key West, Florida, prohibiting the sale of sunscreen containing the organic UV filters oxybenzone and/or octinoxate within city limits.1 On June 25, 2019, a similar law that also included octocrylene was passed in the US Virgin Islands.2 In so doing, these areas joined Hawaii, the Republic of Palau, and parts of Mexico in restricting chemical sunscreen sales.1 Although the Key West ordinance is set to take effect in January 2021, opponents, including dermatologists who believe it will discourage sunscreen use, currently are trying to overturn the ban.3 In the US Virgin Islands, part of the ban went into effect in September 2019, with the rest of the ban set to start in March 2020.2 Companies have started to follow suit. On August 1, 2019, CVS Pharmacy announced that, by the end of 2020, it will remove oxybenzone and octinoxate from some of its store-brand chemical sunscreens.4

On February 26, 2019, the US Food and Drug Administration (FDA) proposed that there are insufficient data to determine if 12 organic UV filters—including the aforementioned oxybenzone, octinoxate, and octocrylene—are generally recognized as safe and effective (GRASE).5 Although these ingredients were listed as GRASE by the FDA in 2011, the rise in sunscreen use since then, as well as changes in sunscreen formulations, prompted the FDA to ask manufacturers to perform additional studies on safety parameters such as systemic absorption.5,6 One study conducted by the FDA itself was published in May 2019 and showed that maximal use of 4 sunscreens resulted in systemic absorption of 4 organic UV filters above 0.5 ng/mL, the FDA’s threshold for requiring nonclinical toxicology assessment. The study authors concluded that “further studies [are needed] to determine the clinical significance of these findings. [But] These results do not indicate that individuals should refrain from the use of sunscreen.”7 Some in the industry have suggested it may take at least 5 years to generate all the data the FDA has requested.6

End of the New York City Measles Outbreak

On September 3, 2019, New York City’s largest measles outbreak in nearly 30 years was declared over. This announcement reflected the fact that 2 incubation periods for measles—42 days—had passed since the last measles patient was considered contagious. In total, there were 654 cases of measles and 52 associated hospitalizations, including 16 admissions to the intensive care unit. Most patients were younger than 18 years and unvaccinated.8

The outbreak began in October 2018 after Orthodox Jewish children from Brooklyn became infected while visiting Israel and imported the measles virus upon their return home.8,9 All 5 boroughs in New York City were ultimately affected, although 4 zip codes in Williamsburg, a neighborhood in Brooklyn with an undervaccinated Orthodox Jewish community, accounted for 72% of cases.8,10 As part of a $6 million effort to stop the outbreak, an emergency order was placed on these 4 zip codes, posing potential fines on people living or working there if they were unvaccinated.8 In addition, a bill was passed and signed into law in New York State that eliminated religious exemptions for immunizations.11 In collaboration with Jewish leaders, these efforts increased the administration of measles-mumps-rubella vaccines by 41% compared with the year before in Williamsburg and Borough Park, another heavily Orthodox Jewish neighborhood in Brooklyn.8

Drug Approvals for Pediatric Dermatology

On March 11, 2019, the IL-4/IL-13 inhibitor dupilumab became the third biologic with a pediatric dermatology indication when the FDA extended its approval to adolescents for the treatment of atopic dermatitis.12 The FDA approval was based on a randomized, double-blind, placebo-controlled trial in which 42% (34/82) of adolescents treated with dupilumab monotherapy every other week achieved 75% or more improvement in the Eczema Area and Severity Index at week 16 compared with 8% (7/85) in the placebo group (P<.001).13

In October 2019, trifarotene cream and minocycline foam were approved by the FDA for the treatment of acne in patients 9 years and older.14,15 As such, both became the first acne therapies to include patients as young as 9 years in their studies and indication—a milestone, considering the fact that children have historically been excluded from clinical trials.16 The 2 topical treatments also are noteworthy for being first in class: trifarotene cream is the only topical retinoid to selectively target the retinoic acid receptor γ and to have been studied specifically for both facial and truncal acne,14,17 and minocycline foam is the first topical tetracycline.15

 

 

Drug Approvals for Rare Dermatologic Diseases

On July 19, 2019, apremilast, a phosphodiesterase 4 inhibitor, became the first medication approved by the FDA for the treatment of adults with oral ulcers due to Behçet disease, a rare multisystem inflammatory disease.18 The FDA approval was based on a double-blind, randomized, placebo-controlled trial in which 53% (55/104) of patients receiving apremilast monotherapy were ulcer free at week 12 compared to 22% (23/103) receiving placebo (P<.0001)(ClinicalTrials.gov Identifier NCT02307513).19

On October 8, 2019, afamelanotide was approved by the FDA to increase pain-free light exposure in adults with erythropoietic protoporphyria, a rare metabolic disorder associated with photosensitivity.20 A melanocortin receptor agonist, afamelanotide is believed to confer photoprotection by increasing the production of eumelanin in the epidermis. The FDA approval was based on 2 randomized, double-blind, placebo-controlled trials, both of which found that patients given afamelanotide spent significantly more time in direct sunlight without pain compared to patients in the placebo group (P=.005 and P=.04).21

Recalls of Popular Skin Products

On July 5, 2019, Neutrogena recalled its cult-favorite Light Therapy Acne Mask. The recall was driven by rare reports of transient visual side effects due to insufficient eye protection from the mask’s light-emitting diodes.22,23 Reported in association with 0.02% of masks sold at the time of the recall, these side effects included eye pain, irritation, tearing, blurry vision, seeing spots, and changes in color vision.24 In addition, a risk for potentially irreversible eye injury from the mask was cited in people taking photosensitizing medications, such as doxycycline, and people with certain underlying eye conditions, such as retinitis pigmentosa and ocular albinism.22,24,25

Following decades of asbestos-related controversy, 1 lot of the iconic Johnson’s Baby Powder was recalled for the first time on October 18, 2019, after the FDA found subtrace levels of asbestos in 1 of the lot’s bottles.26 After the recall, Johnson & Johnson reported that 2 third-party laboratories did not ultimately find asbestos when they tested the bottle of interest as well as other bottles from the recalled lot. Three of 5 samples prepared in 1 room by the third-party laboratories initially did test positive for asbestos, but this result was attributed to the room’s air conditioner, which was found to be contaminated with asbestos. When the same samples were prepared in another room, no asbestos was detected.27 The FDA maintained there was “no indication of cross-contamination” when they originally tested the implicated bottle.28

References
  1. Zraick K. Key West bans sunscreen containing chemicals believed to harm coral reefs. New York Times. February 7, 2019. https://www.nytimes.com/2019/02/07/us/sunscreen-coral-reef-key-west.html. Accessed December 23, 2019.
  2. Gies H. The U.S. Virigin Islands becomes the first American jurisdiction to ban common chemical sunscreens. Pacific Standard. July 18, 2019. https://psmag.com/environment/sunscreen-is-corals-biggest-anemone. Accessed December 23, 2019.
  3. Luscombe R. Republicans seek to overturn Key West ban on coral-damaging sunscreens. The Guardian. November 9, 2019. https://www.theguardian.com/us-news/2019/nov/09/key-west-sunscreen-coral-reef-backlash-skin-cancer. Accessed December 23, 2019.
  4. Salazar D. CVS to remove 2 chemicals from 60 store-brand sunscreens. Drug Store News. August 2, 2019. https://drugstorenews.com/retail-news/cvs-to-remove-2-chemicals-from-60-store-brand-sunscreens. Accessed December 23, 2019.
  5. Sunscreen drug products for over-the-counter human use. Fed Registr. 2019;84(38):6204-6275. To be codified at 21 CFR §201, 310, 347, and 352.
  6. DeLeo VA. Sunscreen regulations and advice for your patients. Cutis. 2019;103:251-253.
  7. Matta MK, Zusterzeel R, Pilli NR, et al. Effect of sunscreen application under maximal use conditions on plasma concentration of sunscreen active ingredients: a randomized clinical trial. JAMA. 2019;321:2082-2091.
  8. Mayor de Blasio, health officials declare end of measles outbreak in New York City [news release]. New York, NY: City of New York; September 3, 2019. https://www1.nyc.gov/office-of-the-mayor/news/409-19/mayor-de-blasio-health-officials-declare-end-measles-outbreak-new-york-city. Accessed December 23, 2019.
  9. Health department reports eleven new cases of measles in Brooklyn’s Orthodox Jewish community, urges on time vaccination for all children, especially before traveling to Israel and other countries experiencing measles outbreaks [news release]. New York, NY: City of New York; November 2, 2018. https://www1.nyc.gov/site/doh/about/press/pr2018/pr091-18.page. Accessed December 23, 2019.
  10. Centers for Disease Control and Prevention. Measles elimination. https://www.cdc.gov/measles/elimination.html. Updated October 4, 2019. Accessed December 23, 2019.
  11. McKinley J. Measles outbreak: N.Y. eliminates religious exemptions for vaccinations. New York Times. June 13, 2019. https://www.nytimes.com/2019/06/13/nyregion/measles-vaccinations-new-york.html. Accessed December 23, 2019.
  12. FDA approves Dupixent® (dupilumab) for moderate-to-severe atopic dermatitis in adolescents [news release]. Cambridge, MA: Sanofi; March 11, 2019. http://www.news.sanofi.us/2019-03-11-FDA-approves-Dupixent-R-dupilumab-for-moderate-to-severe-atopic-dermatitis-in-adolescents. Accessed December 23, 2019.
  13. Simpson EL, Paller AS, Siegfried EC, et al. Efficacy and safety of dupilumab in adolescents with uncontrolled moderate to severe atopic dermatitis: a phase 3 randomized clinical trial [published online ahead of print November 6, 2019]. JAMA Dermatol. doi:10.1001/jamadermatol.2019.3336.
  14. Galderma receives FDA approval for AKLIEF® (trifarotene) cream, 0.005%, the first new retinoid molecule for the treatment of acne in over 20 years [news release]. Fort Worth, TX: Galderma Laboratories, LP; October 4, 2019. https://www.multivu.com/players/English/8613051-galderma-aklief-retinoid-molecule-acne-treatment/. Accessed December 23, 2019.
  15. Update—Foamix receives FDA approval of AMZEEQ™ topical minocycline treatment for millions of moderate to severe acne sufferers [news release]. Bridgewater, NJ: Foamix Pharmaceuticals Ltd; October 18, 2019. http://www.foamix.com/news-releases/news-release-details/update-foamix-receives-fda-approval-amzeeqtm-topical-minocycline. Accessed December 23, 2019.
  16. Redfearn S. Clinical trial patient inclusion and exclusion criteria need an overhaul, say experts. CenterWatch website. April 23, 2018. https://www.centerwatch.com/cwweekly/2018/04/23/clinical-trial-patient-inclusion-and-exclusion-criteria-need-an-overhaul-say-experts. Accessed December 23, 2019.
  17. Tan J, Thiboutot D, Popp G, et al. Randomized phase 3 evaluation of trifarotene 50 mug/g cream treatment of moderate facial and truncal acne. J Am Acad Dermatol. 2019;80:1691-1699.
  18. FDA approves OTEZLA® (apremilast) for the treatment of oral ulcers associated with Behçet’s disease [news release]. Summit, NJ: Celgene; July 19, 2019. https://ir.celgene.com/press-releases/press-release-details/2019/FDA-Approves-OTEZLA-apremilast-for-the-Treatment-of-Oral-Ulcers-Associated-with-Behets-Disease/default.aspx. Accessed December 23, 2019.
  19. Apremilast [package insert]. Summit, NJ: Celgene Corporation; 2019.
  20. FDA approves first treatment to increase pain-free light exposure in patients with a rare disorder [news release]. Silver Spring, MD: US Food and Drug Administration; October 8, 2019. https://www.fda.gov/news-events/press-announcements/fda-approves-first-treatment-increase-pain-free-light-exposure-patients-rare-disorder. Accessed December 23, 2019.
  21. Langendonk JG, Balwani M, Anderson KE, et al. Afamelanotide for erythropoietic protoporphyria. N Engl J Med. 2015;373:48-59.
  22. Light Therapy Mask recall statement. Neutrogena website. https://www.neutrogena.com/light-therapy-statement.html. Accessed December 23, 2019.
  23. Bromwich JE. Neutrogena recalls Light Therapy Masks, citing risk of eye injury. New York Times. July 18, 2019. https://www.nytimes.com/2019/07/18/style/neutrogena-light-therapy-mask-recall.html. Accessed December 23, 2019, 2019.
  24. Nguyen T. Neutrogena recalls acne mask over concerns about blue light. Chemical & Engineering News. August 6, 2019. https://cen.acs.org/safety/lab-safety/Neutrogena-recalls-acne-mask-over-concerns-about-blue-light/97/web/2019/08. Accessed November 16, 2019.
  25. Australian Government Department of Health, Therapeutic Goods Administration. Neutrogena Visibly Clear Light Therapy Acne Mask and Activator: Recall - potential for eye damage. https://www.tga.gov.au/alert/neutrogena-visibly-clear-light-therapy-acne-mask-and-activator. Published July 17, 2019. Accessed December 23, 2019.
  26. Johnson & Johnson Consumer Inc. to voluntarily recall a single lot of Johnson’s Baby Powder in the United States [press release]. New Brunswick, NJ: Johnson & Johnson Consumer Inc; October 18, 2019. https://www.factsabouttalc.com/_document/15-new-tests-from-the-same-bottle-of-johnsons-baby-powder-previously-tested-by-fda-find-no-asbestos?id=0000016e-1915-dc68-af7e-df3f147c0000. Accessed December 23, 2019.
  27. 15 new tests from the same bottle of Johnson’s Baby Powder previously tested by FDA find no asbestos [press release]. New Brunswick, NJ: Johnson & Johnson Consumer Inc; October 29, 2019. https://www.factsabouttalc.com/_document/johnson-johnson-consumer-inc-to-voluntarily-recall-a-single-lot-of-johnsons-baby-powder-in-the-united-states?id=0000016d-debf-d71d-a77d-dfbfebeb0000. Accessed December 23, 2019.
  28. Hsu T. Johnson & Johnson says recalled baby powder doesn’t have asbestos. New York Times. October 29, 2019. https://www.nytimes.com/2019/10/29/business/johnson-baby-powder-asbestos.html. Accessed December 23, 2019.
References
  1. Zraick K. Key West bans sunscreen containing chemicals believed to harm coral reefs. New York Times. February 7, 2019. https://www.nytimes.com/2019/02/07/us/sunscreen-coral-reef-key-west.html. Accessed December 23, 2019.
  2. Gies H. The U.S. Virigin Islands becomes the first American jurisdiction to ban common chemical sunscreens. Pacific Standard. July 18, 2019. https://psmag.com/environment/sunscreen-is-corals-biggest-anemone. Accessed December 23, 2019.
  3. Luscombe R. Republicans seek to overturn Key West ban on coral-damaging sunscreens. The Guardian. November 9, 2019. https://www.theguardian.com/us-news/2019/nov/09/key-west-sunscreen-coral-reef-backlash-skin-cancer. Accessed December 23, 2019.
  4. Salazar D. CVS to remove 2 chemicals from 60 store-brand sunscreens. Drug Store News. August 2, 2019. https://drugstorenews.com/retail-news/cvs-to-remove-2-chemicals-from-60-store-brand-sunscreens. Accessed December 23, 2019.
  5. Sunscreen drug products for over-the-counter human use. Fed Registr. 2019;84(38):6204-6275. To be codified at 21 CFR §201, 310, 347, and 352.
  6. DeLeo VA. Sunscreen regulations and advice for your patients. Cutis. 2019;103:251-253.
  7. Matta MK, Zusterzeel R, Pilli NR, et al. Effect of sunscreen application under maximal use conditions on plasma concentration of sunscreen active ingredients: a randomized clinical trial. JAMA. 2019;321:2082-2091.
  8. Mayor de Blasio, health officials declare end of measles outbreak in New York City [news release]. New York, NY: City of New York; September 3, 2019. https://www1.nyc.gov/office-of-the-mayor/news/409-19/mayor-de-blasio-health-officials-declare-end-measles-outbreak-new-york-city. Accessed December 23, 2019.
  9. Health department reports eleven new cases of measles in Brooklyn’s Orthodox Jewish community, urges on time vaccination for all children, especially before traveling to Israel and other countries experiencing measles outbreaks [news release]. New York, NY: City of New York; November 2, 2018. https://www1.nyc.gov/site/doh/about/press/pr2018/pr091-18.page. Accessed December 23, 2019.
  10. Centers for Disease Control and Prevention. Measles elimination. https://www.cdc.gov/measles/elimination.html. Updated October 4, 2019. Accessed December 23, 2019.
  11. McKinley J. Measles outbreak: N.Y. eliminates religious exemptions for vaccinations. New York Times. June 13, 2019. https://www.nytimes.com/2019/06/13/nyregion/measles-vaccinations-new-york.html. Accessed December 23, 2019.
  12. FDA approves Dupixent® (dupilumab) for moderate-to-severe atopic dermatitis in adolescents [news release]. Cambridge, MA: Sanofi; March 11, 2019. http://www.news.sanofi.us/2019-03-11-FDA-approves-Dupixent-R-dupilumab-for-moderate-to-severe-atopic-dermatitis-in-adolescents. Accessed December 23, 2019.
  13. Simpson EL, Paller AS, Siegfried EC, et al. Efficacy and safety of dupilumab in adolescents with uncontrolled moderate to severe atopic dermatitis: a phase 3 randomized clinical trial [published online ahead of print November 6, 2019]. JAMA Dermatol. doi:10.1001/jamadermatol.2019.3336.
  14. Galderma receives FDA approval for AKLIEF® (trifarotene) cream, 0.005%, the first new retinoid molecule for the treatment of acne in over 20 years [news release]. Fort Worth, TX: Galderma Laboratories, LP; October 4, 2019. https://www.multivu.com/players/English/8613051-galderma-aklief-retinoid-molecule-acne-treatment/. Accessed December 23, 2019.
  15. Update—Foamix receives FDA approval of AMZEEQ™ topical minocycline treatment for millions of moderate to severe acne sufferers [news release]. Bridgewater, NJ: Foamix Pharmaceuticals Ltd; October 18, 2019. http://www.foamix.com/news-releases/news-release-details/update-foamix-receives-fda-approval-amzeeqtm-topical-minocycline. Accessed December 23, 2019.
  16. Redfearn S. Clinical trial patient inclusion and exclusion criteria need an overhaul, say experts. CenterWatch website. April 23, 2018. https://www.centerwatch.com/cwweekly/2018/04/23/clinical-trial-patient-inclusion-and-exclusion-criteria-need-an-overhaul-say-experts. Accessed December 23, 2019.
  17. Tan J, Thiboutot D, Popp G, et al. Randomized phase 3 evaluation of trifarotene 50 mug/g cream treatment of moderate facial and truncal acne. J Am Acad Dermatol. 2019;80:1691-1699.
  18. FDA approves OTEZLA® (apremilast) for the treatment of oral ulcers associated with Behçet’s disease [news release]. Summit, NJ: Celgene; July 19, 2019. https://ir.celgene.com/press-releases/press-release-details/2019/FDA-Approves-OTEZLA-apremilast-for-the-Treatment-of-Oral-Ulcers-Associated-with-Behets-Disease/default.aspx. Accessed December 23, 2019.
  19. Apremilast [package insert]. Summit, NJ: Celgene Corporation; 2019.
  20. FDA approves first treatment to increase pain-free light exposure in patients with a rare disorder [news release]. Silver Spring, MD: US Food and Drug Administration; October 8, 2019. https://www.fda.gov/news-events/press-announcements/fda-approves-first-treatment-increase-pain-free-light-exposure-patients-rare-disorder. Accessed December 23, 2019.
  21. Langendonk JG, Balwani M, Anderson KE, et al. Afamelanotide for erythropoietic protoporphyria. N Engl J Med. 2015;373:48-59.
  22. Light Therapy Mask recall statement. Neutrogena website. https://www.neutrogena.com/light-therapy-statement.html. Accessed December 23, 2019.
  23. Bromwich JE. Neutrogena recalls Light Therapy Masks, citing risk of eye injury. New York Times. July 18, 2019. https://www.nytimes.com/2019/07/18/style/neutrogena-light-therapy-mask-recall.html. Accessed December 23, 2019, 2019.
  24. Nguyen T. Neutrogena recalls acne mask over concerns about blue light. Chemical & Engineering News. August 6, 2019. https://cen.acs.org/safety/lab-safety/Neutrogena-recalls-acne-mask-over-concerns-about-blue-light/97/web/2019/08. Accessed November 16, 2019.
  25. Australian Government Department of Health, Therapeutic Goods Administration. Neutrogena Visibly Clear Light Therapy Acne Mask and Activator: Recall - potential for eye damage. https://www.tga.gov.au/alert/neutrogena-visibly-clear-light-therapy-acne-mask-and-activator. Published July 17, 2019. Accessed December 23, 2019.
  26. Johnson & Johnson Consumer Inc. to voluntarily recall a single lot of Johnson’s Baby Powder in the United States [press release]. New Brunswick, NJ: Johnson & Johnson Consumer Inc; October 18, 2019. https://www.factsabouttalc.com/_document/15-new-tests-from-the-same-bottle-of-johnsons-baby-powder-previously-tested-by-fda-find-no-asbestos?id=0000016e-1915-dc68-af7e-df3f147c0000. Accessed December 23, 2019.
  27. 15 new tests from the same bottle of Johnson’s Baby Powder previously tested by FDA find no asbestos [press release]. New Brunswick, NJ: Johnson & Johnson Consumer Inc; October 29, 2019. https://www.factsabouttalc.com/_document/johnson-johnson-consumer-inc-to-voluntarily-recall-a-single-lot-of-johnsons-baby-powder-in-the-united-states?id=0000016d-debf-d71d-a77d-dfbfebeb0000. Accessed December 23, 2019.
  28. Hsu T. Johnson & Johnson says recalled baby powder doesn’t have asbestos. New York Times. October 29, 2019. https://www.nytimes.com/2019/10/29/business/johnson-baby-powder-asbestos.html. Accessed December 23, 2019.
Issue
Cutis - 104(6)
Issue
Cutis - 104(6)
Page Number
E28-E30
Page Number
E28-E30
Publications
Publications
Topics
Article Type
Display Headline
Annual Skin Check: Examining the Dermatology Headlines of 2019
Display Headline
Annual Skin Check: Examining the Dermatology Headlines of 2019
Sections
Inside the Article

Resident Pearls

  • Chemical sunscreen made headlines in 2019 due to concerns over coral reef toxicity and systemic absorption in humans.
  • With a total of 654 cases, New York City’s largest measles outbreak in nearly 30 years ended in September 2019.
  • From dupilumab for adolescent atopic dermatitis to apremilast for Behçet disease, the US Food and Drug Administration approved several therapies for pediatric dermatology and rare dermatologic conditions in 2019.
  • Two popular skin care products—the Neutrogena Light Therapy Acne Mask and Johnson’s Baby Powderwere involved in recalls in 2019.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Article PDF Media

Skin Scores: A Review of Clinical Scoring Systems in Dermatology

Article Type
Changed
Tue, 02/07/2023 - 16:51

The practice of dermatology is rife with bedside tools: swabs, smears, and scoring systems. First popularized in specialties such as emergency medicine and internal medicine, clinical scoring systems are now emerging in dermatology. These evidence-based scores can be calculated quickly at the bedside—often through a free smartphone app—to help guide clinical decision-making regarding diagnosis, prognosis, and management. As with any medical tool, scoring systems have limitations and should be used as a supplement, not substitute, for one’s clinical judgement. This article reviews 4 clinical scoring systems practical for dermatology residents.

