Moving Beyond Traditional Methods for Treatment of Acne Keloidalis Nuchae

Article Type
Changed
Thu, 09/19/2024 - 15:10

Acne keloidalis nuchae (AKN) is a chronic inflammatory condition commonly affecting the occipital scalp and posterior neck. It causes discrete or extensive fibrosing papules that may coalesce to form pronounced ­tumorlike masses1,2 with scarring alopecia (Figure, A–C).3 Pustules, hair tufts, secondary bacterial infections, abscesses, and sinus tracts also may occur.1 The pathogenesis of AKN has been characterized as varying stages of follicular inflammation at the infundibular and isthmus levels followed by fibrotic occlusion of the ­follicular lumen.4 Pruritus, pain, bleeding, oozing, and a feeling of scalp tightness may occur.1,5

Umar et al6 performed a retrospective review of 108 men with AKN—58% of African descent, 37% Hispanic, 3% Asian, and 2% Middle Eastern—and proposed a 3-tier classification system for AKN. Tier 1 focused on the distribution and sagittal spread of AKN lesions between the clinical demarcation lines of the occipital notch and posterior hairline. Tier 2 focused on the type of lesions present—discrete papules or nodules, coalescing/abutting lesions, plaques (raised, atrophic, or indurated), or dome-shaped tumoral masses. Tier 3 focused on the presence or absence of co-existing dissecting cellulitis or folliculitis decalvans.6

Epidemiology

Acne keloidalis nuchae primarily manifests in adolescent and adult men of African or Afro-Caribbean descent.7 Among African American men, the prevalence of AKN ranges from 0.5% to 13.6%.8 Similar ranges have been reported among Nigerian, South African, and West African men.1 Acne keloidalis nuchae also affects Asian and Hispanic men but rarely is seen in non-Hispanic White men or in women of any ethnicity.9,10 The male to female ratio is 20:1.1,11 Hair texture, hairstyling practices such as closely shaved or faded haircuts, and genetics likely contribute to development of AKN. Sports and occupations that require the use of headgear or a tight collar may increase the risk for AKN.12

Key clinical features in people with darker skin tones

  • The lesions of AKN range in color from pink to dark brown or black. Postinflammatory hyperpigmentation or hyperchromia may be present around AKN lesions.
  • Chronicity of AKN may lead to extended use of high-potency topical or intralesional corticosteroids, which causes transient or long-lasting hypopigmentation, especially in those with darker skin tones.

Worth noting

  • Acne keloidalis nuchae can be disfiguring, which negatively impacts quality of life and self-esteem.12
  • Some occupations (eg, military, police) have hair policies that may not be favorable to those with or at risk for AKN.
  • Patients with AKN are 2 to 3 times more likely to present with metabolic syndrome, hypertension, type 2 diabetes mellitus, or obesity.13
 

 

Treatment

There are no treatments approved by the US Food and Drug Administration specifically for AKN. Treatment approaches are based on the pathophysiology, secondary impacts on the skin, and disease severity. Growing out the hair may prevent worsening and/or decrease the risk for new lesions.6

  • Options include but are not limited to topical and systemic therapies (eg, topical corticosteroids, oral or topical antibiotics, isotretinoin, topical retinoids, imiquimod, pimecrolimus), light devices (eg, phototherapy, laser), ablative therapies (eg, laser, cryotherapy, radiotherapy), and surgery (eg, excision, follicular unit excision), often in combination.6,14,15
  • Intralesional triamcinolone injections are considered standard of care. Adotama et al found that injecting ­triamcinolone into the deep dermis in the area of flat or papular AKN yielded better control of inflammation and decreased appearance of lesions compared with injecting individual lesions.16
  • For extensive AKN lesions that do not respond to ­less-invasive therapies, consider surgical techniques,6,17 such as follicular unit excision18 and more extensive surgical excisions building on approaches from pioneers Drs. John Kenney and Harold Pierce.19 An innovative surgical approach for removal of large AKNs is the bat excision technique—wound shape resembles a bat in a spread-eagled position—with secondary intention healing with or without debridement and/or tension sutures. The resulting linear scar acts as a new posterior hair line.20
 

 

Health disparity highlights

Access to a dermatologic or plastic surgeon with expertise in the surgical treatment of large AKNs may be challenging but is needed to reduce risk for recurrence and adverse events.

Close-cropped haircuts on the occipital scalp, which are particularly popular among men of African descent, increase the risk for AKN.5 Although this grooming style may be a personal preference, other hairstyles commonly worn by those with tightly coiled hair may be deemed “unprofessional” in society or the workplace, which leads to hairstyling practices that may increase the risk for AKN.21

Acne keloidalis nuchae remains an understudied entity that adversely affects patients with skin of color.

References
  1. Ogunbiyi A. Acne keloidalis nuchae: prevalence, impact, and management challenges. Clin Cosmet Investig Dermatol. 2016;9:483-489. doi:10.2147/CCID.S99225 
  2. Al Aboud DM, Badri T. Acne keloidalis nuchae. In: StatPearls [Internet]. Updated July 31, 2023. Accessed August 2, 2024. https://www.ncbi.nlm.nih.gov/books/NBK459135/
  3. Sperling LC, Homoky C, Pratt L, et al. Acne keloidalis is a form of primary scarring alopecia. Arch Dermatol. 2000;136:479-484.
  4. Herzberg AJ, Dinehart SM, Kerns BJ, et al. Acne keloidalis: transverse microscopy, immunohistochemistry, and electron microscopy. Am J Dermatopathol. 1990;12:109-121. doi:10.1097/00000372-199004000-00001
  5. Saka B, Akakpo A-S, Téclessou JN, et al. Risk factors associated with acne keloidalis nuchae in black subjects: a case-control study. Ann Dermatol Venereol. 2020;147:350-354. doi:10.1016/j.annder.2020.01.007
  6. Umar S, Lee DJ, Lullo JJ. A retrospective cohort study and clinical classification system of acne keloidalis nuchae. J Clin Aesthet Dermatol. 2021;14:E61-E67.
  7. Reja M, Silverberg NB. Acne keloidalis nuchae. In: Silverberg NB, Durán-McKinster C, Tay YK, eds. Pediatric Skin of Color. Springer; 2015:141-145. doi:10.1007/978-1-4614-6654-3_16
  8. Knable AL Jr, Hanke CW, Gonin R. Prevalence of acne keloidalis nuchae in football players. J Am Acad Dermatol. 1997;37:570-574. doi:10.1016/s0190-9622(97)70173-7
  9. Umar S, Ton D, Carter MJ, et al. Unveiling a shared precursor condition for acne keloidalis nuchae and primary cicatricial alopecias. Clin Cosmet Investig Dermatol. 2023;16:2315-2327. doi:10.2147/CCID.S422310
  10. Na K, Oh SH, Kim SK. Acne keloidalis nuchae in Asian: a single institutional experience. PLoS One. 2017;12:e0189790. doi:10.1371/journal.pone.0189790
  11. Ogunbiyi A, George A. Acne keloidalis in females: case report and review of literature. J Natl Med Assoc. 2005;97:736-738. 
  12. Alexis A, Heath CR, Halder RM. Folliculitis keloidalis nuchae and pseudofolliculitis barbae: are prevention and effective treatment within reach? Dermatol Clin. 2014;32:183-191. doi:10.1016/j.det.2013.12.001
  13. Kridin K, Solomon A, Tzur-Bitan D, et al. Acne keloidalis nuchae and the metabolic syndrome: a population-based study. Am J Clin Dermatol. 2020;21:733-739. doi:10.1007/s40257-020-00541-z
  14. Smart K, Rodriguez I, Worswick S. Comorbidities and treatment options for acne keloidalis nuchae. Dermatol Ther. Published online May 25, 2024. doi:10.1155/2024/8336926
  15. Callender VD, Young CM, Haverstock CL, et al. An open label study of clobetasol propionate 0.05% and betamethasone valerate 0.12% foams in the treatment of mild to moderate acne keloidalis. Cutis. 2005;75:317-321.
  16. Adotama P, Grullon K, Ali S, et al. How we do it: our method for triamcinolone injections of acne keloidalis nuchae. Dermatol Surg. 2023;49:713-714. doi:10.1097/DSS.0000000000003803
  17. Beckett N, Lawson C, Cohen G. Electrosurgical excision of acne keloidalis nuchae with secondary intention healing. J Clin Aesthet Dermatol. 2011;4:36-39.
  18. Esmat SM, Abdel Hay RM, Abu Zeid OM, et al. The efficacy of laser-assisted hair removal in the treatment of acne keloidalis nuchae; a pilot study. Eur J Dermatol. 2012;22:645-650. doi:10.1684/ejd.2012.1830
  19. Dillard AD, Quarles FN. African-American pioneers in dermatology. In: Taylor SC, Kelly AP, Lim HW, et al, eds. Dermatology for Skin of Color. 2nd ed. McGraw-Hill Education; 2016:717-730.
  20. Umar S, David CV, Castillo JR, et al. Innovative surgical approaches and selection criteria of large acne keloidalis nuchae lesions. Plast Reconstr Surg Glob Open. 2019;7:E2215. doi:10.1097/GOX.0000000000002215
  21. Lee MS, Nambudiri VE. The CROWN act and dermatology: taking a stand against race-based hair discrimination. J Am Acad Dermatol. 2021;84:1181-1182. doi:10.1016/j.jaad.2020.11.065
Article PDF
Issue
Federal Practitioner - 41(9)a
Publications
Topics
Page Number
312-313
Sections
Article PDF
Article PDF

Acne keloidalis nuchae (AKN) is a chronic inflammatory condition commonly affecting the occipital scalp and posterior neck. It causes discrete or extensive fibrosing papules that may coalesce to form pronounced ­tumorlike masses1,2 with scarring alopecia (Figure, A–C).3 Pustules, hair tufts, secondary bacterial infections, abscesses, and sinus tracts also may occur.1 The pathogenesis of AKN has been characterized as varying stages of follicular inflammation at the infundibular and isthmus levels followed by fibrotic occlusion of the ­follicular lumen.4 Pruritus, pain, bleeding, oozing, and a feeling of scalp tightness may occur.1,5

Umar et al6 performed a retrospective review of 108 men with AKN—58% of African descent, 37% Hispanic, 3% Asian, and 2% Middle Eastern—and proposed a 3-tier classification system for AKN. Tier 1 focused on the distribution and sagittal spread of AKN lesions between the clinical demarcation lines of the occipital notch and posterior hairline. Tier 2 focused on the type of lesions present—discrete papules or nodules, coalescing/abutting lesions, plaques (raised, atrophic, or indurated), or dome-shaped tumoral masses. Tier 3 focused on the presence or absence of co-existing dissecting cellulitis or folliculitis decalvans.6

Epidemiology

Acne keloidalis nuchae primarily manifests in adolescent and adult men of African or Afro-Caribbean descent.7 Among African American men, the prevalence of AKN ranges from 0.5% to 13.6%.8 Similar ranges have been reported among Nigerian, South African, and West African men.1 Acne keloidalis nuchae also affects Asian and Hispanic men but rarely is seen in non-Hispanic White men or in women of any ethnicity.9,10 The male to female ratio is 20:1.1,11 Hair texture, hairstyling practices such as closely shaved or faded haircuts, and genetics likely contribute to development of AKN. Sports and occupations that require the use of headgear or a tight collar may increase the risk for AKN.12

Key clinical features in people with darker skin tones

  • The lesions of AKN range in color from pink to dark brown or black. Postinflammatory hyperpigmentation or hyperchromia may be present around AKN lesions.
  • Chronicity of AKN may lead to extended use of high-potency topical or intralesional corticosteroids, which causes transient or long-lasting hypopigmentation, especially in those with darker skin tones.

Worth noting

  • Acne keloidalis nuchae can be disfiguring, which negatively impacts quality of life and self-esteem.12
  • Some occupations (eg, military, police) have hair policies that may not be favorable to those with or at risk for AKN.
  • Patients with AKN are 2 to 3 times more likely to present with metabolic syndrome, hypertension, type 2 diabetes mellitus, or obesity.13
 

 

Treatment

There are no treatments approved by the US Food and Drug Administration specifically for AKN. Treatment approaches are based on the pathophysiology, secondary impacts on the skin, and disease severity. Growing out the hair may prevent worsening and/or decrease the risk for new lesions.6

  • Options include but are not limited to topical and systemic therapies (eg, topical corticosteroids, oral or topical antibiotics, isotretinoin, topical retinoids, imiquimod, pimecrolimus), light devices (eg, phototherapy, laser), ablative therapies (eg, laser, cryotherapy, radiotherapy), and surgery (eg, excision, follicular unit excision), often in combination.6,14,15
  • Intralesional triamcinolone injections are considered standard of care. Adotama et al found that injecting ­triamcinolone into the deep dermis in the area of flat or papular AKN yielded better control of inflammation and decreased appearance of lesions compared with injecting individual lesions.16
  • For extensive AKN lesions that do not respond to ­less-invasive therapies, consider surgical techniques,6,17 such as follicular unit excision18 and more extensive surgical excisions building on approaches from pioneers Drs. John Kenney and Harold Pierce.19 An innovative surgical approach for removal of large AKNs is the bat excision technique—wound shape resembles a bat in a spread-eagled position—with secondary intention healing with or without debridement and/or tension sutures. The resulting linear scar acts as a new posterior hair line.20
 

 

Health disparity highlights

Access to a dermatologic or plastic surgeon with expertise in the surgical treatment of large AKNs may be challenging but is needed to reduce risk for recurrence and adverse events.

Close-cropped haircuts on the occipital scalp, which are particularly popular among men of African descent, increase the risk for AKN.5 Although this grooming style may be a personal preference, other hairstyles commonly worn by those with tightly coiled hair may be deemed “unprofessional” in society or the workplace, which leads to hairstyling practices that may increase the risk for AKN.21

Acne keloidalis nuchae remains an understudied entity that adversely affects patients with skin of color.

Acne keloidalis nuchae (AKN) is a chronic inflammatory condition commonly affecting the occipital scalp and posterior neck. It causes discrete or extensive fibrosing papules that may coalesce to form pronounced ­tumorlike masses1,2 with scarring alopecia (Figure, A–C).3 Pustules, hair tufts, secondary bacterial infections, abscesses, and sinus tracts also may occur.1 The pathogenesis of AKN has been characterized as varying stages of follicular inflammation at the infundibular and isthmus levels followed by fibrotic occlusion of the ­follicular lumen.4 Pruritus, pain, bleeding, oozing, and a feeling of scalp tightness may occur.1,5

Umar et al6 performed a retrospective review of 108 men with AKN—58% of African descent, 37% Hispanic, 3% Asian, and 2% Middle Eastern—and proposed a 3-tier classification system for AKN. Tier 1 focused on the distribution and sagittal spread of AKN lesions between the clinical demarcation lines of the occipital notch and posterior hairline. Tier 2 focused on the type of lesions present—discrete papules or nodules, coalescing/abutting lesions, plaques (raised, atrophic, or indurated), or dome-shaped tumoral masses. Tier 3 focused on the presence or absence of co-existing dissecting cellulitis or folliculitis decalvans.6

Epidemiology

Acne keloidalis nuchae primarily manifests in adolescent and adult men of African or Afro-Caribbean descent.7 Among African American men, the prevalence of AKN ranges from 0.5% to 13.6%.8 Similar ranges have been reported among Nigerian, South African, and West African men.1 Acne keloidalis nuchae also affects Asian and Hispanic men but rarely is seen in non-Hispanic White men or in women of any ethnicity.9,10 The male to female ratio is 20:1.1,11 Hair texture, hairstyling practices such as closely shaved or faded haircuts, and genetics likely contribute to development of AKN. Sports and occupations that require the use of headgear or a tight collar may increase the risk for AKN.12

Key clinical features in people with darker skin tones

  • The lesions of AKN range in color from pink to dark brown or black. Postinflammatory hyperpigmentation or hyperchromia may be present around AKN lesions.
  • Chronicity of AKN may lead to extended use of high-potency topical or intralesional corticosteroids, which causes transient or long-lasting hypopigmentation, especially in those with darker skin tones.

Worth noting

  • Acne keloidalis nuchae can be disfiguring, which negatively impacts quality of life and self-esteem.12
  • Some occupations (eg, military, police) have hair policies that may not be favorable to those with or at risk for AKN.
  • Patients with AKN are 2 to 3 times more likely to present with metabolic syndrome, hypertension, type 2 diabetes mellitus, or obesity.13
 

 

Treatment

There are no treatments approved by the US Food and Drug Administration specifically for AKN. Treatment approaches are based on the pathophysiology, secondary impacts on the skin, and disease severity. Growing out the hair may prevent worsening and/or decrease the risk for new lesions.6

  • Options include but are not limited to topical and systemic therapies (eg, topical corticosteroids, oral or topical antibiotics, isotretinoin, topical retinoids, imiquimod, pimecrolimus), light devices (eg, phototherapy, laser), ablative therapies (eg, laser, cryotherapy, radiotherapy), and surgery (eg, excision, follicular unit excision), often in combination.6,14,15
  • Intralesional triamcinolone injections are considered standard of care. Adotama et al found that injecting ­triamcinolone into the deep dermis in the area of flat or papular AKN yielded better control of inflammation and decreased appearance of lesions compared with injecting individual lesions.16
  • For extensive AKN lesions that do not respond to ­less-invasive therapies, consider surgical techniques,6,17 such as follicular unit excision18 and more extensive surgical excisions building on approaches from pioneers Drs. John Kenney and Harold Pierce.19 An innovative surgical approach for removal of large AKNs is the bat excision technique—wound shape resembles a bat in a spread-eagled position—with secondary intention healing with or without debridement and/or tension sutures. The resulting linear scar acts as a new posterior hair line.20
 

 

Health disparity highlights

Access to a dermatologic or plastic surgeon with expertise in the surgical treatment of large AKNs may be challenging but is needed to reduce risk for recurrence and adverse events.

Close-cropped haircuts on the occipital scalp, which are particularly popular among men of African descent, increase the risk for AKN.5 Although this grooming style may be a personal preference, other hairstyles commonly worn by those with tightly coiled hair may be deemed “unprofessional” in society or the workplace, which leads to hairstyling practices that may increase the risk for AKN.21

Acne keloidalis nuchae remains an understudied entity that adversely affects patients with skin of color.

References
  1. Ogunbiyi A. Acne keloidalis nuchae: prevalence, impact, and management challenges. Clin Cosmet Investig Dermatol. 2016;9:483-489. doi:10.2147/CCID.S99225 
  2. Al Aboud DM, Badri T. Acne keloidalis nuchae. In: StatPearls [Internet]. Updated July 31, 2023. Accessed August 2, 2024. https://www.ncbi.nlm.nih.gov/books/NBK459135/
  3. Sperling LC, Homoky C, Pratt L, et al. Acne keloidalis is a form of primary scarring alopecia. Arch Dermatol. 2000;136:479-484.
  4. Herzberg AJ, Dinehart SM, Kerns BJ, et al. Acne keloidalis: transverse microscopy, immunohistochemistry, and electron microscopy. Am J Dermatopathol. 1990;12:109-121. doi:10.1097/00000372-199004000-00001
  5. Saka B, Akakpo A-S, Téclessou JN, et al. Risk factors associated with acne keloidalis nuchae in black subjects: a case-control study. Ann Dermatol Venereol. 2020;147:350-354. doi:10.1016/j.annder.2020.01.007
  6. Umar S, Lee DJ, Lullo JJ. A retrospective cohort study and clinical classification system of acne keloidalis nuchae. J Clin Aesthet Dermatol. 2021;14:E61-E67.
  7. Reja M, Silverberg NB. Acne keloidalis nuchae. In: Silverberg NB, Durán-McKinster C, Tay YK, eds. Pediatric Skin of Color. Springer; 2015:141-145. doi:10.1007/978-1-4614-6654-3_16
  8. Knable AL Jr, Hanke CW, Gonin R. Prevalence of acne keloidalis nuchae in football players. J Am Acad Dermatol. 1997;37:570-574. doi:10.1016/s0190-9622(97)70173-7
  9. Umar S, Ton D, Carter MJ, et al. Unveiling a shared precursor condition for acne keloidalis nuchae and primary cicatricial alopecias. Clin Cosmet Investig Dermatol. 2023;16:2315-2327. doi:10.2147/CCID.S422310
  10. Na K, Oh SH, Kim SK. Acne keloidalis nuchae in Asian: a single institutional experience. PLoS One. 2017;12:e0189790. doi:10.1371/journal.pone.0189790
  11. Ogunbiyi A, George A. Acne keloidalis in females: case report and review of literature. J Natl Med Assoc. 2005;97:736-738. 
  12. Alexis A, Heath CR, Halder RM. Folliculitis keloidalis nuchae and pseudofolliculitis barbae: are prevention and effective treatment within reach? Dermatol Clin. 2014;32:183-191. doi:10.1016/j.det.2013.12.001
  13. Kridin K, Solomon A, Tzur-Bitan D, et al. Acne keloidalis nuchae and the metabolic syndrome: a population-based study. Am J Clin Dermatol. 2020;21:733-739. doi:10.1007/s40257-020-00541-z
  14. Smart K, Rodriguez I, Worswick S. Comorbidities and treatment options for acne keloidalis nuchae. Dermatol Ther. Published online May 25, 2024. doi:10.1155/2024/8336926
  15. Callender VD, Young CM, Haverstock CL, et al. An open label study of clobetasol propionate 0.05% and betamethasone valerate 0.12% foams in the treatment of mild to moderate acne keloidalis. Cutis. 2005;75:317-321.
  16. Adotama P, Grullon K, Ali S, et al. How we do it: our method for triamcinolone injections of acne keloidalis nuchae. Dermatol Surg. 2023;49:713-714. doi:10.1097/DSS.0000000000003803
  17. Beckett N, Lawson C, Cohen G. Electrosurgical excision of acne keloidalis nuchae with secondary intention healing. J Clin Aesthet Dermatol. 2011;4:36-39.
  18. Esmat SM, Abdel Hay RM, Abu Zeid OM, et al. The efficacy of laser-assisted hair removal in the treatment of acne keloidalis nuchae; a pilot study. Eur J Dermatol. 2012;22:645-650. doi:10.1684/ejd.2012.1830
  19. Dillard AD, Quarles FN. African-American pioneers in dermatology. In: Taylor SC, Kelly AP, Lim HW, et al, eds. Dermatology for Skin of Color. 2nd ed. McGraw-Hill Education; 2016:717-730.
  20. Umar S, David CV, Castillo JR, et al. Innovative surgical approaches and selection criteria of large acne keloidalis nuchae lesions. Plast Reconstr Surg Glob Open. 2019;7:E2215. doi:10.1097/GOX.0000000000002215
  21. Lee MS, Nambudiri VE. The CROWN act and dermatology: taking a stand against race-based hair discrimination. J Am Acad Dermatol. 2021;84:1181-1182. doi:10.1016/j.jaad.2020.11.065
References
  1. Ogunbiyi A. Acne keloidalis nuchae: prevalence, impact, and management challenges. Clin Cosmet Investig Dermatol. 2016;9:483-489. doi:10.2147/CCID.S99225 
  2. Al Aboud DM, Badri T. Acne keloidalis nuchae. In: StatPearls [Internet]. Updated July 31, 2023. Accessed August 2, 2024. https://www.ncbi.nlm.nih.gov/books/NBK459135/
  3. Sperling LC, Homoky C, Pratt L, et al. Acne keloidalis is a form of primary scarring alopecia. Arch Dermatol. 2000;136:479-484.
  4. Herzberg AJ, Dinehart SM, Kerns BJ, et al. Acne keloidalis: transverse microscopy, immunohistochemistry, and electron microscopy. Am J Dermatopathol. 1990;12:109-121. doi:10.1097/00000372-199004000-00001
  5. Saka B, Akakpo A-S, Téclessou JN, et al. Risk factors associated with acne keloidalis nuchae in black subjects: a case-control study. Ann Dermatol Venereol. 2020;147:350-354. doi:10.1016/j.annder.2020.01.007
  6. Umar S, Lee DJ, Lullo JJ. A retrospective cohort study and clinical classification system of acne keloidalis nuchae. J Clin Aesthet Dermatol. 2021;14:E61-E67.
  7. Reja M, Silverberg NB. Acne keloidalis nuchae. In: Silverberg NB, Durán-McKinster C, Tay YK, eds. Pediatric Skin of Color. Springer; 2015:141-145. doi:10.1007/978-1-4614-6654-3_16
  8. Knable AL Jr, Hanke CW, Gonin R. Prevalence of acne keloidalis nuchae in football players. J Am Acad Dermatol. 1997;37:570-574. doi:10.1016/s0190-9622(97)70173-7
  9. Umar S, Ton D, Carter MJ, et al. Unveiling a shared precursor condition for acne keloidalis nuchae and primary cicatricial alopecias. Clin Cosmet Investig Dermatol. 2023;16:2315-2327. doi:10.2147/CCID.S422310
  10. Na K, Oh SH, Kim SK. Acne keloidalis nuchae in Asian: a single institutional experience. PLoS One. 2017;12:e0189790. doi:10.1371/journal.pone.0189790
  11. Ogunbiyi A, George A. Acne keloidalis in females: case report and review of literature. J Natl Med Assoc. 2005;97:736-738. 
  12. Alexis A, Heath CR, Halder RM. Folliculitis keloidalis nuchae and pseudofolliculitis barbae: are prevention and effective treatment within reach? Dermatol Clin. 2014;32:183-191. doi:10.1016/j.det.2013.12.001
  13. Kridin K, Solomon A, Tzur-Bitan D, et al. Acne keloidalis nuchae and the metabolic syndrome: a population-based study. Am J Clin Dermatol. 2020;21:733-739. doi:10.1007/s40257-020-00541-z
  14. Smart K, Rodriguez I, Worswick S. Comorbidities and treatment options for acne keloidalis nuchae. Dermatol Ther. Published online May 25, 2024. doi:10.1155/2024/8336926
  15. Callender VD, Young CM, Haverstock CL, et al. An open label study of clobetasol propionate 0.05% and betamethasone valerate 0.12% foams in the treatment of mild to moderate acne keloidalis. Cutis. 2005;75:317-321.
  16. Adotama P, Grullon K, Ali S, et al. How we do it: our method for triamcinolone injections of acne keloidalis nuchae. Dermatol Surg. 2023;49:713-714. doi:10.1097/DSS.0000000000003803
  17. Beckett N, Lawson C, Cohen G. Electrosurgical excision of acne keloidalis nuchae with secondary intention healing. J Clin Aesthet Dermatol. 2011;4:36-39.
  18. Esmat SM, Abdel Hay RM, Abu Zeid OM, et al. The efficacy of laser-assisted hair removal in the treatment of acne keloidalis nuchae; a pilot study. Eur J Dermatol. 2012;22:645-650. doi:10.1684/ejd.2012.1830
  19. Dillard AD, Quarles FN. African-American pioneers in dermatology. In: Taylor SC, Kelly AP, Lim HW, et al, eds. Dermatology for Skin of Color. 2nd ed. McGraw-Hill Education; 2016:717-730.
  20. Umar S, David CV, Castillo JR, et al. Innovative surgical approaches and selection criteria of large acne keloidalis nuchae lesions. Plast Reconstr Surg Glob Open. 2019;7:E2215. doi:10.1097/GOX.0000000000002215
  21. Lee MS, Nambudiri VE. The CROWN act and dermatology: taking a stand against race-based hair discrimination. J Am Acad Dermatol. 2021;84:1181-1182. doi:10.1016/j.jaad.2020.11.065
Issue
Federal Practitioner - 41(9)a
Issue
Federal Practitioner - 41(9)a
Page Number
312-313
Page Number
312-313
Publications
Publications
Topics
Article Type
Sections
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.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