SCORTEN Prognosticates Cases of Stevens-Johnson Syndrome/Toxic Epidermal Necrolysis

Perhaps the best-known scoring system in dermatology, the SCORTEN is widely used to predict hospital mortality from Stevens-Johnson syndrome/toxic epidermal necrolysis. The SCORTEN includes 7 variables of equal weight—age of 40 years or older, heart rate of 120 beats per minute or more, cancer/hematologic malignancy, involved body surface area (BSA) greater than 10%, serum urea greater than 10 mmol/L, serum bicarbonate less than 20 mmol/L, and serum glucose greater than 14 mmol/L—each contributing 1 point to the overall score if present.1 The involved BSA is defined as the sum of detached and detachable epidermis.1

The SCORTEN was developed and prospectively validated to be calculated at the end of the first 24 hours of admission; for this calculation, use the BSA affected at that time, and use the most abnormal values during the first 24 hours of admission for the other variables.1 In addition, a follow-up study including some of the original coauthors recommends recalculating the SCORTEN at the end of hospital day 3, having found that the score’s predictive value was better on this day than hospital days 1, 2, 4, or 5.2 Based on the original study, a SCORTEN of 0 to 1 corresponds to a mortality rate of 3.2%, 2 to 12.1%, 3 to 35.3%, 4 to 58.3%, and 5 or greater to 90.0%.1



Limitations of the SCORTEN include its ability to overestimate or underestimate mortality as demonstrated by 2 multi-institutional cohorts.3,4 Recently, the ABCD-10 score was developed as an alternative to the SCORTEN and was found to predict mortality similarly when validated in an internal cohort.5

PEST Screens for Psoriatic Arthritis

Dermatologists play an important role in screening for psoriatic arthritis, as an estimated 1 in 5 patients with psoriasis have psoriatic arthritis.6 To this end, several screening tools have been developed to help differentiate psoriatic arthritis from other arthritides. Joint guidelines from the American Academy of Dermatology and the National Psoriasis Foundation acknowledge that “. . . these screening tools have tended to perform less well when tested in groups of people other than those for which they were originally developed. As such, their usefulness in routine clinical practice remains controversial.”7 Nevertheless, the guidelines state, “[b]ecause screening and early detection of inflammatory arthritis are essential to optimize patient [quality of life] and reduce morbidity, providers may consider using a formal screening tool of their choice.”7

 

 

With these limitations in mind, I have found the Psoriasis Epidemiology Screening Tool (PEST) to be the most useful psoriatic arthritis screening tool. One study determined that the PEST has the best trade-off between sensitivity and specificity compared to 2 other psoriatic arthritis screening tools, the Psoriatic Arthritis Screening and Evaluation (PASE) and the Early Arthritis for Psoriatic Patients (EARP).8



The PEST is comprised of 5 questions: (1) Have you ever had a swollen joint (or joints)? (2) Has a doctor ever told you that you have arthritis? (3) Do your fingernails or toenails have holes or pits? (4) Have you had pain in your heel? (5) Have you had a finger or toe that was completely swollen and painful for no apparent reason? According to the PEST, a referral to a rheumatologist should be considered for patients answering yes to 3 or more questions, which is 97% sensitive and 79% specific for psoriatic arthritis.9 Patients who answer yes to fewer than 3 questions should still be referred to a rheumatologist if there is a strong clinical suspicion of psoriatic arthritis.10

The PEST can be accessed for free in 13 languages via the GRAPPA (Group for Research and Assessment of Psoriasis and Psoriatic Arthritis) app as well as downloaded for free from the National Psoriasis Foundation’s website (https://www.psoriasis.org/psa-screening/providers).

ALT-70 Differentiates Cellulitis From Pseudocellulitis

Overdiagnosing cellulitis in the United States has been estimated to result in up to 130,000 unnecessary hospitalizations and up to $515 million in avoidable health care spending.11 Dermatologists are in a unique position to help fix this issue. In one retrospective study of 1430 inpatient dermatology consultations, 74.32% of inpatients evaluated for presumed cellulitis by a dermatologist were instead diagnosed with a cellulitis mimicker (ie, pseudocellulitis), such as stasis dermatitis or contact dermatitis.12

The ALT-70 score was developed and prospectively validated to help differentiate lower extremity cellulitis from pseudocellulitis in adult patients in the emergency department (ED).13 In addition, the score has retrospectively been shown to function similarly in the inpatient setting when calculated at 24 and 48 hours after ED presentation.14 Although the ALT-70 score was designed for use by frontline clinicians prior to dermatology consultation, I also have found it helpful to calculate as a consultant, as it provides an objective measure of risk to communicate to the primary team in support of one diagnosis or another.

 

 


ALT-70 is an acronym for the score’s 4 variables: asymmetry, leukocytosis, tachycardia, and age of 70 years or older.15 If present, each variable confers a certain number of points to the final score: 3 points for asymmetry (defined as unilateral leg involvement), 1 point for leukocytosis (white blood cell count ≥10,000/μL), 1 point for tachycardia (≥90 beats per minute), and 2 points for age of 70 years or older. An ALT-70 score of 0 to 2 corresponds to an 83.3% or greater chance of pseudocellulitis, suggesting that the diagnosis of cellulitis be reconsidered. A score of 3 to 4 is indeterminate, and additional information such as a dermatology consultation should be pursued. A score of 5 to 7 corresponds to an 82.2% or greater chance of cellulitis, signifying that empiric treatment with antibiotics be considered.15



The ALT-70 score does not apply to cases involving areas other than the lower extremities; intravenous antibiotic use within 48 hours before ED presentation; surgery within the last 30 days; abscess; penetrating trauma; burn; or known history of osteomyelitis, diabetic ulcer, or indwelling hardware at the site of infection.15 The ALT-70 score is available for free via the MDCalc app and website (https://www.mdcalc.com/alt-70-score-cellulitis).

Mohs AUC Determines the Appropriateness of Mohs Micrographic Surgery

In 2012, the American Academy of Dermatology, American College of Mohs Surgery, American Society for Dermatologic Surgery Association, and American Society for Mohs Surgery published appropriate use criteria (AUC) to guide the decision to pursue Mohs micrographic surgery (MMS) in the United States.16 Based on various tumor and patient characteristics, the Mohs AUC assign scores to 270 different clinical scenarios. A score of 1 to 3 signifies that MMS is inappropriate and generally not considered acceptable. A score 4 to 6 indicates that the appropriateness of MMS is uncertain. A score 7 to 9 means that MMS is appropriate and generally considered acceptable.16

Since publication, the Mohs AUC have been criticized for classifying most primary superficial basal cell carcinomas as appropriate for MMS17 (which an AUC coauthor18 and others19,20 have defended), excluding certain reasons for performing MMS (such as operating on multiple tumors on the same day),21 including counterintuitive scores,22 and omitting trials from Europe23 (which AUC coauthors also have defended24). As with any clinical scoring system, the Mohs AUC has limitations; the creators acknowledge that “. . . these criteria should not be interpreted as setting a standard of care, or be deemed inclusive of all proper methods of care nor exclusive of other methods of care reasonably directed to obtaining the same results, even for those indications scored as inappropriate.”16 The Mohs AUC app (https://www.aad.org/members/aad-apps/mohs-auc) is free and allows users to enter tumor and patient characteristics to determine the score for their specific scenario.

Final Thoughts

Scoring systems are emerging in dermatology as evidence-based bedside tools to help guide clinical decision-making. Despite their limitations, these scores have the potential to make a meaningful impact in dermatology as they have in other specialties.

References
  1. Bastuji-Garin S, Fouchard N, Bertocchi M, et al. SCORTEN: a severity-of-illness score for toxic epidermal necrolysis. J Invest Dermatol. 2000;115:149-153.
  2. Guegan S, Bastuji-Garin S, Poszepczynska-Guigne E, et al. Performance of the SCORTEN during the first five days of hospitalization to predict the prognosis of epidermal necrolysis. J Invest Dermatol. 2006;126:272-276.
  3. Micheletti RG, Chiesa-Fuxench Z, Noe MH, et al. Stevens-Johnson syndrome/toxic epidermal necrolysis: a multicenter retrospective study of 377 adult patients from the United States. J Invest Dermatol. 2018;138:2315-2321.
  4. Sekula P, Liss Y, Davidovici B, et al. Evaluation of SCORTEN on a cohort of patients with Stevens-Johnson syndrome and toxic epidermal necrolysis included in the RegiSCAR study. J Burn Care Res. 2011;32:237-245.
  5. Noe MH, Rosenbach M, Hubbard RA, et al. Development and validation of a risk prediction model for in-hospital mortality among patients with Stevens-Johnson syndrome/toxic epidermal necrolysis-ABCD-10. JAMA Dermatol. 2019;155:448-454.
  6. Alinaghi F, Calov M, Kristensen LE, et al. Prevalence of psoriatic arthritis in patients with psoriasis: a systematic review and meta-analysis of observational and clinical studies. J Am Acad Dermatol. 2019;80:251-265.e219.
  7. Elmets CA, Leonardi CL, Davis DMR, et al. Joint AAD-NPF guidelines of care for the management and treatment of psoriasis with awareness and attention to comorbidities. J Am Acad Dermatol. 2019;80:1073-1113.
  8. Karreman MC, Weel A, van der Ven M, et al. Performance of screening tools for psoriatic arthritis: a cross-sectional study in primary care. Rheumatology (Oxford). 2017;56:597-602.
     

     

  9. Ibrahim GH, Buch MH, Lawson C, et al. Evaluation of an existing screening tool for psoriatic arthritis in people with psoriasis and the development of a new instrument: the Psoriasis Epidemiology Screening Tool (PEST) questionnaire. Clin Exp Rheumatol. 2009;27:469-474.
  10. Zhang A, Kurtzman DJB, Perez-Chada LM, et al. Psoriatic arthritis and the dermatologist: an approach to screening and clinical evaluation. Clin Dermatol. 2018;36:551-560.
  11. Weng QY, Raff AB, Cohen JM, et al. Costs and consequences associated with misdiagnosed lower extremity cellulitis. JAMA Dermatol. 2017;153:141-146.
  12. Strazzula L, Cotliar J, Fox LP, et al. Inpatient dermatology consultation aids diagnosis of cellulitis among hospitalized patients: a multi-institutional analysis. J Am Acad Dermatol. 2015;73:70-75.
  13. Li DG, Dewan AK, Xia FD, et al. The ALT-70 predictive model outperforms thermal imaging for the diagnosis of lower extremity cellulitis: a prospective evaluation. J Am Acad Dermatol. 2018;79:1076-1080.e1071.
  14. Singer S, Li DG, Gunasekera N, et al. The ALT-70 predictive model maintains predictive value at 24 and 48 hours after presentation [published online March 23, 2019]. J Am Acad Dermatol. doi:10.1016/j.jaad.2019.03.050.
  15. Raff AB, Weng QY, Cohen JM, et al. A predictive model for diagnosis of lower extremity cellulitis: a cross-sectional study. J Am Acad Dermatol. 2017;76:618-625.e2.
  16. Connolly SM, Baker DR, Coldiron BM, et al. AAD/ACMS/ASDSA/ASMS 2012 appropriate use criteria for Mohs micrographic surgery: a report of the American Academy of Dermatology, American College of Mohs Surgery, American Society for Dermatologic Surgery Association, and the American Society for Mohs Surgery. J Am Acad Dermatol. 2012;67:531-550.
  17. Steinman HK, Dixon A, Zachary CB. Reevaluating Mohs surgery appropriate use criteria for primary superficial basal cell carcinoma. JAMA Dermatol. 2018;154:755-756.
  18. Montuno MA, Coldiron BM. Mohs appropriate use criteria for superficial basal cell carcinoma. JAMA Dermatol. 2019;155:394-395.
  19. MacFarlane DF, Perlis C. Mohs appropriate use criteria for superficial basal cell carcinoma. JAMA Dermatol. 2019;155:395-396.
  20. Kantor J. Mohs appropriate use criteria for superficial basal cell carcinoma. JAMA Dermatol. 2019;155:395.
  21. Ruiz ES, Karia PS, Morgan FC, et al. Multiple Mohs micrographic surgery is the most common reason for divergence from the appropriate use criteria: a single institution retrospective cohort study. J Am Acad Dermatol. 2016;75:830-831.
  22. Croley JA, Joseph AK, Wagner RF Jr. Discrepancies in the Mohs Micrographic Surgery appropriate use criteria [published online December 23, 2018]. J Am Acad Dermatol. doi:10.1016/j.jaad.2018.11.064.
  23. Kelleners-Smeets NW, Mosterd K. Comment on 2012 appropriate use criteria for Mohs micrographic surgery. J Am Acad Dermatol. 2013;69:317-318.
  24. Connolly S, Baker D, Coldiron B, et al. Reply to “comment on 2012 appropriate use criteria for Mohs micrographic surgery.” J Am Acad Dermatol. 2013;69:318.
Article PDF
Author and Disclosure Information

From the Department of Dermatology, State University of New York Downstate Medical Center, Brooklyn.

The author reports no conflict of interest.

Correspondence: Daniel R. Mazori, MD, Department of Dermatology, State University of New York Downstate Medical Center, 450 Clarkson Ave, Box 46, Brooklyn, NY 11203 (daniel.mazori@downstate.edu).

Issue
Cutis - 104(3)
Publications
Topics
Page Number
E37-E39
Sections
Author and Disclosure Information

From the Department of Dermatology, State University of New York Downstate Medical Center, Brooklyn.

The author reports no conflict of interest.

Correspondence: Daniel R. Mazori, MD, Department of Dermatology, State University of New York Downstate Medical Center, 450 Clarkson Ave, Box 46, Brooklyn, NY 11203 (daniel.mazori@downstate.edu).

Author and Disclosure Information

From the Department of Dermatology, State University of New York Downstate Medical Center, Brooklyn.

The author reports no conflict of interest.

Correspondence: Daniel R. Mazori, MD, Department of Dermatology, State University of New York Downstate Medical Center, 450 Clarkson Ave, Box 46, Brooklyn, NY 11203 (daniel.mazori@downstate.edu).

Article PDF
Article PDF

The practice of dermatology is rife with bedside tools: swabs, smears, and scoring systems. First popularized in specialties such as emergency medicine and internal medicine, clinical scoring systems are now emerging in dermatology. These evidence-based scores can be calculated quickly at the bedside—often through a free smartphone app—to help guide clinical decision-making regarding diagnosis, prognosis, and management. As with any medical tool, scoring systems have limitations and should be used as a supplement, not substitute, for one’s clinical judgement. This article reviews 4 clinical scoring systems practical for dermatology residents.

SCORTEN Prognosticates Cases of Stevens-Johnson Syndrome/Toxic Epidermal Necrolysis

Perhaps the best-known scoring system in dermatology, the SCORTEN is widely used to predict hospital mortality from Stevens-Johnson syndrome/toxic epidermal necrolysis. The SCORTEN includes 7 variables of equal weight—age of 40 years or older, heart rate of 120 beats per minute or more, cancer/hematologic malignancy, involved body surface area (BSA) greater than 10%, serum urea greater than 10 mmol/L, serum bicarbonate less than 20 mmol/L, and serum glucose greater than 14 mmol/L—each contributing 1 point to the overall score if present.1 The involved BSA is defined as the sum of detached and detachable epidermis.1

The SCORTEN was developed and prospectively validated to be calculated at the end of the first 24 hours of admission; for this calculation, use the BSA affected at that time, and use the most abnormal values during the first 24 hours of admission for the other variables.1 In addition, a follow-up study including some of the original coauthors recommends recalculating the SCORTEN at the end of hospital day 3, having found that the score’s predictive value was better on this day than hospital days 1, 2, 4, or 5.2 Based on the original study, a SCORTEN of 0 to 1 corresponds to a mortality rate of 3.2%, 2 to 12.1%, 3 to 35.3%, 4 to 58.3%, and 5 or greater to 90.0%.1



Limitations of the SCORTEN include its ability to overestimate or underestimate mortality as demonstrated by 2 multi-institutional cohorts.3,4 Recently, the ABCD-10 score was developed as an alternative to the SCORTEN and was found to predict mortality similarly when validated in an internal cohort.5

PEST Screens for Psoriatic Arthritis

Dermatologists play an important role in screening for psoriatic arthritis, as an estimated 1 in 5 patients with psoriasis have psoriatic arthritis.6 To this end, several screening tools have been developed to help differentiate psoriatic arthritis from other arthritides. Joint guidelines from the American Academy of Dermatology and the National Psoriasis Foundation acknowledge that “. . . these screening tools have tended to perform less well when tested in groups of people other than those for which they were originally developed. As such, their usefulness in routine clinical practice remains controversial.”7 Nevertheless, the guidelines state, “[b]ecause screening and early detection of inflammatory arthritis are essential to optimize patient [quality of life] and reduce morbidity, providers may consider using a formal screening tool of their choice.”7

 

 

With these limitations in mind, I have found the Psoriasis Epidemiology Screening Tool (PEST) to be the most useful psoriatic arthritis screening tool. One study determined that the PEST has the best trade-off between sensitivity and specificity compared to 2 other psoriatic arthritis screening tools, the Psoriatic Arthritis Screening and Evaluation (PASE) and the Early Arthritis for Psoriatic Patients (EARP).8



The PEST is comprised of 5 questions: (1) Have you ever had a swollen joint (or joints)? (2) Has a doctor ever told you that you have arthritis? (3) Do your fingernails or toenails have holes or pits? (4) Have you had pain in your heel? (5) Have you had a finger or toe that was completely swollen and painful for no apparent reason? According to the PEST, a referral to a rheumatologist should be considered for patients answering yes to 3 or more questions, which is 97% sensitive and 79% specific for psoriatic arthritis.9 Patients who answer yes to fewer than 3 questions should still be referred to a rheumatologist if there is a strong clinical suspicion of psoriatic arthritis.10

The PEST can be accessed for free in 13 languages via the GRAPPA (Group for Research and Assessment of Psoriasis and Psoriatic Arthritis) app as well as downloaded for free from the National Psoriasis Foundation’s website (https://www.psoriasis.org/psa-screening/providers).

ALT-70 Differentiates Cellulitis From Pseudocellulitis

Overdiagnosing cellulitis in the United States has been estimated to result in up to 130,000 unnecessary hospitalizations and up to $515 million in avoidable health care spending.11 Dermatologists are in a unique position to help fix this issue. In one retrospective study of 1430 inpatient dermatology consultations, 74.32% of inpatients evaluated for presumed cellulitis by a dermatologist were instead diagnosed with a cellulitis mimicker (ie, pseudocellulitis), such as stasis dermatitis or contact dermatitis.12

The ALT-70 score was developed and prospectively validated to help differentiate lower extremity cellulitis from pseudocellulitis in adult patients in the emergency department (ED).13 In addition, the score has retrospectively been shown to function similarly in the inpatient setting when calculated at 24 and 48 hours after ED presentation.14 Although the ALT-70 score was designed for use by frontline clinicians prior to dermatology consultation, I also have found it helpful to calculate as a consultant, as it provides an objective measure of risk to communicate to the primary team in support of one diagnosis or another.

 

 


ALT-70 is an acronym for the score’s 4 variables: asymmetry, leukocytosis, tachycardia, and age of 70 years or older.15 If present, each variable confers a certain number of points to the final score: 3 points for asymmetry (defined as unilateral leg involvement), 1 point for leukocytosis (white blood cell count ≥10,000/μL), 1 point for tachycardia (≥90 beats per minute), and 2 points for age of 70 years or older. An ALT-70 score of 0 to 2 corresponds to an 83.3% or greater chance of pseudocellulitis, suggesting that the diagnosis of cellulitis be reconsidered. A score of 3 to 4 is indeterminate, and additional information such as a dermatology consultation should be pursued. A score of 5 to 7 corresponds to an 82.2% or greater chance of cellulitis, signifying that empiric treatment with antibiotics be considered.15



The ALT-70 score does not apply to cases involving areas other than the lower extremities; intravenous antibiotic use within 48 hours before ED presentation; surgery within the last 30 days; abscess; penetrating trauma; burn; or known history of osteomyelitis, diabetic ulcer, or indwelling hardware at the site of infection.15 The ALT-70 score is available for free via the MDCalc app and website (https://www.mdcalc.com/alt-70-score-cellulitis).

Mohs AUC Determines the Appropriateness of Mohs Micrographic Surgery

In 2012, the American Academy of Dermatology, American College of Mohs Surgery, American Society for Dermatologic Surgery Association, and American Society for Mohs Surgery published appropriate use criteria (AUC) to guide the decision to pursue Mohs micrographic surgery (MMS) in the United States.16 Based on various tumor and patient characteristics, the Mohs AUC assign scores to 270 different clinical scenarios. A score of 1 to 3 signifies that MMS is inappropriate and generally not considered acceptable. A score 4 to 6 indicates that the appropriateness of MMS is uncertain. A score 7 to 9 means that MMS is appropriate and generally considered acceptable.16

Since publication, the Mohs AUC have been criticized for classifying most primary superficial basal cell carcinomas as appropriate for MMS17 (which an AUC coauthor18 and others19,20 have defended), excluding certain reasons for performing MMS (such as operating on multiple tumors on the same day),21 including counterintuitive scores,22 and omitting trials from Europe23 (which AUC coauthors also have defended24). As with any clinical scoring system, the Mohs AUC has limitations; the creators acknowledge that “. . . these criteria should not be interpreted as setting a standard of care, or be deemed inclusive of all proper methods of care nor exclusive of other methods of care reasonably directed to obtaining the same results, even for those indications scored as inappropriate.”16 The Mohs AUC app (https://www.aad.org/members/aad-apps/mohs-auc) is free and allows users to enter tumor and patient characteristics to determine the score for their specific scenario.

Final Thoughts

Scoring systems are emerging in dermatology as evidence-based bedside tools to help guide clinical decision-making. Despite their limitations, these scores have the potential to make a meaningful impact in dermatology as they have in other specialties.

The practice of dermatology is rife with bedside tools: swabs, smears, and scoring systems. First popularized in specialties such as emergency medicine and internal medicine, clinical scoring systems are now emerging in dermatology. These evidence-based scores can be calculated quickly at the bedside—often through a free smartphone app—to help guide clinical decision-making regarding diagnosis, prognosis, and management. As with any medical tool, scoring systems have limitations and should be used as a supplement, not substitute, for one’s clinical judgement. This article reviews 4 clinical scoring systems practical for dermatology residents.