Prurigo Nodularis Mechanisms and Current Management Options

Article Type
Changed
Tue, 09/03/2024 - 15:53
Display Headline
Prurigo Nodularis Mechanisms and Current Management Options

Prurigo nodularis (PN)(also called chronic nodular prurigo, prurigo nodularis of Hyde, or picker’s nodules) was first characterized by James Hyde in 1909.1-3 Prurigo nodularis manifests with symmetrical, intensely pruritic, eroded, or hyperkeratotic nodules or papules on the extremities and trunk.1,2,4,5 Studies have shown that individuals with PN experience pruritus, sleep loss, decreased social functioning from the appearance of the nodules, and a higher incidence of anxiety and depression, causing a negative impact on their quality of life.2,6 In addition, the manifestation of PN has been linked to neurologic and psychiatric disorders; however, PN also can be idiopathic and manifest without underlying illnesses.2,6,7

Prurigo nodularis has been associated with other dermatologic conditions such as atopic dermatitis (up to 50%), lichen planus, keratoacanthomas (KAs), and bullous pemphigoid.7-9 It also has been linked to systemic diseases in 38% to 50% of cases, including chronic kidney disease, liver disease, type 2 diabetes mellitus, malignancies (hematopoietic, liver, and skin), and HIV infection.6,8,10

The pathophysiology of PN is highly complex and has yet to be fully elucidated. It is thought to be due to dysregulation and interaction of the increase in neural and immunologic responses of proinflammatory and pruritogenic cytokines.2,11 Treatments aim to break the itch-scratch cycle that perpetuates this disorder; however, this proves difficult, as PN is associated with a higher itch intensity than atopic dermatitis and psoriasis.10 Therefore, most patients attempt multiple forms of treatment for PN, ranging from topical therapies, oral immunosuppressants, and phototherapy to the newest and only medication approved by the US Food and Drug Administration for the treatment of PN—dupilumab.1,7,11 Herein, we provide an updated review of PN with a focus on its epidemiology, histopathology and pathophysiology, comorbidities, clinical presentation, differential diagnosis, and current treatment options.

Epidemiology

There are few studies on the epidemiology of PN; however, middle-aged populations with underlying dermatologic or psychiatric disorders tend to be impacted most frequently.2,12,13 In 2016, it was estimated that almost 88,000 individuals had PN in the United States, with the majority being female; however, this estimate only took into account those aged 18 to 64 years and utilized data from IBM MarketScan Commercial Claims and Encounters Database (IBM Watson Health) from October 2015 to December 2016.14 More recently, a retrospective database analysis estimated the prevalence of PN in the United States to be anywhere from 36.7 to 43.9 cases per 100,000 individuals. However, this retrospective review utilized the International Classification of Diseases, Tenth Revision code; PN has 2 codes associated with the diagnosis, and the coding accuracy is unknown.15 Sutaria et al16 looked at racial disparities in patients with PN utilizing data from TriNetX and found that patients who received a diagnosis of PN were more likely to be women, non-Hispanic, and Black compared with control patients. However, these estimates are restricted to the health care organizations within this database.

In 2018, Poland reported an annual prevalence of 6.52 cases per 100,000 individuals,17 while England reported a yearly prevalence of 3.27 cases per 100,000 individuals.18 Both countries reported most cases were female. However, these studies are not without limitations. Poland only uses the primary diagnosis code for medical billing to simplify clinical coding, thus underestimating the actual prevalence; furthermore, clinical codes more often than not are assigned by someone other than the diagnosing physician, leaving room for error.17 In addition, England’s PN estimate utilized diagnosis data from primary care and inpatient datasets, leaving out outpatient datasets in which patients with PN may have been referred and obtained the diagnosis, potentially underestimating the prevalence in this population.18

In contrast, Korea estimated the annual prevalence of PN to be 4.82 cases per 1000 dermatology outpatients, with the majority being men, based on results from a cross-sectional study among outpatients from the Catholic Medical Center. Although this is the largest health organization in Korea, the scope of this study is limited and lacks data from other medical centers in Korea.19

Histopathology and Pathophysiology

Almost all cells in the skin are involved in PN: keratinocytes, mast cells, dendritic cells, endothelial cells, lymphocytes, eosinophils, collagen fibers, and nerve fibers.11,20 Classically, PN manifests as a dome-shaped lesion with hyperkeratosis, hypergranulosis, and psoriasiform epidermal hyperplasia with increased thickness of the papillary dermis consisting of coarse collagen with compact interstitial and circumvascular infiltration as well as increased lymphocytes and histocytes in the superficial dermis (Figure 1).20 Hyperkeratosis is thought to be due to either the alteration of keratinocyte structures from scratching or keratinocyte abnormalities triggering PN.21 However, the increase in keratinocytes, which secrete nerve growth factor, allows for neuronal hyperplasia within the dermis.22 Nerve growth factor can stimulate keratinocyte proliferation23 in addition to the upregulation of substance P (SP), a tachykinin that triggers vascular dilation and pruritus in the skin.24 The density of SP nerve fibers in the dermis increases in PN, causing proinflammatory effects, upregulating the immune response to promote endothelial hyperplasia and increased vascularization.25 The increase in these fibers may lead to pruritus associated with PN.2,26

FIGURE 1. A and B, Histopathology of prurigo nodularis lesions reveals hyperkeratosis, hypergranulosis, and psoriasiform hyperplasia with increased thickness of the papillary dermis and a superficial perivascular lymphohistiocytic infiltrate (H&E, original magnifications ×2 and ×10).

Many inflammatory cytokines and mediators also have been implicated in PN. Increased messenger RNA expression of IL-4, IL-17, IL-22, and IL-31 has been described in PN lesions.3,27 Furthermore, studies also have reported increased helper T cell (TH2) cytokines, including IL-4, IL-5, IL-10, and IL-13, in the dermis of PN lesions in patients without a history of atopy.3,28 These pruritogenic cytokines in conjunction with the SP fibers may create an intractable itch for those with PN. The interaction and culmination of the neural and immune responses make PN a complex condition to treat with the multifactorial interaction of systems. 

 

 

Comorbidities

Prurigo nodularis has been associated with a wide array of comorbidities; however, the direction of the relationship between PN and these conditions makes it difficult to discern if PN is a primary or secondary condition.29 Prurigo nodularis commonly has been connected to other inflammatory dermatoses, with a link to atopic dermatitis being the strongest.5,29 However, PN also has been linked to other pruritic inflammatory cutaneous disorders, including psoriasis, cutaneous T-cell lymphoma, lichen planus, and dermatitis herpetiformis.14,29

Huang et al14 found an increased likelihood of psychiatric illnesses in patients with PN, including eating disorders, nonsuicidal self-injury disorder, attention-deficit/hyperactivity disorder, schizophrenia, mood disorders, anxiety, and substance abuse disorders. Treatments directed at the neural aspect of PN have included selective serotonin reuptake inhibitors (SSRIs), which also are utilized to treat these mental health disorders.

Furthermore, systemic diseases also have been found to be associated with PN, including hypertension, type 2 diabetes mellitus, chronic kidney disease, heart failure, cerebrovascular disease, coronary heart disease, and chronic obstructive pulmonary disease.14 The relationship between PN and systemic conditions may be due to increased systemic inflammation and dysregulation of neural and metabolic functions implicated in these conditions from increased pruritic manifestations.29,30 However, studies also have connected PN to infectious conditions such as HIV. One study found that patients with PN had 2.68 higher odds of infection with HIV compared to age- and sex-matched controls.14 It is unknown if these conditions contributed to the development of PN or PN contributed to the development of these disorders.

Clinical Presentations

Prurigo nodularis is a chronic inflammatory skin disease that typically manifests with multiple severely pruritic, dome-shaped, firm, hyperpigmented papulonodules with central scale or crust, often with erosion, due to chronic repetitive scratching and picking secondary to pruritic systemic or dermatologic diseases or psychological disorders (Figure 2).1,2,4,5,8,31 Most often, diagnosis of PN is based on history and physical examination of the lesion; however, biopsies may be performed. These nodules commonly manifest with ulceration distributed symmetrically on extensor extremities in easy-to-reach places, sparing the mid back (called the butterfly sign).8 Lesions—either a few or hundreds—can range from a few millimeters to 2 to 3 cm.8,32 The lesions differ in appearance depending on the pigment in the patient’s skin. In patients with darker skin tones, hyperpigmented or hypopigmented papulonodules are not uncommon, while those with fairer skin tones tend to present with erythema.31

FIGURE 2. Prurigo nodularis lesions. A, Dome-shaped nodules with central ulceration on the right side of the trunk. B, Centrally ulcerated papulonodules distributed symmetrically on the chest. C, Domeshaped papulonodule with ulceration on the neck.

Differential Diagnosis

Because of the variation in manifestation of PN, these lesions may resemble other cutaneous conditions. If the lesions are hyperkeratotic, they can mimic hypertrophic lichen planus, which mainfests with hyperkeratotic plaques or nodules on the lower extremities.8,29 In addition, the histopathology of lichen planus resembles the appearance of PN, with epidermal hyperplasia, hypergranulosis, hyperkeratosis, and increased fibroblasts and capillaries.8,29

Pemphigoid nodularis is a rare subtype of bullous pemphigoid that exhibits characteristics of PN with pruritic plaques and erosions.8,29,33 The patient population for pemphigoid nodularis tends to be aged 50 to 60 years, and females are affected more frequently than males. However, pemphigoid nodularis may manifest with blistering and large plaques, which are not seen commonly with PN.29 On histopathology, pemphigoid nodularis deposits IgG and C3 on the basement membrane and has subepidermal clefting, unlike PN.7,29

Actinic prurigo manifests with pruritic papules or nodules post–UV exposure to unprotected skin.8,29,33 This rare condition usually manifests with cheilitis and conjunctivitis. Unlike PN, which commonly affects elderly populations, actinic prurigo typically is found in young females.8,29 Cytologic examination shows hyperkeratosis, spongiosis, and acanthosis of the epidermis with lymphocytic perivascular infiltration of the dermis.34

Neurotic excoriations also tend to mimic PN with raised excoriated lesions; however, this disorder is due to neurotic picking of the skin without associated pruritus or true hyperkeratosis.8,29,33 Histopathology shows epidermal crusting with inflammation of the upper dermis.35

Infiltrative cutaneous squamous cell carcinoma (SCC) may imitate PN in appearance. It manifests as tender, ulcerated, scaly plaques or nodules. Histopathology shows cytologic atypia with an infiltrative architectural pattern and presence of collections of compact keratin and parakeratin (called keratin pearls).

Keratoacanthomas can resemble PN lesions. They usually manifest as nodules measuring 1 to 2 cm in diameter and 0.5 cm thick, resembling crateriform tumors.36 On histopathology, KAs can resemble SCCs; however, KAs tend to manifest more frequently with a keratin-filled crater with a ground-glass appearance.36

Inverted follicular keratosis commonly manifests on the face in elderly men as a single, flesh-colored, verrucous papule that may resemble PN. However, cytology of inverted follicular keratosis is characterized by proliferation and squamous eddies.37 Consideration of the histologic findings and clinical appearance are important to differentiate between PN and cutaneous SCC.

Pseudoepitheliomatous hyperplasia is a benign condition that manifests as a plaque or nodule with crust, scale, or ulceration. Histologically, this condition presents with hyperplastic proliferation of the epidermis and adnexal epithelium.38 The clinical and histologic appearance can mimic PN and other cutaneous eruptions with epidermal hyperplasia. 

In clinical cases that are resistant to treatment, biopsy is the best approach to diagnose the lesion. Due to similarities in physical appearance and superficial histologic presentation of PN, KAs from SCC, hypertrophic lichen planus, and other hyperkeratotic lesions, the biopsy should be taken at the base of the lesion to sample deeper layers of skin to differentiate these dermatologic disorders.

 

 

Management

Current treatments for PN yield varied results. Many patients with moderate to severe PN attempt multiple therapies before seeing improvement.31 Treatments include topical, oral, and injectable medications and are either directed at the neural or immune components of PN due to the interplay between increased nerve fibers in the lesions (neural axis) as well as increases in cytokines and other immunologic mediators (immune axis) of this condition. However, the FDA recently approved the first treatment for PN—dupilumab—which is an injectable IL-4 receptor antagonist directed at the immunologic interactions affiliated with PN.

Immune-Mediated Topical Therapies—Immunologic topical therapies include corticosteroids, calcipotriol, and calcineurin inhibitors. Studies that have analyzed these treatments are limited to case reports and small intraindividual and randomized controlled trials (Table 1). Topical therapies usually are first-line agents for most patients. Adverse effects include transient irritation of the skin.40,42,43



Cryotherapy is another topical and immunologic therapy for those with PN; however, this treatment is more appropriate for patients with fewer lesions due to the pain that accompanies lesions treated with liquid nitrogen. In addition, this therapy can cause dyspigmentation of the skin in the treated areas.41

Similar to cryotherapy, intralesional corticosteroid injections are appropriate for patients with few PN lesions. A recent report described intralesional corticosteroid injections of 2.5 mg/mL for a PN nodule with high efficacy.46,47 This treatment has not undergone trials, but success with this modality has been documented, with adverse effects including hyperpigmentation or hypopigmentation in the treated area and transient pain.46

Neural-Mediated Topical Therapies—Neural topical therapies include capsaicin and neurokinin-1 receptor antagonists, aprepitant43 and serlopitant. These treatment studies are limited to small open-label and randomized controlled trials. Adverse effects of these treatments include transient cutaneous pain at the site of topical administration. In addition, neural-mediated topical therapies have shown either limited improvements from baseline or return of symptoms after treatment cessation.42,43

Supplements—N-acetyl cysteine is an over-the-counter supplement that has been reported to improve symptoms in patients with skin-picking disorders.48 The mechanism of action includes antioxidant effects such as decreasing reactive oxygen species, decreasing inflammatory markers, regulating neurotransmitters, and inhibiting hyperkeratosis.49 N-acetyl cysteine has been poorly studied for its application in PN. A small study of 3 patients with subacute PN receiving 1200 mg of oral N-acetyl cysteine reported varying levels of improvement in skin appearance and reduction in skin picking.50

Phototherapy—Phototherapy, a typical first- or second-line treatment modality for PN, targets both the neural- and immune-mediated aspects associated with pruritus in PN (Table 1).51 UV light can penetrate through the epidermal layer of the skin and reach the keratinocytes, which play a role in the immune-related response of PN. In addition, the cutaneous sensory nerves are located in the upper dermal layer, from which nerve fibers grow and penetrate into the epidermis, thereby interacting with the keratinocytes where pruritic signals are transmitted from the periphery up to the brain.51

Studies analyzing the effects of phototherapy on PN are limited to case series and a small randomized controlled trial. However, this trial has shown improvements in pruritus in the participants. Adverse effects include transient burning and erythema at the treated sites.44,45

Immune-Mediated Oral Therapies—Immunologic-targeted oral therapies include bilastine, methotrexate, and cyclosporine (Table 2).52,53 Bilastine efficacy was analyzed in a small phase 3, open-label, multicenter study in Japan; however, patients were allowed to use topical steroids in conjunction with the oral antihistamine.54 Methotrexate and cyclosporine are immunosuppressive medications and were analyzed in small retrospective studies. Both treatments yielded notable relief for patients; however, 38.5% (15/39) of patients receiving methotrexate experienced adverse events, and 50.0% (4/8) experienced adverse events with cyclosporine.52,53



Neural-Mediated Oral Therapies—Neural-targeted oral therapies include pregabalin, serlopitant, aprepitant, naltrexone, nalbuphine, SSRIs (paroxetine and fluvoxamine), amitriptyline, and thalidomide. The research on these treatments ranges from case reviews to randomized controlled trials and open-label trials (Table 2).55-63


Thalidomide was studied in a small retrospective case review that showed notable improvement in PN. Dosages of thalidomide varied, but on average the dose was 100 mg/d. However, greater than 50% of patients experienced at least 1 adverse effect with this treatment.63

A study performed in Italy showed promising results for patients treated with pregabalin, with 70.0% (21/30) continuing to take pregabalin for almost 2 years following completion of the initial 3-month trial.55 Naltrexone decreased pruritus in more than half of patients (9/17).59 Amitriptyline yielded improvements in patients with PN; however, disease recurred in 5 patients (29%) after 7 months.62 A study performed in Germany reported promising results for paroxetine and fluvoxamine; however, some patients enrolled in the study had some form of psychiatric disorder.61

Serlopitant, aprepitant, and nalbuphine were studied in randomized controlled trials. The serlopitant trials were the largest of the neurally mediated oral medication studies; one showed substantial improvement in patients with PN,56 while the most recent trial did not show significant improvement (ClinicalTrials.gov identifier NCT03546816).57 On the other hand, aprepitant showed no major difference between the experimental and placebo groups.58 Nalbuphine 162 mg twice daily showed greater improvement in PN than nalbuphine 81 mg twice daily.60

Immune-Mediated Injectable Therapies—Immune-targeted injectables include nemolizumab and dupilumab (Table 2). Nemolizumab is an IL-31 antagonist that has been studied in a small randomized controlled trial that showed great success in decreasing pruritus associated with PN.64 IL-31 has been implicated in PN, and inhibition of the IL-31 receptor has been shown to disrupt the itch-scratch cycle of PN. Dupilumab is a monoclonal antibody against the IL-4 and IL-13 receptors, and it is the only FDA-approved treatment for PN.65 Blockage of these protein receptors decreases type 2 inflammation and chronic pruritus.66,67 Dupilumab is FDA approved for the treatment of atopic dermatitis and recently was approved for adults with PN. Dupilumab acts to block the shared α-subunit of the pruritogenic cytokines IL-4 and IL-13 pathways,29 thereby breaking the itch-scratch cycle associated with PN and allowing for the healing of these lesions. Results from 2 clinical trials showed substantially reduced itch in patients with PN.65 Dupilumab also was approved by the European Medicines Agency for moderate to severe PN.68

Conclusion

Prurigo nodularis is a chronic condition that affects patient quality of life and can mimic various dermatologic conditions. The epidemiology and pathophysiology of PN have not been fully expounded. More research should be conducted to determine the underpinnings of PN to help identify more consistently effective therapies for this complex condition.

References
  1. Durmaz K, Ataseven A, Ozer I, et al. Prurigo nodularis responding to intravenous immunoglobulins. Przegl Dermatol. 2022;109:159-162. doi:10.5114/dr.2022.117988
  2. Kowalski EH, Kneiber D, Valdebran M, et al. Treatment-resistant prurigo nodularis: challenges and solutions. Clin Cosmet Investig Dermatol. 2019;12:163-172. doi:10.2147/CCID.S188070
  3. Wong LS, Yen YT. Chronic nodular prurigo: an update on the pathogenesis and treatment. Int J Mol Sci. 2022;23:12390. doi:10.3390/ijms232012390
  4. Janmohamed SR, Gwillim EC, Yousaf M, et al. The impact of prurigo nodularis on quality of life: a systematic review and meta-analysis. Arch Dermatol Res. 2021;313:669-677. doi:10.1007/s00403-020-02148-0
  5. Zeidler C, Ständer S. The pathogenesis of prurigo nodularis - ‘super-itch’ in exploration. Eur J Pain. 2016;20:37-40. doi:10.1002/ejp.767
  6. Kwatra SG. Breaking the itch–scratch cycle in prurigo nodularis. N Engl J Med. 2020;382:757-758. doi:10.1056/NEJMe1916733
  7. Frølunde AS, Wiis MAK, Ben Abdallah H, et al. Non-atopic chronic nodular prurigo (prurigo nodularis hyde): a systematic review of best-evidenced treatment options. Dermatology. 2022;238:950-960. doi:10.1159/000523700
  8. Kwon CD, Khanna R, Williams KA, et al. Diagnostic workup and evaluation of patients with prurigo nodularis. Medicines (Basel). 2019;6:97. doi:10.3390/medicines6040097
  9. Kowalski EH, Kneiber D, Valdebran M, et al. Distinguishing truly recalcitrant prurigo nodularis from poor treatment adherence: a response to treatment-resistant prurigo nodularis [Response to letter]. Clin Cosmet Investig Dermatol. 2019;12:371-372. doi:10.2147/CCID.S214195
  10. Whang KA, Le TK, Khanna R, et al. Health-related quality of life and economic burden of prurigo nodularis. J Am Acad Dermatol. 2022;86:573-580. doi:10.1016/j.jaad.2021.05.036
  11. Labib A, Ju T, Vander Does A, et al. Immunotargets and therapy for prurigo nodularis. Immunotargets Ther. 2022;11:11-21. doi:10.2147/ITT.S316602
  12. Belzberg M, Alphonse MP, Brown I, et al. Prurigo nodularis is characterized by systemic and cutaneous T helper 22 immune polarization. J Invest Dermatol. 2021;141:2208-2218.e14. doi:10.1016/j.jid.2021.02.749
  13. Ständer S, Pereira MP, Berger T, et al. IFSI-guideline on chronic prurigo including prurigo nodularis. Itch. 2020;5:e42. doi:10.1097/itx.0000000000000042
  14. Huang AH, Canner JK, Khanna R, et al. Real-world prevalence of prurigo nodularis and burden of associated diseases. J Invest Dermatol. 2020;140:480-483.e4. doi:10.1016/j.jid.2019.07.697
  15. Ständer S, Augustin M, Berger T, et al. Prevalence of prurigo nodularis in the United States of America: a retrospective database analysis. JAAD Int. 2021;2:28-30. doi:10.1016/j.jdin.2020.10.009
  16. Sutaria N, Adawi W, Brown I, et al. Racial disparities in mortality among patients with prurigo nodularis: a multi-center cohort study. J Am Acad Dermatol. 2022;86:487-490. doi:10.1016/j.jaad.2021.09.028
  17. Ryczek A, Reich A. Prevalence of prurigo nodularis in Poland. Acta Derm Venereol. 2020;100:adv00155. doi:10.2340/00015555-3518
  18. Morgan CL, Thomas M, Ständer S, et al. Epidemiology of prurigo nodularis in England: a retrospective database analysis. Br J Dermatol. 2022;187:188-195. doi:10.1111/bjd.21032
  19. Woo YR, Wang S, Sohn KA, et al. Epidemiology, comorbidities, and prescription patterns of Korean prurigo nodularis patients: a multi-institution study. J Clin Med Res. 2021;11:95. doi:10.3390/jcm11010095
  20. Weigelt N, Metze D, Ständer S. Prurigo nodularis: systematic analysis of 58 histological criteria in 136 patients. J Cutan Pathol. 2010;37:578-586. doi:10.1111/j.1600-0560.2009.01484.x
  21. Yang LL, Jiang B, Chen SH, et al. Abnormal keratin expression pattern in prurigo nodularis epidermis. Skin Health Dis. 2022;2:e75. doi:10.1002/ski2.75
  22. Nockher WA, Renz H. Neurotrophins in allergic diseases: from neuronal growth factors to intercellular signaling molecules. J Allergy Clin Immunol. 2006;117:583-589. doi:10.1016/j.jaci.2005.11.049
  23. Di Marco E, Mathor M, Bondanza S, et al. Nerve growth factor binds to normal human keratinocytes through high and low affinity receptors and stimulates their growth by a novel autocrine loop. J Biol Chem. 1993;268:22838-22846.
  24. Hägermark O, Hökfelt T, Pernow B. Flare and itch induced by substance P in human skin. J Invest Dermatol. 1978;71:233-235. doi:10.1111/1523-1747.ep12515092
  25. Choi JE, Di Nardo A. Skin neurogenic inflammation. Semin Immunopathol. 2018;40:249-259. doi:10.1007/s00281-018-0675-z
  26. Haas S, Capellino S, Phan NQ, et al. Low density of sympathetic nerve fibers relative to substance P-positive nerve fibers in lesional skin of chronic pruritus and prurigo nodularis. J Dermatol Sci. 2010;58:193-197. doi:10.1016/j.jdermsci.2010.03.020
  27. Park K, Mori T, Nakamura M, et al. Increased expression of mRNAs for IL-4, IL-17, IL-22 and IL-31 in skin lesions of subacute and chronic forms of prurigo. Eur J Dermatol. 2011;21:135-136.
  28. Tokura Y, Yagi H, Hanaoka K, et al. Subacute and chronic prurigo effectively treated with recombination interferon-gamma: implications for participation of Th2 cells in the pathogenesis of prurigo. Acta Derm Venereol. 1997;77:231-234. doi:10.2340/0001555577231234
  29. Williams KA, Roh YS, Brown I, et al. Pathophysiology, diagnosis, and pharmacological treatment of prurigo nodularis. Expert Rev Clin Pharmacol. 2021;14:67-77. doi:10.1080/17512433.2021.1852080
  30. Huang AH, Williams KA, Kwatra SG. Prurigo nodularis: epidemiology and clinical features. J Am Acad Dermatol. 2020;83:1559-1565. doi:10.1016/j.jaad.2020.04.183
  31. Bewley A, Homey B, Pink A. Prurigo nodularis: a review of IL-31RA blockade and other potential treatments. Dermatol Ther. 2022;12:2039-2048. doi:10.1007/s13555-022-00782-2
  32. Zeidler C, Yosipovitch G, Ständer S. Prurigo nodularis and its management. Dermatol Clin. 2018;36:189-197. doi:10.1016/j.det.2018.02.003
  33. Siepmann D, Lotts T, Blome C, et al. Evaluation of the antipruritic effects of topical pimecrolimus in non-atopic prurigo nodularis: results of a randomized, hydrocortisone-controlled, double-blind phase II trial. Dermatology. 2013;227:353-360. doi:10.1159/000355671
  34. Valbuena MC, Muvdi S, Lim HW. Actinic prurigo. Dermatol Clin. 2014;32:335-344, viii. doi:10.1016/j.det.2014.03.010
  35. Aldhahwani R, Al Hawsawi KA. Neurotic excoriation presenting as solitary papule: case report. J Dermatol Dermatolog Surg. 2022;26:45. doi:10.4103/jdds.jdds_59_21
  36. Kwiek B, Schwartz RA. Keratoacanthoma (KA): an update and review. J Am Acad Dermatol. 2016;74:1220-1233. doi:10.1016/j.jaad.2015.11.033
  37. Karadag AS, Ozlu E, Uzuncakmak TK, et al. Inverted follicular keratosis successfully treated with imiquimod. Indian Dermatol Online J. 2016;7:177-179. doi:10.4103/2229-5178.182354
  38. Nayak VN, Uma K, Girish HC, et al. Pseudoepitheliomatous hyperplasia in oral lesions: a review. J Int Oral Health. 2015;7:148-152.
  39. Saraceno R, Chiricozzi A, Nisticò SP, et al. An occlusive dressing containing betamethasone valerate 0.1% for the treatment of prurigo nodularis. J Dermatolog Treat. 2010;21:363-366. doi:10.3109/09546630903386606
  40. Wong SS, Goh CL. Double-blind, right/left comparison of calcipotriol ointment and betamethasone ointment in the treatment of prurigo nodularis. Arch Dermatol. 2000;136:807-808. doi:10.1001/archderm.136.6.807
  41. Waldinger TP, Wong RC, Taylor WB, et al. Cryotherapy improves prurigo nodularis. Arch Dermatol. 1984;120:1598-1600.
  42. Ständer S, Luger T, Metze D. Treatment of prurigo nodularis with topical capsaicin. J Am Acad Dermatol. 2001;44:471-478. doi:10.1067/mjd.2001.110059
  43. Ohanyan T, Schoepke N, Eirefelt S, et al. Role of substance P and its receptor neurokinin 1 in chronic prurigo: a randomized, proof-of-concept, controlled trial with topical aprepitant. Acta Derm Venereol. 2018;98:26-31. doi:10.2340/00015555-2780
  44. Tamagawa-Mineoka R, Katoh N, Ueda E, et al. Narrow-band ultraviolet B phototherapy in patients with recalcitrant nodular prurigo. J Dermatol. 2007;34:691-695. doi:10.1111/j.1346-8138.2007.00360.x
  45. Hammes S, Hermann J, Roos S, et al. UVB 308-nm excimer light and bath PUVA: combination therapy is very effective in the treatment of prurigo nodularis. J Eur Acad Dermatol Venereol. 2011;25:799-803. doi:10.1111/j.1468-3083.2010.03865.x
  46. Richards RN. Update on intralesional steroid: focus on dermatoses. J Cutan Med Surg. 2010;14:19-23. doi:10.2310/7750.2009.08082
  47. Elmariah S, Kim B, Berger T, et al. Practical approaches for diagnosis and management of prurigo nodularis: United States expert panel consensus. J Am Acad Dermatol. 2021;84:747-760. doi:10.1016/j.jaad.2020.07.025
  48. Grant JE, Chamberlain SR, Redden SA, et al. N-Acetylcysteine in the treatment of excoriation disorder: a randomized clinical trial. JAMA Psychiatry. 2016;73:490-496. doi:10.1001/jamapsychiatry.2016.0060
  49. Adil M, Amin SS, Mohtashim M. N-acetylcysteine in dermatology. Indian J Dermatol Venereol Leprol. 2018;84:652-659. doi: 10.4103/ijdvl.IJDVL_33_18.
  50. Taylor M, Bhagwandas K. Trichotillosis, skin picking and N-acetylcysteine. J Am Acad Dermatol. 2015;72(suppl 1):AB117. https://doi.org/10.1016/j.jaad.2015.02.482
  51. Legat FJ. The antipruritic effect of phototherapy. Front Med (Lausanne). 2018;5:333. doi:10.3389/fmed.2018.00333
  52. Klejtman T, Beylot-Barry M, Joly P, et al. Treatment of prurigo with methotrexate: a multicentre retrospective study of 39 cases. J Eur Acad Dermatol Venereol. 2018;32:437-440. doi:10.1111/jdv.14646
  53. Wiznia LE, Callahan SW, Cohen DE, et al. Rapid improvement of prurigo nodularis with cyclosporine treatment. J Am Acad Dermatol. 2018;78:1209-1211. doi:10.1016/j.jaad.2018.02.024
  54. Yagami A, Furue M, Togawa M, et al. One-year safety and efficacy study of bilastine treatment in Japanese patients with chronic spontaneous urticaria or pruritus associated with skin diseases. J Dermatol. 2017;44:375-385. doi:10.1111/1346-8138.13644
  55. Mazza M, Guerriero G, Marano G, et al. Treatment of prurigo nodularis with pregabalin. J Clin Pharm Ther. 2013;38:16-18. doi:10.1111/jcpt.12005
  56. Ständer S, Kwon P, Hirman J, et al. Serlopitant reduced pruritus in patients with prurigo nodularis in a phase 2, randomized, placebo-controlled trial. J Am Acad Dermatol. 2019;80:1395-1402. doi:10.1016/j.jaad.2019.01.052
  57. Study of the efficacy, safety and tolerability of serlopitant for the treatment of pruritus (itch) with prurigo nodularis. ClinicalTrials.gov identifier: NCT03546816. Updated May 20, 2021. Accessed August 8, 2024. https://clinicaltrials.gov/study/NCT03546816
  58. Tsianakas A, Zeidler C, Riepe C, et al. Aprepitant in anti-histamine-refractory chronic nodular prurigo: a multicentre, randomized, double-blind, placebo-controlled, cross-over, phase-II trial (APREPRU). Acta Derm Venereol. 2019;99:379-385. doi:10.2340/00015555-3120
  59. Metze D, Reimann S, Beissert S, et al. Efficacy and safety of naltrexone, an oral opiate receptor antagonist, in the treatment of pruritus in internal and dermatological diseases. J Am Acad Dermatol. 1999;41:533-539.
  60. Weisshaar E, Szepietowski JC, Bernhard JD, et al. Efficacy and safety of oral nalbuphine extended release in prurigo nodularis: results of a phase 2 randomized controlled trial with an open‐label extension phase. J Eur Acad Dermatol Venereol. 2022;36:453-461. doi:10.1111/jdv.17816
  61. Ständer S, Böckenholt B, Schürmeyer-Horst F, et al. Treatment of chronic pruritus with the selective serotonin re-uptake inhibitors paroxetine and fluvoxamine: results of an open-labelled, two-arm proof-of-concept study. Acta Derm Venereol. 2009;89:45-51. doi:10.2340/00015555-0553
  62. Zalaudek I, Petrillo G, Baldassarre MA, et al. Amitriptyline as therapeutic and not symptomatic approach in the treatment of prurigo nodularis. G Ital Dermatol Venereol. 2006;141:433-437.
  63. Andersen TP, Fogh K. Thalidomide in 42 patients with prurigo nodularis Hyde. Dermatology. 2011;223:107-112. doi:10.1159/000331577
  64. Ständer S, Yosipovitch G, Legat FJ, et al. Trial of nemolizumab in moderate-to-severe prurigo nodularis. N Engl J Med. 2020;382:706-716. doi:10.1056/NEJMoa1908316
  65. Yosipovitch G, Mollanazar N, Ständer S, et al. Dupilumab in patients with prurigo nodularis: two randomized, double-blind, placebo-controlled phase 3 trials. Nat Med. 2023;29:1180-1190. doi:10.1038/s41591-023-02320-9
  66. Mastorino L, Rosset F, Gelato F, et al. Chronic pruritus in atopic patients treated with dupilumab: real life response and related parameters in 354 patients. Pharmaceuticals (Basel). 2022;15:883. doi: 10.3390/ph15070883
  67. Kishi R, Toyama S, Tominaga M, et al. Effects of dupilumab on itch-related events in atopic dermatitis: implications for assessing treatment efficacy in clinical practice. Cells. 2023;12:239. doi: 10.3390/cells12020239
  68. Dupixent. European Medicines Agency website. Updated July 15, 2024. Accessed August 27, 2024. https://www.ema.europa.eu/en/medicines/human/EPAR/dupixent
Article PDF
Author and Disclosure Information

Taylor A. Brown is from the Washington State University Elson S. Floyd College of Medicine, Spokane. Dr. Khachemoune is from the Department of Dermatology, Brooklyn VA Medical Center, New York, and SUNY Downstate Dermatology Service, Brooklyn.

The authors report no conflict of interest.

Correspondence: Amor Khachemoune, MD, Brooklyn VA Medical Center, 800 Poly Place, Brooklyn, NY 11209 (amorkh@gmail.com).

Cutis. 2024 August;114(2):E43-E52. doi:10.12788/cutis.1085

Issue
Cutis - 114(2)
Publications
Topics
Page Number
E43-E52
Sections
Author and Disclosure Information

Taylor A. Brown is from the Washington State University Elson S. Floyd College of Medicine, Spokane. Dr. Khachemoune is from the Department of Dermatology, Brooklyn VA Medical Center, New York, and SUNY Downstate Dermatology Service, Brooklyn.

The authors report no conflict of interest.

Correspondence: Amor Khachemoune, MD, Brooklyn VA Medical Center, 800 Poly Place, Brooklyn, NY 11209 (amorkh@gmail.com).

Cutis. 2024 August;114(2):E43-E52. doi:10.12788/cutis.1085

Author and Disclosure Information

Taylor A. Brown is from the Washington State University Elson S. Floyd College of Medicine, Spokane. Dr. Khachemoune is from the Department of Dermatology, Brooklyn VA Medical Center, New York, and SUNY Downstate Dermatology Service, Brooklyn.

The authors report no conflict of interest.

Correspondence: Amor Khachemoune, MD, Brooklyn VA Medical Center, 800 Poly Place, Brooklyn, NY 11209 (amorkh@gmail.com).

Cutis. 2024 August;114(2):E43-E52. doi:10.12788/cutis.1085

Article PDF
Article PDF

Prurigo nodularis (PN)(also called chronic nodular prurigo, prurigo nodularis of Hyde, or picker’s nodules) was first characterized by James Hyde in 1909.1-3 Prurigo nodularis manifests with symmetrical, intensely pruritic, eroded, or hyperkeratotic nodules or papules on the extremities and trunk.1,2,4,5 Studies have shown that individuals with PN experience pruritus, sleep loss, decreased social functioning from the appearance of the nodules, and a higher incidence of anxiety and depression, causing a negative impact on their quality of life.2,6 In addition, the manifestation of PN has been linked to neurologic and psychiatric disorders; however, PN also can be idiopathic and manifest without underlying illnesses.2,6,7

Prurigo nodularis has been associated with other dermatologic conditions such as atopic dermatitis (up to 50%), lichen planus, keratoacanthomas (KAs), and bullous pemphigoid.7-9 It also has been linked to systemic diseases in 38% to 50% of cases, including chronic kidney disease, liver disease, type 2 diabetes mellitus, malignancies (hematopoietic, liver, and skin), and HIV infection.6,8,10

The pathophysiology of PN is highly complex and has yet to be fully elucidated. It is thought to be due to dysregulation and interaction of the increase in neural and immunologic responses of proinflammatory and pruritogenic cytokines.2,11 Treatments aim to break the itch-scratch cycle that perpetuates this disorder; however, this proves difficult, as PN is associated with a higher itch intensity than atopic dermatitis and psoriasis.10 Therefore, most patients attempt multiple forms of treatment for PN, ranging from topical therapies, oral immunosuppressants, and phototherapy to the newest and only medication approved by the US Food and Drug Administration for the treatment of PN—dupilumab.1,7,11 Herein, we provide an updated review of PN with a focus on its epidemiology, histopathology and pathophysiology, comorbidities, clinical presentation, differential diagnosis, and current treatment options.

Epidemiology

There are few studies on the epidemiology of PN; however, middle-aged populations with underlying dermatologic or psychiatric disorders tend to be impacted most frequently.2,12,13 In 2016, it was estimated that almost 88,000 individuals had PN in the United States, with the majority being female; however, this estimate only took into account those aged 18 to 64 years and utilized data from IBM MarketScan Commercial Claims and Encounters Database (IBM Watson Health) from October 2015 to December 2016.14 More recently, a retrospective database analysis estimated the prevalence of PN in the United States to be anywhere from 36.7 to 43.9 cases per 100,000 individuals. However, this retrospective review utilized the International Classification of Diseases, Tenth Revision code; PN has 2 codes associated with the diagnosis, and the coding accuracy is unknown.15 Sutaria et al16 looked at racial disparities in patients with PN utilizing data from TriNetX and found that patients who received a diagnosis of PN were more likely to be women, non-Hispanic, and Black compared with control patients. However, these estimates are restricted to the health care organizations within this database.

In 2018, Poland reported an annual prevalence of 6.52 cases per 100,000 individuals,17 while England reported a yearly prevalence of 3.27 cases per 100,000 individuals.18 Both countries reported most cases were female. However, these studies are not without limitations. Poland only uses the primary diagnosis code for medical billing to simplify clinical coding, thus underestimating the actual prevalence; furthermore, clinical codes more often than not are assigned by someone other than the diagnosing physician, leaving room for error.17 In addition, England’s PN estimate utilized diagnosis data from primary care and inpatient datasets, leaving out outpatient datasets in which patients with PN may have been referred and obtained the diagnosis, potentially underestimating the prevalence in this population.18

In contrast, Korea estimated the annual prevalence of PN to be 4.82 cases per 1000 dermatology outpatients, with the majority being men, based on results from a cross-sectional study among outpatients from the Catholic Medical Center. Although this is the largest health organization in Korea, the scope of this study is limited and lacks data from other medical centers in Korea.19

Histopathology and Pathophysiology

Almost all cells in the skin are involved in PN: keratinocytes, mast cells, dendritic cells, endothelial cells, lymphocytes, eosinophils, collagen fibers, and nerve fibers.11,20 Classically, PN manifests as a dome-shaped lesion with hyperkeratosis, hypergranulosis, and psoriasiform epidermal hyperplasia with increased thickness of the papillary dermis consisting of coarse collagen with compact interstitial and circumvascular infiltration as well as increased lymphocytes and histocytes in the superficial dermis (Figure 1).20 Hyperkeratosis is thought to be due to either the alteration of keratinocyte structures from scratching or keratinocyte abnormalities triggering PN.21 However, the increase in keratinocytes, which secrete nerve growth factor, allows for neuronal hyperplasia within the dermis.22 Nerve growth factor can stimulate keratinocyte proliferation23 in addition to the upregulation of substance P (SP), a tachykinin that triggers vascular dilation and pruritus in the skin.24 The density of SP nerve fibers in the dermis increases in PN, causing proinflammatory effects, upregulating the immune response to promote endothelial hyperplasia and increased vascularization.25 The increase in these fibers may lead to pruritus associated with PN.2,26

FIGURE 1. A and B, Histopathology of prurigo nodularis lesions reveals hyperkeratosis, hypergranulosis, and psoriasiform hyperplasia with increased thickness of the papillary dermis and a superficial perivascular lymphohistiocytic infiltrate (H&E, original magnifications ×2 and ×10).

Many inflammatory cytokines and mediators also have been implicated in PN. Increased messenger RNA expression of IL-4, IL-17, IL-22, and IL-31 has been described in PN lesions.3,27 Furthermore, studies also have reported increased helper T cell (TH2) cytokines, including IL-4, IL-5, IL-10, and IL-13, in the dermis of PN lesions in patients without a history of atopy.3,28 These pruritogenic cytokines in conjunction with the SP fibers may create an intractable itch for those with PN. The interaction and culmination of the neural and immune responses make PN a complex condition to treat with the multifactorial interaction of systems. 

 

 

Comorbidities

Prurigo nodularis has been associated with a wide array of comorbidities; however, the direction of the relationship between PN and these conditions makes it difficult to discern if PN is a primary or secondary condition.29 Prurigo nodularis commonly has been connected to other inflammatory dermatoses, with a link to atopic dermatitis being the strongest.5,29 However, PN also has been linked to other pruritic inflammatory cutaneous disorders, including psoriasis, cutaneous T-cell lymphoma, lichen planus, and dermatitis herpetiformis.14,29

Huang et al14 found an increased likelihood of psychiatric illnesses in patients with PN, including eating disorders, nonsuicidal self-injury disorder, attention-deficit/hyperactivity disorder, schizophrenia, mood disorders, anxiety, and substance abuse disorders. Treatments directed at the neural aspect of PN have included selective serotonin reuptake inhibitors (SSRIs), which also are utilized to treat these mental health disorders.

Furthermore, systemic diseases also have been found to be associated with PN, including hypertension, type 2 diabetes mellitus, chronic kidney disease, heart failure, cerebrovascular disease, coronary heart disease, and chronic obstructive pulmonary disease.14 The relationship between PN and systemic conditions may be due to increased systemic inflammation and dysregulation of neural and metabolic functions implicated in these conditions from increased pruritic manifestations.29,30 However, studies also have connected PN to infectious conditions such as HIV. One study found that patients with PN had 2.68 higher odds of infection with HIV compared to age- and sex-matched controls.14 It is unknown if these conditions contributed to the development of PN or PN contributed to the development of these disorders.

Clinical Presentations

Prurigo nodularis is a chronic inflammatory skin disease that typically manifests with multiple severely pruritic, dome-shaped, firm, hyperpigmented papulonodules with central scale or crust, often with erosion, due to chronic repetitive scratching and picking secondary to pruritic systemic or dermatologic diseases or psychological disorders (Figure 2).1,2,4,5,8,31 Most often, diagnosis of PN is based on history and physical examination of the lesion; however, biopsies may be performed. These nodules commonly manifest with ulceration distributed symmetrically on extensor extremities in easy-to-reach places, sparing the mid back (called the butterfly sign).8 Lesions—either a few or hundreds—can range from a few millimeters to 2 to 3 cm.8,32 The lesions differ in appearance depending on the pigment in the patient’s skin. In patients with darker skin tones, hyperpigmented or hypopigmented papulonodules are not uncommon, while those with fairer skin tones tend to present with erythema.31

FIGURE 2. Prurigo nodularis lesions. A, Dome-shaped nodules with central ulceration on the right side of the trunk. B, Centrally ulcerated papulonodules distributed symmetrically on the chest. C, Domeshaped papulonodule with ulceration on the neck.

Differential Diagnosis

Because of the variation in manifestation of PN, these lesions may resemble other cutaneous conditions. If the lesions are hyperkeratotic, they can mimic hypertrophic lichen planus, which mainfests with hyperkeratotic plaques or nodules on the lower extremities.8,29 In addition, the histopathology of lichen planus resembles the appearance of PN, with epidermal hyperplasia, hypergranulosis, hyperkeratosis, and increased fibroblasts and capillaries.8,29

Pemphigoid nodularis is a rare subtype of bullous pemphigoid that exhibits characteristics of PN with pruritic plaques and erosions.8,29,33 The patient population for pemphigoid nodularis tends to be aged 50 to 60 years, and females are affected more frequently than males. However, pemphigoid nodularis may manifest with blistering and large plaques, which are not seen commonly with PN.29 On histopathology, pemphigoid nodularis deposits IgG and C3 on the basement membrane and has subepidermal clefting, unlike PN.7,29

Actinic prurigo manifests with pruritic papules or nodules post–UV exposure to unprotected skin.8,29,33 This rare condition usually manifests with cheilitis and conjunctivitis. Unlike PN, which commonly affects elderly populations, actinic prurigo typically is found in young females.8,29 Cytologic examination shows hyperkeratosis, spongiosis, and acanthosis of the epidermis with lymphocytic perivascular infiltration of the dermis.34

Neurotic excoriations also tend to mimic PN with raised excoriated lesions; however, this disorder is due to neurotic picking of the skin without associated pruritus or true hyperkeratosis.8,29,33 Histopathology shows epidermal crusting with inflammation of the upper dermis.35

Infiltrative cutaneous squamous cell carcinoma (SCC) may imitate PN in appearance. It manifests as tender, ulcerated, scaly plaques or nodules. Histopathology shows cytologic atypia with an infiltrative architectural pattern and presence of collections of compact keratin and parakeratin (called keratin pearls).

Keratoacanthomas can resemble PN lesions. They usually manifest as nodules measuring 1 to 2 cm in diameter and 0.5 cm thick, resembling crateriform tumors.36 On histopathology, KAs can resemble SCCs; however, KAs tend to manifest more frequently with a keratin-filled crater with a ground-glass appearance.36

Inverted follicular keratosis commonly manifests on the face in elderly men as a single, flesh-colored, verrucous papule that may resemble PN. However, cytology of inverted follicular keratosis is characterized by proliferation and squamous eddies.37 Consideration of the histologic findings and clinical appearance are important to differentiate between PN and cutaneous SCC.

Pseudoepitheliomatous hyperplasia is a benign condition that manifests as a plaque or nodule with crust, scale, or ulceration. Histologically, this condition presents with hyperplastic proliferation of the epidermis and adnexal epithelium.38 The clinical and histologic appearance can mimic PN and other cutaneous eruptions with epidermal hyperplasia. 

In clinical cases that are resistant to treatment, biopsy is the best approach to diagnose the lesion. Due to similarities in physical appearance and superficial histologic presentation of PN, KAs from SCC, hypertrophic lichen planus, and other hyperkeratotic lesions, the biopsy should be taken at the base of the lesion to sample deeper layers of skin to differentiate these dermatologic disorders.

 

 

Management

Current treatments for PN yield varied results. Many patients with moderate to severe PN attempt multiple therapies before seeing improvement.31 Treatments include topical, oral, and injectable medications and are either directed at the neural or immune components of PN due to the interplay between increased nerve fibers in the lesions (neural axis) as well as increases in cytokines and other immunologic mediators (immune axis) of this condition. However, the FDA recently approved the first treatment for PN—dupilumab—which is an injectable IL-4 receptor antagonist directed at the immunologic interactions affiliated with PN.