SCORTEN Prognosticates Cases of Stevens-Johnson Syndrome/Toxic Epidermal Necrolysis

Perhaps the best-known scoring system in dermatology, the SCORTEN is widely used to predict hospital mortality from Stevens-Johnson syndrome/toxic epidermal necrolysis. The SCORTEN includes 7 variables of equal weight—age of 40 years or older, heart rate of 120 beats per minute or more, cancer/hematologic malignancy, involved body surface area (BSA) greater than 10%, serum urea greater than 10 mmol/L, serum bicarbonate less than 20 mmol/L, and serum glucose greater than 14 mmol/L—each contributing 1 point to the overall score if present.1 The involved BSA is defined as the sum of detached and detachable epidermis.1

The SCORTEN was developed and prospectively validated to be calculated at the end of the first 24 hours of admission; for this calculation, use the BSA affected at that time, and use the most abnormal values during the first 24 hours of admission for the other variables.1 In addition, a follow-up study including some of the original coauthors recommends recalculating the SCORTEN at the end of hospital day 3, having found that the score’s predictive value was better on this day than hospital days 1, 2, 4, or 5.2 Based on the original study, a SCORTEN of 0 to 1 corresponds to a mortality rate of 3.2%, 2 to 12.1%, 3 to 35.3%, 4 to 58.3%, and 5 or greater to 90.0%.1



Limitations of the SCORTEN include its ability to overestimate or underestimate mortality as demonstrated by 2 multi-institutional cohorts.3,4 Recently, the ABCD-10 score was developed as an alternative to the SCORTEN and was found to predict mortality similarly when validated in an internal cohort.5

PEST Screens for Psoriatic Arthritis

Dermatologists play an important role in screening for psoriatic arthritis, as an estimated 1 in 5 patients with psoriasis have psoriatic arthritis.6 To this end, several screening tools have been developed to help differentiate psoriatic arthritis from other arthritides. Joint guidelines from the American Academy of Dermatology and the National Psoriasis Foundation acknowledge that “. . . these screening tools have tended to perform less well when tested in groups of people other than those for which they were originally developed. As such, their usefulness in routine clinical practice remains controversial.”7 Nevertheless, the guidelines state, “[b]ecause screening and early detection of inflammatory arthritis are essential to optimize patient [quality of life] and reduce morbidity, providers may consider using a formal screening tool of their choice.”7

 

 

With these limitations in mind, I have found the Psoriasis Epidemiology Screening Tool (PEST) to be the most useful psoriatic arthritis screening tool. One study determined that the PEST has the best trade-off between sensitivity and specificity compared to 2 other psoriatic arthritis screening tools, the Psoriatic Arthritis Screening and Evaluation (PASE) and the Early Arthritis for Psoriatic Patients (EARP).8



The PEST is comprised of 5 questions: (1) Have you ever had a swollen joint (or joints)? (2) Has a doctor ever told you that you have arthritis? (3) Do your fingernails or toenails have holes or pits? (4) Have you had pain in your heel? (5) Have you had a finger or toe that was completely swollen and painful for no apparent reason? According to the PEST, a referral to a rheumatologist should be considered for patients answering yes to 3 or more questions, which is 97% sensitive and 79% specific for psoriatic arthritis.9 Patients who answer yes to fewer than 3 questions should still be referred to a rheumatologist if there is a strong clinical suspicion of psoriatic arthritis.10

The PEST can be accessed for free in 13 languages via the GRAPPA (Group for Research and Assessment of Psoriasis and Psoriatic Arthritis) app as well as downloaded for free from the National Psoriasis Foundation’s website (https://www.psoriasis.org/psa-screening/providers).

ALT-70 Differentiates Cellulitis From Pseudocellulitis

Overdiagnosing cellulitis in the United States has been estimated to result in up to 130,000 unnecessary hospitalizations and up to $515 million in avoidable health care spending.11 Dermatologists are in a unique position to help fix this issue. In one retrospective study of 1430 inpatient dermatology consultations, 74.32% of inpatients evaluated for presumed cellulitis by a dermatologist were instead diagnosed with a cellulitis mimicker (ie, pseudocellulitis), such as stasis dermatitis or contact dermatitis.12

The ALT-70 score was developed and prospectively validated to help differentiate lower extremity cellulitis from pseudocellulitis in adult patients in the emergency department (ED).13 In addition, the score has retrospectively been shown to function similarly in the inpatient setting when calculated at 24 and 48 hours after ED presentation.14 Although the ALT-70 score was designed for use by frontline clinicians prior to dermatology consultation, I also have found it helpful to calculate as a consultant, as it provides an objective measure of risk to communicate to the primary team in support of one diagnosis or another.

 

 


ALT-70 is an acronym for the score’s 4 variables: asymmetry, leukocytosis, tachycardia, and age of 70 years or older.15 If present, each variable confers a certain number of points to the final score: 3 points for asymmetry (defined as unilateral leg involvement), 1 point for leukocytosis (white blood cell count ≥10,000/μL), 1 point for tachycardia (≥90 beats per minute), and 2 points for age of 70 years or older. An ALT-70 score of 0 to 2 corresponds to an 83.3% or greater chance of pseudocellulitis, suggesting that the diagnosis of cellulitis be reconsidered. A score of 3 to 4 is indeterminate, and additional information such as a dermatology consultation should be pursued. A score of 5 to 7 corresponds to an 82.2% or greater chance of cellulitis, signifying that empiric treatment with antibiotics be considered.15



The ALT-70 score does not apply to cases involving areas other than the lower extremities; intravenous antibiotic use within 48 hours before ED presentation; surgery within the last 30 days; abscess; penetrating trauma; burn; or known history of osteomyelitis, diabetic ulcer, or indwelling hardware at the site of infection.15 The ALT-70 score is available for free via the MDCalc app and website (https://www.mdcalc.com/alt-70-score-cellulitis).

Mohs AUC Determines the Appropriateness of Mohs Micrographic Surgery

In 2012, the American Academy of Dermatology, American College of Mohs Surgery, American Society for Dermatologic Surgery Association, and American Society for Mohs Surgery published appropriate use criteria (AUC) to guide the decision to pursue Mohs micrographic surgery (MMS) in the United States.16 Based on various tumor and patient characteristics, the Mohs AUC assign scores to 270 different clinical scenarios. A score of 1 to 3 signifies that MMS is inappropriate and generally not considered acceptable. A score 4 to 6 indicates that the appropriateness of MMS is uncertain. A score 7 to 9 means that MMS is appropriate and generally considered acceptable.16

Since publication, the Mohs AUC have been criticized for classifying most primary superficial basal cell carcinomas as appropriate for MMS17 (which an AUC coauthor18 and others19,20 have defended), excluding certain reasons for performing MMS (such as operating on multiple tumors on the same day),21 including counterintuitive scores,22 and omitting trials from Europe23 (which AUC coauthors also have defended24). As with any clinical scoring system, the Mohs AUC has limitations; the creators acknowledge that “. . . these criteria should not be interpreted as setting a standard of care, or be deemed inclusive of all proper methods of care nor exclusive of other methods of care reasonably directed to obtaining the same results, even for those indications scored as inappropriate.”16 The Mohs AUC app (https://www.aad.org/members/aad-apps/mohs-auc) is free and allows users to enter tumor and patient characteristics to determine the score for their specific scenario.

Final Thoughts

Scoring systems are emerging in dermatology as evidence-based bedside tools to help guide clinical decision-making. Despite their limitations, these scores have the potential to make a meaningful impact in dermatology as they have in other specialties.

References
  1. Bastuji-Garin S, Fouchard N, Bertocchi M, et al. SCORTEN: a severity-of-illness score for toxic epidermal necrolysis. J Invest Dermatol. 2000;115:149-153.
  2. Guegan S, Bastuji-Garin S, Poszepczynska-Guigne E, et al. Performance of the SCORTEN during the first five days of hospitalization to predict the prognosis of epidermal necrolysis. J Invest Dermatol. 2006;126:272-276.
  3. Micheletti RG, Chiesa-Fuxench Z, Noe MH, et al. Stevens-Johnson syndrome/toxic epidermal necrolysis: a multicenter retrospective study of 377 adult patients from the United States. J Invest Dermatol. 2018;138:2315-2321.
  4. Sekula P, Liss Y, Davidovici B, et al. Evaluation of SCORTEN on a cohort of patients with Stevens-Johnson syndrome and toxic epidermal necrolysis included in the RegiSCAR study. J Burn Care Res. 2011;32:237-245.
  5. Noe MH, Rosenbach M, Hubbard RA, et al. Development and validation of a risk prediction model for in-hospital mortality among patients with Stevens-Johnson syndrome/toxic epidermal necrolysis-ABCD-10. JAMA Dermatol. 2019;155:448-454.
  6. Alinaghi F, Calov M, Kristensen LE, et al. Prevalence of psoriatic arthritis in patients with psoriasis: a systematic review and meta-analysis of observational and clinical studies. J Am Acad Dermatol. 2019;80:251-265.e219.
  7. Elmets CA, Leonardi CL, Davis DMR, et al. Joint AAD-NPF guidelines of care for the management and treatment of psoriasis with awareness and attention to comorbidities. J Am Acad Dermatol. 2019;80:1073-1113.
  8. Karreman MC, Weel A, van der Ven M, et al. Performance of screening tools for psoriatic arthritis: a cross-sectional study in primary care. Rheumatology (Oxford). 2017;56:597-602.
     

     

  9. Ibrahim GH, Buch MH, Lawson C, et al. Evaluation of an existing screening tool for psoriatic arthritis in people with psoriasis and the development of a new instrument: the Psoriasis Epidemiology Screening Tool (PEST) questionnaire. Clin Exp Rheumatol. 2009;27:469-474.
  10. Zhang A, Kurtzman DJB, Perez-Chada LM, et al. Psoriatic arthritis and the dermatologist: an approach to screening and clinical evaluation. Clin Dermatol. 2018;36:551-560.
  11. Weng QY, Raff AB, Cohen JM, et al. Costs and consequences associated with misdiagnosed lower extremity cellulitis. JAMA Dermatol. 2017;153:141-146.
  12. Strazzula L, Cotliar J, Fox LP, et al. Inpatient dermatology consultation aids diagnosis of cellulitis among hospitalized patients: a multi-institutional analysis. J Am Acad Dermatol. 2015;73:70-75.
  13. Li DG, Dewan AK, Xia FD, et al. The ALT-70 predictive model outperforms thermal imaging for the diagnosis of lower extremity cellulitis: a prospective evaluation. J Am Acad Dermatol. 2018;79:1076-1080.e1071.
  14. Singer S, Li DG, Gunasekera N, et al. The ALT-70 predictive model maintains predictive value at 24 and 48 hours after presentation [published online March 23, 2019]. J Am Acad Dermatol. doi:10.1016/j.jaad.2019.03.050.
  15. Raff AB, Weng QY, Cohen JM, et al. A predictive model for diagnosis of lower extremity cellulitis: a cross-sectional study. J Am Acad Dermatol. 2017;76:618-625.e2.
  16. Connolly SM, Baker DR, Coldiron BM, et al. AAD/ACMS/ASDSA/ASMS 2012 appropriate use criteria for Mohs micrographic surgery: a report of the American Academy of Dermatology, American College of Mohs Surgery, American Society for Dermatologic Surgery Association, and the American Society for Mohs Surgery. J Am Acad Dermatol. 2012;67:531-550.
  17. Steinman HK, Dixon A, Zachary CB. Reevaluating Mohs surgery appropriate use criteria for primary superficial basal cell carcinoma. JAMA Dermatol. 2018;154:755-756.
  18. Montuno MA, Coldiron BM. Mohs appropriate use criteria for superficial basal cell carcinoma. JAMA Dermatol. 2019;155:394-395.
  19. MacFarlane DF, Perlis C. Mohs appropriate use criteria for superficial basal cell carcinoma. JAMA Dermatol. 2019;155:395-396.
  20. Kantor J. Mohs appropriate use criteria for superficial basal cell carcinoma. JAMA Dermatol. 2019;155:395.
  21. Ruiz ES, Karia PS, Morgan FC, et al. Multiple Mohs micrographic surgery is the most common reason for divergence from the appropriate use criteria: a single institution retrospective cohort study. J Am Acad Dermatol. 2016;75:830-831.
  22. Croley JA, Joseph AK, Wagner RF Jr. Discrepancies in the Mohs Micrographic Surgery appropriate use criteria [published online December 23, 2018]. J Am Acad Dermatol. doi:10.1016/j.jaad.2018.11.064.
  23. Kelleners-Smeets NW, Mosterd K. Comment on 2012 appropriate use criteria for Mohs micrographic surgery. J Am Acad Dermatol. 2013;69:317-318.
  24. Connolly S, Baker D, Coldiron B, et al. Reply to “comment on 2012 appropriate use criteria for Mohs micrographic surgery.” J Am Acad Dermatol. 2013;69:318.
References
  1. Bastuji-Garin S, Fouchard N, Bertocchi M, et al. SCORTEN: a severity-of-illness score for toxic epidermal necrolysis. J Invest Dermatol. 2000;115:149-153.
  2. Guegan S, Bastuji-Garin S, Poszepczynska-Guigne E, et al. Performance of the SCORTEN during the first five days of hospitalization to predict the prognosis of epidermal necrolysis. J Invest Dermatol. 2006;126:272-276.
  3. Micheletti RG, Chiesa-Fuxench Z, Noe MH, et al. Stevens-Johnson syndrome/toxic epidermal necrolysis: a multicenter retrospective study of 377 adult patients from the United States. J Invest Dermatol. 2018;138:2315-2321.
  4. Sekula P, Liss Y, Davidovici B, et al. Evaluation of SCORTEN on a cohort of patients with Stevens-Johnson syndrome and toxic epidermal necrolysis included in the RegiSCAR study. J Burn Care Res. 2011;32:237-245.
  5. Noe MH, Rosenbach M, Hubbard RA, et al. Development and validation of a risk prediction model for in-hospital mortality among patients with Stevens-Johnson syndrome/toxic epidermal necrolysis-ABCD-10. JAMA Dermatol. 2019;155:448-454.
  6. Alinaghi F, Calov M, Kristensen LE, et al. Prevalence of psoriatic arthritis in patients with psoriasis: a systematic review and meta-analysis of observational and clinical studies. J Am Acad Dermatol. 2019;80:251-265.e219.
  7. Elmets CA, Leonardi CL, Davis DMR, et al. Joint AAD-NPF guidelines of care for the management and treatment of psoriasis with awareness and attention to comorbidities. J Am Acad Dermatol. 2019;80:1073-1113.
  8. Karreman MC, Weel A, van der Ven M, et al. Performance of screening tools for psoriatic arthritis: a cross-sectional study in primary care. Rheumatology (Oxford). 2017;56:597-602.
     

     

  9. Ibrahim GH, Buch MH, Lawson C, et al. Evaluation of an existing screening tool for psoriatic arthritis in people with psoriasis and the development of a new instrument: the Psoriasis Epidemiology Screening Tool (PEST) questionnaire. Clin Exp Rheumatol. 2009;27:469-474.
  10. Zhang A, Kurtzman DJB, Perez-Chada LM, et al. Psoriatic arthritis and the dermatologist: an approach to screening and clinical evaluation. Clin Dermatol. 2018;36:551-560.
  11. Weng QY, Raff AB, Cohen JM, et al. Costs and consequences associated with misdiagnosed lower extremity cellulitis. JAMA Dermatol. 2017;153:141-146.
  12. Strazzula L, Cotliar J, Fox LP, et al. Inpatient dermatology consultation aids diagnosis of cellulitis among hospitalized patients: a multi-institutional analysis. J Am Acad Dermatol. 2015;73:70-75.
  13. Li DG, Dewan AK, Xia FD, et al. The ALT-70 predictive model outperforms thermal imaging for the diagnosis of lower extremity cellulitis: a prospective evaluation. J Am Acad Dermatol. 2018;79:1076-1080.e1071.
  14. Singer S, Li DG, Gunasekera N, et al. The ALT-70 predictive model maintains predictive value at 24 and 48 hours after presentation [published online March 23, 2019]. J Am Acad Dermatol. doi:10.1016/j.jaad.2019.03.050.
  15. Raff AB, Weng QY, Cohen JM, et al. A predictive model for diagnosis of lower extremity cellulitis: a cross-sectional study. J Am Acad Dermatol. 2017;76:618-625.e2.
  16. Connolly SM, Baker DR, Coldiron BM, et al. AAD/ACMS/ASDSA/ASMS 2012 appropriate use criteria for Mohs micrographic surgery: a report of the American Academy of Dermatology, American College of Mohs Surgery, American Society for Dermatologic Surgery Association, and the American Society for Mohs Surgery. J Am Acad Dermatol. 2012;67:531-550.
  17. Steinman HK, Dixon A, Zachary CB. Reevaluating Mohs surgery appropriate use criteria for primary superficial basal cell carcinoma. JAMA Dermatol. 2018;154:755-756.
  18. Montuno MA, Coldiron BM. Mohs appropriate use criteria for superficial basal cell carcinoma. JAMA Dermatol. 2019;155:394-395.
  19. MacFarlane DF, Perlis C. Mohs appropriate use criteria for superficial basal cell carcinoma. JAMA Dermatol. 2019;155:395-396.
  20. Kantor J. Mohs appropriate use criteria for superficial basal cell carcinoma. JAMA Dermatol. 2019;155:395.
  21. Ruiz ES, Karia PS, Morgan FC, et al. Multiple Mohs micrographic surgery is the most common reason for divergence from the appropriate use criteria: a single institution retrospective cohort study. J Am Acad Dermatol. 2016;75:830-831.
  22. Croley JA, Joseph AK, Wagner RF Jr. Discrepancies in the Mohs Micrographic Surgery appropriate use criteria [published online December 23, 2018]. J Am Acad Dermatol. doi:10.1016/j.jaad.2018.11.064.
  23. Kelleners-Smeets NW, Mosterd K. Comment on 2012 appropriate use criteria for Mohs micrographic surgery. J Am Acad Dermatol. 2013;69:317-318.
  24. Connolly S, Baker D, Coldiron B, et al. Reply to “comment on 2012 appropriate use criteria for Mohs micrographic surgery.” J Am Acad Dermatol. 2013;69:318.
Issue
Cutis - 104(3)
Issue
Cutis - 104(3)
Page Number
E37-E39
Page Number
E37-E39
Publications
Publications
Topics
Article Type
Sections
Inside the Article

Resident Pearls

  • Mortality from Stevens-Johnson syndrome/toxic epidermal necrolysis can be estimated by calculating the SCORTEN at the end of days 1 and 3 of hospitalization.
  • The Psoriasis Epidemiology Screening Tool (PEST) assists with triaging which patients with psoriasis should be evaluated for psoriatic arthritis by a rheumatologist.
  • The ALT-70 score is helpful to support one’s diagnosis of cellulitis or pseudocellulitis.
  • The Mohs appropriate use criteria (AUC) score 270 different clinical scenarios as appropriate, uncertain, or inappropriate for Mohs micrographic surgery.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Article PDF Media

The ABCs of COCs: A Guide for Dermatology Residents on Combined Oral Contraceptives

Article Type
Changed
Tue, 01/07/2020 - 09:10
Display Headline
The ABCs of COCs: A Guide for Dermatology Residents on Combined Oral Contraceptives

The American Academy of Dermatology confers combined oral contraceptives (COCs) a strength A recommendation for the treatment of acne based on level I evidence, and 4 COCs are approved for the treatment of acne by the US Food and Drug Administration (FDA).1 Furthermore, when dermatologists prescribe isotretinoin and thalidomide to women of reproductive potential, the iPLEDGE and THALOMID Risk Evaluation and Mitigation Strategy (REMS) programs require 2 concurrent methods of contraception, one of which may be a COC. In addition, COCs have several potential off-label indications in dermatology including idiopathic hirsutism, female pattern hair loss, hidradenitis suppurativa, and autoimmune progesterone dermatitis.

Despite this evidence and opportunity, research suggests that dermatologists underprescribe COCs. The National Ambulatory Medical Care Survey found that between 1993 and 2008, dermatologists in the United States prescribed COCs to only 2.03% of women presenting for acne treatment, which was less often than obstetricians/gynecologists (36.03%) and internists (10.76%).2 More recently, in a survey of 130 US dermatologists conducted from 2014 to 2015, only 55.4% reported prescribing COCs. This survey also found that only 45.8% of dermatologists who prescribed COCs felt very comfortable counseling on how to begin taking them, only 48.6% felt very comfortable counseling patients on side effects, and only 22.2% felt very comfortable managing side effects.3

In light of these data, this article reviews the basics of COCs for dermatology residents, from assessing patient eligibility and selecting a COC to counseling on use and managing risks and side effects. Because there are different approaches to prescribing COCs, readers are encouraged to integrate the information in this article with what they have learned from other sources.

Assess Patient Eligibility

In general, patients should be at least 14 years of age and have waited 2 years after menarche to start COCs. They can be taken until menopause.1,4 Contraindications can be screened for by taking a medical history and measuring a baseline blood pressure (Tables 1 and 2).5 In addition, pregnancy should be excluded with a urine or serum pregnancy test or criteria provided in Box 2 of the 2016 US Selected Practice Recommendations for Contraceptive Use from the Centers for Disease Control and Prevention (CDC).4 Although important for women’s overall health, a pelvic examination is not required to start COCs according to the CDC and the American Academy of Dermatology.1,4

Select the COC

Combined oral contraceptives combine estrogen, usually in the form of ethinyl estradiol, with a progestin. Data suggest that all COCs effectively treat acne, but 4 are specifically FDA approved for acne: ethinyl estradiol–norethindrone acetate–ferrous fumarate, ethinyl estradiol–norgestimate, ethinyl estradiol–drospirenone, and ethinyl estradiol–drospirenone–levomefolate.1 Ethinyl estradiol–desogestrel and ethinyl estradiol–drospirenone are 2 go-to COCs for some of the attending physicians at my residency program. All COCs are FDA approved for contraception. When selecting a COC, one approach is to start with the patient’s drug formulary, then consider the following characteristics.

 

 

Monophasic vs Multiphasic
All the hormonally active pills in a monophasic formulation contain the same dose of estrogen and progestin; however, these doses change per pill in a multiphasic formulation, which requires that patients take the pills in a specific order. Given this greater complexity and the fact that multiphasic formulations often are more expensive and lack evidence of superiority, a 2011 Cochrane review recommended monophasic formulations as first line.6 In addition, monophasic formulations are preferred for autoimmune progesterone dermatitis because of the stable progestin dose.



Hormone-Free Interval
Some COCs include placebo pills during which hormone withdrawal symptoms such as bleeding, pelvic pain, mood changes, and headache may occur. If a patient is concerned about these symptoms, choose a COC with no or fewer placebo pills, or have the patient skip the hormone-free interval altogether and start the next pack early7; in this case, the prescription should be written with instructions to allow the patient to get earlier refills from the pharmacy.

Estrogen Dose
To minimize estrogen-related side effects, the lowest possible dose of ethinyl estradiol that is effective and tolerable should be prescribed7,8; 20 μg of ethinyl estradiol generally is the lowest dose available, but it may be associated with more frequent breakthrough bleeding.9 The International Planned Parenthood Federation recommends starting with COCs that contain 30 to 35 μg of estrogen.10 Synthesizing this information, one option is to start with 20 μg of ethinyl estradiol and increase the dose if breakthrough bleeding persists after 3 cycles.

Progestin Type
First-generation progestins (eg, norethindrone), second-generation progestins (eg, norgestrel, levonorgestrel), and third-generation progestins (eg, norgestimate, desogestrel) are derived from testosterone and therefore are variably androgenic; second-generation progestins are the most androgenic, and third-generation progestins are the least. On the other hand, drospirenone, the fourth-generation progestin available in the United States, is derived from 17α-spironolactone and thus is mildly antiandrogenic (3 mg of drospirenone is considered equivalent to 25 mg of spironolactone).

Although COCs with less androgenic progestins should theoretically treat acne better, a 2012 Cochrane review of COCs and acne concluded that “differences in the comparative effectiveness of COCs containing varying progestin types and dosages were less clear, and data were limited for any particular comparison.”11 As a result, regardless of the progestin, all COCs are believed to have a net antiandrogenic effect due to their estrogen component.1

Counsel on Use

Combined oral contraceptives can be started on any day of the menstrual cycle, including the day the prescription is given. If a patient begins a COC within 5 days of the first day of her most recent period, backup contraception is not needed.4 If she begins the COC more than 5 days after the first day of her most recent period, she needs to use backup contraception or abstain from sexual intercourse for the next 7 days.4 In general, at least 3 months of therapy are required to evaluate the effectiveness of COCs for acne.1

Manage Risks and Side Effects

Breakthrough Bleeding
The most common side effect of breakthrough bleeding can be minimized by taking COCs at approximately the same time every day and avoiding missed pills. If breakthrough bleeding does not stop after 3 cycles, consider increasing the estrogen dose to 30 to 35 μg and/or referring to an obstetrician/gynecologist to rule out other etiologies of bleeding.7,8

 

 

Nausea, Headache, Bloating, and Breast Tenderness
These symptoms typically resolve after the first 3 months. To minimize nausea, patients should take COCs in the early evening and eat breakfast the next morning.7,8 For headaches that occur during the hormone-free interval, consider skipping the placebo pills and starting the next pack early. Switching the progestin to drospirenone, which has a mild diuretic effect, can help with bloating as well as breast tenderness.7 For persistent symptoms, consider a lower estrogen dose.7,8



Changes in Libido
In a systemic review including 8422 COC users, 64% reported no change in libido, 22% reported an increase, and 15% reported a decrease.12

Weight Gain
Although patients may be concerned that COCs cause weight gain, a 2014 Cochrane review concluded that “available evidence is insufficient to determine the effect of combination contraceptives on weight, but no large effect is evident.”13 If weight gain does occur, anecdotal evidence suggests it tends to be not more than 5 pounds. If weight gain is an issue, consider a less androgenic progestin.8

Venous Thromboembolism
Use the 3-6-9-12 model to contextualize venous thromboembolism (VTE) risk: a woman’s annual VTE risk is 3 per 10,000 women at baseline, 6 per 10,000 women with nondrospirenone COCs, 9 per 10,000 women with drospirenone-containing COCs, and 12 per 10,000 women when pregnant.14 Patients should be counseled on the signs and symptoms of VTE such as unilateral or bilateral leg or arm swelling, pain, warmth, redness, and/or shortness of breath. The British Society for Haematology recommends maintaining mobility as a reasonable precaution when traveling for more than 3 hours.15

Cardiovascular Disease
A 2015 Cochrane review found that the risk for myocardial infarction or ischemic stroke is increased 1.6‐fold in COC users.16 Despite this increased relative risk, the increased absolute annual risk of myocardial infarction in nonsmoking women remains low: increased from 0.83 to 3.53 per 10,000,000 women younger than 35 years and from 9.45 to 40.4 per 10,000,000 women 35 years and older.17

Breast Cancer and Cervical Cancer
Data are mixed on the effect of COCs on the risk for breast cancer and cervical cancer.1 According to the CDC, COC use for 5 or more years might increase the risk of cervical carcinoma in situ and invasive cervical carcinoma in women with persistent human papillomavirus infection.5 Regardless of COC use, women should undergo age-appropriate screening for breast cancer and cervical cancer.



Melasma
Melasma is an estrogen-mediated side effect of COCs.8 A study from 1967 found that 29% of COC users (N=212) developed melasma; however, they were taking COCs with much higher ethinyl estradiol doses (50–100 μg) than typically used today.18 Nevertheless, as part of an overall skin care regimen, photoprotection should be encouraged with a broad-spectrum, water-resistant sunscreen that has a sun protection factor of at least 30. In addition, sunscreens with iron oxides have been shown to better prevent melasma relapse by protecting against the shorter wavelengths of visible light.19

References
  1. Zaenglein AL, Pathy AL, Schlosser BJ, et al. Guidelines of care for the management of acne vulgaris. J Am Acad Dermatol. 2016;74:945-973.e933.
  2. Landis ET, Levender MM, Davis SA, et al. Isotretinoin and oral contraceptive use in female acne patients varies by physician specialty: analysis of data from the National Ambulatory Medical Care Survey. J Dermatolog Treat. 2012;23:272-277.
  3. Fitzpatrick L, Mauer E, Chen CL. Oral contraceptives for acne treatment: US dermatologists’ knowledge, comfort, and prescribing practices. Cutis. 2017;99:195-201.
  4. Curtis KM, Jatlaoui TC, Tepper NK, et al. U.S. Selected Practice Recommendations for Contraceptive Use, 2016. MMWR Recomm Rep. 2016;65:1-66.
  5. Curtis KM, Tepper NK, Jatlaoui TC, et al. U.S. Medical Eligibility Criteria for Contraceptive Use, 2016. MMWR Recomm Rep. 2016;65:1-103.
  6. Van Vliet HA, Grimes DA, Lopez LM, et al. Triphasic versus monophasic oral contraceptives for contraception. Cochrane Database Syst Rev. 2011:CD003553.
  7. Stewart M, Black K. Choosing a combined oral contraceptive pill. Aust Prescr. 2015;38:6-11.
  8. McKinney K. Understanding the options: a guide to oral contraceptives. https://www.cecentral.com/assets/2097/022%20Oral%20Contraceptives%2010-26-09.pdf. Published November 5, 2009. Accessed June 20, 2019.
  9. Gallo MF, Nanda K, Grimes DA, et al. 20 microg versus >20 microg estrogen combined oral contraceptives for contraception. Cochrane Database Syst Rev. 2013:CD003989.
  10. Terki F, Malhotra U. Medical and Service Delivery Guidelines for Sexual and Reproductive Health Services. London, United Kingdom: International Planned Parenthood Federation; 2004.
  11. Arowojolu AO, Gallo MF, Lopez LM, et al. Combined oral contraceptive pills for treatment of acne. Cochrane Database Syst Rev. 2012:CD004425.
  12. Pastor Z, Holla K, Chmel R. The influence of combined oral contraceptives on female sexual desire: a systematic review. Eur J Contracept Reprod Health Care. 2013;18:27-43.
  13. Gallo MF, Lopez LM, Grimes DA, et al. Combination contraceptives: effects on weight. Cochrane Database Syst Rev. 2014:CD003987.
  14. Birth control pills for acne: tips from Julie Harper at the Summer AAD. Cutis. https://www.mdedge.com/dermatology/article/144550/acne/birth-control-pills-acne-tips-julie-harper-summer-aad. Published August 14, 2017. Accessed June 24, 2019.
  15. Watson HG, Baglin TP. Guidelines on travel-related venous thrombosis. Br J Haematol. 2011;152:31-34.
  16. Roach RE, Helmerhorst FM, Lijfering WM, et al. Combined oral contraceptives: the risk of myocardial infarction and ischemic stroke. Cochrane Database Syst Rev. 2015:CD011054.
  17. Acute myocardial infarction and combined oral contraceptives: results of an international multicentre case-control study. WHO Collaborative Study of Cardiovascular Disease and Steroid Hormone Contraception. Lancet. 1997;349:1202-1209.
  18. Resnik S. Melasma induced by oral contraceptive drugs. JAMA. 1967;199:601-605.
  19. Boukari F, Jourdan E, Fontas E, et al. Prevention of melasma relapses with sunscreen combining protection against UV and short wavelengths of visible light: a prospective randomized comparative trial. J Am Acad Dermatol. 2015;72:189-190.e181.
Article PDF
Author and Disclosure Information

From the Department of Dermatology, State University of New York Downstate Medical Center, Brooklyn.

The author reports no conflict of interest.

Correspondence: Daniel R. Mazori, MD, Department of Dermatology, State University of New York Downstate Medical Center, 450 Clarkson Ave, Box 46, Brooklyn, NY 11203 (daniel.mazori@downstate.edu).

Issue
Cutis - 104(1)
Publications
Topics
Page Number
E10-E14
Sections
Author and Disclosure Information

From the Department of Dermatology, State University of New York Downstate Medical Center, Brooklyn.

The author reports no conflict of interest.

Correspondence: Daniel R. Mazori, MD, Department of Dermatology, State University of New York Downstate Medical Center, 450 Clarkson Ave, Box 46, Brooklyn, NY 11203 (daniel.mazori@downstate.edu).

Author and Disclosure Information

From the Department of Dermatology, State University of New York Downstate Medical Center, Brooklyn.

The author reports no conflict of interest.

Correspondence: Daniel R. Mazori, MD, Department of Dermatology, State University of New York Downstate Medical Center, 450 Clarkson Ave, Box 46, Brooklyn, NY 11203 (daniel.mazori@downstate.edu).

Article PDF
Article PDF

The American Academy of Dermatology confers combined oral contraceptives (COCs) a strength A recommendation for the treatment of acne based on level I evidence, and 4 COCs are approved for the treatment of acne by the US Food and Drug Administration (FDA).1 Furthermore, when dermatologists prescribe isotretinoin and thalidomide to women of reproductive potential, the iPLEDGE and THALOMID Risk Evaluation and Mitigation Strategy (REMS) programs require 2 concurrent methods of contraception, one of which may be a COC. In addition, COCs have several potential off-label indications in dermatology including idiopathic hirsutism, female pattern hair loss, hidradenitis suppurativa, and autoimmune progesterone dermatitis.

Despite this evidence and opportunity, research suggests that dermatologists underprescribe COCs. The National Ambulatory Medical Care Survey found that between 1993 and 2008, dermatologists in the United States prescribed COCs to only 2.03% of women presenting for acne treatment, which was less often than obstetricians/gynecologists (36.03%) and internists (10.76%).2 More recently, in a survey of 130 US dermatologists conducted from 2014 to 2015, only 55.4% reported prescribing COCs. This survey also found that only 45.8% of dermatologists who prescribed COCs felt very comfortable counseling on how to begin taking them, only 48.6% felt very comfortable counseling patients on side effects, and only 22.2% felt very comfortable managing side effects.3

In light of these data, this article reviews the basics of COCs for dermatology residents, from assessing patient eligibility and selecting a COC to counseling on use and managing risks and side effects. Because there are different approaches to prescribing COCs, readers are encouraged to integrate the information in this article with what they have learned from other sources.

Assess Patient Eligibility

In general, patients should be at least 14 years of age and have waited 2 years after menarche to start COCs. They can be taken until menopause.1,4 Contraindications can be screened for by taking a medical history and measuring a baseline blood pressure (Tables 1 and 2).5 In addition, pregnancy should be excluded with a urine or serum pregnancy test or criteria provided in Box 2 of the 2016 US Selected Practice Recommendations for Contraceptive Use from the Centers for Disease Control and Prevention (CDC).4 Although important for women’s overall health, a pelvic examination is not required to start COCs according to the CDC and the American Academy of Dermatology.1,4

Select the COC

Combined oral contraceptives combine estrogen, usually in the form of ethinyl estradiol, with a progestin. Data suggest that all COCs effectively treat acne, but 4 are specifically FDA approved for acne: ethinyl estradiol–norethindrone acetate–ferrous fumarate, ethinyl estradiol–norgestimate, ethinyl estradiol–drospirenone, and ethinyl estradiol–drospirenone–levomefolate.1 Ethinyl estradiol–desogestrel and ethinyl estradiol–drospirenone are 2 go-to COCs for some of the attending physicians at my residency program. All COCs are FDA approved for contraception. When selecting a COC, one approach is to start with the patient’s drug formulary, then consider the following characteristics.

 

 

Monophasic vs Multiphasic
All the hormonally active pills in a monophasic formulation contain the same dose of estrogen and progestin; however, these doses change per pill in a multiphasic formulation, which requires that patients take the pills in a specific order. Given this greater complexity and the fact that multiphasic formulations often are more expensive and lack evidence of superiority, a 2011 Cochrane review recommended monophasic formulations as first line.6 In addition, monophasic formulations are preferred for autoimmune progesterone dermatitis because of the stable progestin dose.



Hormone-Free Interval
Some COCs include placebo pills during which hormone withdrawal symptoms such as bleeding, pelvic pain, mood changes, and headache may occur. If a patient is concerned about these symptoms, choose a COC with no or fewer placebo pills, or have the patient skip the hormone-free interval altogether and start the next pack early7; in this case, the prescription should be written with instructions to allow the patient to get earlier refills from the pharmacy.

Estrogen Dose
To minimize estrogen-related side effects, the lowest possible dose of ethinyl estradiol that is effective and tolerable should be prescribed7,8; 20 μg of ethinyl estradiol generally is the lowest dose available, but it may be associated with more frequent breakthrough bleeding.9 The International Planned Parenthood Federation recommends starting with COCs that contain 30 to 35 μg of estrogen.10 Synthesizing this information, one option is to start with 20 μg of ethinyl estradiol and increase the dose if breakthrough bleeding persists after 3 cycles.

Progestin Type
First-generation progestins (eg, norethindrone), second-generation progestins (eg, norgestrel, levonorgestrel), and third-generation progestins (eg, norgestimate, desogestrel) are derived from testosterone and therefore are variably androgenic; second-generation progestins are the most androgenic, and third-generation progestins are the least. On the other hand, drospirenone, the fourth-generation progestin available in the United States, is derived from 17α-spironolactone and thus is mildly antiandrogenic (3 mg of drospirenone is considered equivalent to 25 mg of spironolactone).

Although COCs with less androgenic progestins should theoretically treat acne better, a 2012 Cochrane review of COCs and acne concluded that “differences in the comparative effectiveness of COCs containing varying progestin types and dosages were less clear, and data were limited for any particular comparison.”11 As a result, regardless of the progestin, all COCs are believed to have a net antiandrogenic effect due to their estrogen component.1

Counsel on Use

Combined oral contraceptives can be started on any day of the menstrual cycle, including the day the prescription is given. If a patient begins a COC within 5 days of the first day of her most recent period, backup contraception is not needed.4 If she begins the COC more than 5 days after the first day of her most recent period, she needs to use backup contraception or abstain from sexual intercourse for the next 7 days.4 In general, at least 3 months of therapy are required to evaluate the effectiveness of COCs for acne.1

Manage Risks and Side Effects

Breakthrough Bleeding
The most common side effect of breakthrough bleeding can be minimized by taking COCs at approximately the same time every day and avoiding missed pills. If breakthrough bleeding does not stop after 3 cycles, consider increasing the estrogen dose to 30 to 35 μg and/or referring to an obstetrician/gynecologist to rule out other etiologies of bleeding.7,8

 

 

Nausea, Headache, Bloating, and Breast Tenderness
These symptoms typically resolve after the first 3 months. To minimize nausea, patients should take COCs in the early evening and eat breakfast the next morning.7,8 For headaches that occur during the hormone-free interval, consider skipping the placebo pills and starting the next pack early. Switching the progestin to drospirenone, which has a mild diuretic effect, can help with bloating as well as breast tenderness.7 For persistent symptoms, consider a lower estrogen dose.7,8



Changes in Libido
In a systemic review including 8422 COC users, 64% reported no change in libido, 22% reported an increase, and 15% reported a decrease.12

Weight Gain
Although patients may be concerned that COCs cause weight gain, a 2014 Cochrane review concluded that “available evidence is insufficient to determine the effect of combination contraceptives on weight, but no large effect is evident.”13 If weight gain does occur, anecdotal evidence suggests it tends to be not more than 5 pounds. If weight gain is an issue, consider a less androgenic progestin.8

Venous Thromboembolism
Use the 3-6-9-12 model to contextualize venous thromboembolism (VTE) risk: a woman’s annual VTE risk is 3 per 10,000 women at baseline, 6 per 10,000 women with nondrospirenone COCs, 9 per 10,000 women with drospirenone-containing COCs, and 12 per 10,000 women when pregnant.14 Patients should be counseled on the signs and symptoms of VTE such as unilateral or bilateral leg or arm swelling, pain, warmth, redness, and/or shortness of breath. The British Society for Haematology recommends maintaining mobility as a reasonable precaution when traveling for more than 3 hours.15

Cardiovascular Disease
A 2015 Cochrane review found that the risk for myocardial infarction or ischemic stroke is increased 1.6‐fold in COC users.16 Despite this increased relative risk, the increased absolute annual risk of myocardial infarction in nonsmoking women remains low: increased from 0.83 to 3.53 per 10,000,000 women younger than 35 years and from 9.45 to 40.4 per 10,000,000 women 35 years and older.17

Breast Cancer and Cervical Cancer
Data are mixed on the effect of COCs on the risk for breast cancer and cervical cancer.1 According to the CDC, COC use for 5 or more years might increase the risk of cervical carcinoma in situ and invasive cervical carcinoma in women with persistent human papillomavirus infection.5 Regardless of COC use, women should undergo age-appropriate screening for breast cancer and cervical cancer.



Melasma
Melasma is an estrogen-mediated side effect of COCs.8 A study from 1967 found that 29% of COC users (N=212) developed melasma; however, they were taking COCs with much higher ethinyl estradiol doses (50–100 μg) than typically used today.18 Nevertheless, as part of an overall skin care regimen, photoprotection should be encouraged with a broad-spectrum, water-resistant sunscreen that has a sun protection factor of at least 30. In addition, sunscreens with iron oxides have been shown to better prevent melasma relapse by protecting against the shorter wavelengths of visible light.19

The American Academy of Dermatology confers combined oral contraceptives (COCs) a strength A recommendation for the treatment of acne based on level I evidence, and 4 COCs are approved for the treatment of acne by the US Food and Drug Administration (FDA).1 Furthermore, when dermatologists prescribe isotretinoin and thalidomide to women of reproductive potential, the iPLEDGE and THALOMID Risk Evaluation and Mitigation Strategy (REMS) programs require 2 concurrent methods of contraception, one of which may be a COC. In addition, COCs have several potential off-label indications in dermatology including idiopathic hirsutism, female pattern hair loss, hidradenitis suppurativa, and autoimmune progesterone dermatitis.

Despite this evidence and opportunity, research suggests that dermatologists underprescribe COCs. The National Ambulatory Medical Care Survey found that between 1993 and 2008, dermatologists in the United States prescribed COCs to only 2.03% of women presenting for acne treatment, which was less often than obstetricians/gynecologists (36.03%) and internists (10.76%).2 More recently, in a survey of 130 US dermatologists conducted from 2014 to 2015, only 55.4% reported prescribing COCs. This survey also found that only 45.8% of dermatologists who prescribed COCs felt very comfortable counseling on how to begin taking them, only 48.6% felt very comfortable counseling patients on side effects, and only 22.2% felt very comfortable managing side effects.3

In light of these data, this article reviews the basics of COCs for dermatology residents, from assessing patient eligibility and selecting a COC to counseling on use and managing risks and side effects. Because there are different approaches to prescribing COCs, readers are encouraged to integrate the information in this article with what they have learned from other sources.

Assess Patient Eligibility

In general, patients should be at least 14 years of age and have waited 2 years after menarche to start COCs. They can be taken until menopause.1,4 Contraindications can be screened for by taking a medical history and measuring a baseline blood pressure (Tables 1 and 2).5 In addition, pregnancy should be excluded with a urine or serum pregnancy test or criteria provided in Box 2 of the 2016 US Selected Practice Recommendations for Contraceptive Use from the Centers for Disease Control and Prevention (CDC).4 Although important for women’s overall health, a pelvic examination is not required to start COCs according to the CDC and the American Academy of Dermatology.1,4

Select the COC

Combined oral contraceptives combine estrogen, usually in the form of ethinyl estradiol, with a progestin. Data suggest that all COCs effectively treat acne, but 4 are specifically FDA approved for acne: ethinyl estradiol–norethindrone acetate–ferrous fumarate, ethinyl estradiol–norgestimate, ethinyl estradiol–drospirenone, and ethinyl estradiol–drospirenone–levomefolate.1 Ethinyl estradiol–desogestrel and ethinyl estradiol–drospirenone are 2 go-to COCs for some of the attending physicians at my residency program. All COCs are FDA approved for contraception. When selecting a COC, one approach is to start with the patient’s drug formulary, then consider the following characteristics.

 

 

Monophasic vs Multiphasic
All the hormonally active pills in a monophasic formulation contain the same dose of estrogen and progestin; however, these doses change per pill in a multiphasic formulation, which requires that patients take the pills in a specific order. Given this greater complexity and the fact that multiphasic formulations often are more expensive and lack evidence of superiority, a 2011 Cochrane review recommended monophasic formulations as first line.6 In addition, monophasic formulations are preferred for autoimmune progesterone dermatitis because of the stable progestin dose.



Hormone-Free Interval
Some COCs include placebo pills during which hormone withdrawal symptoms such as bleeding, pelvic pain, mood changes, and headache may occur. If a patient is concerned about these symptoms, choose a COC with no or fewer placebo pills, or have the patient skip the hormone-free interval altogether and start the next pack early7; in this case, the prescription should be written with instructions to allow the patient to get earlier refills from the pharmacy.

Estrogen Dose
To minimize estrogen-related side effects, the lowest possible dose of ethinyl estradiol that is effective and tolerable should be prescribed7,8; 20 μg of ethinyl estradiol generally is the lowest dose available, but it may be associated with more frequent breakthrough bleeding.9 The International Planned Parenthood Federation recommends starting with COCs that contain 30 to 35 μg of estrogen.10 Synthesizing this information, one option is to start with 20 μg of ethinyl estradiol and increase the dose if breakthrough bleeding persists after 3 cycles.

Progestin Type
First-generation progestins (eg, norethindrone), second-generation progestins (eg, norgestrel, levonorgestrel), and third-generation progestins (eg, norgestimate, desogestrel) are derived from testosterone and therefore are variably androgenic; second-generation progestins are the most androgenic, and third-generation progestins are the least. On the other hand, drospirenone, the fourth-generation progestin available in the United States, is derived from 17α-spironolactone and thus is mildly antiandrogenic (3 mg of drospirenone is considered equivalent to 25 mg of spironolactone).

Although COCs with less androgenic progestins should theoretically treat acne better, a 2012 Cochrane review of COCs and acne concluded that “differences in the comparative effectiveness of COCs containing varying progestin types and dosages were less clear, and data were limited for any particular comparison.”11 As a result, regardless of the progestin, all COCs are believed to have a net antiandrogenic effect due to their estrogen component.1

Counsel on Use

Combined oral contraceptives can be started on any day of the menstrual cycle, including the day the prescription is given. If a patient begins a COC within 5 days of the first day of her most recent period, backup contraception is not needed.4 If she begins the COC more than 5 days after the first day of her most recent period, she needs to use backup contraception or abstain from sexual intercourse for the next 7 days.4 In general, at least 3 months of therapy are required to evaluate the effectiveness of COCs for acne.1

Manage Risks and Side Effects

Breakthrough Bleeding
The most common side effect of breakthrough bleeding can be minimized by taking COCs at approximately the same time every day and avoiding missed pills. If breakthrough bleeding does not stop after 3 cycles, consider increasing the estrogen dose to 30 to 35 μg and/or referring to an obstetrician/gynecologist to rule out other etiologies of bleeding.7,8

 

 

Nausea, Headache, Bloating, and Breast Tenderness
These symptoms typically resolve after the first 3 months. To minimize nausea, patients should take COCs in the early evening and eat breakfast the next morning.7,8 For headaches that occur during the hormone-free interval, consider skipping the placebo pills and starting the next pack early. Switching the progestin to drospirenone, which has a mild diuretic effect, can help with bloating as well as breast tenderness.7 For persistent symptoms, consider a lower estrogen dose.7,8



Changes in Libido
In a systemic review including 8422 COC users, 64% reported no change in libido, 22% reported an increase, and 15% reported a decrease.12

Weight Gain
Although patients may be concerned that COCs cause weight gain, a 2014 Cochrane review concluded that “available evidence is insufficient to determine the effect of combination contraceptives on weight, but no large effect is evident.”13 If weight gain does occur, anecdotal evidence suggests it tends to be not more than 5 pounds. If weight gain is an issue, consider a less androgenic progestin.8

Venous Thromboembolism
Use the 3-6-9-12 model to contextualize venous thromboembolism (VTE) risk: a woman’s annual VTE risk is 3 per 10,000 women at baseline, 6 per 10,000 women with nondrospirenone COCs, 9 per 10,000 women with drospirenone-containing COCs, and 12 per 10,000 women when pregnant.14 Patients should be counseled on the signs and symptoms of VTE such as unilateral or bilateral leg or arm swelling, pain, warmth, redness, and/or shortness of breath. The British Society for Haematology recommends maintaining mobility as a reasonable precaution when traveling for more than 3 hours.15

Cardiovascular Disease
A 2015 Cochrane review found that the risk for myocardial infarction or ischemic stroke is increased 1.6‐fold in COC users.16 Despite this increased relative risk, the increased absolute annual risk of myocardial infarction in nonsmoking women remains low: increased from 0.83 to 3.53 per 10,000,000 women younger than 35 years and from 9.45 to 40.4 per 10,000,000 women 35 years and older.17

Breast Cancer and Cervical Cancer
Data are mixed on the effect of COCs on the risk for breast cancer and cervical cancer.1 According to the CDC, COC use for 5 or more years might increase the risk of cervical carcinoma in situ and invasive cervical carcinoma in women with persistent human papillomavirus infection.5 Regardless of COC use, women should undergo age-appropriate screening for breast cancer and cervical cancer.