Immune-Mediated Topical Therapies—Immunologic topical therapies include corticosteroids, calcipotriol, and calcineurin inhibitors. Studies that have analyzed these treatments are limited to case reports and small intraindividual and randomized controlled trials (Table 1). Topical therapies usually are first-line agents for most patients. Adverse effects include transient irritation of the skin.40,42,43



Cryotherapy is another topical and immunologic therapy for those with PN; however, this treatment is more appropriate for patients with fewer lesions due to the pain that accompanies lesions treated with liquid nitrogen. In addition, this therapy can cause dyspigmentation of the skin in the treated areas.41

Similar to cryotherapy, intralesional corticosteroid injections are appropriate for patients with few PN lesions. A recent report described intralesional corticosteroid injections of 2.5 mg/mL for a PN nodule with high efficacy.46,47 This treatment has not undergone trials, but success with this modality has been documented, with adverse effects including hyperpigmentation or hypopigmentation in the treated area and transient pain.46

Neural-Mediated Topical Therapies—Neural topical therapies include capsaicin and neurokinin-1 receptor antagonists, aprepitant43 and serlopitant. These treatment studies are limited to small open-label and randomized controlled trials. Adverse effects of these treatments include transient cutaneous pain at the site of topical administration. In addition, neural-mediated topical therapies have shown either limited improvements from baseline or return of symptoms after treatment cessation.42,43

Supplements—N-acetyl cysteine is an over-the-counter supplement that has been reported to improve symptoms in patients with skin-picking disorders.48 The mechanism of action includes antioxidant effects such as decreasing reactive oxygen species, decreasing inflammatory markers, regulating neurotransmitters, and inhibiting hyperkeratosis.49 N-acetyl cysteine has been poorly studied for its application in PN. A small study of 3 patients with subacute PN receiving 1200 mg of oral N-acetyl cysteine reported varying levels of improvement in skin appearance and reduction in skin picking.50

Phototherapy—Phototherapy, a typical first- or second-line treatment modality for PN, targets both the neural- and immune-mediated aspects associated with pruritus in PN (Table 1).51 UV light can penetrate through the epidermal layer of the skin and reach the keratinocytes, which play a role in the immune-related response of PN. In addition, the cutaneous sensory nerves are located in the upper dermal layer, from which nerve fibers grow and penetrate into the epidermis, thereby interacting with the keratinocytes where pruritic signals are transmitted from the periphery up to the brain.51

Studies analyzing the effects of phototherapy on PN are limited to case series and a small randomized controlled trial. However, this trial has shown improvements in pruritus in the participants. Adverse effects include transient burning and erythema at the treated sites.44,45

Immune-Mediated Oral Therapies—Immunologic-targeted oral therapies include bilastine, methotrexate, and cyclosporine (Table 2).52,53 Bilastine efficacy was analyzed in a small phase 3, open-label, multicenter study in Japan; however, patients were allowed to use topical steroids in conjunction with the oral antihistamine.54 Methotrexate and cyclosporine are immunosuppressive medications and were analyzed in small retrospective studies. Both treatments yielded notable relief for patients; however, 38.5% (15/39) of patients receiving methotrexate experienced adverse events, and 50.0% (4/8) experienced adverse events with cyclosporine.52,53



Neural-Mediated Oral Therapies—Neural-targeted oral therapies include pregabalin, serlopitant, aprepitant, naltrexone, nalbuphine, SSRIs (paroxetine and fluvoxamine), amitriptyline, and thalidomide. The research on these treatments ranges from case reviews to randomized controlled trials and open-label trials (Table 2).55-63


Thalidomide was studied in a small retrospective case review that showed notable improvement in PN. Dosages of thalidomide varied, but on average the dose was 100 mg/d. However, greater than 50% of patients experienced at least 1 adverse effect with this treatment.63

A study performed in Italy showed promising results for patients treated with pregabalin, with 70.0% (21/30) continuing to take pregabalin for almost 2 years following completion of the initial 3-month trial.55 Naltrexone decreased pruritus in more than half of patients (9/17).59 Amitriptyline yielded improvements in patients with PN; however, disease recurred in 5 patients (29%) after 7 months.62 A study performed in Germany reported promising results for paroxetine and fluvoxamine; however, some patients enrolled in the study had some form of psychiatric disorder.61

Serlopitant, aprepitant, and nalbuphine were studied in randomized controlled trials. The serlopitant trials were the largest of the neurally mediated oral medication studies; one showed substantial improvement in patients with PN,56 while the most recent trial did not show significant improvement (ClinicalTrials.gov identifier NCT03546816).57 On the other hand, aprepitant showed no major difference between the experimental and placebo groups.58 Nalbuphine 162 mg twice daily showed greater improvement in PN than nalbuphine 81 mg twice daily.60

Immune-Mediated Injectable Therapies—Immune-targeted injectables include nemolizumab and dupilumab (Table 2). Nemolizumab is an IL-31 antagonist that has been studied in a small randomized controlled trial that showed great success in decreasing pruritus associated with PN.64 IL-31 has been implicated in PN, and inhibition of the IL-31 receptor has been shown to disrupt the itch-scratch cycle of PN. Dupilumab is a monoclonal antibody against the IL-4 and IL-13 receptors, and it is the only FDA-approved treatment for PN.65 Blockage of these protein receptors decreases type 2 inflammation and chronic pruritus.66,67 Dupilumab is FDA approved for the treatment of atopic dermatitis and recently was approved for adults with PN. Dupilumab acts to block the shared α-subunit of the pruritogenic cytokines IL-4 and IL-13 pathways,29 thereby breaking the itch-scratch cycle associated with PN and allowing for the healing of these lesions. Results from 2 clinical trials showed substantially reduced itch in patients with PN.65 Dupilumab also was approved by the European Medicines Agency for moderate to severe PN.68

Conclusion

Prurigo nodularis is a chronic condition that affects patient quality of life and can mimic various dermatologic conditions. The epidemiology and pathophysiology of PN have not been fully expounded. More research should be conducted to determine the underpinnings of PN to help identify more consistently effective therapies for this complex condition.

Prurigo nodularis (PN)(also called chronic nodular prurigo, prurigo nodularis of Hyde, or picker’s nodules) was first characterized by James Hyde in 1909.1-3 Prurigo nodularis manifests with symmetrical, intensely pruritic, eroded, or hyperkeratotic nodules or papules on the extremities and trunk.1,2,4,5 Studies have shown that individuals with PN experience pruritus, sleep loss, decreased social functioning from the appearance of the nodules, and a higher incidence of anxiety and depression, causing a negative impact on their quality of life.2,6 In addition, the manifestation of PN has been linked to neurologic and psychiatric disorders; however, PN also can be idiopathic and manifest without underlying illnesses.2,6,7

Prurigo nodularis has been associated with other dermatologic conditions such as atopic dermatitis (up to 50%), lichen planus, keratoacanthomas (KAs), and bullous pemphigoid.7-9 It also has been linked to systemic diseases in 38% to 50% of cases, including chronic kidney disease, liver disease, type 2 diabetes mellitus, malignancies (hematopoietic, liver, and skin), and HIV infection.6,8,10

The pathophysiology of PN is highly complex and has yet to be fully elucidated. It is thought to be due to dysregulation and interaction of the increase in neural and immunologic responses of proinflammatory and pruritogenic cytokines.2,11 Treatments aim to break the itch-scratch cycle that perpetuates this disorder; however, this proves difficult, as PN is associated with a higher itch intensity than atopic dermatitis and psoriasis.10 Therefore, most patients attempt multiple forms of treatment for PN, ranging from topical therapies, oral immunosuppressants, and phototherapy to the newest and only medication approved by the US Food and Drug Administration for the treatment of PN—dupilumab.1,7,11 Herein, we provide an updated review of PN with a focus on its epidemiology, histopathology and pathophysiology, comorbidities, clinical presentation, differential diagnosis, and current treatment options.

Epidemiology

There are few studies on the epidemiology of PN; however, middle-aged populations with underlying dermatologic or psychiatric disorders tend to be impacted most frequently.2,12,13 In 2016, it was estimated that almost 88,000 individuals had PN in the United States, with the majority being female; however, this estimate only took into account those aged 18 to 64 years and utilized data from IBM MarketScan Commercial Claims and Encounters Database (IBM Watson Health) from October 2015 to December 2016.14 More recently, a retrospective database analysis estimated the prevalence of PN in the United States to be anywhere from 36.7 to 43.9 cases per 100,000 individuals. However, this retrospective review utilized the International Classification of Diseases, Tenth Revision code; PN has 2 codes associated with the diagnosis, and the coding accuracy is unknown.15 Sutaria et al16 looked at racial disparities in patients with PN utilizing data from TriNetX and found that patients who received a diagnosis of PN were more likely to be women, non-Hispanic, and Black compared with control patients. However, these estimates are restricted to the health care organizations within this database.

In 2018, Poland reported an annual prevalence of 6.52 cases per 100,000 individuals,17 while England reported a yearly prevalence of 3.27 cases per 100,000 individuals.18 Both countries reported most cases were female. However, these studies are not without limitations. Poland only uses the primary diagnosis code for medical billing to simplify clinical coding, thus underestimating the actual prevalence; furthermore, clinical codes more often than not are assigned by someone other than the diagnosing physician, leaving room for error.17 In addition, England’s PN estimate utilized diagnosis data from primary care and inpatient datasets, leaving out outpatient datasets in which patients with PN may have been referred and obtained the diagnosis, potentially underestimating the prevalence in this population.18

In contrast, Korea estimated the annual prevalence of PN to be 4.82 cases per 1000 dermatology outpatients, with the majority being men, based on results from a cross-sectional study among outpatients from the Catholic Medical Center. Although this is the largest health organization in Korea, the scope of this study is limited and lacks data from other medical centers in Korea.19

Histopathology and Pathophysiology

Almost all cells in the skin are involved in PN: keratinocytes, mast cells, dendritic cells, endothelial cells, lymphocytes, eosinophils, collagen fibers, and nerve fibers.11,20 Classically, PN manifests as a dome-shaped lesion with hyperkeratosis, hypergranulosis, and psoriasiform epidermal hyperplasia with increased thickness of the papillary dermis consisting of coarse collagen with compact interstitial and circumvascular infiltration as well as increased lymphocytes and histocytes in the superficial dermis (Figure 1).20 Hyperkeratosis is thought to be due to either the alteration of keratinocyte structures from scratching or keratinocyte abnormalities triggering PN.21 However, the increase in keratinocytes, which secrete nerve growth factor, allows for neuronal hyperplasia within the dermis.22 Nerve growth factor can stimulate keratinocyte proliferation23 in addition to the upregulation of substance P (SP), a tachykinin that triggers vascular dilation and pruritus in the skin.24 The density of SP nerve fibers in the dermis increases in PN, causing proinflammatory effects, upregulating the immune response to promote endothelial hyperplasia and increased vascularization.25 The increase in these fibers may lead to pruritus associated with PN.2,26

FIGURE 1. A and B, Histopathology of prurigo nodularis lesions reveals hyperkeratosis, hypergranulosis, and psoriasiform hyperplasia with increased thickness of the papillary dermis and a superficial perivascular lymphohistiocytic infiltrate (H&E, original magnifications ×2 and ×10).

Many inflammatory cytokines and mediators also have been implicated in PN. Increased messenger RNA expression of IL-4, IL-17, IL-22, and IL-31 has been described in PN lesions.3,27 Furthermore, studies also have reported increased helper T cell (TH2) cytokines, including IL-4, IL-5, IL-10, and IL-13, in the dermis of PN lesions in patients without a history of atopy.3,28 These pruritogenic cytokines in conjunction with the SP fibers may create an intractable itch for those with PN. The interaction and culmination of the neural and immune responses make PN a complex condition to treat with the multifactorial interaction of systems. 

 

 

Comorbidities

Prurigo nodularis has been associated with a wide array of comorbidities; however, the direction of the relationship between PN and these conditions makes it difficult to discern if PN is a primary or secondary condition.29 Prurigo nodularis commonly has been connected to other inflammatory dermatoses, with a link to atopic dermatitis being the strongest.5,29 However, PN also has been linked to other pruritic inflammatory cutaneous disorders, including psoriasis, cutaneous T-cell lymphoma, lichen planus, and dermatitis herpetiformis.14,29

Huang et al14 found an increased likelihood of psychiatric illnesses in patients with PN, including eating disorders, nonsuicidal self-injury disorder, attention-deficit/hyperactivity disorder, schizophrenia, mood disorders, anxiety, and substance abuse disorders. Treatments directed at the neural aspect of PN have included selective serotonin reuptake inhibitors (SSRIs), which also are utilized to treat these mental health disorders.

Furthermore, systemic diseases also have been found to be associated with PN, including hypertension, type 2 diabetes mellitus, chronic kidney disease, heart failure, cerebrovascular disease, coronary heart disease, and chronic obstructive pulmonary disease.14 The relationship between PN and systemic conditions may be due to increased systemic inflammation and dysregulation of neural and metabolic functions implicated in these conditions from increased pruritic manifestations.29,30 However, studies also have connected PN to infectious conditions such as HIV. One study found that patients with PN had 2.68 higher odds of infection with HIV compared to age- and sex-matched controls.14 It is unknown if these conditions contributed to the development of PN or PN contributed to the development of these disorders.

Clinical Presentations

Prurigo nodularis is a chronic inflammatory skin disease that typically manifests with multiple severely pruritic, dome-shaped, firm, hyperpigmented papulonodules with central scale or crust, often with erosion, due to chronic repetitive scratching and picking secondary to pruritic systemic or dermatologic diseases or psychological disorders (Figure 2).1,2,4,5,8,31 Most often, diagnosis of PN is based on history and physical examination of the lesion; however, biopsies may be performed. These nodules commonly manifest with ulceration distributed symmetrically on extensor extremities in easy-to-reach places, sparing the mid back (called the butterfly sign).8 Lesions—either a few or hundreds—can range from a few millimeters to 2 to 3 cm.8,32 The lesions differ in appearance depending on the pigment in the patient’s skin. In patients with darker skin tones, hyperpigmented or hypopigmented papulonodules are not uncommon, while those with fairer skin tones tend to present with erythema.31

FIGURE 2. Prurigo nodularis lesions. A, Dome-shaped nodules with central ulceration on the right side of the trunk. B, Centrally ulcerated papulonodules distributed symmetrically on the chest. C, Domeshaped papulonodule with ulceration on the neck.

Differential Diagnosis

Because of the variation in manifestation of PN, these lesions may resemble other cutaneous conditions. If the lesions are hyperkeratotic, they can mimic hypertrophic lichen planus, which mainfests with hyperkeratotic plaques or nodules on the lower extremities.8,29 In addition, the histopathology of lichen planus resembles the appearance of PN, with epidermal hyperplasia, hypergranulosis, hyperkeratosis, and increased fibroblasts and capillaries.8,29

Pemphigoid nodularis is a rare subtype of bullous pemphigoid that exhibits characteristics of PN with pruritic plaques and erosions.8,29,33 The patient population for pemphigoid nodularis tends to be aged 50 to 60 years, and females are affected more frequently than males. However, pemphigoid nodularis may manifest with blistering and large plaques, which are not seen commonly with PN.29 On histopathology, pemphigoid nodularis deposits IgG and C3 on the basement membrane and has subepidermal clefting, unlike PN.7,29

Actinic prurigo manifests with pruritic papules or nodules post–UV exposure to unprotected skin.8,29,33 This rare condition usually manifests with cheilitis and conjunctivitis. Unlike PN, which commonly affects elderly populations, actinic prurigo typically is found in young females.8,29 Cytologic examination shows hyperkeratosis, spongiosis, and acanthosis of the epidermis with lymphocytic perivascular infiltration of the dermis.34

Neurotic excoriations also tend to mimic PN with raised excoriated lesions; however, this disorder is due to neurotic picking of the skin without associated pruritus or true hyperkeratosis.8,29,33 Histopathology shows epidermal crusting with inflammation of the upper dermis.35

Infiltrative cutaneous squamous cell carcinoma (SCC) may imitate PN in appearance. It manifests as tender, ulcerated, scaly plaques or nodules. Histopathology shows cytologic atypia with an infiltrative architectural pattern and presence of collections of compact keratin and parakeratin (called keratin pearls).

Keratoacanthomas can resemble PN lesions. They usually manifest as nodules measuring 1 to 2 cm in diameter and 0.5 cm thick, resembling crateriform tumors.36 On histopathology, KAs can resemble SCCs; however, KAs tend to manifest more frequently with a keratin-filled crater with a ground-glass appearance.36

Inverted follicular keratosis commonly manifests on the face in elderly men as a single, flesh-colored, verrucous papule that may resemble PN. However, cytology of inverted follicular keratosis is characterized by proliferation and squamous eddies.37 Consideration of the histologic findings and clinical appearance are important to differentiate between PN and cutaneous SCC.

Pseudoepitheliomatous hyperplasia is a benign condition that manifests as a plaque or nodule with crust, scale, or ulceration. Histologically, this condition presents with hyperplastic proliferation of the epidermis and adnexal epithelium.38 The clinical and histologic appearance can mimic PN and other cutaneous eruptions with epidermal hyperplasia. 

In clinical cases that are resistant to treatment, biopsy is the best approach to diagnose the lesion. Due to similarities in physical appearance and superficial histologic presentation of PN, KAs from SCC, hypertrophic lichen planus, and other hyperkeratotic lesions, the biopsy should be taken at the base of the lesion to sample deeper layers of skin to differentiate these dermatologic disorders.

 

 

Management

Current treatments for PN yield varied results. Many patients with moderate to severe PN attempt multiple therapies before seeing improvement.31 Treatments include topical, oral, and injectable medications and are either directed at the neural or immune components of PN due to the interplay between increased nerve fibers in the lesions (neural axis) as well as increases in cytokines and other immunologic mediators (immune axis) of this condition. However, the FDA recently approved the first treatment for PN—dupilumab—which is an injectable IL-4 receptor antagonist directed at the immunologic interactions affiliated with PN.

Immune-Mediated Topical Therapies—Immunologic topical therapies include corticosteroids, calcipotriol, and calcineurin inhibitors. Studies that have analyzed these treatments are limited to case reports and small intraindividual and randomized controlled trials (Table 1). Topical therapies usually are first-line agents for most patients. Adverse effects include transient irritation of the skin.40,42,43



Cryotherapy is another topical and immunologic therapy for those with PN; however, this treatment is more appropriate for patients with fewer lesions due to the pain that accompanies lesions treated with liquid nitrogen. In addition, this therapy can cause dyspigmentation of the skin in the treated areas.41

Similar to cryotherapy, intralesional corticosteroid injections are appropriate for patients with few PN lesions. A recent report described intralesional corticosteroid injections of 2.5 mg/mL for a PN nodule with high efficacy.46,47 This treatment has not undergone trials, but success with this modality has been documented, with adverse effects including hyperpigmentation or hypopigmentation in the treated area and transient pain.46

Neural-Mediated Topical Therapies—Neural topical therapies include capsaicin and neurokinin-1 receptor antagonists, aprepitant43 and serlopitant. These treatment studies are limited to small open-label and randomized controlled trials. Adverse effects of these treatments include transient cutaneous pain at the site of topical administration. In addition, neural-mediated topical therapies have shown either limited improvements from baseline or return of symptoms after treatment cessation.42,43

Supplements—N-acetyl cysteine is an over-the-counter supplement that has been reported to improve symptoms in patients with skin-picking disorders.48 The mechanism of action includes antioxidant effects such as decreasing reactive oxygen species, decreasing inflammatory markers, regulating neurotransmitters, and inhibiting hyperkeratosis.49 N-acetyl cysteine has been poorly studied for its application in PN. A small study of 3 patients with subacute PN receiving 1200 mg of oral N-acetyl cysteine reported varying levels of improvement in skin appearance and reduction in skin picking.50

Phototherapy—Phototherapy, a typical first- or second-line treatment modality for PN, targets both the neural- and immune-mediated aspects associated with pruritus in PN (Table 1).51 UV light can penetrate through the epidermal layer of the skin and reach the keratinocytes, which play a role in the immune-related response of PN. In addition, the cutaneous sensory nerves are located in the upper dermal layer, from which nerve fibers grow and penetrate into the epidermis, thereby interacting with the keratinocytes where pruritic signals are transmitted from the periphery up to the brain.51

Studies analyzing the effects of phototherapy on PN are limited to case series and a small randomized controlled trial. However, this trial has shown improvements in pruritus in the participants. Adverse effects include transient burning and erythema at the treated sites.44,45

Immune-Mediated Oral Therapies—Immunologic-targeted oral therapies include bilastine, methotrexate, and cyclosporine (Table 2).52,53 Bilastine efficacy was analyzed in a small phase 3, open-label, multicenter study in Japan; however, patients were allowed to use topical steroids in conjunction with the oral antihistamine.54 Methotrexate and cyclosporine are immunosuppressive medications and were analyzed in small retrospective studies. Both treatments yielded notable relief for patients; however, 38.5% (15/39) of patients receiving methotrexate experienced adverse events, and 50.0% (4/8) experienced adverse events with cyclosporine.52,53



Neural-Mediated Oral Therapies—Neural-targeted oral therapies include pregabalin, serlopitant, aprepitant, naltrexone, nalbuphine, SSRIs (paroxetine and fluvoxamine), amitriptyline, and thalidomide. The research on these treatments ranges from case reviews to randomized controlled trials and open-label trials (Table 2).55-63


Thalidomide was studied in a small retrospective case review that showed notable improvement in PN. Dosages of thalidomide varied, but on average the dose was 100 mg/d. However, greater than 50% of patients experienced at least 1 adverse effect with this treatment.63

A study performed in Italy showed promising results for patients treated with pregabalin, with 70.0% (21/30) continuing to take pregabalin for almost 2 years following completion of the initial 3-month trial.55 Naltrexone decreased pruritus in more than half of patients (9/17).59 Amitriptyline yielded improvements in patients with PN; however, disease recurred in 5 patients (29%) after 7 months.62 A study performed in Germany reported promising results for paroxetine and fluvoxamine; however, some patients enrolled in the study had some form of psychiatric disorder.61

Serlopitant, aprepitant, and nalbuphine were studied in randomized controlled trials. The serlopitant trials were the largest of the neurally mediated oral medication studies; one showed substantial improvement in patients with PN,56 while the most recent trial did not show significant improvement (ClinicalTrials.gov identifier NCT03546816).57 On the other hand, aprepitant showed no major difference between the experimental and placebo groups.58 Nalbuphine 162 mg twice daily showed greater improvement in PN than nalbuphine 81 mg twice daily.60

Immune-Mediated Injectable Therapies—Immune-targeted injectables include nemolizumab and dupilumab (Table 2). Nemolizumab is an IL-31 antagonist that has been studied in a small randomized controlled trial that showed great success in decreasing pruritus associated with PN.64 IL-31 has been implicated in PN, and inhibition of the IL-31 receptor has been shown to disrupt the itch-scratch cycle of PN. Dupilumab is a monoclonal antibody against the IL-4 and IL-13 receptors, and it is the only FDA-approved treatment for PN.65 Blockage of these protein receptors decreases type 2 inflammation and chronic pruritus.66,67 Dupilumab is FDA approved for the treatment of atopic dermatitis and recently was approved for adults with PN. Dupilumab acts to block the shared α-subunit of the pruritogenic cytokines IL-4 and IL-13 pathways,29 thereby breaking the itch-scratch cycle associated with PN and allowing for the healing of these lesions. Results from 2 clinical trials showed substantially reduced itch in patients with PN.65 Dupilumab also was approved by the European Medicines Agency for moderate to severe PN.68

Conclusion

Prurigo nodularis is a chronic condition that affects patient quality of life and can mimic various dermatologic conditions. The epidemiology and pathophysiology of PN have not been fully expounded. More research should be conducted to determine the underpinnings of PN to help identify more consistently effective therapies for this complex condition.