Melasma
Melasma is an estrogen-mediated side effect of COCs.8 A study from 1967 found that 29% of COC users (N=212) developed melasma; however, they were taking COCs with much higher ethinyl estradiol doses (50–100 μg) than typically used today.18 Nevertheless, as part of an overall skin care regimen, photoprotection should be encouraged with a broad-spectrum, water-resistant sunscreen that has a sun protection factor of at least 30. In addition, sunscreens with iron oxides have been shown to better prevent melasma relapse by protecting against the shorter wavelengths of visible light.19

References
  1. Zaenglein AL, Pathy AL, Schlosser BJ, et al. Guidelines of care for the management of acne vulgaris. J Am Acad Dermatol. 2016;74:945-973.e933.
  2. Landis ET, Levender MM, Davis SA, et al. Isotretinoin and oral contraceptive use in female acne patients varies by physician specialty: analysis of data from the National Ambulatory Medical Care Survey. J Dermatolog Treat. 2012;23:272-277.
  3. Fitzpatrick L, Mauer E, Chen CL. Oral contraceptives for acne treatment: US dermatologists’ knowledge, comfort, and prescribing practices. Cutis. 2017;99:195-201.
  4. Curtis KM, Jatlaoui TC, Tepper NK, et al. U.S. Selected Practice Recommendations for Contraceptive Use, 2016. MMWR Recomm Rep. 2016;65:1-66.
  5. Curtis KM, Tepper NK, Jatlaoui TC, et al. U.S. Medical Eligibility Criteria for Contraceptive Use, 2016. MMWR Recomm Rep. 2016;65:1-103.
  6. Van Vliet HA, Grimes DA, Lopez LM, et al. Triphasic versus monophasic oral contraceptives for contraception. Cochrane Database Syst Rev. 2011:CD003553.
  7. Stewart M, Black K. Choosing a combined oral contraceptive pill. Aust Prescr. 2015;38:6-11.
  8. McKinney K. Understanding the options: a guide to oral contraceptives. https://www.cecentral.com/assets/2097/022%20Oral%20Contraceptives%2010-26-09.pdf. Published November 5, 2009. Accessed June 20, 2019.
  9. Gallo MF, Nanda K, Grimes DA, et al. 20 microg versus >20 microg estrogen combined oral contraceptives for contraception. Cochrane Database Syst Rev. 2013:CD003989.
  10. Terki F, Malhotra U. Medical and Service Delivery Guidelines for Sexual and Reproductive Health Services. London, United Kingdom: International Planned Parenthood Federation; 2004.
  11. Arowojolu AO, Gallo MF, Lopez LM, et al. Combined oral contraceptive pills for treatment of acne. Cochrane Database Syst Rev. 2012:CD004425.
  12. Pastor Z, Holla K, Chmel R. The influence of combined oral contraceptives on female sexual desire: a systematic review. Eur J Contracept Reprod Health Care. 2013;18:27-43.
  13. Gallo MF, Lopez LM, Grimes DA, et al. Combination contraceptives: effects on weight. Cochrane Database Syst Rev. 2014:CD003987.
  14. Birth control pills for acne: tips from Julie Harper at the Summer AAD. Cutis. https://www.mdedge.com/dermatology/article/144550/acne/birth-control-pills-acne-tips-julie-harper-summer-aad. Published August 14, 2017. Accessed June 24, 2019.
  15. Watson HG, Baglin TP. Guidelines on travel-related venous thrombosis. Br J Haematol. 2011;152:31-34.
  16. Roach RE, Helmerhorst FM, Lijfering WM, et al. Combined oral contraceptives: the risk of myocardial infarction and ischemic stroke. Cochrane Database Syst Rev. 2015:CD011054.
  17. Acute myocardial infarction and combined oral contraceptives: results of an international multicentre case-control study. WHO Collaborative Study of Cardiovascular Disease and Steroid Hormone Contraception. Lancet. 1997;349:1202-1209.
  18. Resnik S. Melasma induced by oral contraceptive drugs. JAMA. 1967;199:601-605.
  19. Boukari F, Jourdan E, Fontas E, et al. Prevention of melasma relapses with sunscreen combining protection against UV and short wavelengths of visible light: a prospective randomized comparative trial. J Am Acad Dermatol. 2015;72:189-190.e181.
References
  1. Zaenglein AL, Pathy AL, Schlosser BJ, et al. Guidelines of care for the management of acne vulgaris. J Am Acad Dermatol. 2016;74:945-973.e933.
  2. Landis ET, Levender MM, Davis SA, et al. Isotretinoin and oral contraceptive use in female acne patients varies by physician specialty: analysis of data from the National Ambulatory Medical Care Survey. J Dermatolog Treat. 2012;23:272-277.
  3. Fitzpatrick L, Mauer E, Chen CL. Oral contraceptives for acne treatment: US dermatologists’ knowledge, comfort, and prescribing practices. Cutis. 2017;99:195-201.
  4. Curtis KM, Jatlaoui TC, Tepper NK, et al. U.S. Selected Practice Recommendations for Contraceptive Use, 2016. MMWR Recomm Rep. 2016;65:1-66.
  5. Curtis KM, Tepper NK, Jatlaoui TC, et al. U.S. Medical Eligibility Criteria for Contraceptive Use, 2016. MMWR Recomm Rep. 2016;65:1-103.
  6. Van Vliet HA, Grimes DA, Lopez LM, et al. Triphasic versus monophasic oral contraceptives for contraception. Cochrane Database Syst Rev. 2011:CD003553.
  7. Stewart M, Black K. Choosing a combined oral contraceptive pill. Aust Prescr. 2015;38:6-11.
  8. McKinney K. Understanding the options: a guide to oral contraceptives. https://www.cecentral.com/assets/2097/022%20Oral%20Contraceptives%2010-26-09.pdf. Published November 5, 2009. Accessed June 20, 2019.
  9. Gallo MF, Nanda K, Grimes DA, et al. 20 microg versus >20 microg estrogen combined oral contraceptives for contraception. Cochrane Database Syst Rev. 2013:CD003989.
  10. Terki F, Malhotra U. Medical and Service Delivery Guidelines for Sexual and Reproductive Health Services. London, United Kingdom: International Planned Parenthood Federation; 2004.
  11. Arowojolu AO, Gallo MF, Lopez LM, et al. Combined oral contraceptive pills for treatment of acne. Cochrane Database Syst Rev. 2012:CD004425.
  12. Pastor Z, Holla K, Chmel R. The influence of combined oral contraceptives on female sexual desire: a systematic review. Eur J Contracept Reprod Health Care. 2013;18:27-43.
  13. Gallo MF, Lopez LM, Grimes DA, et al. Combination contraceptives: effects on weight. Cochrane Database Syst Rev. 2014:CD003987.
  14. Birth control pills for acne: tips from Julie Harper at the Summer AAD. Cutis. https://www.mdedge.com/dermatology/article/144550/acne/birth-control-pills-acne-tips-julie-harper-summer-aad. Published August 14, 2017. Accessed June 24, 2019.
  15. Watson HG, Baglin TP. Guidelines on travel-related venous thrombosis. Br J Haematol. 2011;152:31-34.
  16. Roach RE, Helmerhorst FM, Lijfering WM, et al. Combined oral contraceptives: the risk of myocardial infarction and ischemic stroke. Cochrane Database Syst Rev. 2015:CD011054.
  17. Acute myocardial infarction and combined oral contraceptives: results of an international multicentre case-control study. WHO Collaborative Study of Cardiovascular Disease and Steroid Hormone Contraception. Lancet. 1997;349:1202-1209.
  18. Resnik S. Melasma induced by oral contraceptive drugs. JAMA. 1967;199:601-605.
  19. Boukari F, Jourdan E, Fontas E, et al. Prevention of melasma relapses with sunscreen combining protection against UV and short wavelengths of visible light: a prospective randomized comparative trial. J Am Acad Dermatol. 2015;72:189-190.e181.
Issue
Cutis - 104(1)
Issue
Cutis - 104(1)
Page Number
E10-E14
Page Number
E10-E14
Publications
Publications
Topics
Article Type
Display Headline
The ABCs of COCs: A Guide for Dermatology Residents on Combined Oral Contraceptives
Display Headline
The ABCs of COCs: A Guide for Dermatology Residents on Combined Oral Contraceptives
Sections
Inside the Article

Resident Pearls

  • Screen for contraindications to combined oral contraceptives (COCs) by taking a medical history, measuring a baseline blood pressure, and excluding pregnancy. A baseline pelvic examination is unnecessary.
  • Characteristics to consider when selecting a COC include the formulation, hormone-free interval, estrogen dose, and progestin type.
  • Combined oral contraceptives can be initiated on any day of the menstrual cycle, with the need for backup contraception based on the number of days since the first day of the patient’s most recent period.
  • Management of risks and side effects includes simple lifestyle changes, skipping the hormone-free interval, switching the COC, and referring to an obstetrician/gynecologist.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Article PDF Media

Supercharge Your On-Call Bag: 4 Must-Have Items for Dermatology Residents

Article Type
Changed
Tue, 01/07/2020 - 09:13
Display Headline
Supercharge Your On-Call Bag: 4 Must-Have Items for Dermatology Residents

It is no secret that a well-stocked on-call bag is one of the keys to providing inpatient care as a dermatology resident. Beyond the basic items that should never be left at home, there are some lesser-known tools that I have learned about from my book- and street-smart attendings and co-residents in the Department of Dermatology at the State University of New York Downstate Medical Center (referred to here as Downstate). Here are our top 4 items to pack the next time you are on call. (Bonus: you will find them helpful in clinic, too.)

Item 1: WoundSeal Powder

The most valuable player in my on-call bag, WoundSeal Powder (Biolife) is an over-the-counter hemostatic agent that I learned about from Daniel M. Siegel, MD, MS, a Mohs surgeon at Downstate and former president of the American Academy of Dermatology. The powder consists of a hydrophilic polymer and potassium ferrate.1 When poured over a bleeding wound and pressed in place (eg, with a sterile cotton-tipped swab), the hydrophilic polymer absorbs plasma while the iron in potassium ferrate agglomerates blood solids. The result is a scablike seal that is safe to leave in place until the wound has healed.1

Since Dr. Siegel introduced WoundSeal to Downstate about a decade ago, it has become our department’s go-to hemostatic agent for most punch biopsies performed in the inpatient setting. In our experience, achieving hemostasis in the hospital usually is easier, safer, and faster with WoundSeal than suture. Furthermore, using WoundSeal eliminates the need for patients to follow up for suture removal. From a practical perspective, WoundSeal works best when the biopsy defect is positioned parallel to the ground so the powder can be poured directly over and into the defect. From a cosmetic perspective, we have found that WoundSeal and suture have similar outcomes when used for punch biopsies up to 4 mm in size on the trunk and extremities in both adult and pediatric patients. Working with other dermatology attendings such as Sharon A. Glick, MD; Eve Lowenstein, MD, PhD; and Jeannette Jakus, MD, MBA, I also have found WoundSeal helpful when taking care of suture-phobic children or patients with lesions that are less amenable to suture, such as an ulcer or indurated plaque.

Item 2: Purple Surgical Marker

Another tip I have learned from Drs. Siegel and Jakus: If you are ever in a bind for a topical antibacterial or antifungal agent, look no further than a sterile purple surgical marker. These markers are a surprising source of gentian violet, the same purple dye that is the basis of Gram staining and sold as an over-the-counter antiseptic in 1% to 2% concentrations. Purple surgical markers, on the other hand, are 2.5% to 10% gentian violet.2

 

 

Gentian violet has been shown to have antibacterial, antifungal, antiviral, antihelminthic, and antitrypanosomal properties, but its efficacy has been mostly demonstrated against Streptococcus, methicillin-sensitive and methicillin-resistant Staphylococcus aureus, and Candida.3 Given the dermatologic relevance of these organisms, gentian violet is a favorite among attendings at my residency program; it is not uncommon to remove a patient’s dressing and uncover an iatrogenically purple wound. Best of all, pediatric patients are invariably amused when they see someone drawing on their skin with a purple marker.



When using a sterile surgical marker to apply gentian violet to the skin, we use either the marker tip or the ink core, which Dr. Siegel taught me can be easily accessed by snapping most plastic markers in half.

Item 3: Handheld Blacklight

The Wood lamp is a useful tool in the diagnosis of various infectious diseases and pigmentary disorders,4 but it is not always practical to use when on call, as standard ones are relatively large and corded, so they must be plugged into an electric outlet to work. You can therefore imagine the gratitude I have for my co-residents Miriam Lieberman, MD; Jaime Alexander, MD; Nicole Weiler, MD; and Alessandra Haskin, MD, for introducing me to the most convenient Wood lamp: the handheld blacklight. For less than $10, this gadget combines the diagnostic power of UV light with the portability of a pocket-sized, battery-powered flashlight. You will never want to use another Wood lamp again.

Item 4: Normal Saline Flush

Normal saline can be used for more than storing specimens for frozen section or tissue culture; it also can substitute for Michel solution when storing specimens for direct immunofluorescence (DIF) studies. I learned this tip from Edward Heilman, MD, a dermatopathologist at Downstate. For the last 20 years, Dr. Heilman has been successfully storing DIF specimens in refrigerated normal saline for up to 24 hours when Michel solution is unavailable, after which the specimen is processed or transferred to Michel solution for further storage while being transported to an immunofluorescence laboratory.

In 2004, Vodegel et al5 formally studied this technique in 25 patients with autoimmune skin diseases such as pemphigus and pemphigoid. (Thanks to Dr. Lieberman for telling me about this study.) The experiment involved taking 4 punch biopsies from each patient and placing them in either normal saline at −80°C for 24 or 48 hours, room temperature Michel solution for 48 hours, or liquid nitrogen for up to 2 weeks before being processed for DIF and analyzed by a blinded interpreter. Interestingly, specimens stored in normal saline for 24 hours were the most diagnostic, with a conclusive diagnosis reached in 21 of 25 specimens (84%). This result was attributed to the statistically significant reduction (P<.01) in background fluorescence with normal saline compared to Michel solution and liquid nitrogen, which in turn allowed for easier detection of diagnostic immunoreactants. Similar to Dr. Heilman, the authors cautioned against placing DIF specimens in normal saline for more than 24 hours; in their experience, the risk for an artefactual split developing at the dermoepidermal junction increases with this practice.5

References
  1. Biolife. How WoundSeal works. WoundSeal website. http://woundseal.com/how-it-works. Accessed March 7, 2019.
  2. Viscot Medical, LLC. Safety data sheet. http://www.viscot.com/download/MSDS%20Gentian%20Violet%20Ink.pdf. Published September 11, 2014. Accessed March 7, 2019.
  3. Maley AM, Arbiser JL. Gentian violet: a 19th century drug re-emerges in the 21st century. Exp Dermatol. 2013;22:775-780.
  4. Klatte JL, van der Beek N, Kemperman PM. 100 years of Wood’s lamp revised. J Eur Acad Dermatol Venereol. 2015;29:842-847.
  5. Vodegel RM, de Jong MC, Meijer HJ, et al. Enhanced diagnostic immunofluorescence using biopsies transported in saline. BMC Dermatol. 2004;4:10.
Article PDF
Author and Disclosure Information

From the Department of Dermatology, State University of New York Downstate Medical Center, Brooklyn.

The author reports no conflict of interest.

Correspondence: Daniel R. Mazori, MD, Department of Dermatology, State University of New York Downstate Medical Center, 450 Clarkson Ave, Box 46, Brooklyn, NY 11203 (daniel.mazori@downstate.edu).

Issue
Cutis - 103(3)
Publications
Topics
Page Number
E6-E7
Sections
Author and Disclosure Information

From the Department of Dermatology, State University of New York Downstate Medical Center, Brooklyn.

The author reports no conflict of interest.

Correspondence: Daniel R. Mazori, MD, Department of Dermatology, State University of New York Downstate Medical Center, 450 Clarkson Ave, Box 46, Brooklyn, NY 11203 (daniel.mazori@downstate.edu).

Author and Disclosure Information

From the Department of Dermatology, State University of New York Downstate Medical Center, Brooklyn.

The author reports no conflict of interest.

Correspondence: Daniel R. Mazori, MD, Department of Dermatology, State University of New York Downstate Medical Center, 450 Clarkson Ave, Box 46, Brooklyn, NY 11203 (daniel.mazori@downstate.edu).

Article PDF
Article PDF

It is no secret that a well-stocked on-call bag is one of the keys to providing inpatient care as a dermatology resident. Beyond the basic items that should never be left at home, there are some lesser-known tools that I have learned about from my book- and street-smart attendings and co-residents in the Department of Dermatology at the State University of New York Downstate Medical Center (referred to here as Downstate). Here are our top 4 items to pack the next time you are on call. (Bonus: you will find them helpful in clinic, too.)

Item 1: WoundSeal Powder

The most valuable player in my on-call bag, WoundSeal Powder (Biolife) is an over-the-counter hemostatic agent that I learned about from Daniel M. Siegel, MD, MS, a Mohs surgeon at Downstate and former president of the American Academy of Dermatology. The powder consists of a hydrophilic polymer and potassium ferrate.1 When poured over a bleeding wound and pressed in place (eg, with a sterile cotton-tipped swab), the hydrophilic polymer absorbs plasma while the iron in potassium ferrate agglomerates blood solids. The result is a scablike seal that is safe to leave in place until the wound has healed.1

Since Dr. Siegel introduced WoundSeal to Downstate about a decade ago, it has become our department’s go-to hemostatic agent for most punch biopsies performed in the inpatient setting. In our experience, achieving hemostasis in the hospital usually is easier, safer, and faster with WoundSeal than suture. Furthermore, using WoundSeal eliminates the need for patients to follow up for suture removal. From a practical perspective, WoundSeal works best when the biopsy defect is positioned parallel to the ground so the powder can be poured directly over and into the defect. From a cosmetic perspective, we have found that WoundSeal and suture have similar outcomes when used for punch biopsies up to 4 mm in size on the trunk and extremities in both adult and pediatric patients. Working with other dermatology attendings such as Sharon A. Glick, MD; Eve Lowenstein, MD, PhD; and Jeannette Jakus, MD, MBA, I also have found WoundSeal helpful when taking care of suture-phobic children or patients with lesions that are less amenable to suture, such as an ulcer or indurated plaque.

Item 2: Purple Surgical Marker

Another tip I have learned from Drs. Siegel and Jakus: If you are ever in a bind for a topical antibacterial or antifungal agent, look no further than a sterile purple surgical marker. These markers are a surprising source of gentian violet, the same purple dye that is the basis of Gram staining and sold as an over-the-counter antiseptic in 1% to 2% concentrations. Purple surgical markers, on the other hand, are 2.5% to 10% gentian violet.2

 

 

Gentian violet has been shown to have antibacterial, antifungal, antiviral, antihelminthic, and antitrypanosomal properties, but its efficacy has been mostly demonstrated against Streptococcus, methicillin-sensitive and methicillin-resistant Staphylococcus aureus, and Candida.3 Given the dermatologic relevance of these organisms, gentian violet is a favorite among attendings at my residency program; it is not uncommon to remove a patient’s dressing and uncover an iatrogenically purple wound. Best of all, pediatric patients are invariably amused when they see someone drawing on their skin with a purple marker.



When using a sterile surgical marker to apply gentian violet to the skin, we use either the marker tip or the ink core, which Dr. Siegel taught me can be easily accessed by snapping most plastic markers in half.

Item 3: Handheld Blacklight

The Wood lamp is a useful tool in the diagnosis of various infectious diseases and pigmentary disorders,4 but it is not always practical to use when on call, as standard ones are relatively large and corded, so they must be plugged into an electric outlet to work. You can therefore imagine the gratitude I have for my co-residents Miriam Lieberman, MD; Jaime Alexander, MD; Nicole Weiler, MD; and Alessandra Haskin, MD, for introducing me to the most convenient Wood lamp: the handheld blacklight. For less than $10, this gadget combines the diagnostic power of UV light with the portability of a pocket-sized, battery-powered flashlight. You will never want to use another Wood lamp again.

Item 4: Normal Saline Flush

Normal saline can be used for more than storing specimens for frozen section or tissue culture; it also can substitute for Michel solution when storing specimens for direct immunofluorescence (DIF) studies. I learned this tip from Edward Heilman, MD, a dermatopathologist at Downstate. For the last 20 years, Dr. Heilman has been successfully storing DIF specimens in refrigerated normal saline for up to 24 hours when Michel solution is unavailable, after which the specimen is processed or transferred to Michel solution for further storage while being transported to an immunofluorescence laboratory.

In 2004, Vodegel et al5 formally studied this technique in 25 patients with autoimmune skin diseases such as pemphigus and pemphigoid. (Thanks to Dr. Lieberman for telling me about this study.) The experiment involved taking 4 punch biopsies from each patient and placing them in either normal saline at −80°C for 24 or 48 hours, room temperature Michel solution for 48 hours, or liquid nitrogen for up to 2 weeks before being processed for DIF and analyzed by a blinded interpreter. Interestingly, specimens stored in normal saline for 24 hours were the most diagnostic, with a conclusive diagnosis reached in 21 of 25 specimens (84%). This result was attributed to the statistically significant reduction (P<.01) in background fluorescence with normal saline compared to Michel solution and liquid nitrogen, which in turn allowed for easier detection of diagnostic immunoreactants. Similar to Dr. Heilman, the authors cautioned against placing DIF specimens in normal saline for more than 24 hours; in their experience, the risk for an artefactual split developing at the dermoepidermal junction increases with this practice.5

It is no secret that a well-stocked on-call bag is one of the keys to providing inpatient care as a dermatology resident. Beyond the basic items that should never be left at home, there are some lesser-known tools that I have learned about from my book- and street-smart attendings and co-residents in the Department of Dermatology at the State University of New York Downstate Medical Center (referred to here as Downstate). Here are our top 4 items to pack the next time you are on call. (Bonus: you will find them helpful in clinic, too.)

Item 1: WoundSeal Powder

The most valuable player in my on-call bag, WoundSeal Powder (Biolife) is an over-the-counter hemostatic agent that I learned about from Daniel M. Siegel, MD, MS, a Mohs surgeon at Downstate and former president of the American Academy of Dermatology. The powder consists of a hydrophilic polymer and potassium ferrate.1 When poured over a bleeding wound and pressed in place (eg, with a sterile cotton-tipped swab), the hydrophilic polymer absorbs plasma while the iron in potassium ferrate agglomerates blood solids. The result is a scablike seal that is safe to leave in place until the wound has healed.1

Since Dr. Siegel introduced WoundSeal to Downstate about a decade ago, it has become our department’s go-to hemostatic agent for most punch biopsies performed in the inpatient setting. In our experience, achieving hemostasis in the hospital usually is easier, safer, and faster with WoundSeal than suture. Furthermore, using WoundSeal eliminates the need for patients to follow up for suture removal. From a practical perspective, WoundSeal works best when the biopsy defect is positioned parallel to the ground so the powder can be poured directly over and into the defect. From a cosmetic perspective, we have found that WoundSeal and suture have similar outcomes when used for punch biopsies up to 4 mm in size on the trunk and extremities in both adult and pediatric patients. Working with other dermatology attendings such as Sharon A. Glick, MD; Eve Lowenstein, MD, PhD; and Jeannette Jakus, MD, MBA, I also have found WoundSeal helpful when taking care of suture-phobic children or patients with lesions that are less amenable to suture, such as an ulcer or indurated plaque.

Item 2: Purple Surgical Marker

Another tip I have learned from Drs. Siegel and Jakus: If you are ever in a bind for a topical antibacterial or antifungal agent, look no further than a sterile purple surgical marker. These markers are a surprising source of gentian violet, the same purple dye that is the basis of Gram staining and sold as an over-the-counter antiseptic in 1% to 2% concentrations. Purple surgical markers, on the other hand, are 2.5% to 10% gentian violet.2

 

 

Gentian violet has been shown to have antibacterial, antifungal, antiviral, antihelminthic, and antitrypanosomal properties, but its efficacy has been mostly demonstrated against Streptococcus, methicillin-sensitive and methicillin-resistant Staphylococcus aureus, and Candida.3 Given the dermatologic relevance of these organisms, gentian violet is a favorite among attendings at my residency program; it is not uncommon to remove a patient’s dressing and uncover an iatrogenically purple wound. Best of all, pediatric patients are invariably amused when they see someone drawing on their skin with a purple marker.



When using a sterile surgical marker to apply gentian violet to the skin, we use either the marker tip or the ink core, which Dr. Siegel taught me can be easily accessed by snapping most plastic markers in half.

Item 3: Handheld Blacklight

The Wood lamp is a useful tool in the diagnosis of various infectious diseases and pigmentary disorders,4 but it is not always practical to use when on call, as standard ones are relatively large and corded, so they must be plugged into an electric outlet to work. You can therefore imagine the gratitude I have for my co-residents Miriam Lieberman, MD; Jaime Alexander, MD; Nicole Weiler, MD; and Alessandra Haskin, MD, for introducing me to the most convenient Wood lamp: the handheld blacklight. For less than $10, this gadget combines the diagnostic power of UV light with the portability of a pocket-sized, battery-powered flashlight. You will never want to use another Wood lamp again.

Item 4: Normal Saline Flush

Normal saline can be used for more than storing specimens for frozen section or tissue culture; it also can substitute for Michel solution when storing specimens for direct immunofluorescence (DIF) studies. I learned this tip from Edward Heilman, MD, a dermatopathologist at Downstate. For the last 20 years, Dr. Heilman has been successfully storing DIF specimens in refrigerated normal saline for up to 24 hours when Michel solution is unavailable, after which the specimen is processed or transferred to Michel solution for further storage while being transported to an immunofluorescence laboratory.

In 2004, Vodegel et al5 formally studied this technique in 25 patients with autoimmune skin diseases such as pemphigus and pemphigoid. (Thanks to Dr. Lieberman for telling me about this study.) The experiment involved taking 4 punch biopsies from each patient and placing them in either normal saline at −80°C for 24 or 48 hours, room temperature Michel solution for 48 hours, or liquid nitrogen for up to 2 weeks before being processed for DIF and analyzed by a blinded interpreter. Interestingly, specimens stored in normal saline for 24 hours were the most diagnostic, with a conclusive diagnosis reached in 21 of 25 specimens (84%). This result was attributed to the statistically significant reduction (P<.01) in background fluorescence with normal saline compared to Michel solution and liquid nitrogen, which in turn allowed for easier detection of diagnostic immunoreactants. Similar to Dr. Heilman, the authors cautioned against placing DIF specimens in normal saline for more than 24 hours; in their experience, the risk for an artefactual split developing at the dermoepidermal junction increases with this practice.5

References
  1. Biolife. How WoundSeal works. WoundSeal website. http://woundseal.com/how-it-works. Accessed March 7, 2019.
  2. Viscot Medical, LLC. Safety data sheet. http://www.viscot.com/download/MSDS%20Gentian%20Violet%20Ink.pdf. Published September 11, 2014. Accessed March 7, 2019.
  3. Maley AM, Arbiser JL. Gentian violet: a 19th century drug re-emerges in the 21st century. Exp Dermatol. 2013;22:775-780.
  4. Klatte JL, van der Beek N, Kemperman PM. 100 years of Wood’s lamp revised. J Eur Acad Dermatol Venereol. 2015;29:842-847.
  5. Vodegel RM, de Jong MC, Meijer HJ, et al. Enhanced diagnostic immunofluorescence using biopsies transported in saline. BMC Dermatol. 2004;4:10.
References
  1. Biolife. How WoundSeal works. WoundSeal website. http://woundseal.com/how-it-works. Accessed March 7, 2019.
  2. Viscot Medical, LLC. Safety data sheet. http://www.viscot.com/download/MSDS%20Gentian%20Violet%20Ink.pdf. Published September 11, 2014. Accessed March 7, 2019.
  3. Maley AM, Arbiser JL. Gentian violet: a 19th century drug re-emerges in the 21st century. Exp Dermatol. 2013;22:775-780.
  4. Klatte JL, van der Beek N, Kemperman PM. 100 years of Wood’s lamp revised. J Eur Acad Dermatol Venereol. 2015;29:842-847.
  5. Vodegel RM, de Jong MC, Meijer HJ, et al. Enhanced diagnostic immunofluorescence using biopsies transported in saline. BMC Dermatol. 2004;4:10.
Issue
Cutis - 103(3)
Issue
Cutis - 103(3)
Page Number
E6-E7
Page Number
E6-E7
Publications
Publications
Topics
Article Type
Display Headline
Supercharge Your On-Call Bag: 4 Must-Have Items for Dermatology Residents
Display Headline
Supercharge Your On-Call Bag: 4 Must-Have Items for Dermatology Residents
Sections
Inside the Article

Resident Pearl

  • The following unconventional items will come in handy the next time you are on call (or in clinic) and need an alternative to a suture, topical antimicrobial, Wood lamp, or Michel solution.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Article PDF Media

Delayed Cutaneous Reactions to Iodinated Contrast

Article Type
Changed
Thu, 01/10/2019 - 13:51
Display Headline
Delayed Cutaneous Reactions to Iodinated Contrast

Case Report

A 67-year-old woman with a history of allergic rhinitis presented in the spring with a pruritic eruption of 2 days’ duration that began on the abdomen and spread to the chest, back, and bilateral arms. Six days prior to the onset of the symptoms she underwent computed tomography (CT) of the abdomen and pelvis to evaluate abdominal pain and peripheral eosinophilia. Two iodinated contrast (IC) agents were used: intravenous iohexol and oral diatrizoate meglumine–diatrizoate sodium. The eruption was not preceded by fever, malaise, sore throat, rhinorrhea, cough, arthralgia, headache, diarrhea, or new medication or supplement use. The patient denied any history of drug allergy or cutaneous eruptions. Her current medications, which she had been taking long-term, were aspirin, lisinopril, diltiazem, levothyroxine, esomeprazole, paroxetine, gabapentin, and diphenhydramine.

Physical examination was notable for erythematous, blanchable, nontender macules coalescing into patches on the trunk and bilateral arms (Figure). There was slight erythema on the nasolabial folds and ears. The mucosal surfaces and distal legs were clear. The patient was afebrile. Her white blood cell count was 12.5×109/L with 32.3% eosinophils (baseline: white blood cell count, 14.8×109/L; 22% eosinophils)(reference range, 4.8–10.8×109/L; 1%–4% eosinophils). Her comprehensive metabolic panel was within reference range. The human immunodeficiency virus 1/2 antibody immunoassay was nonreactive.

Figure1
Erythematous, blanchable, nontender macules coalescing into patches on the abdomen (A) and left arm (B).

The patient was diagnosed with an exanthematous eruption caused by IC and was treated with oral hydroxyzine and triamcinolone acetonide cream 0.1%. The eruption resolved within 2 weeks without recurrence at 3-month follow-up.

Comment

Delayed cutaneous eruptions caused by IC are underrecognized in medicine and are infrequently described in the dermatology literature.1 Unlike urticaria and other well-known immediate reactions to IC, delayed reactions develop when patients are less likely to be under medical supervision.2 Moreover, only 12% to 33% of patients with delayed reactions to IC seek medical attention.3-6 As a result, these delayed reactions often are attributed to other causes.1 Patients may then be unknowingly reexposed to the offending contrast agent and experience recurrent eruptions, such as in one fatal case of toxic epidermal necrolysis (TEN).7-11 Given the role of dermatologists in the diagnosis and prevention of cutaneous drug reactions, it is important to be mindful of delayed cutaneous eruptions caused by IC.

Clinical Presentation of Delayed Reactions
Most delayed cutaneous reactions to IC present as exanthematous eruptions in the week following a contrast-enhanced CT scan or coronary angiogram.2,12 The reactions tend to resolve within 2 weeks of onset, and the treatment is largely supportive with antihistamines and/or corticosteroids for the associated pruritus.2,5,6 In a study of 98 patients with a history of delayed reactions to IC, delayed-onset urticaria and angioedema also have been reported with incidence rates of 19% and 24%, respectively.2 Other reactions are less common. In the same study, 7% of patients developed palpable purpura; acute generalized exanthematous pustulosis; bullous, flexural, or psoriasislike exanthema; exfoliative eruptions; or purpura and a maculopapular eruption combined with eosinophilia.2 There also have been reports of IC-induced erythema multiforme,3 fixed drug eruptions,10,11 symmetrical drug-related intertriginous and flexural exanthema,13 cutaneous vasculitis,14 drug reactions with eosinophilia and systemic symptoms,15 Stevens-Johnson syndrome/TEN,7,8,16,17 and iododerma.18

IC Agents
Virtually all delayed cutaneous reactions to IC reportedly are due to intravascular rather than oral agents,1,2,19 with the exception of iododerma18 and 1 reported case of TEN.17 Intravenous iohexol was most likely the offending drug in our case. In a prospective cohort study of 539 patients undergoing CT scans, the absolute risk for developing a delayed cutaneous reaction (defined as rash, itching, or skin redness or swelling) to intravascular iohexol was 9.4%.20 Randomized, double-blind studies have found that the risk for delayed cutaneous eruptions is similar among various types of IC, except for iodixanol, which confers a higher risk.5,6,21

Risk Factors
Interestingly, analyses have shown that delayed reactions to IC are more common in atopic patients and during high pollen season.22 Our patient displayed these risk factors, as she had allergic rhinitis and presented for evaluation in late spring when local pollen counts were high. Additionally, patients who develop delayed reactions to IC are notably more likely than controls to have a history of other cutaneous drug reactions, serum creatinine levels greater than 2.0 mg/dL (reference range, 0.6–1.2 mg/dL),3 or history of treatment with recombinant interleukin 2.19

Patients with a history of delayed reactions to IC are not at increased risk for immediate reactions and vice versa.2,3 This finding is consistent with the evidence that delayed and immediate reactions to IC are mechanistically unrelated.23 Additionally, seafood allergy is not a risk factor for either immediate or delayed reactions to IC, despite a common misconception among physicians and patients because seafood is iodinated.24,25

Reexposure to IC
Patients who have had delayed cutaneous reactions to IC are at risk for similar eruptions upon reexposure. Although the reactions are believed to be cell mediated, skin testing with IC is not sensitive enough to reliably identify tolerable alternatives.12 Consequently, gadolinium-based agents have been recommended in patients with a history of reactions to IC if additional contrast-enhanced studies are needed.13,26 Iodinated and gadolinium-based contrast agents do not cross-react, and gadolinium is compatible with studies other than magnetic resonance imaging.1,27

Premedication
Despite the absence of cross-reactivity, the American College of Radiology considers patients with hypersensitivity reactions to IC to be at increased risk for reactions to gadolinium but does not make specific recommendations regarding premedication given the dearth of data.1 As a result, premedication may be considered prior to gadolinium administration depending on the severity of the delayed reaction to IC. Additionally, premedication may be beneficial in cases in which gadolinium is contraindicated and IC must be reused. In a retrospective study, all patients with suspected delayed reactions to IC tolerated IC or gadolinium contrast when pretreated with corticosteroids with or without antihistamines.28 Regimens with corticosteroids and either cyclosporine or intravenous immunoglobulin also have prevented the recurrence of IC-induced exanthematous eruptions and Stevens-Johnson syndrome.29,30 Despite these reports, delayed cutaneous reactions to IC have recurred in other patients receiving corticosteroids, antihistamines, and/or cyclosporine for premedication or concurrent treatment of an underlying condition.16,29-31

Conclusion

It is important for dermatologists to recognize IC as a cause of delayed drug reactions. Current awareness is limited, and as a result, patients often are reexposed to the offending contrast agents unsuspectingly. In addition to diagnosing these eruptions, dermatologists may help prevent their recurrence if future contrast-enhanced studies are required by recommending gadolinium-based agents and/or premedication.

References
  1. Cohan RH, Davenport MS, Dillman JR, et al; ACR Committee on Drugs and Contrast Media. ACR Manual on Contrast Media. 9th ed. Reston, VA: American College of Radiology; 2013.
  2. Brockow K, Romano A, Aberer W, et al; European Network of Drug Allergy and the EAACI Interest Group on Drug Hypersensitivity. Skin testing in patients with hypersensitivity reactions to iodinated contrast media—a European multicenter study. Allergy. 2009;64:234-241.
  3. Hosoya T, Yamaguchi K, Akutsu T, et al. Delayed adverse reactions to iodinated contrast media and their risk factors. Radiat Med. 2000;18:39-45.
  4. Rydberg J, Charles J, Aspelin P. Frequency of late allergy-like adverse reactions following injection of intravascular non-ionic contrast media: a retrospective study comparing a non-ionic monomeric contrast medium with a non-ionic dimeric contrast medium. Acta Radiol. 1998;39:219-222.
  5. Sutton AG, Finn P, Grech ED, et al. Early and late reactions after the use of iopamidol 340, ioxaglate 320, and iodixanol 320 in cardiac catheterization. Am Heart J. 2001;141:677-683.
  6. Sutton AG, Finn P, Campbell PG, et al. Early and late reactions following the use of iopamidol 340, iomeprol 350 and iodixanol 320 in cardiac catheterization. J Invasive Cardiol. 2003;15:133-138.
  7. Brown M, Yowler C, Brandt C. Recurrent toxic epidermal necrolysis secondary to iopromide contrast. J Burn Care Res. 2013;34:E53-E56.
  8. Rosado A, Canto G, Veleiro B, et al. Toxic epidermal necrolysis after repeated injections of iohexol. AJR Am J Roentgenol. 2001;176:262-263.
  9. Peterson A, Katzberg RW, Fung MA, et al. Acute generalized exanthematous pustulosis as a delayed dermatotoxic reaction to IV-administered nonionic contrast media. AJR Am J Roentgenol. 2006;187:W198-W201.
  10. Good AE, Novak E, Sonda LP III. Fixed eruption and fever after urography. South Med J. 1980;73:948-949.
  11. Benson PM, Giblin WJ, Douglas DM. Transient, nonpigmenting fixed drug eruption caused by radiopaque contrast media. J Am Acad Dermatol. 1990;23(2, pt 2):379-381.
  12. Torres MJ, Gomez F, Doña I, et al. Diagnostic evaluation of patients with nonimmediate cutaneous hypersensitivity reactions to iodinated contrast media. Allergy. 2012;67:929-935.
  13. Scherer K, Harr T, Bach S, et al. The role of iodine in hypersensitivity reactions to radio contrast media. Clin Exp Allergy. 2010;40:468-475.
  14. Reynolds NJ, Wallington TB, Burton JL. Hydralazine predisposes to acute cutaneous vasculitis following urography with iopamidol. Br J Dermatol. 1993;129:82-85.
  15. Belhadjali H, Bouzgarrou L, Youssef M, et al. DRESS syndrome induced by sodium meglumine ioxitalamate. Allergy. 2008;63:786-787.
  16. Baldwin BT, Lien MH, Khan H, et al. Case of fatal toxic epidermal necrolysis due to cardiac catheterization dye. J Drugs Dermatol. 2010;9:837-840.
  17. Schmidt BJ, Foley WD, Bohorfoush AG. Toxic epidermal necrolysis related to oral administration of diluted diatrizoate meglumine and diatrizoate sodium. AJR Am J Roentgenol. 1998;171:1215-1216.
  18. Young AL, Grossman ME. Acute iododerma secondary to iodinated contrast media. Br J Dermatol. 2014;170:1377-1379.
  19. Choyke PL, Miller DL, Lotze MT, et al. Delayed reactions to contrast media after interleukin-2 immunotherapy. Radiology. 1992;183:111-114.
  20. Loh S, Bagheri S, Katzberg RW, et al. Delayed adverse reaction to contrast-enhanced CT: a prospective single-center study comparison to control group without enhancement. Radiology. 2010;255:764-771.
  21. Bertrand P, Delhommais A, Alison D, et al. Immediate and delayed tolerance of iohexol and ioxaglate in lower limb phlebography: a double-blind comparative study in humans. Acad Radiol. 1995;2:683-686.
  22. Munechika H, Hiramatsu Y, Kudo S, et al. A prospective survey of delayed adverse reactions to iohexol in urography and computed tomography. Eur Radiol. 2003;13:185-194.
  23. Guéant-Rodriguez RM, Romano A, Barbaud A, et al. Hypersensitivity reactions to iodinated contrast media. Curr Pharm Des. 2006;12:3359-3372.
  24. Huang SW. Seafood and iodine: an analysis of a medical myth. Allergy Asthma Proc. 2005;26:468-469.
  25. Baig M, Farag A, Sajid J, et al. Shellfish allergy and relation to iodinated contrast media: United Kingdom survey. World J Cardiol. 2014;6:107-111.
  26. Böhm I, Schild HH. A practical guide to diagnose lesser-known immediate and delayed contrast media-induced adverse cutaneous reactions. Eur Radiol. 2006;16:1570-1579.
  27. Ose K, Doue T, Zen K, et al. “Gadolinium” as an alternative to iodinated contrast media for X-ray angiography in patients with severe allergy. Circ J. 2005;69:507-509.
  28. Jingu A, Fukuda J, Taketomi-Takahashi A, et al. Breakthrough reactions of iodinated and gadolinium contrast media after oral steroid premedication protocol. BMC Med Imaging. 2014;14:34.
  29. Romano A, Artesani MC, Andriolo M, et al. Effective prophylactic protocol in delayed hypersensitivity to contrast media: report of a case involving lymphocyte transformation studies with different compounds. Radiology. 2002;225:466-470.
  30. Hebert AA, Bogle MA. Intravenous immunoglobulin prophylaxis for recurrent Stevens-Johnson syndrome. J Am Acad Dermatol. 2004;50:286-288.
  31. Hasdenteufel F, Waton J, Cordebar V, et al. Delayed hypersensitivity reactions caused by iodixanol: an assessment of cross-reactivity in 22 patients. J Allergy Clin Immunol. 2011;128:1356-1357.
Article PDF
Author and Disclosure Information

From the Ronald O. Perelman Department of Dermatology, New York University School of Medicine, New York.

The authors report no conflict of interest.

Correspondence: Miriam Keltz Pomeranz, MD, 240 E 38th St, 12th Floor, New York, NY 10016 (miriam.pomeranz@nyumc.org).

Issue
Cutis - 101(6)
Publications
Topics
Page Number
433-435
Sections
Author and Disclosure Information

From the Ronald O. Perelman Department of Dermatology, New York University School of Medicine, New York.

The authors report no conflict of interest.

Correspondence: Miriam Keltz Pomeranz, MD, 240 E 38th St, 12th Floor, New York, NY 10016 (miriam.pomeranz@nyumc.org).

Author and Disclosure Information

From the Ronald O. Perelman Department of Dermatology, New York University School of Medicine, New York.

The authors report no conflict of interest.

Correspondence: Miriam Keltz Pomeranz, MD, 240 E 38th St, 12th Floor, New York, NY 10016 (miriam.pomeranz@nyumc.org).

Article PDF
Article PDF

Case Report

A 67-year-old woman with a history of allergic rhinitis presented in the spring with a pruritic eruption of 2 days’ duration that began on the abdomen and spread to the chest, back, and bilateral arms. Six days prior to the onset of the symptoms she underwent computed tomography (CT) of the abdomen and pelvis to evaluate abdominal pain and peripheral eosinophilia. Two iodinated contrast (IC) agents were used: intravenous iohexol and oral diatrizoate meglumine–diatrizoate sodium. The eruption was not preceded by fever, malaise, sore throat, rhinorrhea, cough, arthralgia, headache, diarrhea, or new medication or supplement use. The patient denied any history of drug allergy or cutaneous eruptions. Her current medications, which she had been taking long-term, were aspirin, lisinopril, diltiazem, levothyroxine, esomeprazole, paroxetine, gabapentin, and diphenhydramine.

Physical examination was notable for erythematous, blanchable, nontender macules coalescing into patches on the trunk and bilateral arms (Figure). There was slight erythema on the nasolabial folds and ears. The mucosal surfaces and distal legs were clear. The patient was afebrile. Her white blood cell count was 12.5×109/L with 32.3% eosinophils (baseline: white blood cell count, 14.8×109/L; 22% eosinophils)(reference range, 4.8–10.8×109/L; 1%–4% eosinophils). Her comprehensive metabolic panel was within reference range. The human immunodeficiency virus 1/2 antibody immunoassay was nonreactive.

Figure1
Erythematous, blanchable, nontender macules coalescing into patches on the abdomen (A) and left arm (B).

The patient was diagnosed with an exanthematous eruption caused by IC and was treated with oral hydroxyzine and triamcinolone acetonide cream 0.1%. The eruption resolved within 2 weeks without recurrence at 3-month follow-up.

Comment

Delayed cutaneous eruptions caused by IC are underrecognized in medicine and are infrequently described in the dermatology literature.1 Unlike urticaria and other well-known immediate reactions to IC, delayed reactions develop when patients are less likely to be under medical supervision.2 Moreover, only 12% to 33% of patients with delayed reactions to IC seek medical attention.3-6 As a result, these delayed reactions often are attributed to other causes.1 Patients may then be unknowingly reexposed to the offending contrast agent and experience recurrent eruptions, such as in one fatal case of toxic epidermal necrolysis (TEN).7-11 Given the role of dermatologists in the diagnosis and prevention of cutaneous drug reactions, it is important to be mindful of delayed cutaneous eruptions caused by IC.