References
  1. Durmaz K, Ataseven A, Ozer I, et al. Prurigo nodularis responding to intravenous immunoglobulins. Przegl Dermatol. 2022;109:159-162. doi:10.5114/dr.2022.117988
  2. Kowalski EH, Kneiber D, Valdebran M, et al. Treatment-resistant prurigo nodularis: challenges and solutions. Clin Cosmet Investig Dermatol. 2019;12:163-172. doi:10.2147/CCID.S188070
  3. Wong LS, Yen YT. Chronic nodular prurigo: an update on the pathogenesis and treatment. Int J Mol Sci. 2022;23:12390. doi:10.3390/ijms232012390
  4. Janmohamed SR, Gwillim EC, Yousaf M, et al. The impact of prurigo nodularis on quality of life: a systematic review and meta-analysis. Arch Dermatol Res. 2021;313:669-677. doi:10.1007/s00403-020-02148-0
  5. Zeidler C, Ständer S. The pathogenesis of prurigo nodularis - ‘super-itch’ in exploration. Eur J Pain. 2016;20:37-40. doi:10.1002/ejp.767
  6. Kwatra SG. Breaking the itch–scratch cycle in prurigo nodularis. N Engl J Med. 2020;382:757-758. doi:10.1056/NEJMe1916733
  7. Frølunde AS, Wiis MAK, Ben Abdallah H, et al. Non-atopic chronic nodular prurigo (prurigo nodularis hyde): a systematic review of best-evidenced treatment options. Dermatology. 2022;238:950-960. doi:10.1159/000523700
  8. Kwon CD, Khanna R, Williams KA, et al. Diagnostic workup and evaluation of patients with prurigo nodularis. Medicines (Basel). 2019;6:97. doi:10.3390/medicines6040097
  9. Kowalski EH, Kneiber D, Valdebran M, et al. Distinguishing truly recalcitrant prurigo nodularis from poor treatment adherence: a response to treatment-resistant prurigo nodularis [Response to letter]. Clin Cosmet Investig Dermatol. 2019;12:371-372. doi:10.2147/CCID.S214195
  10. Whang KA, Le TK, Khanna R, et al. Health-related quality of life and economic burden of prurigo nodularis. J Am Acad Dermatol. 2022;86:573-580. doi:10.1016/j.jaad.2021.05.036
  11. Labib A, Ju T, Vander Does A, et al. Immunotargets and therapy for prurigo nodularis. Immunotargets Ther. 2022;11:11-21. doi:10.2147/ITT.S316602
  12. Belzberg M, Alphonse MP, Brown I, et al. Prurigo nodularis is characterized by systemic and cutaneous T helper 22 immune polarization. J Invest Dermatol. 2021;141:2208-2218.e14. doi:10.1016/j.jid.2021.02.749
  13. Ständer S, Pereira MP, Berger T, et al. IFSI-guideline on chronic prurigo including prurigo nodularis. Itch. 2020;5:e42. doi:10.1097/itx.0000000000000042
  14. Huang AH, Canner JK, Khanna R, et al. Real-world prevalence of prurigo nodularis and burden of associated diseases. J Invest Dermatol. 2020;140:480-483.e4. doi:10.1016/j.jid.2019.07.697
  15. Ständer S, Augustin M, Berger T, et al. Prevalence of prurigo nodularis in the United States of America: a retrospective database analysis. JAAD Int. 2021;2:28-30. doi:10.1016/j.jdin.2020.10.009
  16. Sutaria N, Adawi W, Brown I, et al. Racial disparities in mortality among patients with prurigo nodularis: a multi-center cohort study. J Am Acad Dermatol. 2022;86:487-490. doi:10.1016/j.jaad.2021.09.028
  17. Ryczek A, Reich A. Prevalence of prurigo nodularis in Poland. Acta Derm Venereol. 2020;100:adv00155. doi:10.2340/00015555-3518
  18. Morgan CL, Thomas M, Ständer S, et al. Epidemiology of prurigo nodularis in England: a retrospective database analysis. Br J Dermatol. 2022;187:188-195. doi:10.1111/bjd.21032
  19. Woo YR, Wang S, Sohn KA, et al. Epidemiology, comorbidities, and prescription patterns of Korean prurigo nodularis patients: a multi-institution study. J Clin Med Res. 2021;11:95. doi:10.3390/jcm11010095
  20. Weigelt N, Metze D, Ständer S. Prurigo nodularis: systematic analysis of 58 histological criteria in 136 patients. J Cutan Pathol. 2010;37:578-586. doi:10.1111/j.1600-0560.2009.01484.x
  21. Yang LL, Jiang B, Chen SH, et al. Abnormal keratin expression pattern in prurigo nodularis epidermis. Skin Health Dis. 2022;2:e75. doi:10.1002/ski2.75
  22. Nockher WA, Renz H. Neurotrophins in allergic diseases: from neuronal growth factors to intercellular signaling molecules. J Allergy Clin Immunol. 2006;117:583-589. doi:10.1016/j.jaci.2005.11.049
  23. Di Marco E, Mathor M, Bondanza S, et al. Nerve growth factor binds to normal human keratinocytes through high and low affinity receptors and stimulates their growth by a novel autocrine loop. J Biol Chem. 1993;268:22838-22846.
  24. Hägermark O, Hökfelt T, Pernow B. Flare and itch induced by substance P in human skin. J Invest Dermatol. 1978;71:233-235. doi:10.1111/1523-1747.ep12515092
  25. Choi JE, Di Nardo A. Skin neurogenic inflammation. Semin Immunopathol. 2018;40:249-259. doi:10.1007/s00281-018-0675-z
  26. Haas S, Capellino S, Phan NQ, et al. Low density of sympathetic nerve fibers relative to substance P-positive nerve fibers in lesional skin of chronic pruritus and prurigo nodularis. J Dermatol Sci. 2010;58:193-197. doi:10.1016/j.jdermsci.2010.03.020
  27. Park K, Mori T, Nakamura M, et al. Increased expression of mRNAs for IL-4, IL-17, IL-22 and IL-31 in skin lesions of subacute and chronic forms of prurigo. Eur J Dermatol. 2011;21:135-136.
  28. Tokura Y, Yagi H, Hanaoka K, et al. Subacute and chronic prurigo effectively treated with recombination interferon-gamma: implications for participation of Th2 cells in the pathogenesis of prurigo. Acta Derm Venereol. 1997;77:231-234. doi:10.2340/0001555577231234
  29. Williams KA, Roh YS, Brown I, et al. Pathophysiology, diagnosis, and pharmacological treatment of prurigo nodularis. Expert Rev Clin Pharmacol. 2021;14:67-77. doi:10.1080/17512433.2021.1852080
  30. Huang AH, Williams KA, Kwatra SG. Prurigo nodularis: epidemiology and clinical features. J Am Acad Dermatol. 2020;83:1559-1565. doi:10.1016/j.jaad.2020.04.183
  31. Bewley A, Homey B, Pink A. Prurigo nodularis: a review of IL-31RA blockade and other potential treatments. Dermatol Ther. 2022;12:2039-2048. doi:10.1007/s13555-022-00782-2
  32. Zeidler C, Yosipovitch G, Ständer S. Prurigo nodularis and its management. Dermatol Clin. 2018;36:189-197. doi:10.1016/j.det.2018.02.003
  33. Siepmann D, Lotts T, Blome C, et al. Evaluation of the antipruritic effects of topical pimecrolimus in non-atopic prurigo nodularis: results of a randomized, hydrocortisone-controlled, double-blind phase II trial. Dermatology. 2013;227:353-360. doi:10.1159/000355671
  34. Valbuena MC, Muvdi S, Lim HW. Actinic prurigo. Dermatol Clin. 2014;32:335-344, viii. doi:10.1016/j.det.2014.03.010
  35. Aldhahwani R, Al Hawsawi KA. Neurotic excoriation presenting as solitary papule: case report. J Dermatol Dermatolog Surg. 2022;26:45. doi:10.4103/jdds.jdds_59_21
  36. Kwiek B, Schwartz RA. Keratoacanthoma (KA): an update and review. J Am Acad Dermatol. 2016;74:1220-1233. doi:10.1016/j.jaad.2015.11.033
  37. Karadag AS, Ozlu E, Uzuncakmak TK, et al. Inverted follicular keratosis successfully treated with imiquimod. Indian Dermatol Online J. 2016;7:177-179. doi:10.4103/2229-5178.182354
  38. Nayak VN, Uma K, Girish HC, et al. Pseudoepitheliomatous hyperplasia in oral lesions: a review. J Int Oral Health. 2015;7:148-152.
  39. Saraceno R, Chiricozzi A, Nisticò SP, et al. An occlusive dressing containing betamethasone valerate 0.1% for the treatment of prurigo nodularis. J Dermatolog Treat. 2010;21:363-366. doi:10.3109/09546630903386606
  40. Wong SS, Goh CL. Double-blind, right/left comparison of calcipotriol ointment and betamethasone ointment in the treatment of prurigo nodularis. Arch Dermatol. 2000;136:807-808. doi:10.1001/archderm.136.6.807
  41. Waldinger TP, Wong RC, Taylor WB, et al. Cryotherapy improves prurigo nodularis. Arch Dermatol. 1984;120:1598-1600.
  42. Ständer S, Luger T, Metze D. Treatment of prurigo nodularis with topical capsaicin. J Am Acad Dermatol. 2001;44:471-478. doi:10.1067/mjd.2001.110059
  43. Ohanyan T, Schoepke N, Eirefelt S, et al. Role of substance P and its receptor neurokinin 1 in chronic prurigo: a randomized, proof-of-concept, controlled trial with topical aprepitant. Acta Derm Venereol. 2018;98:26-31. doi:10.2340/00015555-2780
  44. Tamagawa-Mineoka R, Katoh N, Ueda E, et al. Narrow-band ultraviolet B phototherapy in patients with recalcitrant nodular prurigo. J Dermatol. 2007;34:691-695. doi:10.1111/j.1346-8138.2007.00360.x
  45. Hammes S, Hermann J, Roos S, et al. UVB 308-nm excimer light and bath PUVA: combination therapy is very effective in the treatment of prurigo nodularis. J Eur Acad Dermatol Venereol. 2011;25:799-803. doi:10.1111/j.1468-3083.2010.03865.x
  46. Richards RN. Update on intralesional steroid: focus on dermatoses. J Cutan Med Surg. 2010;14:19-23. doi:10.2310/7750.2009.08082
  47. Elmariah S, Kim B, Berger T, et al. Practical approaches for diagnosis and management of prurigo nodularis: United States expert panel consensus. J Am Acad Dermatol. 2021;84:747-760. doi:10.1016/j.jaad.2020.07.025
  48. Grant JE, Chamberlain SR, Redden SA, et al. N-Acetylcysteine in the treatment of excoriation disorder: a randomized clinical trial. JAMA Psychiatry. 2016;73:490-496. doi:10.1001/jamapsychiatry.2016.0060
  49. Adil M, Amin SS, Mohtashim M. N-acetylcysteine in dermatology. Indian J Dermatol Venereol Leprol. 2018;84:652-659. doi: 10.4103/ijdvl.IJDVL_33_18.
  50. Taylor M, Bhagwandas K. Trichotillosis, skin picking and N-acetylcysteine. J Am Acad Dermatol. 2015;72(suppl 1):AB117. https://doi.org/10.1016/j.jaad.2015.02.482
  51. Legat FJ. The antipruritic effect of phototherapy. Front Med (Lausanne). 2018;5:333. doi:10.3389/fmed.2018.00333
  52. Klejtman T, Beylot-Barry M, Joly P, et al. Treatment of prurigo with methotrexate: a multicentre retrospective study of 39 cases. J Eur Acad Dermatol Venereol. 2018;32:437-440. doi:10.1111/jdv.14646
  53. Wiznia LE, Callahan SW, Cohen DE, et al. Rapid improvement of prurigo nodularis with cyclosporine treatment. J Am Acad Dermatol. 2018;78:1209-1211. doi:10.1016/j.jaad.2018.02.024
  54. Yagami A, Furue M, Togawa M, et al. One-year safety and efficacy study of bilastine treatment in Japanese patients with chronic spontaneous urticaria or pruritus associated with skin diseases. J Dermatol. 2017;44:375-385. doi:10.1111/1346-8138.13644
  55. Mazza M, Guerriero G, Marano G, et al. Treatment of prurigo nodularis with pregabalin. J Clin Pharm Ther. 2013;38:16-18. doi:10.1111/jcpt.12005
  56. Ständer S, Kwon P, Hirman J, et al. Serlopitant reduced pruritus in patients with prurigo nodularis in a phase 2, randomized, placebo-controlled trial. J Am Acad Dermatol. 2019;80:1395-1402. doi:10.1016/j.jaad.2019.01.052
  57. Study of the efficacy, safety and tolerability of serlopitant for the treatment of pruritus (itch) with prurigo nodularis. ClinicalTrials.gov identifier: NCT03546816. Updated May 20, 2021. Accessed August 8, 2024. https://clinicaltrials.gov/study/NCT03546816
  58. Tsianakas A, Zeidler C, Riepe C, et al. Aprepitant in anti-histamine-refractory chronic nodular prurigo: a multicentre, randomized, double-blind, placebo-controlled, cross-over, phase-II trial (APREPRU). Acta Derm Venereol. 2019;99:379-385. doi:10.2340/00015555-3120
  59. Metze D, Reimann S, Beissert S, et al. Efficacy and safety of naltrexone, an oral opiate receptor antagonist, in the treatment of pruritus in internal and dermatological diseases. J Am Acad Dermatol. 1999;41:533-539.
  60. Weisshaar E, Szepietowski JC, Bernhard JD, et al. Efficacy and safety of oral nalbuphine extended release in prurigo nodularis: results of a phase 2 randomized controlled trial with an open‐label extension phase. J Eur Acad Dermatol Venereol. 2022;36:453-461. doi:10.1111/jdv.17816
  61. Ständer S, Böckenholt B, Schürmeyer-Horst F, et al. Treatment of chronic pruritus with the selective serotonin re-uptake inhibitors paroxetine and fluvoxamine: results of an open-labelled, two-arm proof-of-concept study. Acta Derm Venereol. 2009;89:45-51. doi:10.2340/00015555-0553
  62. Zalaudek I, Petrillo G, Baldassarre MA, et al. Amitriptyline as therapeutic and not symptomatic approach in the treatment of prurigo nodularis. G Ital Dermatol Venereol. 2006;141:433-437.
  63. Andersen TP, Fogh K. Thalidomide in 42 patients with prurigo nodularis Hyde. Dermatology. 2011;223:107-112. doi:10.1159/000331577
  64. Ständer S, Yosipovitch G, Legat FJ, et al. Trial of nemolizumab in moderate-to-severe prurigo nodularis. N Engl J Med. 2020;382:706-716. doi:10.1056/NEJMoa1908316
  65. Yosipovitch G, Mollanazar N, Ständer S, et al. Dupilumab in patients with prurigo nodularis: two randomized, double-blind, placebo-controlled phase 3 trials. Nat Med. 2023;29:1180-1190. doi:10.1038/s41591-023-02320-9
  66. Mastorino L, Rosset F, Gelato F, et al. Chronic pruritus in atopic patients treated with dupilumab: real life response and related parameters in 354 patients. Pharmaceuticals (Basel). 2022;15:883. doi: 10.3390/ph15070883
  67. Kishi R, Toyama S, Tominaga M, et al. Effects of dupilumab on itch-related events in atopic dermatitis: implications for assessing treatment efficacy in clinical practice. Cells. 2023;12:239. doi: 10.3390/cells12020239
  68. Dupixent. European Medicines Agency website. Updated July 15, 2024. Accessed August 27, 2024. https://www.ema.europa.eu/en/medicines/human/EPAR/dupixent
References
  1. Durmaz K, Ataseven A, Ozer I, et al. Prurigo nodularis responding to intravenous immunoglobulins. Przegl Dermatol. 2022;109:159-162. doi:10.5114/dr.2022.117988
  2. Kowalski EH, Kneiber D, Valdebran M, et al. Treatment-resistant prurigo nodularis: challenges and solutions. Clin Cosmet Investig Dermatol. 2019;12:163-172. doi:10.2147/CCID.S188070
  3. Wong LS, Yen YT. Chronic nodular prurigo: an update on the pathogenesis and treatment. Int J Mol Sci. 2022;23:12390. doi:10.3390/ijms232012390
  4. Janmohamed SR, Gwillim EC, Yousaf M, et al. The impact of prurigo nodularis on quality of life: a systematic review and meta-analysis. Arch Dermatol Res. 2021;313:669-677. doi:10.1007/s00403-020-02148-0
  5. Zeidler C, Ständer S. The pathogenesis of prurigo nodularis - ‘super-itch’ in exploration. Eur J Pain. 2016;20:37-40. doi:10.1002/ejp.767
  6. Kwatra SG. Breaking the itch–scratch cycle in prurigo nodularis. N Engl J Med. 2020;382:757-758. doi:10.1056/NEJMe1916733
  7. Frølunde AS, Wiis MAK, Ben Abdallah H, et al. Non-atopic chronic nodular prurigo (prurigo nodularis hyde): a systematic review of best-evidenced treatment options. Dermatology. 2022;238:950-960. doi:10.1159/000523700
  8. Kwon CD, Khanna R, Williams KA, et al. Diagnostic workup and evaluation of patients with prurigo nodularis. Medicines (Basel). 2019;6:97. doi:10.3390/medicines6040097
  9. Kowalski EH, Kneiber D, Valdebran M, et al. Distinguishing truly recalcitrant prurigo nodularis from poor treatment adherence: a response to treatment-resistant prurigo nodularis [Response to letter]. Clin Cosmet Investig Dermatol. 2019;12:371-372. doi:10.2147/CCID.S214195
  10. Whang KA, Le TK, Khanna R, et al. Health-related quality of life and economic burden of prurigo nodularis. J Am Acad Dermatol. 2022;86:573-580. doi:10.1016/j.jaad.2021.05.036
  11. Labib A, Ju T, Vander Does A, et al. Immunotargets and therapy for prurigo nodularis. Immunotargets Ther. 2022;11:11-21. doi:10.2147/ITT.S316602
  12. Belzberg M, Alphonse MP, Brown I, et al. Prurigo nodularis is characterized by systemic and cutaneous T helper 22 immune polarization. J Invest Dermatol. 2021;141:2208-2218.e14. doi:10.1016/j.jid.2021.02.749
  13. Ständer S, Pereira MP, Berger T, et al. IFSI-guideline on chronic prurigo including prurigo nodularis. Itch. 2020;5:e42. doi:10.1097/itx.0000000000000042
  14. Huang AH, Canner JK, Khanna R, et al. Real-world prevalence of prurigo nodularis and burden of associated diseases. J Invest Dermatol. 2020;140:480-483.e4. doi:10.1016/j.jid.2019.07.697
  15. Ständer S, Augustin M, Berger T, et al. Prevalence of prurigo nodularis in the United States of America: a retrospective database analysis. JAAD Int. 2021;2:28-30. doi:10.1016/j.jdin.2020.10.009
  16. Sutaria N, Adawi W, Brown I, et al. Racial disparities in mortality among patients with prurigo nodularis: a multi-center cohort study. J Am Acad Dermatol. 2022;86:487-490. doi:10.1016/j.jaad.2021.09.028
  17. Ryczek A, Reich A. Prevalence of prurigo nodularis in Poland. Acta Derm Venereol. 2020;100:adv00155. doi:10.2340/00015555-3518
  18. Morgan CL, Thomas M, Ständer S, et al. Epidemiology of prurigo nodularis in England: a retrospective database analysis. Br J Dermatol. 2022;187:188-195. doi:10.1111/bjd.21032
  19. Woo YR, Wang S, Sohn KA, et al. Epidemiology, comorbidities, and prescription patterns of Korean prurigo nodularis patients: a multi-institution study. J Clin Med Res. 2021;11:95. doi:10.3390/jcm11010095
  20. Weigelt N, Metze D, Ständer S. Prurigo nodularis: systematic analysis of 58 histological criteria in 136 patients. J Cutan Pathol. 2010;37:578-586. doi:10.1111/j.1600-0560.2009.01484.x
  21. Yang LL, Jiang B, Chen SH, et al. Abnormal keratin expression pattern in prurigo nodularis epidermis. Skin Health Dis. 2022;2:e75. doi:10.1002/ski2.75
  22. Nockher WA, Renz H. Neurotrophins in allergic diseases: from neuronal growth factors to intercellular signaling molecules. J Allergy Clin Immunol. 2006;117:583-589. doi:10.1016/j.jaci.2005.11.049
  23. Di Marco E, Mathor M, Bondanza S, et al. Nerve growth factor binds to normal human keratinocytes through high and low affinity receptors and stimulates their growth by a novel autocrine loop. J Biol Chem. 1993;268:22838-22846.
  24. Hägermark O, Hökfelt T, Pernow B. Flare and itch induced by substance P in human skin. J Invest Dermatol. 1978;71:233-235. doi:10.1111/1523-1747.ep12515092
  25. Choi JE, Di Nardo A. Skin neurogenic inflammation. Semin Immunopathol. 2018;40:249-259. doi:10.1007/s00281-018-0675-z
  26. Haas S, Capellino S, Phan NQ, et al. Low density of sympathetic nerve fibers relative to substance P-positive nerve fibers in lesional skin of chronic pruritus and prurigo nodularis. J Dermatol Sci. 2010;58:193-197. doi:10.1016/j.jdermsci.2010.03.020
  27. Park K, Mori T, Nakamura M, et al. Increased expression of mRNAs for IL-4, IL-17, IL-22 and IL-31 in skin lesions of subacute and chronic forms of prurigo. Eur J Dermatol. 2011;21:135-136.
  28. Tokura Y, Yagi H, Hanaoka K, et al. Subacute and chronic prurigo effectively treated with recombination interferon-gamma: implications for participation of Th2 cells in the pathogenesis of prurigo. Acta Derm Venereol. 1997;77:231-234. doi:10.2340/0001555577231234
  29. Williams KA, Roh YS, Brown I, et al. Pathophysiology, diagnosis, and pharmacological treatment of prurigo nodularis. Expert Rev Clin Pharmacol. 2021;14:67-77. doi:10.1080/17512433.2021.1852080
  30. Huang AH, Williams KA, Kwatra SG. Prurigo nodularis: epidemiology and clinical features. J Am Acad Dermatol. 2020;83:1559-1565. doi:10.1016/j.jaad.2020.04.183
  31. Bewley A, Homey B, Pink A. Prurigo nodularis: a review of IL-31RA blockade and other potential treatments. Dermatol Ther. 2022;12:2039-2048. doi:10.1007/s13555-022-00782-2
  32. Zeidler C, Yosipovitch G, Ständer S. Prurigo nodularis and its management. Dermatol Clin. 2018;36:189-197. doi:10.1016/j.det.2018.02.003
  33. Siepmann D, Lotts T, Blome C, et al. Evaluation of the antipruritic effects of topical pimecrolimus in non-atopic prurigo nodularis: results of a randomized, hydrocortisone-controlled, double-blind phase II trial. Dermatology. 2013;227:353-360. doi:10.1159/000355671
  34. Valbuena MC, Muvdi S, Lim HW. Actinic prurigo. Dermatol Clin. 2014;32:335-344, viii. doi:10.1016/j.det.2014.03.010
  35. Aldhahwani R, Al Hawsawi KA. Neurotic excoriation presenting as solitary papule: case report. J Dermatol Dermatolog Surg. 2022;26:45. doi:10.4103/jdds.jdds_59_21
  36. Kwiek B, Schwartz RA. Keratoacanthoma (KA): an update and review. J Am Acad Dermatol. 2016;74:1220-1233. doi:10.1016/j.jaad.2015.11.033
  37. Karadag AS, Ozlu E, Uzuncakmak TK, et al. Inverted follicular keratosis successfully treated with imiquimod. Indian Dermatol Online J. 2016;7:177-179. doi:10.4103/2229-5178.182354
  38. Nayak VN, Uma K, Girish HC, et al. Pseudoepitheliomatous hyperplasia in oral lesions: a review. J Int Oral Health. 2015;7:148-152.
  39. Saraceno R, Chiricozzi A, Nisticò SP, et al. An occlusive dressing containing betamethasone valerate 0.1% for the treatment of prurigo nodularis. J Dermatolog Treat. 2010;21:363-366. doi:10.3109/09546630903386606
  40. Wong SS, Goh CL. Double-blind, right/left comparison of calcipotriol ointment and betamethasone ointment in the treatment of prurigo nodularis. Arch Dermatol. 2000;136:807-808. doi:10.1001/archderm.136.6.807
  41. Waldinger TP, Wong RC, Taylor WB, et al. Cryotherapy improves prurigo nodularis. Arch Dermatol. 1984;120:1598-1600.
  42. Ständer S, Luger T, Metze D. Treatment of prurigo nodularis with topical capsaicin. J Am Acad Dermatol. 2001;44:471-478. doi:10.1067/mjd.2001.110059
  43. Ohanyan T, Schoepke N, Eirefelt S, et al. Role of substance P and its receptor neurokinin 1 in chronic prurigo: a randomized, proof-of-concept, controlled trial with topical aprepitant. Acta Derm Venereol. 2018;98:26-31. doi:10.2340/00015555-2780
  44. Tamagawa-Mineoka R, Katoh N, Ueda E, et al. Narrow-band ultraviolet B phototherapy in patients with recalcitrant nodular prurigo. J Dermatol. 2007;34:691-695. doi:10.1111/j.1346-8138.2007.00360.x
  45. Hammes S, Hermann J, Roos S, et al. UVB 308-nm excimer light and bath PUVA: combination therapy is very effective in the treatment of prurigo nodularis. J Eur Acad Dermatol Venereol. 2011;25:799-803. doi:10.1111/j.1468-3083.2010.03865.x
  46. Richards RN. Update on intralesional steroid: focus on dermatoses. J Cutan Med Surg. 2010;14:19-23. doi:10.2310/7750.2009.08082
  47. Elmariah S, Kim B, Berger T, et al. Practical approaches for diagnosis and management of prurigo nodularis: United States expert panel consensus. J Am Acad Dermatol. 2021;84:747-760. doi:10.1016/j.jaad.2020.07.025
  48. Grant JE, Chamberlain SR, Redden SA, et al. N-Acetylcysteine in the treatment of excoriation disorder: a randomized clinical trial. JAMA Psychiatry. 2016;73:490-496. doi:10.1001/jamapsychiatry.2016.0060
  49. Adil M, Amin SS, Mohtashim M. N-acetylcysteine in dermatology. Indian J Dermatol Venereol Leprol. 2018;84:652-659. doi: 10.4103/ijdvl.IJDVL_33_18.
  50. Taylor M, Bhagwandas K. Trichotillosis, skin picking and N-acetylcysteine. J Am Acad Dermatol. 2015;72(suppl 1):AB117. https://doi.org/10.1016/j.jaad.2015.02.482
  51. Legat FJ. The antipruritic effect of phototherapy. Front Med (Lausanne). 2018;5:333. doi:10.3389/fmed.2018.00333
  52. Klejtman T, Beylot-Barry M, Joly P, et al. Treatment of prurigo with methotrexate: a multicentre retrospective study of 39 cases. J Eur Acad Dermatol Venereol. 2018;32:437-440. doi:10.1111/jdv.14646
  53. Wiznia LE, Callahan SW, Cohen DE, et al. Rapid improvement of prurigo nodularis with cyclosporine treatment. J Am Acad Dermatol. 2018;78:1209-1211. doi:10.1016/j.jaad.2018.02.024
  54. Yagami A, Furue M, Togawa M, et al. One-year safety and efficacy study of bilastine treatment in Japanese patients with chronic spontaneous urticaria or pruritus associated with skin diseases. J Dermatol. 2017;44:375-385. doi:10.1111/1346-8138.13644
  55. Mazza M, Guerriero G, Marano G, et al. Treatment of prurigo nodularis with pregabalin. J Clin Pharm Ther. 2013;38:16-18. doi:10.1111/jcpt.12005
  56. Ständer S, Kwon P, Hirman J, et al. Serlopitant reduced pruritus in patients with prurigo nodularis in a phase 2, randomized, placebo-controlled trial. J Am Acad Dermatol. 2019;80:1395-1402. doi:10.1016/j.jaad.2019.01.052
  57. Study of the efficacy, safety and tolerability of serlopitant for the treatment of pruritus (itch) with prurigo nodularis. ClinicalTrials.gov identifier: NCT03546816. Updated May 20, 2021. Accessed August 8, 2024. https://clinicaltrials.gov/study/NCT03546816
  58. Tsianakas A, Zeidler C, Riepe C, et al. Aprepitant in anti-histamine-refractory chronic nodular prurigo: a multicentre, randomized, double-blind, placebo-controlled, cross-over, phase-II trial (APREPRU). Acta Derm Venereol. 2019;99:379-385. doi:10.2340/00015555-3120
  59. Metze D, Reimann S, Beissert S, et al. Efficacy and safety of naltrexone, an oral opiate receptor antagonist, in the treatment of pruritus in internal and dermatological diseases. J Am Acad Dermatol. 1999;41:533-539.
  60. Weisshaar E, Szepietowski JC, Bernhard JD, et al. Efficacy and safety of oral nalbuphine extended release in prurigo nodularis: results of a phase 2 randomized controlled trial with an open‐label extension phase. J Eur Acad Dermatol Venereol. 2022;36:453-461. doi:10.1111/jdv.17816
  61. Ständer S, Böckenholt B, Schürmeyer-Horst F, et al. Treatment of chronic pruritus with the selective serotonin re-uptake inhibitors paroxetine and fluvoxamine: results of an open-labelled, two-arm proof-of-concept study. Acta Derm Venereol. 2009;89:45-51. doi:10.2340/00015555-0553
  62. Zalaudek I, Petrillo G, Baldassarre MA, et al. Amitriptyline as therapeutic and not symptomatic approach in the treatment of prurigo nodularis. G Ital Dermatol Venereol. 2006;141:433-437.
  63. Andersen TP, Fogh K. Thalidomide in 42 patients with prurigo nodularis Hyde. Dermatology. 2011;223:107-112. doi:10.1159/000331577
  64. Ständer S, Yosipovitch G, Legat FJ, et al. Trial of nemolizumab in moderate-to-severe prurigo nodularis. N Engl J Med. 2020;382:706-716. doi:10.1056/NEJMoa1908316
  65. Yosipovitch G, Mollanazar N, Ständer S, et al. Dupilumab in patients with prurigo nodularis: two randomized, double-blind, placebo-controlled phase 3 trials. Nat Med. 2023;29:1180-1190. doi:10.1038/s41591-023-02320-9
  66. Mastorino L, Rosset F, Gelato F, et al. Chronic pruritus in atopic patients treated with dupilumab: real life response and related parameters in 354 patients. Pharmaceuticals (Basel). 2022;15:883. doi: 10.3390/ph15070883
  67. Kishi R, Toyama S, Tominaga M, et al. Effects of dupilumab on itch-related events in atopic dermatitis: implications for assessing treatment efficacy in clinical practice. Cells. 2023;12:239. doi: 10.3390/cells12020239
  68. Dupixent. European Medicines Agency website. Updated July 15, 2024. Accessed August 27, 2024. https://www.ema.europa.eu/en/medicines/human/EPAR/dupixent
Issue
Cutis - 114(2)
Issue
Cutis - 114(2)
Page Number
E43-E52
Page Number
E43-E52
Publications
Publications
Topics
Article Type
Display Headline
Prurigo Nodularis Mechanisms and Current Management Options
Display Headline
Prurigo Nodularis Mechanisms and Current Management Options
Sections
Inside the Article

Practice Points

  • Clinically, prurigo nodularis can mimic an array of dermatologic skin conditions and may be diagnosed more frequently in patients with comorbidities.
  • Dupilumab is the first and only treatment for prurigo nodularis approved by the US Food and Drug Administration; however, many topical treatments are currently used as first-line therapies.
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.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

The Use of Tranexamic Acid and Microneedling in the Treatment of Melasma: A Systematic Review

Article Type
Changed
Tue, 09/03/2024 - 15:52
Display Headline
The Use of Tranexamic Acid and Microneedling in the Treatment of Melasma: A Systematic Review

Melasma (also known as chloasma faciei) is a common chronic skin disorder that results in well-demarcated, hyperpigmented, tan to dark patches that mostly appear in sun-exposed areas such as the face and neck and sometimes the arms. The exact prevalence or incidence is not known but is estimated to be 1% to 50% overall depending on the ethnic population and geographic location.1,2 Melasma predominantly affects women, but research has shown that approximately 10% to 20% of men are affected by this condition.3,4 Although melasma can affect patients of all skin types, it primarily affects those with darker skin tones.5 The groups most often affected are women of Black, Hispanic, Middle Eastern, and Southeast Asian ethnicity. Although the pathogenesis is complex and not fully understood, multiple pathways and etiologies have been theorized to cause melasma. Potential causes include exposure to UV radiation, oral contraceptives, hormonal changes, medications, thyroid dysfunction, genetics, and pregnancy.6,7 Cytokines and growth factors, including adipokine and angiopoietin, synthesized by sebaceous glands play a role in the pathogenic mechanism of melasma. Cytokines and growth factors are hypothesized to modulate the function of melanocytes.8 Both melanocytes and sebocytes are controlled by α–melanocyte-stimulating hormone. Therefore, overexpression of α–melanocyte-stimulating hormone will result in overproduction of these 2 cell types, resulting in melasma. Melasma can be classified into 4 subtypes using Wood lamp examination: epidermal, dermal, mixed, or indeterminate.3 Furthermore, melasma is divided into subgroups based on the location: malar region, mandibular region, and centrofacial patch pattern.9,10 The involvement of sebaceous glands in the pathogenesis of melasma may explain the predilection for the centrofacial region, which is the most common pattern.

The severity of melasma can be assessed using the melasma area and severity index (MASI), which is calculated by subjective assessment of 3 main factors: (1) facial area of involvement; (2) darkness of affected region; and (3) homogeneity, with the extent of melasma indicated by a score ranging from 0 to 48.11 The modified MASI (mMASI) subsequently was introduced to assist with assessing the severity of melasma and creating distinct ranges for mild, moderate, and severe cases, ranging from 0 (mild) to 24 (severe).12 Both indices are used in research to assess the improvement of melasma with treatment.

Patients with melasma report a decrease in quality of life, increased emotional stress, and lower self-esteem due to cosmesis.13 Treatment of melasma can be highly challenging and often is complicated by relapsing. Historically, the treatment of melasma has included the use of chemical lightening agents. Additional treatment options include the use of lasers and complex chemical peels,9,10 but these interventions may result in adverse outcomes for individuals with darker skin tones. The current gold-standard treatment is topical hydroquinone and broad-spectrum sunscreen. Although hydroquinone is effective in the treatment of melasma, relapse is common. The goal of melasma management is not only to treat acute hyperpigmentation but also to prevent relapse. Other therapies that currently are being explored for the clinically sustained treatment of melasma include tranexamic acid (TXA)(trans-4-[aminomethyl]cyclohexanecarboxylic acid),9,10 an antifibrinolytic agent routinely used to prevent blood loss during surgery and in the management of menorrhagia. It is a synthetic derivative of lysine and serves as a potent plasmin inhibitor by blocking the lysine-binding sites of plasminogen molecules, thus preventing the conversion of plasminogen to plasmin. It also prevents fibrinolysis and blood loss.

In addition to its hemostatic properties, TXA has been found to have hypopigmentation properties.14,15 Plasminogen also can be found in human epidermal basal cells and human keratinocytes, and it is postulated that TXA’s interaction with these cells explains its hypopigmentation properties. Both UV radiation and hormones activate plasminogen into plasmin, resulting in the activation of tyrosinase and melanogenesis.14,15 Tranexamic acid is postulated to inhibit the keratinocyte-plasminogen pathway, thus leading to the inhibition of UV-induced and hormone-induced pigmentation. Also, TXA serves as a competitive inhibitor for tyrosinase due to its structural similarity to tyrosine.15 The combination of these 2 mechanisms contributes to the skin-lightening effects of TXA, making it a potential treatment for melasma.

Furthermore, the use of microneedling is being explored as a treatment option for melasma. Microneedling creates microscopic punctures in the skin using tiny needles, resulting in a wound-healing response and skin resurfacing. The microneedling technique is utilized to create small holes in the skin, with needle depths that can be adjusted from 0.5 to 3.5 mm to target different layers of the dermis and allow for discreet application of TXA.16 We sought to look at the current literature on the use and effectiveness of microneedling in combination with TXA to treat melasma and prevent relapse.

 

 

Methods

A systematic review was performed of PubMed articles indexed for MEDLINE and Embase in November 2021 to compile available articles that studied TXA and microneedling as a treatment for melasma. The PubMed search terms were (melasma) AND (microneedling* OR ‘tranexamic acid’ OR TXA or TA). The Embase search terms were (cholasma OR melasma) AND (tranexamic acid OR TXA) AND (microneedling)(Figure). The search was then limited to ”randomized controlled trial” and ”clinical trial” in English-language journals. Duplicates were excluded. After thorough evaluation, articles that discussed the use of TXA in combination with treatment options other than microneedling also were excluded.

Flow diagram of study selection. Asterisk indicates platelet-rich plasma, vitamin C, kojic acid, niacinamide, Kligman’s therapy (fluocinolone + hydroquinone + tretinoin), retinoic acid, and cysteamine.

Results

The literature search yielded a total of 12 articles that assessed the effectiveness of TXA and microneedling for the treatment of melasma (Table).17-28 Several articles concluded that TXA was equally effective at reducing melasma lesions when compared with the standard treatment of hydroquinone. Some of the reviewed articles also demonstrated the effectiveness of microneedling in improving melasma lesions as a stand-alone treatment. These studies highlighted the enhanced efficacy of the combined treatment of TXA and microneedling compared with their individual uses.17-28

Comment

Melasma is a common chronic hyperpigmentation disorder, making its treatment clinically challenging. Many patients experience symptom relapses, and limited effective treatment options make achieving complete clearance difficult, underscoring the need for improved therapeutic approaches. Recently, researchers have explored alternative treatments to address the challenges of melasma management. Tranexamic acid is an antifibrinolytic used to prevent blood loss and has emerged as a potential treatment for melasma. Similarly, microneedling—a technique in which multiple punctures are made in the skin to activate and stimulate wound healing and skin rejuvenation—shows promise for melasma.

Oral TXA for Melasma—Oral TXA has been shown to reduce melasma lesions. Del Rosario et al17 recruited 44 women (39 of whom completed the study) with moderate to severe melasma and randomized them into 2 groups: oral TXA and placebo. This study demonstrated a 49% reduction in the mMASI score in all participants taking oral TXA (250 mg twice daily [BID]) compared with an 18% reduction in the control group (placebo capsule BID) after 3 months of treatment. In patients with moderate and severe melasma, 45% and 51% mMASI score reductions were reported in the treatment group, respectively, vs 16% and 19% score reductions in placebo group, respectively. These researchers concluded that oral TXA may be effective at treating moderate to severe melasma. Although patients with severe melasma had a better response to treatment, their improvement was not sustained compared with patients with moderate melasma after a 3-month posttreatment follow-up.17

Microneedling Plus TXA for Melasma—Microneedling alone has been shown to be effective for melasma. El Attar et al18 conducted a split-face study of microneedling (1.5-mm depth) plus topical TXA (0.5 mL)(right side of the face[treatment arm]) compared with microneedling (1.5-mm depth) plus topical vitamin C (0.5 mL)(left side of the face [control group]) in 20 women with melasma. The sessions were repeated every 2 weeks for a total of 6 sessions. Although researchers found no statistically significant differences between the 2 treatment sides, microneedling plus TXA showed a slight advantage over microneedling plus vitamin C in dermoscopic examination. Both sides showed improvement in pigmented lesions, but vitamin C–treated lesions did not show an improvement in vascularity vs TXA.18

Saleh et al19 further showed that combination treatment with microneedling and TXA may improve clinical outcomes better than microneedling alone. Their study demonstrated a reduction in MASI score that was significantly higher in the combination treatment group compared with the microneedling alone group (P=.001). There was a significant reduction in melanoma antigen recognized by T cells 1 (MART-1)–positive cells in the combination treatment group compared with the microneedling alone group (P=.001). Lastly, combined therapy improved melasma patches better than microneedling alone.19

 

Xu et al20 conducted a split-face study (N=28) exploring the effectiveness of transdermal application of topical TXA using a microarray pen with microneedles (vibration at 3000×/min) plus topical TXA on one side of the face, while the other side received only topical TXA as a control. After 12 weeks of treatment, combination therapy with microneedling and TXA decreased brown spot scores, lowered melanin index (MI) values, improved blinded physician assessment, and improved patient satisfaction vs TXA therapy alone.20

Kaur et al21 conducted a split-face, randomized, controlled trial of microneedling (1-mm depth) with TXA solution 10% vs microneedling (1-mm depth) with distilled water alone for 8 weeks (N=40). They graded participant responses to treatment using reductions in mMASI scores12 at every 2 weeks of follow-up (no response, minimal or poor response=0%–25%; partial or fair response=26%–50%; good response=51%–75%; and excellent response=>75%). They reported an overall reduction in mMASI scores for both the treatment side and the control side in all participants, showing a 65.92% improvement in mean mMASI scores on the treatment side vs 20.75% improvement on the control side at week 8. Both sides showed statistically significant reductions in mean mMASI scores (P<.05). Clinically, 40% (16/40) of participants showed an excellent response to combined treatment compared with 0% (0/40) to microneedling alone. Overall, patient satisfaction was similar across both groups. This study demonstrated that microneedling alone improves melasma, but a combination of microneedling plus TXA showed a better clinical reduction in melasma. However, the researchers did not follow up with participants posttreatment, so it remains unclear if the improved clinical outcomes were sustained long-term.21

Ebrahim et al22 reported that the combination of 0.5 mL TXA (4 mg/mL) and microneedling (0.25- to 1-mm depth) was effective for melasma. Although there was improvement within microneedling and TXA, the study also showed that intradermal injection of TXA was significant in reducing mean mMASI scores and improving melasma (P<.001). The reduction in mMASI scores for the group receiving intradermal injections of TXA (left side; 74.8% reduction in mean mMASI score) vs the group receiving microneedling application of TXA (right side; 73.6% reduction in mean mMASI score) was not statistically significant. These findings suggest that the mode of TXA application may not be critical in determining clinical responses to TXA treatment. Although there was no reported statistically significant difference in clinical outcomes between the 2 treatments, patient satisfaction was higher on the microneedling side. Only 8 of 50 participants (16%) experienced recurrence 3 months posttreatment.22

Saki et al23 compared the efficacy of topical hydroquinone (2%) to intradermal TXA injections in treating melasma. They found intradermal TXA injections to be a clinically effective mode of treatment.23

Sharma et al24 explored the efficacy and safety of oral TXA by randomly assigning 100 Indian patients (20 of whom withdrew before study completion) with melasma into 2 groups: group A received TXA 250 mg twice daily, and group B received intradermal microinjections of TXA (4 mg/mL) every 4 weeks. The MASI scores were assessed at 4-week intervals for a total of 12 weeks. There was a decrease in MASI scores in both groups, and there was no statistically significant difference in mean percentage reduction in MASI scores between the 2 routes of drug administration, further suggesting the effectiveness of TXA independent of administration route. Two patients in group A relapsed at 24 weeks, and there were no relapses in group B, which may suggest a minimal superiority of TXA plus microneedling at providing more sustainable results compared with oral TXA alone. A notable limitation of this study was a high dropout rate as well as lack of long-term follow-up with participants, limiting the generalizability of the conclusions.24

Cassiano et al25 assigned 64 women with melasma to 1 of 3 treatment groups or a control group to compare the effectiveness of microneedling (M group: 1.5 mm; 2 sessions), oral TXA (T group: 250 mg/d twice daily for 60 days), and a combination of microneedling (2 sessions) and oral TXA (MT group: 250 mg/d twice daily for 60 days)with placebo for clinically reducing melasma lesions. The intervention period was 60 days followed by a 60-day maintenance phase for a total study period of 120 days. The researchers evaluated mMASI scores, quality of life, and difference in colorimetric luminosity. All treatment groups showed a reduction in mMASI scores at both 30 days and 60 days, indicating improved melasma severity. The MT and T groups had more significant improvement at 30 days compared with the control group (P<.03), suggesting that microneedling plus TXA and TXA alone promote faster improvement in melasma lesions. By 60 days, the M, T, and MT groups outperformed the control group, with no significant differences between the M, T, and MT groups. However, at the 120-day maintenance follow-up, the T group did not maintain its improvement compared with the control group. The M and MT groups showed no significance difference in effectiveness at 120 days, suggesting that microneedling may promote less frequent relapse and sustained remission compared to TXA alone.25

Hydroquinone for Melasma—Additional studies on the use of TXA treatments show that TXA may be an equally effective alternative to the standard use of hydroquinone treatment. Shamsi Meymandi et al26 did not find a statistically significant difference in treatment with TXA plus microneedling vs the standard regimen of hydroquinone. More importantly, patient and physician satisfaction assessments were similar between the 2 groups. Compared to hydroquinone, nightly treatment is not necessary with microneedling and TXA.26

Xing et al27 supported these conclusions with their study. They compared 3 study arms for a duration of 12 weeks: group A received topical 1.8% liposomal TXA BID, group B received stamp-mode electric microneedling with 5% TXA weekly, and group C applied 2% ­hydroquinone cream nightly. The study concluded that all 3 groups showed a significant reduction in mean MI by the end of the study, but a better MI improvement was observed in groups B and C (both P<.001) compared with group A (P<.01).27

Zaky et al28 showed that both hydroquinone and combination treatment of TXA plus microneedling are effective at improving melasma lesions. Further studies are needed to definitively conclude if combination treatment is more efficacious than hydroquinone; if the combination is more effective, it provides a treatment option for patients with melasma who may not be good candidates for hydroquinone treatment.

Study Limitations—One limitation in all the studies evaluated is the sample size. Because they all had small sample sizes, it is difficult to definitively conclude that the combination TXA and microneedling is an effective and appropriate treatment for patients with melasma. Furthermore, the quality of these studies was mostly dependent on subjectivity of the mMASI scores. Future large randomized controlled trials with a diverse participant population are needed to assess the effectiveness of TXA and microneedling in melasma treatment.

Another limitation is that many of the studies did not follow the patients longitudinally, which did not allow for an evaluation of whether patients had a relapse of melasma. Due to the chronic nature of melasma and frequent disease recurrence, future longitudinal studies are needed to monitor for disease recurrence.

Conclusion

Tranexamic acid and microneedling are potential treatment options for patients with melasma, and combination therapy appears more effective than either TXA or microneedling alone at providing sustained improvement of melasma lesions. Combination therapy appears safe and well tolerated, but its effect on reducing long-term disease recurrence is yet to be established.

References
  1. Neagu N, Conforti C, Agozzino M, et al. Melasma treatment: a systematic review. J Dermatolog Treat. 2022;33:1816-1837. doi:10.1080/09546634.2021.1914313
  2. Ogbechie-Godec OA, Elbuluk N. Melasma: an up-to-date comprehensive review. Dermatol Ther (Heidelb). 2017;7:305-318. doi:10.1007/s13555-017-0194-1
  3. Mahajan VK, Patil A, Blicharz L, et al. Medical therapies for melasma. J Cosmet Dermatol. 2022;21:3707-3728. doi:10.1111/jocd.15242
  4. Rigopoulos D, Gregoriou S, Katsambas A. Hyperpigmentation and melasma. J Cosmet Dermatol. 2007;6:195-202. doi:10.1111/j.1473-2165.2007.00321.x
  5. Kagha K, Fabi S, Goldman M. Melasma’s impact on quality of life. J Drugs Dermatol. 2020;19:184-187. doi:10.36849/JDD.2020.4663
  6. Lutfi RJ, Fridmanis M, Misiunas AL, et al. Association of melasma with thyroid autoimmunity and other thyroidal abnormalities and their relationship to the origin of the melasma. J Clin Endocrinol Metab. 1985;61:28-31. doi:10.1210/jcem-61-1-28
  7. Handel AC, Lima PB, Tonolli VM, et al. Risk factors for facial melasma in women: a case-control study. Br J Dermatol. 2014;171:588-594. doi:10.1111/bjd.13059
  8. Filoni A, Mariano M, Cameli N. Melasma: how hormones can modulate skin pigmentation. J Cosmet Dermatol. 2019;18:458-463. doi:10.1111/jocd.12877
  9. Rodrigues M, Pandya AG. Melasma: clinical diagnosis and management options. Australasian J Dermatol. 2015;56:151-163.
  10. Huerth KA, Hassan S, Callender VD. Therapeutic insights in melasma and hyperpigmentation management. J Drugs Dermatol. 2019;18:718-727.
  11. Pandya AG, Hynan LS, Bhore R, et al. Reliability assessment and validation of the Melasma Area and Severity Index (MASI) and a new modified MASI scoring method. J Am Acad Dermatol. 2011;64:78-­83.e832. doi:10.1016/j.jaad.2009.10.051
  12. Rodrigues M, Ayala-Cortés AS, Rodríguez-Arámbula A, et al. Interpretability of the modified Melasma Area and Severity Index (mMASI). JAMA Dermatol. 2016;152:1051-1052. doi:10.1001/jamadermatol.2016.1006
  13. Ikino JK, Nunes DH, da Silva VPM, et al. Melasma and assessment of the quality of life in Brazilian women. An Bras Dermatol. 2015;90:196-200. doi:10.1590/abd1806-4841.20152771
  14. Taraz M, Niknam S, Ehsani AH. Tranexamic acid in treatment of melasma: a comprehensive review of clinical studies. Dermatolog Ther. 2017;30:E12465. doi:10.1111/dth.12465
  15. Bala HR, Lee S, Wong C, et al. Oral tranexamic acid for the treatment of melasma: a review. Dermatol Surg. 2018;44:814-825. doi:10.1097/DSS.0000000000001518
  16. Singh A, Yadav S. Microneedling: advances and widening horizons. Indian Dermatol Online J. 2016;7:244-254. doi:10.4103/2229-5178.185468
  17. Del Rosario E, Florez-Pollack S, Zapata L, et al. Randomized, placebo-controlled, double-blind study of oral tranexamic acid in the treatment of moderate-to-severe melasma. J Am Acad Dermatol. 2018;78:363-369. doi:10.1016/j.jaad.2017.09.053
  18. El Attar Y, Doghaim N, El Far N, et al. Efficacy and safety of tranexamic acid versus vitamin C after microneedling in treatment of melasma: clinical and dermoscopic study. J Cosmet Dermatol. 2022;21:2817-2825. doi:10.1111/jocd.14538
  19. Saleh FY, Abdel-Azim ES, Ragaie MH, et al. Topical tranexamic acid with microneedling versus microneedling alone in treatment of melasma: clinical, histopathologic, and immunohistochemical study. J Egyptian Womens Dermatolog Soc. 2019;16:89-96. doi:10.4103/jewd.jewd_25_19
  20. Xu Y, Ma R, Juliandri J, et al. Efficacy of functional microarray of microneedles combined with topical tranexamic acid for melasma: a randomized, self-controlled, split-face study. Medicine (Baltimore). 2017;96:e6897. doi:10.1097/MD.0000000000006897
  21. Kaur A, Bhalla M, Pal Thami G, et al. Clinical efficacy of topical tranexamic acid with microneedling in melasma. Dermatol Surg. 2020;46:E96-E101. doi:10.1097/DSS.0000000000002520
  22. Ebrahim HM, Said Abdelshafy A, Khattab F, et al. Tranexamic acid for melasma treatment: a split-face study. Dermatol Surg. 2020;46:E102-E107. doi:10.1097/DSS.0000000000002449
  23. Saki N, Darayesh M, Heiran A. Comparing the efficacy of topical hydroquinone 2% versus intradermal tranexamic acid microinjections in treating melasma: a split-face controlled trial. J Dermatolog Treat. 2018;29:405-410. doi:10.1080/09546634.2017.1392476
  24. Sharma R, Mahajan VK, Mehta KS, et al. Therapeutic efficacy and safety of oral tranexamic acid and that of tranexamic acid local infiltration with microinjections in patients with melasma: a comparative study. Clin Exp Dermatol. 2017;42:728-734. doi:10.1111/ced.13164
  25. Cassiano D, Esposito ACC, Hassun K, et al. Efficacy and safety of microneedling and oral tranexamic acid in the treatment of facial melasma in women: an open, evaluator-blinded, randomized clinical trial. J Am Acad Dermatol. 2020;83:1176-1178. doi:10.1016/j.jaad.2020.02.002
  26. Shamsi Meymandi S, Mozayyeni A, Shamsi Meymandi M, et al. Efficacy of microneedling plus topical 4% tranexamic acid solution vs 4% hydroquinone in the treatment of melasma: a single-blind randomized clinical trial. J Cosmet Dermatol. 2020;19:2906-2911. doi:10.1111/jocd.13392
  27. Xing X, Chen L, Xu Z, et al. The efficacy and safety of topical tranexamic acid (liposomal or lotion with microneedling) versus conventional hydroquinone in the treatment of melasma. J Cosmet Dermatol. 2020;19:3238-3244. doi:10.1111/jocd.13810
  28. Zaky MS, Obaid ZM, Khalil EA, et al. Microneedling-assisted topical tranexamic acid solution versus 4% hydroquinone for treating melasma: a split-face randomized study. J Cosmet Dermatol. 2021;20:4011-4016. doi:10.1111/jocd.14440
Article PDF
Author and Disclosure Information

Idowu D. Olugbade is from the Warren Alpert Medical School of Brown University, Providence, Rhode Island. Dr. Negbenebor is from the Department of Dermatology, University of Iowa, Iowa City.