Clinical Presentation of Delayed Reactions
Most delayed cutaneous reactions to IC present as exanthematous eruptions in the week following a contrast-enhanced CT scan or coronary angiogram.2,12 The reactions tend to resolve within 2 weeks of onset, and the treatment is largely supportive with antihistamines and/or corticosteroids for the associated pruritus.2,5,6 In a study of 98 patients with a history of delayed reactions to IC, delayed-onset urticaria and angioedema also have been reported with incidence rates of 19% and 24%, respectively.2 Other reactions are less common. In the same study, 7% of patients developed palpable purpura; acute generalized exanthematous pustulosis; bullous, flexural, or psoriasislike exanthema; exfoliative eruptions; or purpura and a maculopapular eruption combined with eosinophilia.2 There also have been reports of IC-induced erythema multiforme,3 fixed drug eruptions,10,11 symmetrical drug-related intertriginous and flexural exanthema,13 cutaneous vasculitis,14 drug reactions with eosinophilia and systemic symptoms,15 Stevens-Johnson syndrome/TEN,7,8,16,17 and iododerma.18

IC Agents
Virtually all delayed cutaneous reactions to IC reportedly are due to intravascular rather than oral agents,1,2,19 with the exception of iododerma18 and 1 reported case of TEN.17 Intravenous iohexol was most likely the offending drug in our case. In a prospective cohort study of 539 patients undergoing CT scans, the absolute risk for developing a delayed cutaneous reaction (defined as rash, itching, or skin redness or swelling) to intravascular iohexol was 9.4%.20 Randomized, double-blind studies have found that the risk for delayed cutaneous eruptions is similar among various types of IC, except for iodixanol, which confers a higher risk.5,6,21

Risk Factors
Interestingly, analyses have shown that delayed reactions to IC are more common in atopic patients and during high pollen season.22 Our patient displayed these risk factors, as she had allergic rhinitis and presented for evaluation in late spring when local pollen counts were high. Additionally, patients who develop delayed reactions to IC are notably more likely than controls to have a history of other cutaneous drug reactions, serum creatinine levels greater than 2.0 mg/dL (reference range, 0.6–1.2 mg/dL),3 or history of treatment with recombinant interleukin 2.19

Patients with a history of delayed reactions to IC are not at increased risk for immediate reactions and vice versa.2,3 This finding is consistent with the evidence that delayed and immediate reactions to IC are mechanistically unrelated.23 Additionally, seafood allergy is not a risk factor for either immediate or delayed reactions to IC, despite a common misconception among physicians and patients because seafood is iodinated.24,25

Reexposure to IC
Patients who have had delayed cutaneous reactions to IC are at risk for similar eruptions upon reexposure. Although the reactions are believed to be cell mediated, skin testing with IC is not sensitive enough to reliably identify tolerable alternatives.12 Consequently, gadolinium-based agents have been recommended in patients with a history of reactions to IC if additional contrast-enhanced studies are needed.13,26 Iodinated and gadolinium-based contrast agents do not cross-react, and gadolinium is compatible with studies other than magnetic resonance imaging.1,27

Premedication
Despite the absence of cross-reactivity, the American College of Radiology considers patients with hypersensitivity reactions to IC to be at increased risk for reactions to gadolinium but does not make specific recommendations regarding premedication given the dearth of data.1 As a result, premedication may be considered prior to gadolinium administration depending on the severity of the delayed reaction to IC. Additionally, premedication may be beneficial in cases in which gadolinium is contraindicated and IC must be reused. In a retrospective study, all patients with suspected delayed reactions to IC tolerated IC or gadolinium contrast when pretreated with corticosteroids with or without antihistamines.28 Regimens with corticosteroids and either cyclosporine or intravenous immunoglobulin also have prevented the recurrence of IC-induced exanthematous eruptions and Stevens-Johnson syndrome.29,30 Despite these reports, delayed cutaneous reactions to IC have recurred in other patients receiving corticosteroids, antihistamines, and/or cyclosporine for premedication or concurrent treatment of an underlying condition.16,29-31

Conclusion

It is important for dermatologists to recognize IC as a cause of delayed drug reactions. Current awareness is limited, and as a result, patients often are reexposed to the offending contrast agents unsuspectingly. In addition to diagnosing these eruptions, dermatologists may help prevent their recurrence if future contrast-enhanced studies are required by recommending gadolinium-based agents and/or premedication.

Case Report

A 67-year-old woman with a history of allergic rhinitis presented in the spring with a pruritic eruption of 2 days’ duration that began on the abdomen and spread to the chest, back, and bilateral arms. Six days prior to the onset of the symptoms she underwent computed tomography (CT) of the abdomen and pelvis to evaluate abdominal pain and peripheral eosinophilia. Two iodinated contrast (IC) agents were used: intravenous iohexol and oral diatrizoate meglumine–diatrizoate sodium. The eruption was not preceded by fever, malaise, sore throat, rhinorrhea, cough, arthralgia, headache, diarrhea, or new medication or supplement use. The patient denied any history of drug allergy or cutaneous eruptions. Her current medications, which she had been taking long-term, were aspirin, lisinopril, diltiazem, levothyroxine, esomeprazole, paroxetine, gabapentin, and diphenhydramine.

Physical examination was notable for erythematous, blanchable, nontender macules coalescing into patches on the trunk and bilateral arms (Figure). There was slight erythema on the nasolabial folds and ears. The mucosal surfaces and distal legs were clear. The patient was afebrile. Her white blood cell count was 12.5×109/L with 32.3% eosinophils (baseline: white blood cell count, 14.8×109/L; 22% eosinophils)(reference range, 4.8–10.8×109/L; 1%–4% eosinophils). Her comprehensive metabolic panel was within reference range. The human immunodeficiency virus 1/2 antibody immunoassay was nonreactive.

Figure1
Erythematous, blanchable, nontender macules coalescing into patches on the abdomen (A) and left arm (B).

The patient was diagnosed with an exanthematous eruption caused by IC and was treated with oral hydroxyzine and triamcinolone acetonide cream 0.1%. The eruption resolved within 2 weeks without recurrence at 3-month follow-up.

Comment

Delayed cutaneous eruptions caused by IC are underrecognized in medicine and are infrequently described in the dermatology literature.1 Unlike urticaria and other well-known immediate reactions to IC, delayed reactions develop when patients are less likely to be under medical supervision.2 Moreover, only 12% to 33% of patients with delayed reactions to IC seek medical attention.3-6 As a result, these delayed reactions often are attributed to other causes.1 Patients may then be unknowingly reexposed to the offending contrast agent and experience recurrent eruptions, such as in one fatal case of toxic epidermal necrolysis (TEN).7-11 Given the role of dermatologists in the diagnosis and prevention of cutaneous drug reactions, it is important to be mindful of delayed cutaneous eruptions caused by IC.

Clinical Presentation of Delayed Reactions
Most delayed cutaneous reactions to IC present as exanthematous eruptions in the week following a contrast-enhanced CT scan or coronary angiogram.2,12 The reactions tend to resolve within 2 weeks of onset, and the treatment is largely supportive with antihistamines and/or corticosteroids for the associated pruritus.2,5,6 In a study of 98 patients with a history of delayed reactions to IC, delayed-onset urticaria and angioedema also have been reported with incidence rates of 19% and 24%, respectively.2 Other reactions are less common. In the same study, 7% of patients developed palpable purpura; acute generalized exanthematous pustulosis; bullous, flexural, or psoriasislike exanthema; exfoliative eruptions; or purpura and a maculopapular eruption combined with eosinophilia.2 There also have been reports of IC-induced erythema multiforme,3 fixed drug eruptions,10,11 symmetrical drug-related intertriginous and flexural exanthema,13 cutaneous vasculitis,14 drug reactions with eosinophilia and systemic symptoms,15 Stevens-Johnson syndrome/TEN,7,8,16,17 and iododerma.18

IC Agents
Virtually all delayed cutaneous reactions to IC reportedly are due to intravascular rather than oral agents,1,2,19 with the exception of iododerma18 and 1 reported case of TEN.17 Intravenous iohexol was most likely the offending drug in our case. In a prospective cohort study of 539 patients undergoing CT scans, the absolute risk for developing a delayed cutaneous reaction (defined as rash, itching, or skin redness or swelling) to intravascular iohexol was 9.4%.20 Randomized, double-blind studies have found that the risk for delayed cutaneous eruptions is similar among various types of IC, except for iodixanol, which confers a higher risk.5,6,21

Risk Factors
Interestingly, analyses have shown that delayed reactions to IC are more common in atopic patients and during high pollen season.22 Our patient displayed these risk factors, as she had allergic rhinitis and presented for evaluation in late spring when local pollen counts were high. Additionally, patients who develop delayed reactions to IC are notably more likely than controls to have a history of other cutaneous drug reactions, serum creatinine levels greater than 2.0 mg/dL (reference range, 0.6–1.2 mg/dL),3 or history of treatment with recombinant interleukin 2.19

Patients with a history of delayed reactions to IC are not at increased risk for immediate reactions and vice versa.2,3 This finding is consistent with the evidence that delayed and immediate reactions to IC are mechanistically unrelated.23 Additionally, seafood allergy is not a risk factor for either immediate or delayed reactions to IC, despite a common misconception among physicians and patients because seafood is iodinated.24,25

Reexposure to IC
Patients who have had delayed cutaneous reactions to IC are at risk for similar eruptions upon reexposure. Although the reactions are believed to be cell mediated, skin testing with IC is not sensitive enough to reliably identify tolerable alternatives.12 Consequently, gadolinium-based agents have been recommended in patients with a history of reactions to IC if additional contrast-enhanced studies are needed.13,26 Iodinated and gadolinium-based contrast agents do not cross-react, and gadolinium is compatible with studies other than magnetic resonance imaging.1,27

Premedication
Despite the absence of cross-reactivity, the American College of Radiology considers patients with hypersensitivity reactions to IC to be at increased risk for reactions to gadolinium but does not make specific recommendations regarding premedication given the dearth of data.1 As a result, premedication may be considered prior to gadolinium administration depending on the severity of the delayed reaction to IC. Additionally, premedication may be beneficial in cases in which gadolinium is contraindicated and IC must be reused. In a retrospective study, all patients with suspected delayed reactions to IC tolerated IC or gadolinium contrast when pretreated with corticosteroids with or without antihistamines.28 Regimens with corticosteroids and either cyclosporine or intravenous immunoglobulin also have prevented the recurrence of IC-induced exanthematous eruptions and Stevens-Johnson syndrome.29,30 Despite these reports, delayed cutaneous reactions to IC have recurred in other patients receiving corticosteroids, antihistamines, and/or cyclosporine for premedication or concurrent treatment of an underlying condition.16,29-31

Conclusion

It is important for dermatologists to recognize IC as a cause of delayed drug reactions. Current awareness is limited, and as a result, patients often are reexposed to the offending contrast agents unsuspectingly. In addition to diagnosing these eruptions, dermatologists may help prevent their recurrence if future contrast-enhanced studies are required by recommending gadolinium-based agents and/or premedication.

References
  1. Cohan RH, Davenport MS, Dillman JR, et al; ACR Committee on Drugs and Contrast Media. ACR Manual on Contrast Media. 9th ed. Reston, VA: American College of Radiology; 2013.
  2. Brockow K, Romano A, Aberer W, et al; European Network of Drug Allergy and the EAACI Interest Group on Drug Hypersensitivity. Skin testing in patients with hypersensitivity reactions to iodinated contrast media—a European multicenter study. Allergy. 2009;64:234-241.
  3. Hosoya T, Yamaguchi K, Akutsu T, et al. Delayed adverse reactions to iodinated contrast media and their risk factors. Radiat Med. 2000;18:39-45.
  4. Rydberg J, Charles J, Aspelin P. Frequency of late allergy-like adverse reactions following injection of intravascular non-ionic contrast media: a retrospective study comparing a non-ionic monomeric contrast medium with a non-ionic dimeric contrast medium. Acta Radiol. 1998;39:219-222.
  5. Sutton AG, Finn P, Grech ED, et al. Early and late reactions after the use of iopamidol 340, ioxaglate 320, and iodixanol 320 in cardiac catheterization. Am Heart J. 2001;141:677-683.
  6. Sutton AG, Finn P, Campbell PG, et al. Early and late reactions following the use of iopamidol 340, iomeprol 350 and iodixanol 320 in cardiac catheterization. J Invasive Cardiol. 2003;15:133-138.
  7. Brown M, Yowler C, Brandt C. Recurrent toxic epidermal necrolysis secondary to iopromide contrast. J Burn Care Res. 2013;34:E53-E56.
  8. Rosado A, Canto G, Veleiro B, et al. Toxic epidermal necrolysis after repeated injections of iohexol. AJR Am J Roentgenol. 2001;176:262-263.
  9. Peterson A, Katzberg RW, Fung MA, et al. Acute generalized exanthematous pustulosis as a delayed dermatotoxic reaction to IV-administered nonionic contrast media. AJR Am J Roentgenol. 2006;187:W198-W201.
  10. Good AE, Novak E, Sonda LP III. Fixed eruption and fever after urography. South Med J. 1980;73:948-949.
  11. Benson PM, Giblin WJ, Douglas DM. Transient, nonpigmenting fixed drug eruption caused by radiopaque contrast media. J Am Acad Dermatol. 1990;23(2, pt 2):379-381.
  12. Torres MJ, Gomez F, Doña I, et al. Diagnostic evaluation of patients with nonimmediate cutaneous hypersensitivity reactions to iodinated contrast media. Allergy. 2012;67:929-935.
  13. Scherer K, Harr T, Bach S, et al. The role of iodine in hypersensitivity reactions to radio contrast media. Clin Exp Allergy. 2010;40:468-475.
  14. Reynolds NJ, Wallington TB, Burton JL. Hydralazine predisposes to acute cutaneous vasculitis following urography with iopamidol. Br J Dermatol. 1993;129:82-85.
  15. Belhadjali H, Bouzgarrou L, Youssef M, et al. DRESS syndrome induced by sodium meglumine ioxitalamate. Allergy. 2008;63:786-787.
  16. Baldwin BT, Lien MH, Khan H, et al. Case of fatal toxic epidermal necrolysis due to cardiac catheterization dye. J Drugs Dermatol. 2010;9:837-840.
  17. Schmidt BJ, Foley WD, Bohorfoush AG. Toxic epidermal necrolysis related to oral administration of diluted diatrizoate meglumine and diatrizoate sodium. AJR Am J Roentgenol. 1998;171:1215-1216.
  18. Young AL, Grossman ME. Acute iododerma secondary to iodinated contrast media. Br J Dermatol. 2014;170:1377-1379.
  19. Choyke PL, Miller DL, Lotze MT, et al. Delayed reactions to contrast media after interleukin-2 immunotherapy. Radiology. 1992;183:111-114.
  20. Loh S, Bagheri S, Katzberg RW, et al. Delayed adverse reaction to contrast-enhanced CT: a prospective single-center study comparison to control group without enhancement. Radiology. 2010;255:764-771.
  21. Bertrand P, Delhommais A, Alison D, et al. Immediate and delayed tolerance of iohexol and ioxaglate in lower limb phlebography: a double-blind comparative study in humans. Acad Radiol. 1995;2:683-686.
  22. Munechika H, Hiramatsu Y, Kudo S, et al. A prospective survey of delayed adverse reactions to iohexol in urography and computed tomography. Eur Radiol. 2003;13:185-194.
  23. Guéant-Rodriguez RM, Romano A, Barbaud A, et al. Hypersensitivity reactions to iodinated contrast media. Curr Pharm Des. 2006;12:3359-3372.
  24. Huang SW. Seafood and iodine: an analysis of a medical myth. Allergy Asthma Proc. 2005;26:468-469.
  25. Baig M, Farag A, Sajid J, et al. Shellfish allergy and relation to iodinated contrast media: United Kingdom survey. World J Cardiol. 2014;6:107-111.
  26. Böhm I, Schild HH. A practical guide to diagnose lesser-known immediate and delayed contrast media-induced adverse cutaneous reactions. Eur Radiol. 2006;16:1570-1579.
  27. Ose K, Doue T, Zen K, et al. “Gadolinium” as an alternative to iodinated contrast media for X-ray angiography in patients with severe allergy. Circ J. 2005;69:507-509.
  28. Jingu A, Fukuda J, Taketomi-Takahashi A, et al. Breakthrough reactions of iodinated and gadolinium contrast media after oral steroid premedication protocol. BMC Med Imaging. 2014;14:34.
  29. Romano A, Artesani MC, Andriolo M, et al. Effective prophylactic protocol in delayed hypersensitivity to contrast media: report of a case involving lymphocyte transformation studies with different compounds. Radiology. 2002;225:466-470.
  30. Hebert AA, Bogle MA. Intravenous immunoglobulin prophylaxis for recurrent Stevens-Johnson syndrome. J Am Acad Dermatol. 2004;50:286-288.
  31. Hasdenteufel F, Waton J, Cordebar V, et al. Delayed hypersensitivity reactions caused by iodixanol: an assessment of cross-reactivity in 22 patients. J Allergy Clin Immunol. 2011;128:1356-1357.
References
  1. Cohan RH, Davenport MS, Dillman JR, et al; ACR Committee on Drugs and Contrast Media. ACR Manual on Contrast Media. 9th ed. Reston, VA: American College of Radiology; 2013.
  2. Brockow K, Romano A, Aberer W, et al; European Network of Drug Allergy and the EAACI Interest Group on Drug Hypersensitivity. Skin testing in patients with hypersensitivity reactions to iodinated contrast media—a European multicenter study. Allergy. 2009;64:234-241.
  3. Hosoya T, Yamaguchi K, Akutsu T, et al. Delayed adverse reactions to iodinated contrast media and their risk factors. Radiat Med. 2000;18:39-45.
  4. Rydberg J, Charles J, Aspelin P. Frequency of late allergy-like adverse reactions following injection of intravascular non-ionic contrast media: a retrospective study comparing a non-ionic monomeric contrast medium with a non-ionic dimeric contrast medium. Acta Radiol. 1998;39:219-222.
  5. Sutton AG, Finn P, Grech ED, et al. Early and late reactions after the use of iopamidol 340, ioxaglate 320, and iodixanol 320 in cardiac catheterization. Am Heart J. 2001;141:677-683.
  6. Sutton AG, Finn P, Campbell PG, et al. Early and late reactions following the use of iopamidol 340, iomeprol 350 and iodixanol 320 in cardiac catheterization. J Invasive Cardiol. 2003;15:133-138.
  7. Brown M, Yowler C, Brandt C. Recurrent toxic epidermal necrolysis secondary to iopromide contrast. J Burn Care Res. 2013;34:E53-E56.
  8. Rosado A, Canto G, Veleiro B, et al. Toxic epidermal necrolysis after repeated injections of iohexol. AJR Am J Roentgenol. 2001;176:262-263.
  9. Peterson A, Katzberg RW, Fung MA, et al. Acute generalized exanthematous pustulosis as a delayed dermatotoxic reaction to IV-administered nonionic contrast media. AJR Am J Roentgenol. 2006;187:W198-W201.
  10. Good AE, Novak E, Sonda LP III. Fixed eruption and fever after urography. South Med J. 1980;73:948-949.
  11. Benson PM, Giblin WJ, Douglas DM. Transient, nonpigmenting fixed drug eruption caused by radiopaque contrast media. J Am Acad Dermatol. 1990;23(2, pt 2):379-381.
  12. Torres MJ, Gomez F, Doña I, et al. Diagnostic evaluation of patients with nonimmediate cutaneous hypersensitivity reactions to iodinated contrast media. Allergy. 2012;67:929-935.
  13. Scherer K, Harr T, Bach S, et al. The role of iodine in hypersensitivity reactions to radio contrast media. Clin Exp Allergy. 2010;40:468-475.
  14. Reynolds NJ, Wallington TB, Burton JL. Hydralazine predisposes to acute cutaneous vasculitis following urography with iopamidol. Br J Dermatol. 1993;129:82-85.
  15. Belhadjali H, Bouzgarrou L, Youssef M, et al. DRESS syndrome induced by sodium meglumine ioxitalamate. Allergy. 2008;63:786-787.
  16. Baldwin BT, Lien MH, Khan H, et al. Case of fatal toxic epidermal necrolysis due to cardiac catheterization dye. J Drugs Dermatol. 2010;9:837-840.
  17. Schmidt BJ, Foley WD, Bohorfoush AG. Toxic epidermal necrolysis related to oral administration of diluted diatrizoate meglumine and diatrizoate sodium. AJR Am J Roentgenol. 1998;171:1215-1216.
  18. Young AL, Grossman ME. Acute iododerma secondary to iodinated contrast media. Br J Dermatol. 2014;170:1377-1379.
  19. Choyke PL, Miller DL, Lotze MT, et al. Delayed reactions to contrast media after interleukin-2 immunotherapy. Radiology. 1992;183:111-114.
  20. Loh S, Bagheri S, Katzberg RW, et al. Delayed adverse reaction to contrast-enhanced CT: a prospective single-center study comparison to control group without enhancement. Radiology. 2010;255:764-771.
  21. Bertrand P, Delhommais A, Alison D, et al. Immediate and delayed tolerance of iohexol and ioxaglate in lower limb phlebography: a double-blind comparative study in humans. Acad Radiol. 1995;2:683-686.
  22. Munechika H, Hiramatsu Y, Kudo S, et al. A prospective survey of delayed adverse reactions to iohexol in urography and computed tomography. Eur Radiol. 2003;13:185-194.
  23. Guéant-Rodriguez RM, Romano A, Barbaud A, et al. Hypersensitivity reactions to iodinated contrast media. Curr Pharm Des. 2006;12:3359-3372.
  24. Huang SW. Seafood and iodine: an analysis of a medical myth. Allergy Asthma Proc. 2005;26:468-469.
  25. Baig M, Farag A, Sajid J, et al. Shellfish allergy and relation to iodinated contrast media: United Kingdom survey. World J Cardiol. 2014;6:107-111.
  26. Böhm I, Schild HH. A practical guide to diagnose lesser-known immediate and delayed contrast media-induced adverse cutaneous reactions. Eur Radiol. 2006;16:1570-1579.
  27. Ose K, Doue T, Zen K, et al. “Gadolinium” as an alternative to iodinated contrast media for X-ray angiography in patients with severe allergy. Circ J. 2005;69:507-509.
  28. Jingu A, Fukuda J, Taketomi-Takahashi A, et al. Breakthrough reactions of iodinated and gadolinium contrast media after oral steroid premedication protocol. BMC Med Imaging. 2014;14:34.
  29. Romano A, Artesani MC, Andriolo M, et al. Effective prophylactic protocol in delayed hypersensitivity to contrast media: report of a case involving lymphocyte transformation studies with different compounds. Radiology. 2002;225:466-470.
  30. Hebert AA, Bogle MA. Intravenous immunoglobulin prophylaxis for recurrent Stevens-Johnson syndrome. J Am Acad Dermatol. 2004;50:286-288.
  31. Hasdenteufel F, Waton J, Cordebar V, et al. Delayed hypersensitivity reactions caused by iodixanol: an assessment of cross-reactivity in 22 patients. J Allergy Clin Immunol. 2011;128:1356-1357.
Issue
Cutis - 101(6)
Issue
Cutis - 101(6)
Page Number
433-435
Page Number
433-435
Publications
Publications
Topics
Article Type
Display Headline
Delayed Cutaneous Reactions to Iodinated Contrast
Display Headline
Delayed Cutaneous Reactions to Iodinated Contrast
Sections
Inside the Article

Practice Points

  • Delayed cutaneous reactions to iodinated contrast (IC) are common, but patients frequently are misdiagnosed and inadvertently readministered the offending agent.
  • The most common IC-induced delayed reactions are self-limited exanthematous eruptions that develop within 1 week of exposure.
  • Risk factors for delayed reactions to IC include atopy, contrast exposure during high pollen season, use of the agent iodixanol, a history of other cutaneous drug eruptions, elevated serum creatinine levels, and treatment with recombinant interleukin 2.
  • Dermatologists can help prevent recurrent reactions in patients who require repeated exposure to IC by recommending gadolinium-based contrast agents and/or premedication.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Article PDF Media

Black Eschars on the Face and Body

Article Type
Changed
Thu, 01/10/2019 - 13:46
Display Headline
Black Eschars on the Face and Body

The Diagnosis: Lymphomatoid Papulosis

Histopathologic and immunohistochemical examination of the ulcer revealed a dense nodular and diffuse infiltrate in the papillary and reticular dermis comprised predominantly of atypical, CD30+, small T cells and large lymphoid cells admixed with neutrophils and eosinophils (Figures 1 and 2). Tissue cultures and infectious stains were negative. The complete blood cell count, metabolic panel, serum lactate dehydrogenase level, and peripheral blood flow cytometry were normal. Correlation of the lesions' self-healing nature with the histopathologic and immunohistochemical findings led to a diagnosis of lymphomatoid papulosis (LyP). In light of this diagnosis, a shave biopsy was obtained of one of the patient's poikilodermatous patches and was found to be consistent with poikilodermatous mycosis fungoides (MF).

Figure 1. Lymphomatoid papulosis histopathology of an ulcer on the chest showed a dense nodular and diffuse infiltrate in the papillary and reticular dermis comprised predominantly of atypical small T cells and large lymphoid cells admixed with neutrophils and eosinophils (H&E, original magnification ×400).

Figure 2. Lymphomatoid papulosis immunohistochemistry of a chest ulcer showed CD30 T cells (original magnification ×400).

At 4-month follow-up, the patient reported that she continued to develop crops of 1 to 3 LyP lesions each month. She continued to deny systemic concerns, and the poikilodermatous MF appeared unchanged. As part of a hematologic workup, a positron emission tomography-computed tomography scan revealed glucose-avid lymph nodes in the axillary, supraclavicular, abdominal, and inguinal regions. These findings raised concern for possible lymphomatous involvement of the patient's MF. Systemic therapy may be required pending further surveillance.

Lymphomatoid papulosis is a chronic papulonecrotic disease characterized clinically by recurrent crops of self-healing papules. Histopathologically, LyP features a perivascular infiltrate with atypical dermal T cells. Macaulay1 first described LyP in 1968 in a 41-year-old woman with a several-year history of continuously self-resolving crops of necrotic papules, noting the paradox between the patient's benign clinical course and malignant-seeming histology featuring "an alarming infiltrate of anaplastic cells." Since this report, LyP has continued to spur debate regarding its malignant potential but is now recognized as an indolent cutaneous T-cell lymphoma with an excellent prognosis.2

There are several histopathologic subtypes of LyP, the most common of which are type A, resembling Hodgkin lymphoma; type B, resembling MF; type C, resembling primary cutaneous anaplastic large cell lymphoma (C-ALCL); and type D, resembling aggressive epidermotropic CD8+ cutaneous T-cell lymphoma.2 

The multifocal ulcers and eschars of LyP may appropriately raise suspicion for an infectious process, as in the present case. Numerous reports show that LyP may be initially misdiagnosed as an infection, such as cellulitis,3 furunculosis,4 parapoxvirus Orf,5 and ecthyma.6 Furthermore, several cutaneous infections have histopathologic features indistinguishable from LyP.7 For example, herpes simplex virus infection, molluscum contagiosum, Milker nodule, syphilis, and leishmaniasis may contain an appreciable number of large CD30+ T cells, which is compatible with both LyP type C and C-ALCL.7 As in the present case, the final diagnosis rests on clinicopathologic correlation, with LyP often distinguished by its invariable self-resolution, unlike its numerous infectious mimickers. The self-regressing nature of LyP also helps differentiate LyP occurring in the setting of MF from MF that has underwent CD30+ large cell transformation. In addition, the diagnosis of MF-associated LyP is favored over transformed MF when, as in the present case, CD30+ lesions develop on skin distinct from MF-affected skin.

Although isolated LyP is benign, 18% (11/61) of patients will subsequently develop lymphoma. More commonly, lymphomas may precede or occur concomitantly with the onset of LyP. In a retrospective study of 84 LyP patients, for example, 40% (34/84) had prior or concomitant lymphoma.8 Owing to the well-established link between LyP and lymphoma, there is appropriate emphasis on close monitoring of these patients. In addition, a careful history and physical examination are necessary to evaluate for a preceding, previously undiagnosed lymphoma. In point of fact, our patient had undiagnosed poikilodermatous MF prior to developing LyP, which was proven by biopsy at the time of LyP diagnosis. A distinct clinical variant of MF, poikilodermatous MF is characterized by hyperpigmented and hypopigmented patches, atrophy, and telangiectasia. A study of 49 patients with poikilodermatous MF found that this variant had an earlier age of onset compared with other types of MF. The study also showed that 18% (9/49) of patients had coexistent LyP, suggesting that poikilodermatous MF and LyP may be more frequently associated than previously believed.9

Treatment of LyP is unnecessary beyond basic wound care to avoid bacterial superinfection.2,10 Therapy for poikilodermatous MF, similar to other types of MF, is based on disease stage. Topical therapy may be utilized for localized disease, while systemic therapies are reserved for recalcitrant cases and internal involvement.9
 
Acknowledgments
We thank David L. Ramsay, MD, for obtaining aspects of the patient's history, and Shane A. Meehan, MD, and Adnan Mir, MD, PhD, as well as Cynthia M. Magro, MD, (all from New York, New York) for performing the histopathologic and immunohistochemical analyses.

References
  1. Macaulay WL. Lymphomatoid papulosis. a continuing self-healing eruption, clinically benign--histologically malignant. Arch Dermatol. 1968;97:23-30.
  2. Willemze R, Jaffe ES, Burg G, et al. WHO-EORTC classification for cutaneous lymphomas. Blood. 2005;105:3768-3785.
  3. Meena M, Martin PA, Abouseif C, et al. Lymphomatoid papulosis type C of the eyelid in a young girl: a case report and review of literature. Orbit. 2014;3:395-398.
  4. Dinotta F, Lacarrubba F, Micali G. Sixteen-year-old girl with papules and nodules on the face and upper limbs. Pediatr Dermatol. 2014;31:103-104.
  5. Eminger LA, Shinohara MM, Kim EJ, et al. Clinicopathologic challenge: acral lymphomatoid papulosis. Int J Dermatol. 2012;51:531-534.
  6. Harder D, Kuhn A, Mahrle G. Lymphomatoid papulosis resembling ecthyma. a case report. Z Hautkr. 1989;64:593-595.
  7. Werner B, Massone C, Kerl H, et al. Large CD30-positive cells in benign, atypical lymphoid infiltrates of the skin. J Cutan Pathol. 2008;35:1100-1107.
  8. Kunishige JH, McDonald H, Alvarez G, et al. Lymphomatoid papulosis and associated lymphomas: a retrospective case series of 84 patients. Clin Exp Dermatol. 2009;34:576-581.
  9. Abbott RA, Sahni D, Robson A, et al. Poikilodermatous mycosis fungoides: a study of its clinicopathological, immunophenotypic, and prognostic features. J Am Acad Dermatol. 2011;65:313-319.
  10. Kempf W, Pfaltz K, Vermeer MH, et al. EORTC, ISCL, and USCLC consensus recommendations for the treatment of primary cutaneous CD30-positive lymphoproliferative disorders: lymphomatoid papulosis and primary cutaneous anaplastic large-cell lymphoma. Blood. 2011;118:4024-4035.
Article PDF
Author and Disclosure Information

From the Ronald O. Perelman Department of Dermatology, New York University Langone Medical Center, New York.

The authors report no conflict of interest. Dr. Femia's career has been in part supported by funding from the Noah-Sadie K. Wachtel Foundation, which had no role in study design, data collection, data analysis, manuscript preparation, or publication decisions.

Correspondence: Alisa N. Femia, MD, Ronald O. Perelman Department of Dermatology, New York University Langone Medical Center, 240 E 38th St, 11th Floor, New York, NY 10016 (alisa.femia@nyumc.org).

Issue
Cutis - 100(5)
Publications
Topics
Page Number
281, 297-298
Sections
Author and Disclosure Information

From the Ronald O. Perelman Department of Dermatology, New York University Langone Medical Center, New York.

The authors report no conflict of interest. Dr. Femia's career has been in part supported by funding from the Noah-Sadie K. Wachtel Foundation, which had no role in study design, data collection, data analysis, manuscript preparation, or publication decisions.

Correspondence: Alisa N. Femia, MD, Ronald O. Perelman Department of Dermatology, New York University Langone Medical Center, 240 E 38th St, 11th Floor, New York, NY 10016 (alisa.femia@nyumc.org).

Author and Disclosure Information

From the Ronald O. Perelman Department of Dermatology, New York University Langone Medical Center, New York.

The authors report no conflict of interest. Dr. Femia's career has been in part supported by funding from the Noah-Sadie K. Wachtel Foundation, which had no role in study design, data collection, data analysis, manuscript preparation, or publication decisions.

Correspondence: Alisa N. Femia, MD, Ronald O. Perelman Department of Dermatology, New York University Langone Medical Center, 240 E 38th St, 11th Floor, New York, NY 10016 (alisa.femia@nyumc.org).

Article PDF
Article PDF
Related Articles

The Diagnosis: Lymphomatoid Papulosis

Histopathologic and immunohistochemical examination of the ulcer revealed a dense nodular and diffuse infiltrate in the papillary and reticular dermis comprised predominantly of atypical, CD30+, small T cells and large lymphoid cells admixed with neutrophils and eosinophils (Figures 1 and 2). Tissue cultures and infectious stains were negative. The complete blood cell count, metabolic panel, serum lactate dehydrogenase level, and peripheral blood flow cytometry were normal. Correlation of the lesions' self-healing nature with the histopathologic and immunohistochemical findings led to a diagnosis of lymphomatoid papulosis (LyP). In light of this diagnosis, a shave biopsy was obtained of one of the patient's poikilodermatous patches and was found to be consistent with poikilodermatous mycosis fungoides (MF).

Figure 1. Lymphomatoid papulosis histopathology of an ulcer on the chest showed a dense nodular and diffuse infiltrate in the papillary and reticular dermis comprised predominantly of atypical small T cells and large lymphoid cells admixed with neutrophils and eosinophils (H&E, original magnification ×400).

Figure 2. Lymphomatoid papulosis immunohistochemistry of a chest ulcer showed CD30 T cells (original magnification ×400).

At 4-month follow-up, the patient reported that she continued to develop crops of 1 to 3 LyP lesions each month. She continued to deny systemic concerns, and the poikilodermatous MF appeared unchanged. As part of a hematologic workup, a positron emission tomography-computed tomography scan revealed glucose-avid lymph nodes in the axillary, supraclavicular, abdominal, and inguinal regions. These findings raised concern for possible lymphomatous involvement of the patient's MF. Systemic therapy may be required pending further surveillance.

Lymphomatoid papulosis is a chronic papulonecrotic disease characterized clinically by recurrent crops of self-healing papules. Histopathologically, LyP features a perivascular infiltrate with atypical dermal T cells. Macaulay1 first described LyP in 1968 in a 41-year-old woman with a several-year history of continuously self-resolving crops of necrotic papules, noting the paradox between the patient's benign clinical course and malignant-seeming histology featuring "an alarming infiltrate of anaplastic cells." Since this report, LyP has continued to spur debate regarding its malignant potential but is now recognized as an indolent cutaneous T-cell lymphoma with an excellent prognosis.2

There are several histopathologic subtypes of LyP, the most common of which are type A, resembling Hodgkin lymphoma; type B, resembling MF; type C, resembling primary cutaneous anaplastic large cell lymphoma (C-ALCL); and type D, resembling aggressive epidermotropic CD8+ cutaneous T-cell lymphoma.2 

The multifocal ulcers and eschars of LyP may appropriately raise suspicion for an infectious process, as in the present case. Numerous reports show that LyP may be initially misdiagnosed as an infection, such as cellulitis,3 furunculosis,4 parapoxvirus Orf,5 and ecthyma.6 Furthermore, several cutaneous infections have histopathologic features indistinguishable from LyP.7 For example, herpes simplex virus infection, molluscum contagiosum, Milker nodule, syphilis, and leishmaniasis may contain an appreciable number of large CD30+ T cells, which is compatible with both LyP type C and C-ALCL.7 As in the present case, the final diagnosis rests on clinicopathologic correlation, with LyP often distinguished by its invariable self-resolution, unlike its numerous infectious mimickers. The self-regressing nature of LyP also helps differentiate LyP occurring in the setting of MF from MF that has underwent CD30+ large cell transformation. In addition, the diagnosis of MF-associated LyP is favored over transformed MF when, as in the present case, CD30+ lesions develop on skin distinct from MF-affected skin.

Although isolated LyP is benign, 18% (11/61) of patients will subsequently develop lymphoma. More commonly, lymphomas may precede or occur concomitantly with the onset of LyP. In a retrospective study of 84 LyP patients, for example, 40% (34/84) had prior or concomitant lymphoma.8 Owing to the well-established link between LyP and lymphoma, there is appropriate emphasis on close monitoring of these patients. In addition, a careful history and physical examination are necessary to evaluate for a preceding, previously undiagnosed lymphoma. In point of fact, our patient had undiagnosed poikilodermatous MF prior to developing LyP, which was proven by biopsy at the time of LyP diagnosis. A distinct clinical variant of MF, poikilodermatous MF is characterized by hyperpigmented and hypopigmented patches, atrophy, and telangiectasia. A study of 49 patients with poikilodermatous MF found that this variant had an earlier age of onset compared with other types of MF. The study also showed that 18% (9/49) of patients had coexistent LyP, suggesting that poikilodermatous MF and LyP may be more frequently associated than previously believed.9

Treatment of LyP is unnecessary beyond basic wound care to avoid bacterial superinfection.2,10 Therapy for poikilodermatous MF, similar to other types of MF, is based on disease stage. Topical therapy may be utilized for localized disease, while systemic therapies are reserved for recalcitrant cases and internal involvement.9
 
Acknowledgments
We thank David L. Ramsay, MD, for obtaining aspects of the patient's history, and Shane A. Meehan, MD, and Adnan Mir, MD, PhD, as well as Cynthia M. Magro, MD, (all from New York, New York) for performing the histopathologic and immunohistochemical analyses.

The Diagnosis: Lymphomatoid Papulosis

Histopathologic and immunohistochemical examination of the ulcer revealed a dense nodular and diffuse infiltrate in the papillary and reticular dermis comprised predominantly of atypical, CD30+, small T cells and large lymphoid cells admixed with neutrophils and eosinophils (Figures 1 and 2). Tissue cultures and infectious stains were negative. The complete blood cell count, metabolic panel, serum lactate dehydrogenase level, and peripheral blood flow cytometry were normal. Correlation of the lesions' self-healing nature with the histopathologic and immunohistochemical findings led to a diagnosis of lymphomatoid papulosis (LyP). In light of this diagnosis, a shave biopsy was obtained of one of the patient's poikilodermatous patches and was found to be consistent with poikilodermatous mycosis fungoides (MF).

Figure 1. Lymphomatoid papulosis histopathology of an ulcer on the chest showed a dense nodular and diffuse infiltrate in the papillary and reticular dermis comprised predominantly of atypical small T cells and large lymphoid cells admixed with neutrophils and eosinophils (H&E, original magnification ×400).

Figure 2. Lymphomatoid papulosis immunohistochemistry of a chest ulcer showed CD30 T cells (original magnification ×400).

At 4-month follow-up, the patient reported that she continued to develop crops of 1 to 3 LyP lesions each month. She continued to deny systemic concerns, and the poikilodermatous MF appeared unchanged. As part of a hematologic workup, a positron emission tomography-computed tomography scan revealed glucose-avid lymph nodes in the axillary, supraclavicular, abdominal, and inguinal regions. These findings raised concern for possible lymphomatous involvement of the patient's MF. Systemic therapy may be required pending further surveillance.

Lymphomatoid papulosis is a chronic papulonecrotic disease characterized clinically by recurrent crops of self-healing papules. Histopathologically, LyP features a perivascular infiltrate with atypical dermal T cells. Macaulay1 first described LyP in 1968 in a 41-year-old woman with a several-year history of continuously self-resolving crops of necrotic papules, noting the paradox between the patient's benign clinical course and malignant-seeming histology featuring "an alarming infiltrate of anaplastic cells." Since this report, LyP has continued to spur debate regarding its malignant potential but is now recognized as an indolent cutaneous T-cell lymphoma with an excellent prognosis.2

There are several histopathologic subtypes of LyP, the most common of which are type A, resembling Hodgkin lymphoma; type B, resembling MF; type C, resembling primary cutaneous anaplastic large cell lymphoma (C-ALCL); and type D, resembling aggressive epidermotropic CD8+ cutaneous T-cell lymphoma.2 

The multifocal ulcers and eschars of LyP may appropriately raise suspicion for an infectious process, as in the present case. Numerous reports show that LyP may be initially misdiagnosed as an infection, such as cellulitis,3 furunculosis,4 parapoxvirus Orf,5 and ecthyma.6 Furthermore, several cutaneous infections have histopathologic features indistinguishable from LyP.7 For example, herpes simplex virus infection, molluscum contagiosum, Milker nodule, syphilis, and leishmaniasis may contain an appreciable number of large CD30+ T cells, which is compatible with both LyP type C and C-ALCL.7 As in the present case, the final diagnosis rests on clinicopathologic correlation, with LyP often distinguished by its invariable self-resolution, unlike its numerous infectious mimickers. The self-regressing nature of LyP also helps differentiate LyP occurring in the setting of MF from MF that has underwent CD30+ large cell transformation. In addition, the diagnosis of MF-associated LyP is favored over transformed MF when, as in the present case, CD30+ lesions develop on skin distinct from MF-affected skin.

Although isolated LyP is benign, 18% (11/61) of patients will subsequently develop lymphoma. More commonly, lymphomas may precede or occur concomitantly with the onset of LyP. In a retrospective study of 84 LyP patients, for example, 40% (34/84) had prior or concomitant lymphoma.8 Owing to the well-established link between LyP and lymphoma, there is appropriate emphasis on close monitoring of these patients. In addition, a careful history and physical examination are necessary to evaluate for a preceding, previously undiagnosed lymphoma. In point of fact, our patient had undiagnosed poikilodermatous MF prior to developing LyP, which was proven by biopsy at the time of LyP diagnosis. A distinct clinical variant of MF, poikilodermatous MF is characterized by hyperpigmented and hypopigmented patches, atrophy, and telangiectasia. A study of 49 patients with poikilodermatous MF found that this variant had an earlier age of onset compared with other types of MF. The study also showed that 18% (9/49) of patients had coexistent LyP, suggesting that poikilodermatous MF and LyP may be more frequently associated than previously believed.9

Treatment of LyP is unnecessary beyond basic wound care to avoid bacterial superinfection.2,10 Therapy for poikilodermatous MF, similar to other types of MF, is based on disease stage. Topical therapy may be utilized for localized disease, while systemic therapies are reserved for recalcitrant cases and internal involvement.9
 
Acknowledgments
We thank David L. Ramsay, MD, for obtaining aspects of the patient's history, and Shane A. Meehan, MD, and Adnan Mir, MD, PhD, as well as Cynthia M. Magro, MD, (all from New York, New York) for performing the histopathologic and immunohistochemical analyses.

References
  1. Macaulay WL. Lymphomatoid papulosis. a continuing self-healing eruption, clinically benign--histologically malignant. Arch Dermatol. 1968;97:23-30.
  2. Willemze R, Jaffe ES, Burg G, et al. WHO-EORTC classification for cutaneous lymphomas. Blood. 2005;105:3768-3785.
  3. Meena M, Martin PA, Abouseif C, et al. Lymphomatoid papulosis type C of the eyelid in a young girl: a case report and review of literature. Orbit. 2014;3:395-398.
  4. Dinotta F, Lacarrubba F, Micali G. Sixteen-year-old girl with papules and nodules on the face and upper limbs. Pediatr Dermatol. 2014;31:103-104.
  5. Eminger LA, Shinohara MM, Kim EJ, et al. Clinicopathologic challenge: acral lymphomatoid papulosis. Int J Dermatol. 2012;51:531-534.
  6. Harder D, Kuhn A, Mahrle G. Lymphomatoid papulosis resembling ecthyma. a case report. Z Hautkr. 1989;64:593-595.
  7. Werner B, Massone C, Kerl H, et al. Large CD30-positive cells in benign, atypical lymphoid infiltrates of the skin. J Cutan Pathol. 2008;35:1100-1107.
  8. Kunishige JH, McDonald H, Alvarez G, et al. Lymphomatoid papulosis and associated lymphomas: a retrospective case series of 84 patients. Clin Exp Dermatol. 2009;34:576-581.
  9. Abbott RA, Sahni D, Robson A, et al. Poikilodermatous mycosis fungoides: a study of its clinicopathological, immunophenotypic, and prognostic features. J Am Acad Dermatol. 2011;65:313-319.
  10. Kempf W, Pfaltz K, Vermeer MH, et al. EORTC, ISCL, and USCLC consensus recommendations for the treatment of primary cutaneous CD30-positive lymphoproliferative disorders: lymphomatoid papulosis and primary cutaneous anaplastic large-cell lymphoma. Blood. 2011;118:4024-4035.
References
  1. Macaulay WL. Lymphomatoid papulosis. a continuing self-healing eruption, clinically benign--histologically malignant. Arch Dermatol. 1968;97:23-30.
  2. Willemze R, Jaffe ES, Burg G, et al. WHO-EORTC classification for cutaneous lymphomas. Blood. 2005;105:3768-3785.
  3. Meena M, Martin PA, Abouseif C, et al. Lymphomatoid papulosis type C of the eyelid in a young girl: a case report and review of literature. Orbit. 2014;3:395-398.
  4. Dinotta F, Lacarrubba F, Micali G. Sixteen-year-old girl with papules and nodules on the face and upper limbs. Pediatr Dermatol. 2014;31:103-104.
  5. Eminger LA, Shinohara MM, Kim EJ, et al. Clinicopathologic challenge: acral lymphomatoid papulosis. Int J Dermatol. 2012;51:531-534.
  6. Harder D, Kuhn A, Mahrle G. Lymphomatoid papulosis resembling ecthyma. a case report. Z Hautkr. 1989;64:593-595.
  7. Werner B, Massone C, Kerl H, et al. Large CD30-positive cells in benign, atypical lymphoid infiltrates of the skin. J Cutan Pathol. 2008;35:1100-1107.
  8. Kunishige JH, McDonald H, Alvarez G, et al. Lymphomatoid papulosis and associated lymphomas: a retrospective case series of 84 patients. Clin Exp Dermatol. 2009;34:576-581.
  9. Abbott RA, Sahni D, Robson A, et al. Poikilodermatous mycosis fungoides: a study of its clinicopathological, immunophenotypic, and prognostic features. J Am Acad Dermatol. 2011;65:313-319.
  10. Kempf W, Pfaltz K, Vermeer MH, et al. EORTC, ISCL, and USCLC consensus recommendations for the treatment of primary cutaneous CD30-positive lymphoproliferative disorders: lymphomatoid papulosis and primary cutaneous anaplastic large-cell lymphoma. Blood. 2011;118:4024-4035.
Issue
Cutis - 100(5)
Issue
Cutis - 100(5)
Page Number
281, 297-298
Page Number
281, 297-298
Publications
Publications
Topics
Article Type
Display Headline
Black Eschars on the Face and Body
Display Headline
Black Eschars on the Face and Body
Sections
Questionnaire Body

A 50-year-old woman presented for evaluation of black eschars on the face and body. Over the preceding 8 weeks she had developed several asymptomatic papules that gradually enlarged, ulcerated, and formed a black eschar, prior to gradually self-resolving over the course of several weeks. During this time, new lesions were forming. The resulting skin revealed dyspigmentation and scar formation. Prior to presentation, antimicrobial therapy had been initiated for a presumed infectious etiology; however, the eruption continued to progress. The patient denied sick contacts, livestock exposure, or recent travel. A complete review of systems, including fever, chills, or lymphadenopathy, was negative. Physical examination revealed 6 circular necrotic ulcers with an overlying black eschar on the face (top), trunk (bottom), hands, and thighs, all in various stages of healing. In addition, large, reticulated, poikilodermatous patches were incidentally noted in areas free of ulcers and eschars on the trunk (bottom) and bilateral arms and legs. Upon questioning, the patient said these patches had been present for more than 30 years. A punch biopsy from an ulcer on the chest was obtained and sent for histopathologic and immunohistochemical examination.

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Article PDF Media