The authors report no conflict of interest.

Correspondence: Nicole A. Negbenebor, MD (nicole-negbenebor@uiowa.edu).

Cutis. 2024 August;114(2):E15-E23. doi:10.12788/cutis.1080

Issue
Cutis - 114(2)
Publications
Topics
Page Number
E15-E23
Sections
Author and Disclosure Information

Idowu D. Olugbade is from the Warren Alpert Medical School of Brown University, Providence, Rhode Island. Dr. Negbenebor is from the Department of Dermatology, University of Iowa, Iowa City.

The authors report no conflict of interest.

Correspondence: Nicole A. Negbenebor, MD (nicole-negbenebor@uiowa.edu).

Cutis. 2024 August;114(2):E15-E23. doi:10.12788/cutis.1080

Author and Disclosure Information

Idowu D. Olugbade is from the Warren Alpert Medical School of Brown University, Providence, Rhode Island. Dr. Negbenebor is from the Department of Dermatology, University of Iowa, Iowa City.

The authors report no conflict of interest.

Correspondence: Nicole A. Negbenebor, MD (nicole-negbenebor@uiowa.edu).

Cutis. 2024 August;114(2):E15-E23. doi:10.12788/cutis.1080

Article PDF
Article PDF

Melasma (also known as chloasma faciei) is a common chronic skin disorder that results in well-demarcated, hyperpigmented, tan to dark patches that mostly appear in sun-exposed areas such as the face and neck and sometimes the arms. The exact prevalence or incidence is not known but is estimated to be 1% to 50% overall depending on the ethnic population and geographic location.1,2 Melasma predominantly affects women, but research has shown that approximately 10% to 20% of men are affected by this condition.3,4 Although melasma can affect patients of all skin types, it primarily affects those with darker skin tones.5 The groups most often affected are women of Black, Hispanic, Middle Eastern, and Southeast Asian ethnicity. Although the pathogenesis is complex and not fully understood, multiple pathways and etiologies have been theorized to cause melasma. Potential causes include exposure to UV radiation, oral contraceptives, hormonal changes, medications, thyroid dysfunction, genetics, and pregnancy.6,7 Cytokines and growth factors, including adipokine and angiopoietin, synthesized by sebaceous glands play a role in the pathogenic mechanism of melasma. Cytokines and growth factors are hypothesized to modulate the function of melanocytes.8 Both melanocytes and sebocytes are controlled by α–melanocyte-stimulating hormone. Therefore, overexpression of α–melanocyte-stimulating hormone will result in overproduction of these 2 cell types, resulting in melasma. Melasma can be classified into 4 subtypes using Wood lamp examination: epidermal, dermal, mixed, or indeterminate.3 Furthermore, melasma is divided into subgroups based on the location: malar region, mandibular region, and centrofacial patch pattern.9,10 The involvement of sebaceous glands in the pathogenesis of melasma may explain the predilection for the centrofacial region, which is the most common pattern.

The severity of melasma can be assessed using the melasma area and severity index (MASI), which is calculated by subjective assessment of 3 main factors: (1) facial area of involvement; (2) darkness of affected region; and (3) homogeneity, with the extent of melasma indicated by a score ranging from 0 to 48.11 The modified MASI (mMASI) subsequently was introduced to assist with assessing the severity of melasma and creating distinct ranges for mild, moderate, and severe cases, ranging from 0 (mild) to 24 (severe).12 Both indices are used in research to assess the improvement of melasma with treatment.

Patients with melasma report a decrease in quality of life, increased emotional stress, and lower self-esteem due to cosmesis.13 Treatment of melasma can be highly challenging and often is complicated by relapsing. Historically, the treatment of melasma has included the use of chemical lightening agents. Additional treatment options include the use of lasers and complex chemical peels,9,10 but these interventions may result in adverse outcomes for individuals with darker skin tones. The current gold-standard treatment is topical hydroquinone and broad-spectrum sunscreen. Although hydroquinone is effective in the treatment of melasma, relapse is common. The goal of melasma management is not only to treat acute hyperpigmentation but also to prevent relapse. Other therapies that currently are being explored for the clinically sustained treatment of melasma include tranexamic acid (TXA)(trans-4-[aminomethyl]cyclohexanecarboxylic acid),9,10 an antifibrinolytic agent routinely used to prevent blood loss during surgery and in the management of menorrhagia. It is a synthetic derivative of lysine and serves as a potent plasmin inhibitor by blocking the lysine-binding sites of plasminogen molecules, thus preventing the conversion of plasminogen to plasmin. It also prevents fibrinolysis and blood loss.

In addition to its hemostatic properties, TXA has been found to have hypopigmentation properties.14,15 Plasminogen also can be found in human epidermal basal cells and human keratinocytes, and it is postulated that TXA’s interaction with these cells explains its hypopigmentation properties. Both UV radiation and hormones activate plasminogen into plasmin, resulting in the activation of tyrosinase and melanogenesis.14,15 Tranexamic acid is postulated to inhibit the keratinocyte-plasminogen pathway, thus leading to the inhibition of UV-induced and hormone-induced pigmentation. Also, TXA serves as a competitive inhibitor for tyrosinase due to its structural similarity to tyrosine.15 The combination of these 2 mechanisms contributes to the skin-lightening effects of TXA, making it a potential treatment for melasma.

Furthermore, the use of microneedling is being explored as a treatment option for melasma. Microneedling creates microscopic punctures in the skin using tiny needles, resulting in a wound-healing response and skin resurfacing. The microneedling technique is utilized to create small holes in the skin, with needle depths that can be adjusted from 0.5 to 3.5 mm to target different layers of the dermis and allow for discreet application of TXA.16 We sought to look at the current literature on the use and effectiveness of microneedling in combination with TXA to treat melasma and prevent relapse.

 

 

Methods

A systematic review was performed of PubMed articles indexed for MEDLINE and Embase in November 2021 to compile available articles that studied TXA and microneedling as a treatment for melasma. The PubMed search terms were (melasma) AND (microneedling* OR ‘tranexamic acid’ OR TXA or TA). The Embase search terms were (cholasma OR melasma) AND (tranexamic acid OR TXA) AND (microneedling)(Figure). The search was then limited to ”randomized controlled trial” and ”clinical trial” in English-language journals. Duplicates were excluded. After thorough evaluation, articles that discussed the use of TXA in combination with treatment options other than microneedling also were excluded.

Flow diagram of study selection. Asterisk indicates platelet-rich plasma, vitamin C, kojic acid, niacinamide, Kligman’s therapy (fluocinolone + hydroquinone + tretinoin), retinoic acid, and cysteamine.

Results

The literature search yielded a total of 12 articles that assessed the effectiveness of TXA and microneedling for the treatment of melasma (Table).17-28 Several articles concluded that TXA was equally effective at reducing melasma lesions when compared with the standard treatment of hydroquinone. Some of the reviewed articles also demonstrated the effectiveness of microneedling in improving melasma lesions as a stand-alone treatment. These studies highlighted the enhanced efficacy of the combined treatment of TXA and microneedling compared with their individual uses.17-28

Comment

Melasma is a common chronic hyperpigmentation disorder, making its treatment clinically challenging. Many patients experience symptom relapses, and limited effective treatment options make achieving complete clearance difficult, underscoring the need for improved therapeutic approaches. Recently, researchers have explored alternative treatments to address the challenges of melasma management. Tranexamic acid is an antifibrinolytic used to prevent blood loss and has emerged as a potential treatment for melasma. Similarly, microneedling—a technique in which multiple punctures are made in the skin to activate and stimulate wound healing and skin rejuvenation—shows promise for melasma.

Oral TXA for Melasma—Oral TXA has been shown to reduce melasma lesions. Del Rosario et al17 recruited 44 women (39 of whom completed the study) with moderate to severe melasma and randomized them into 2 groups: oral TXA and placebo. This study demonstrated a 49% reduction in the mMASI score in all participants taking oral TXA (250 mg twice daily [BID]) compared with an 18% reduction in the control group (placebo capsule BID) after 3 months of treatment. In patients with moderate and severe melasma, 45% and 51% mMASI score reductions were reported in the treatment group, respectively, vs 16% and 19% score reductions in placebo group, respectively. These researchers concluded that oral TXA may be effective at treating moderate to severe melasma. Although patients with severe melasma had a better response to treatment, their improvement was not sustained compared with patients with moderate melasma after a 3-month posttreatment follow-up.17

Microneedling Plus TXA for Melasma—Microneedling alone has been shown to be effective for melasma. El Attar et al18 conducted a split-face study of microneedling (1.5-mm depth) plus topical TXA (0.5 mL)(right side of the face[treatment arm]) compared with microneedling (1.5-mm depth) plus topical vitamin C (0.5 mL)(left side of the face [control group]) in 20 women with melasma. The sessions were repeated every 2 weeks for a total of 6 sessions. Although researchers found no statistically significant differences between the 2 treatment sides, microneedling plus TXA showed a slight advantage over microneedling plus vitamin C in dermoscopic examination. Both sides showed improvement in pigmented lesions, but vitamin C–treated lesions did not show an improvement in vascularity vs TXA.18

Saleh et al19 further showed that combination treatment with microneedling and TXA may improve clinical outcomes better than microneedling alone. Their study demonstrated a reduction in MASI score that was significantly higher in the combination treatment group compared with the microneedling alone group (P=.001). There was a significant reduction in melanoma antigen recognized by T cells 1 (MART-1)–positive cells in the combination treatment group compared with the microneedling alone group (P=.001). Lastly, combined therapy improved melasma patches better than microneedling alone.19

 

Xu et al20 conducted a split-face study (N=28) exploring the effectiveness of transdermal application of topical TXA using a microarray pen with microneedles (vibration at 3000×/min) plus topical TXA on one side of the face, while the other side received only topical TXA as a control. After 12 weeks of treatment, combination therapy with microneedling and TXA decreased brown spot scores, lowered melanin index (MI) values, improved blinded physician assessment, and improved patient satisfaction vs TXA therapy alone.20

Kaur et al21 conducted a split-face, randomized, controlled trial of microneedling (1-mm depth) with TXA solution 10% vs microneedling (1-mm depth) with distilled water alone for 8 weeks (N=40). They graded participant responses to treatment using reductions in mMASI scores12 at every 2 weeks of follow-up (no response, minimal or poor response=0%–25%; partial or fair response=26%–50%; good response=51%–75%; and excellent response=>75%). They reported an overall reduction in mMASI scores for both the treatment side and the control side in all participants, showing a 65.92% improvement in mean mMASI scores on the treatment side vs 20.75% improvement on the control side at week 8. Both sides showed statistically significant reductions in mean mMASI scores (P<.05). Clinically, 40% (16/40) of participants showed an excellent response to combined treatment compared with 0% (0/40) to microneedling alone. Overall, patient satisfaction was similar across both groups. This study demonstrated that microneedling alone improves melasma, but a combination of microneedling plus TXA showed a better clinical reduction in melasma. However, the researchers did not follow up with participants posttreatment, so it remains unclear if the improved clinical outcomes were sustained long-term.21

Ebrahim et al22 reported that the combination of 0.5 mL TXA (4 mg/mL) and microneedling (0.25- to 1-mm depth) was effective for melasma. Although there was improvement within microneedling and TXA, the study also showed that intradermal injection of TXA was significant in reducing mean mMASI scores and improving melasma (P<.001). The reduction in mMASI scores for the group receiving intradermal injections of TXA (left side; 74.8% reduction in mean mMASI score) vs the group receiving microneedling application of TXA (right side; 73.6% reduction in mean mMASI score) was not statistically significant. These findings suggest that the mode of TXA application may not be critical in determining clinical responses to TXA treatment. Although there was no reported statistically significant difference in clinical outcomes between the 2 treatments, patient satisfaction was higher on the microneedling side. Only 8 of 50 participants (16%) experienced recurrence 3 months posttreatment.22

Saki et al23 compared the efficacy of topical hydroquinone (2%) to intradermal TXA injections in treating melasma. They found intradermal TXA injections to be a clinically effective mode of treatment.23

Sharma et al24 explored the efficacy and safety of oral TXA by randomly assigning 100 Indian patients (20 of whom withdrew before study completion) with melasma into 2 groups: group A received TXA 250 mg twice daily, and group B received intradermal microinjections of TXA (4 mg/mL) every 4 weeks. The MASI scores were assessed at 4-week intervals for a total of 12 weeks. There was a decrease in MASI scores in both groups, and there was no statistically significant difference in mean percentage reduction in MASI scores between the 2 routes of drug administration, further suggesting the effectiveness of TXA independent of administration route. Two patients in group A relapsed at 24 weeks, and there were no relapses in group B, which may suggest a minimal superiority of TXA plus microneedling at providing more sustainable results compared with oral TXA alone. A notable limitation of this study was a high dropout rate as well as lack of long-term follow-up with participants, limiting the generalizability of the conclusions.24

Cassiano et al25 assigned 64 women with melasma to 1 of 3 treatment groups or a control group to compare the effectiveness of microneedling (M group: 1.5 mm; 2 sessions), oral TXA (T group: 250 mg/d twice daily for 60 days), and a combination of microneedling (2 sessions) and oral TXA (MT group: 250 mg/d twice daily for 60 days)with placebo for clinically reducing melasma lesions. The intervention period was 60 days followed by a 60-day maintenance phase for a total study period of 120 days. The researchers evaluated mMASI scores, quality of life, and difference in colorimetric luminosity. All treatment groups showed a reduction in mMASI scores at both 30 days and 60 days, indicating improved melasma severity. The MT and T groups had more significant improvement at 30 days compared with the control group (P<.03), suggesting that microneedling plus TXA and TXA alone promote faster improvement in melasma lesions. By 60 days, the M, T, and MT groups outperformed the control group, with no significant differences between the M, T, and MT groups. However, at the 120-day maintenance follow-up, the T group did not maintain its improvement compared with the control group. The M and MT groups showed no significance difference in effectiveness at 120 days, suggesting that microneedling may promote less frequent relapse and sustained remission compared to TXA alone.25

Hydroquinone for Melasma—Additional studies on the use of TXA treatments show that TXA may be an equally effective alternative to the standard use of hydroquinone treatment. Shamsi Meymandi et al26 did not find a statistically significant difference in treatment with TXA plus microneedling vs the standard regimen of hydroquinone. More importantly, patient and physician satisfaction assessments were similar between the 2 groups. Compared to hydroquinone, nightly treatment is not necessary with microneedling and TXA.26

Xing et al27 supported these conclusions with their study. They compared 3 study arms for a duration of 12 weeks: group A received topical 1.8% liposomal TXA BID, group B received stamp-mode electric microneedling with 5% TXA weekly, and group C applied 2% ­hydroquinone cream nightly. The study concluded that all 3 groups showed a significant reduction in mean MI by the end of the study, but a better MI improvement was observed in groups B and C (both P<.001) compared with group A (P<.01).27

Zaky et al28 showed that both hydroquinone and combination treatment of TXA plus microneedling are effective at improving melasma lesions. Further studies are needed to definitively conclude if combination treatment is more efficacious than hydroquinone; if the combination is more effective, it provides a treatment option for patients with melasma who may not be good candidates for hydroquinone treatment.

Study Limitations—One limitation in all the studies evaluated is the sample size. Because they all had small sample sizes, it is difficult to definitively conclude that the combination TXA and microneedling is an effective and appropriate treatment for patients with melasma. Furthermore, the quality of these studies was mostly dependent on subjectivity of the mMASI scores. Future large randomized controlled trials with a diverse participant population are needed to assess the effectiveness of TXA and microneedling in melasma treatment.

Another limitation is that many of the studies did not follow the patients longitudinally, which did not allow for an evaluation of whether patients had a relapse of melasma. Due to the chronic nature of melasma and frequent disease recurrence, future longitudinal studies are needed to monitor for disease recurrence.

Conclusion

Tranexamic acid and microneedling are potential treatment options for patients with melasma, and combination therapy appears more effective than either TXA or microneedling alone at providing sustained improvement of melasma lesions. Combination therapy appears safe and well tolerated, but its effect on reducing long-term disease recurrence is yet to be established.

Melasma (also known as chloasma faciei) is a common chronic skin disorder that results in well-demarcated, hyperpigmented, tan to dark patches that mostly appear in sun-exposed areas such as the face and neck and sometimes the arms. The exact prevalence or incidence is not known but is estimated to be 1% to 50% overall depending on the ethnic population and geographic location.1,2 Melasma predominantly affects women, but research has shown that approximately 10% to 20% of men are affected by this condition.3,4 Although melasma can affect patients of all skin types, it primarily affects those with darker skin tones.5 The groups most often affected are women of Black, Hispanic, Middle Eastern, and Southeast Asian ethnicity. Although the pathogenesis is complex and not fully understood, multiple pathways and etiologies have been theorized to cause melasma. Potential causes include exposure to UV radiation, oral contraceptives, hormonal changes, medications, thyroid dysfunction, genetics, and pregnancy.6,7 Cytokines and growth factors, including adipokine and angiopoietin, synthesized by sebaceous glands play a role in the pathogenic mechanism of melasma. Cytokines and growth factors are hypothesized to modulate the function of melanocytes.8 Both melanocytes and sebocytes are controlled by α–melanocyte-stimulating hormone. Therefore, overexpression of α–melanocyte-stimulating hormone will result in overproduction of these 2 cell types, resulting in melasma. Melasma can be classified into 4 subtypes using Wood lamp examination: epidermal, dermal, mixed, or indeterminate.3 Furthermore, melasma is divided into subgroups based on the location: malar region, mandibular region, and centrofacial patch pattern.9,10 The involvement of sebaceous glands in the pathogenesis of melasma may explain the predilection for the centrofacial region, which is the most common pattern.

The severity of melasma can be assessed using the melasma area and severity index (MASI), which is calculated by subjective assessment of 3 main factors: (1) facial area of involvement; (2) darkness of affected region; and (3) homogeneity, with the extent of melasma indicated by a score ranging from 0 to 48.11 The modified MASI (mMASI) subsequently was introduced to assist with assessing the severity of melasma and creating distinct ranges for mild, moderate, and severe cases, ranging from 0 (mild) to 24 (severe).12 Both indices are used in research to assess the improvement of melasma with treatment.

Patients with melasma report a decrease in quality of life, increased emotional stress, and lower self-esteem due to cosmesis.13 Treatment of melasma can be highly challenging and often is complicated by relapsing. Historically, the treatment of melasma has included the use of chemical lightening agents. Additional treatment options include the use of lasers and complex chemical peels,9,10 but these interventions may result in adverse outcomes for individuals with darker skin tones. The current gold-standard treatment is topical hydroquinone and broad-spectrum sunscreen. Although hydroquinone is effective in the treatment of melasma, relapse is common. The goal of melasma management is not only to treat acute hyperpigmentation but also to prevent relapse. Other therapies that currently are being explored for the clinically sustained treatment of melasma include tranexamic acid (TXA)(trans-4-[aminomethyl]cyclohexanecarboxylic acid),9,10 an antifibrinolytic agent routinely used to prevent blood loss during surgery and in the management of menorrhagia. It is a synthetic derivative of lysine and serves as a potent plasmin inhibitor by blocking the lysine-binding sites of plasminogen molecules, thus preventing the conversion of plasminogen to plasmin. It also prevents fibrinolysis and blood loss.

In addition to its hemostatic properties, TXA has been found to have hypopigmentation properties.14,15 Plasminogen also can be found in human epidermal basal cells and human keratinocytes, and it is postulated that TXA’s interaction with these cells explains its hypopigmentation properties. Both UV radiation and hormones activate plasminogen into plasmin, resulting in the activation of tyrosinase and melanogenesis.14,15 Tranexamic acid is postulated to inhibit the keratinocyte-plasminogen pathway, thus leading to the inhibition of UV-induced and hormone-induced pigmentation. Also, TXA serves as a competitive inhibitor for tyrosinase due to its structural similarity to tyrosine.15 The combination of these 2 mechanisms contributes to the skin-lightening effects of TXA, making it a potential treatment for melasma.

Furthermore, the use of microneedling is being explored as a treatment option for melasma. Microneedling creates microscopic punctures in the skin using tiny needles, resulting in a wound-healing response and skin resurfacing. The microneedling technique is utilized to create small holes in the skin, with needle depths that can be adjusted from 0.5 to 3.5 mm to target different layers of the dermis and allow for discreet application of TXA.16 We sought to look at the current literature on the use and effectiveness of microneedling in combination with TXA to treat melasma and prevent relapse.

 

 

Methods

A systematic review was performed of PubMed articles indexed for MEDLINE and Embase in November 2021 to compile available articles that studied TXA and microneedling as a treatment for melasma. The PubMed search terms were (melasma) AND (microneedling* OR ‘tranexamic acid’ OR TXA or TA). The Embase search terms were (cholasma OR melasma) AND (tranexamic acid OR TXA) AND (microneedling)(Figure). The search was then limited to ”randomized controlled trial” and ”clinical trial” in English-language journals. Duplicates were excluded. After thorough evaluation, articles that discussed the use of TXA in combination with treatment options other than microneedling also were excluded.

Flow diagram of study selection. Asterisk indicates platelet-rich plasma, vitamin C, kojic acid, niacinamide, Kligman’s therapy (fluocinolone + hydroquinone + tretinoin), retinoic acid, and cysteamine.

Results

The literature search yielded a total of 12 articles that assessed the effectiveness of TXA and microneedling for the treatment of melasma (Table).17-28 Several articles concluded that TXA was equally effective at reducing melasma lesions when compared with the standard treatment of hydroquinone. Some of the reviewed articles also demonstrated the effectiveness of microneedling in improving melasma lesions as a stand-alone treatment. These studies highlighted the enhanced efficacy of the combined treatment of TXA and microneedling compared with their individual uses.17-28

Comment

Melasma is a common chronic hyperpigmentation disorder, making its treatment clinically challenging. Many patients experience symptom relapses, and limited effective treatment options make achieving complete clearance difficult, underscoring the need for improved therapeutic approaches. Recently, researchers have explored alternative treatments to address the challenges of melasma management. Tranexamic acid is an antifibrinolytic used to prevent blood loss and has emerged as a potential treatment for melasma. Similarly, microneedling—a technique in which multiple punctures are made in the skin to activate and stimulate wound healing and skin rejuvenation—shows promise for melasma.

Oral TXA for Melasma—Oral TXA has been shown to reduce melasma lesions. Del Rosario et al17 recruited 44 women (39 of whom completed the study) with moderate to severe melasma and randomized them into 2 groups: oral TXA and placebo. This study demonstrated a 49% reduction in the mMASI score in all participants taking oral TXA (250 mg twice daily [BID]) compared with an 18% reduction in the control group (placebo capsule BID) after 3 months of treatment. In patients with moderate and severe melasma, 45% and 51% mMASI score reductions were reported in the treatment group, respectively, vs 16% and 19% score reductions in placebo group, respectively. These researchers concluded that oral TXA may be effective at treating moderate to severe melasma. Although patients with severe melasma had a better response to treatment, their improvement was not sustained compared with patients with moderate melasma after a 3-month posttreatment follow-up.17

Microneedling Plus TXA for Melasma—Microneedling alone has been shown to be effective for melasma. El Attar et al18 conducted a split-face study of microneedling (1.5-mm depth) plus topical TXA (0.5 mL)(right side of the face[treatment arm]) compared with microneedling (1.5-mm depth) plus topical vitamin C (0.5 mL)(left side of the face [control group]) in 20 women with melasma. The sessions were repeated every 2 weeks for a total of 6 sessions. Although researchers found no statistically significant differences between the 2 treatment sides, microneedling plus TXA showed a slight advantage over microneedling plus vitamin C in dermoscopic examination. Both sides showed improvement in pigmented lesions, but vitamin C–treated lesions did not show an improvement in vascularity vs TXA.18

Saleh et al19 further showed that combination treatment with microneedling and TXA may improve clinical outcomes better than microneedling alone. Their study demonstrated a reduction in MASI score that was significantly higher in the combination treatment group compared with the microneedling alone group (P=.001). There was a significant reduction in melanoma antigen recognized by T cells 1 (MART-1)–positive cells in the combination treatment group compared with the microneedling alone group (P=.001). Lastly, combined therapy improved melasma patches better than microneedling alone.19

 

Xu et al20 conducted a split-face study (N=28) exploring the effectiveness of transdermal application of topical TXA using a microarray pen with microneedles (vibration at 3000×/min) plus topical TXA on one side of the face, while the other side received only topical TXA as a control. After 12 weeks of treatment, combination therapy with microneedling and TXA decreased brown spot scores, lowered melanin index (MI) values, improved blinded physician assessment, and improved patient satisfaction vs TXA therapy alone.20

Kaur et al21 conducted a split-face, randomized, controlled trial of microneedling (1-mm depth) with TXA solution 10% vs microneedling (1-mm depth) with distilled water alone for 8 weeks (N=40). They graded participant responses to treatment using reductions in mMASI scores12 at every 2 weeks of follow-up (no response, minimal or poor response=0%–25%; partial or fair response=26%–50%; good response=51%–75%; and excellent response=>75%). They reported an overall reduction in mMASI scores for both the treatment side and the control side in all participants, showing a 65.92% improvement in mean mMASI scores on the treatment side vs 20.75% improvement on the control side at week 8. Both sides showed statistically significant reductions in mean mMASI scores (P<.05). Clinically, 40% (16/40) of participants showed an excellent response to combined treatment compared with 0% (0/40) to microneedling alone. Overall, patient satisfaction was similar across both groups. This study demonstrated that microneedling alone improves melasma, but a combination of microneedling plus TXA showed a better clinical reduction in melasma. However, the researchers did not follow up with participants posttreatment, so it remains unclear if the improved clinical outcomes were sustained long-term.21

Ebrahim et al22 reported that the combination of 0.5 mL TXA (4 mg/mL) and microneedling (0.25- to 1-mm depth) was effective for melasma. Although there was improvement within microneedling and TXA, the study also showed that intradermal injection of TXA was significant in reducing mean mMASI scores and improving melasma (P<.001). The reduction in mMASI scores for the group receiving intradermal injections of TXA (left side; 74.8% reduction in mean mMASI score) vs the group receiving microneedling application of TXA (right side; 73.6% reduction in mean mMASI score) was not statistically significant. These findings suggest that the mode of TXA application may not be critical in determining clinical responses to TXA treatment. Although there was no reported statistically significant difference in clinical outcomes between the 2 treatments, patient satisfaction was higher on the microneedling side. Only 8 of 50 participants (16%) experienced recurrence 3 months posttreatment.22

Saki et al23 compared the efficacy of topical hydroquinone (2%) to intradermal TXA injections in treating melasma. They found intradermal TXA injections to be a clinically effective mode of treatment.23

Sharma et al24 explored the efficacy and safety of oral TXA by randomly assigning 100 Indian patients (20 of whom withdrew before study completion) with melasma into 2 groups: group A received TXA 250 mg twice daily, and group B received intradermal microinjections of TXA (4 mg/mL) every 4 weeks. The MASI scores were assessed at 4-week intervals for a total of 12 weeks. There was a decrease in MASI scores in both groups, and there was no statistically significant difference in mean percentage reduction in MASI scores between the 2 routes of drug administration, further suggesting the effectiveness of TXA independent of administration route. Two patients in group A relapsed at 24 weeks, and there were no relapses in group B, which may suggest a minimal superiority of TXA plus microneedling at providing more sustainable results compared with oral TXA alone. A notable limitation of this study was a high dropout rate as well as lack of long-term follow-up with participants, limiting the generalizability of the conclusions.24

Cassiano et al25 assigned 64 women with melasma to 1 of 3 treatment groups or a control group to compare the effectiveness of microneedling (M group: 1.5 mm; 2 sessions), oral TXA (T group: 250 mg/d twice daily for 60 days), and a combination of microneedling (2 sessions) and oral TXA (MT group: 250 mg/d twice daily for 60 days)with placebo for clinically reducing melasma lesions. The intervention period was 60 days followed by a 60-day maintenance phase for a total study period of 120 days. The researchers evaluated mMASI scores, quality of life, and difference in colorimetric luminosity. All treatment groups showed a reduction in mMASI scores at both 30 days and 60 days, indicating improved melasma severity. The MT and T groups had more significant improvement at 30 days compared with the control group (P<.03), suggesting that microneedling plus TXA and TXA alone promote faster improvement in melasma lesions. By 60 days, the M, T, and MT groups outperformed the control group, with no significant differences between the M, T, and MT groups. However, at the 120-day maintenance follow-up, the T group did not maintain its improvement compared with the control group. The M and MT groups showed no significance difference in effectiveness at 120 days, suggesting that microneedling may promote less frequent relapse and sustained remission compared to TXA alone.25

Hydroquinone for Melasma—Additional studies on the use of TXA treatments show that TXA may be an equally effective alternative to the standard use of hydroquinone treatment. Shamsi Meymandi et al26 did not find a statistically significant difference in treatment with TXA plus microneedling vs the standard regimen of hydroquinone. More importantly, patient and physician satisfaction assessments were similar between the 2 groups. Compared to hydroquinone, nightly treatment is not necessary with microneedling and TXA.26

Xing et al27 supported these conclusions with their study. They compared 3 study arms for a duration of 12 weeks: group A received topical 1.8% liposomal TXA BID, group B received stamp-mode electric microneedling with 5% TXA weekly, and group C applied 2% ­hydroquinone cream nightly. The study concluded that all 3 groups showed a significant reduction in mean MI by the end of the study, but a better MI improvement was observed in groups B and C (both P<.001) compared with group A (P<.01).27

Zaky et al28 showed that both hydroquinone and combination treatment of TXA plus microneedling are effective at improving melasma lesions. Further studies are needed to definitively conclude if combination treatment is more efficacious than hydroquinone; if the combination is more effective, it provides a treatment option for patients with melasma who may not be good candidates for hydroquinone treatment.

Study Limitations—One limitation in all the studies evaluated is the sample size. Because they all had small sample sizes, it is difficult to definitively conclude that the combination TXA and microneedling is an effective and appropriate treatment for patients with melasma. Furthermore, the quality of these studies was mostly dependent on subjectivity of the mMASI scores. Future large randomized controlled trials with a diverse participant population are needed to assess the effectiveness of TXA and microneedling in melasma treatment.

Another limitation is that many of the studies did not follow the patients longitudinally, which did not allow for an evaluation of whether patients had a relapse of melasma. Due to the chronic nature of melasma and frequent disease recurrence, future longitudinal studies are needed to monitor for disease recurrence.

Conclusion

Tranexamic acid and microneedling are potential treatment options for patients with melasma, and combination therapy appears more effective than either TXA or microneedling alone at providing sustained improvement of melasma lesions. Combination therapy appears safe and well tolerated, but its effect on reducing long-term disease recurrence is yet to be established.

References
  1. Neagu N, Conforti C, Agozzino M, et al. Melasma treatment: a systematic review. J Dermatolog Treat. 2022;33:1816-1837. doi:10.1080/09546634.2021.1914313
  2. Ogbechie-Godec OA, Elbuluk N. Melasma: an up-to-date comprehensive review. Dermatol Ther (Heidelb). 2017;7:305-318. doi:10.1007/s13555-017-0194-1
  3. Mahajan VK, Patil A, Blicharz L, et al. Medical therapies for melasma. J Cosmet Dermatol. 2022;21:3707-3728. doi:10.1111/jocd.15242
  4. Rigopoulos D, Gregoriou S, Katsambas A. Hyperpigmentation and melasma. J Cosmet Dermatol. 2007;6:195-202. doi:10.1111/j.1473-2165.2007.00321.x
  5. Kagha K, Fabi S, Goldman M. Melasma’s impact on quality of life. J Drugs Dermatol. 2020;19:184-187. doi:10.36849/JDD.2020.4663
  6. Lutfi RJ, Fridmanis M, Misiunas AL, et al. Association of melasma with thyroid autoimmunity and other thyroidal abnormalities and their relationship to the origin of the melasma. J Clin Endocrinol Metab. 1985;61:28-31. doi:10.1210/jcem-61-1-28
  7. Handel AC, Lima PB, Tonolli VM, et al. Risk factors for facial melasma in women: a case-control study. Br J Dermatol. 2014;171:588-594. doi:10.1111/bjd.13059
  8. Filoni A, Mariano M, Cameli N. Melasma: how hormones can modulate skin pigmentation. J Cosmet Dermatol. 2019;18:458-463. doi:10.1111/jocd.12877
  9. Rodrigues M, Pandya AG. Melasma: clinical diagnosis and management options. Australasian J Dermatol. 2015;56:151-163.
  10. Huerth KA, Hassan S, Callender VD. Therapeutic insights in melasma and hyperpigmentation management. J Drugs Dermatol. 2019;18:718-727.
  11. Pandya AG, Hynan LS, Bhore R, et al. Reliability assessment and validation of the Melasma Area and Severity Index (MASI) and a new modified MASI scoring method. J Am Acad Dermatol. 2011;64:78-­83.e832. doi:10.1016/j.jaad.2009.10.051
  12. Rodrigues M, Ayala-Cortés AS, Rodríguez-Arámbula A, et al. Interpretability of the modified Melasma Area and Severity Index (mMASI). JAMA Dermatol. 2016;152:1051-1052. doi:10.1001/jamadermatol.2016.1006
  13. Ikino JK, Nunes DH, da Silva VPM, et al. Melasma and assessment of the quality of life in Brazilian women. An Bras Dermatol. 2015;90:196-200. doi:10.1590/abd1806-4841.20152771
  14. Taraz M, Niknam S, Ehsani AH. Tranexamic acid in treatment of melasma: a comprehensive review of clinical studies. Dermatolog Ther. 2017;30:E12465. doi:10.1111/dth.12465
  15. Bala HR, Lee S, Wong C, et al. Oral tranexamic acid for the treatment of melasma: a review. Dermatol Surg. 2018;44:814-825. doi:10.1097/DSS.0000000000001518
  16. Singh A, Yadav S. Microneedling: advances and widening horizons. Indian Dermatol Online J. 2016;7:244-254. doi:10.4103/2229-5178.185468
  17. Del Rosario E, Florez-Pollack S, Zapata L, et al. Randomized, placebo-controlled, double-blind study of oral tranexamic acid in the treatment of moderate-to-severe melasma. J Am Acad Dermatol. 2018;78:363-369. doi:10.1016/j.jaad.2017.09.053
  18. El Attar Y, Doghaim N, El Far N, et al. Efficacy and safety of tranexamic acid versus vitamin C after microneedling in treatment of melasma: clinical and dermoscopic study. J Cosmet Dermatol. 2022;21:2817-2825. doi:10.1111/jocd.14538
  19. Saleh FY, Abdel-Azim ES, Ragaie MH, et al. Topical tranexamic acid with microneedling versus microneedling alone in treatment of melasma: clinical, histopathologic, and immunohistochemical study. J Egyptian Womens Dermatolog Soc. 2019;16:89-96. doi:10.4103/jewd.jewd_25_19
  20. Xu Y, Ma R, Juliandri J, et al. Efficacy of functional microarray of microneedles combined with topical tranexamic acid for melasma: a randomized, self-controlled, split-face study. Medicine (Baltimore). 2017;96:e6897. doi:10.1097/MD.0000000000006897
  21. Kaur A, Bhalla M, Pal Thami G, et al. Clinical efficacy of topical tranexamic acid with microneedling in melasma. Dermatol Surg. 2020;46:E96-E101. doi:10.1097/DSS.0000000000002520
  22. Ebrahim HM, Said Abdelshafy A, Khattab F, et al. Tranexamic acid for melasma treatment: a split-face study. Dermatol Surg. 2020;46:E102-E107. doi:10.1097/DSS.0000000000002449
  23. Saki N, Darayesh M, Heiran A. Comparing the efficacy of topical hydroquinone 2% versus intradermal tranexamic acid microinjections in treating melasma: a split-face controlled trial. J Dermatolog Treat. 2018;29:405-410. doi:10.1080/09546634.2017.1392476
  24. Sharma R, Mahajan VK, Mehta KS, et al. Therapeutic efficacy and safety of oral tranexamic acid and that of tranexamic acid local infiltration with microinjections in patients with melasma: a comparative study. Clin Exp Dermatol. 2017;42:728-734. doi:10.1111/ced.13164
  25. Cassiano D, Esposito ACC, Hassun K, et al. Efficacy and safety of microneedling and oral tranexamic acid in the treatment of facial melasma in women: an open, evaluator-blinded, randomized clinical trial. J Am Acad Dermatol. 2020;83:1176-1178. doi:10.1016/j.jaad.2020.02.002
  26. Shamsi Meymandi S, Mozayyeni A, Shamsi Meymandi M, et al. Efficacy of microneedling plus topical 4% tranexamic acid solution vs 4% hydroquinone in the treatment of melasma: a single-blind randomized clinical trial. J Cosmet Dermatol. 2020;19:2906-2911. doi:10.1111/jocd.13392
  27. Xing X, Chen L, Xu Z, et al. The efficacy and safety of topical tranexamic acid (liposomal or lotion with microneedling) versus conventional hydroquinone in the treatment of melasma. J Cosmet Dermatol. 2020;19:3238-3244. doi:10.1111/jocd.13810
  28. Zaky MS, Obaid ZM, Khalil EA, et al. Microneedling-assisted topical tranexamic acid solution versus 4% hydroquinone for treating melasma: a split-face randomized study. J Cosmet Dermatol. 2021;20:4011-4016. doi:10.1111/jocd.14440
References
  1. Neagu N, Conforti C, Agozzino M, et al. Melasma treatment: a systematic review. J Dermatolog Treat. 2022;33:1816-1837. doi:10.1080/09546634.2021.1914313
  2. Ogbechie-Godec OA, Elbuluk N. Melasma: an up-to-date comprehensive review. Dermatol Ther (Heidelb). 2017;7:305-318. doi:10.1007/s13555-017-0194-1
  3. Mahajan VK, Patil A, Blicharz L, et al. Medical therapies for melasma. J Cosmet Dermatol. 2022;21:3707-3728. doi:10.1111/jocd.15242
  4. Rigopoulos D, Gregoriou S, Katsambas A. Hyperpigmentation and melasma. J Cosmet Dermatol. 2007;6:195-202. doi:10.1111/j.1473-2165.2007.00321.x
  5. Kagha K, Fabi S, Goldman M. Melasma’s impact on quality of life. J Drugs Dermatol. 2020;19:184-187. doi:10.36849/JDD.2020.4663
  6. Lutfi RJ, Fridmanis M, Misiunas AL, et al. Association of melasma with thyroid autoimmunity and other thyroidal abnormalities and their relationship to the origin of the melasma. J Clin Endocrinol Metab. 1985;61:28-31. doi:10.1210/jcem-61-1-28
  7. Handel AC, Lima PB, Tonolli VM, et al. Risk factors for facial melasma in women: a case-control study. Br J Dermatol. 2014;171:588-594. doi:10.1111/bjd.13059
  8. Filoni A, Mariano M, Cameli N. Melasma: how hormones can modulate skin pigmentation. J Cosmet Dermatol. 2019;18:458-463. doi:10.1111/jocd.12877
  9. Rodrigues M, Pandya AG. Melasma: clinical diagnosis and management options. Australasian J Dermatol. 2015;56:151-163.
  10. Huerth KA, Hassan S, Callender VD. Therapeutic insights in melasma and hyperpigmentation management. J Drugs Dermatol. 2019;18:718-727.
  11. Pandya AG, Hynan LS, Bhore R, et al. Reliability assessment and validation of the Melasma Area and Severity Index (MASI) and a new modified MASI scoring method. J Am Acad Dermatol. 2011;64:78-­83.e832. doi:10.1016/j.jaad.2009.10.051
  12. Rodrigues M, Ayala-Cortés AS, Rodríguez-Arámbula A, et al. Interpretability of the modified Melasma Area and Severity Index (mMASI). JAMA Dermatol. 2016;152:1051-1052. doi:10.1001/jamadermatol.2016.1006
  13. Ikino JK, Nunes DH, da Silva VPM, et al. Melasma and assessment of the quality of life in Brazilian women. An Bras Dermatol. 2015;90:196-200. doi:10.1590/abd1806-4841.20152771
  14. Taraz M, Niknam S, Ehsani AH. Tranexamic acid in treatment of melasma: a comprehensive review of clinical studies. Dermatolog Ther. 2017;30:E12465. doi:10.1111/dth.12465
  15. Bala HR, Lee S, Wong C, et al. Oral tranexamic acid for the treatment of melasma: a review. Dermatol Surg. 2018;44:814-825. doi:10.1097/DSS.0000000000001518
  16. Singh A, Yadav S. Microneedling: advances and widening horizons. Indian Dermatol Online J. 2016;7:244-254. doi:10.4103/2229-5178.185468
  17. Del Rosario E, Florez-Pollack S, Zapata L, et al. Randomized, placebo-controlled, double-blind study of oral tranexamic acid in the treatment of moderate-to-severe melasma. J Am Acad Dermatol. 2018;78:363-369. doi:10.1016/j.jaad.2017.09.053
  18. El Attar Y, Doghaim N, El Far N, et al. Efficacy and safety of tranexamic acid versus vitamin C after microneedling in treatment of melasma: clinical and dermoscopic study. J Cosmet Dermatol. 2022;21:2817-2825. doi:10.1111/jocd.14538
  19. Saleh FY, Abdel-Azim ES, Ragaie MH, et al. Topical tranexamic acid with microneedling versus microneedling alone in treatment of melasma: clinical, histopathologic, and immunohistochemical study. J Egyptian Womens Dermatolog Soc. 2019;16:89-96. doi:10.4103/jewd.jewd_25_19
  20. Xu Y, Ma R, Juliandri J, et al. Efficacy of functional microarray of microneedles combined with topical tranexamic acid for melasma: a randomized, self-controlled, split-face study. Medicine (Baltimore). 2017;96:e6897. doi:10.1097/MD.0000000000006897
  21. Kaur A, Bhalla M, Pal Thami G, et al. Clinical efficacy of topical tranexamic acid with microneedling in melasma. Dermatol Surg. 2020;46:E96-E101. doi:10.1097/DSS.0000000000002520
  22. Ebrahim HM, Said Abdelshafy A, Khattab F, et al. Tranexamic acid for melasma treatment: a split-face study. Dermatol Surg. 2020;46:E102-E107. doi:10.1097/DSS.0000000000002449
  23. Saki N, Darayesh M, Heiran A. Comparing the efficacy of topical hydroquinone 2% versus intradermal tranexamic acid microinjections in treating melasma: a split-face controlled trial. J Dermatolog Treat. 2018;29:405-410. doi:10.1080/09546634.2017.1392476
  24. Sharma R, Mahajan VK, Mehta KS, et al. Therapeutic efficacy and safety of oral tranexamic acid and that of tranexamic acid local infiltration with microinjections in patients with melasma: a comparative study. Clin Exp Dermatol. 2017;42:728-734. doi:10.1111/ced.13164
  25. Cassiano D, Esposito ACC, Hassun K, et al. Efficacy and safety of microneedling and oral tranexamic acid in the treatment of facial melasma in women: an open, evaluator-blinded, randomized clinical trial. J Am Acad Dermatol. 2020;83:1176-1178. doi:10.1016/j.jaad.2020.02.002
  26. Shamsi Meymandi S, Mozayyeni A, Shamsi Meymandi M, et al. Efficacy of microneedling plus topical 4% tranexamic acid solution vs 4% hydroquinone in the treatment of melasma: a single-blind randomized clinical trial. J Cosmet Dermatol. 2020;19:2906-2911. doi:10.1111/jocd.13392
  27. Xing X, Chen L, Xu Z, et al. The efficacy and safety of topical tranexamic acid (liposomal or lotion with microneedling) versus conventional hydroquinone in the treatment of melasma. J Cosmet Dermatol. 2020;19:3238-3244. doi:10.1111/jocd.13810
  28. Zaky MS, Obaid ZM, Khalil EA, et al. Microneedling-assisted topical tranexamic acid solution versus 4% hydroquinone for treating melasma: a split-face randomized study. J Cosmet Dermatol. 2021;20:4011-4016. doi:10.1111/jocd.14440
Issue
Cutis - 114(2)
Issue
Cutis - 114(2)
Page Number
E15-E23
Page Number
E15-E23
Publications
Publications
Topics
Article Type
Display Headline
The Use of Tranexamic Acid and Microneedling in the Treatment of Melasma: A Systematic Review
Display Headline
The Use of Tranexamic Acid and Microneedling in the Treatment of Melasma: A Systematic Review
Sections
Inside the Article

Practice Points

  • Combination therapy with tranexamic acid (TXA) and microneedling is a safe and effective treatment for melasma.
  • Combining TXA with microneedling may result in decreased melasma relapse rates.
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.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

Epidermal Tumors Arising on Donor Sites From Autologous Skin Grafts: A Systematic Review

Article Type
Changed
Wed, 08/07/2024 - 13:08
Display Headline
Epidermal Tumors Arising on Donor Sites From Autologous Skin Grafts: A Systematic Review

Skin grafting is a surgical technique used to cover skin defects resulting from the removal of skin tumors, ulcers, or burn injuries.1-3 Complications can occur at both donor and recipient sites and may include bleeding, hematoma/seroma formation, postoperative pain, infection, scarring, paresthesia, skin pigmentation, graft contracture, and graft failure.1,2,4,5 The development of epidermal tumors is not commonly reported among the complications of skin grafting; however, cases of epidermal tumor development on skin graft donor sites during the postoperative period have been reported.6-12

We performed a systematic review of the literature for cases of epidermal tumor development on skin graft donor sites in patients undergoing autologous skin graft surgery. We present the clinical characteristics of these cases and discuss the nature of these tumors.

Methods

Search Strategy and Study Selection—A literature search was conducted by 2 independent researchers (Z.P. and V.P.) for articles published before December 2022 in the following databases: MEDLINE/PubMed, Web of Science, Scopus, Cochrane Library, OpenGrey, Google Scholar, and WorldCat. Search terms included all possible combinations of the following: keratoacanthoma, molluscum sebaceum, basal cell carcinoma, squamous cell carcinoma, acanthoma, wart, Merkel cell carcinoma, verruca, Bowen disease, keratosis, skin cancer, cutaneous cancer, skin neoplasia, cutaneous neoplasia, and skin tumor. The literature search terms were selected based on the World Health Organization classification of skin tumors.13 Manual bibliography checks were performed on all eligible search results for possible relevant studies. Discrepancies were resolved through discussion and, if needed, mediation by a third researcher (N.C.). To be included, a study had to report a case(s) of epidermal tumor(s) that was confirmed by histopathology and arose on a graft donor site in a patient receiving autologous skin grafts for any reason. No language, geographic, or report date restrictions were set.

Data Extraction, Quality Assessment, and Statistical Analysis—We adhered to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines.14 Two independent researchers (Z.P. and V.P.) retrieved the data from the included studies. We have used the terms case and patient interchangeably, and 1 month was measured as 4 weeks for simplicity. Disagreements were resolved by discussion and mediation by a third researcher (N.C.). The quality of the included studies was assessed by 2 researchers (M.P. and V.P.) using the tool proposed by Murad et al.15

We used descriptive statistical analysis to analyze clinical characteristics of the included cases. We performed separate descriptive analyses based on the most frequently reported types of epidermal tumors and compared the differences between different groups using the Mann-Whitney U test, χ2 test, and Fisher exact test. The level of significance was set at P<.05. All statistical analyses were conducted using SPSS (version 29).

 

 

Results

Literature Search and Characteristics of Included Studies—The initial literature search identified 1378 studies, which were screened based on title and abstract. After removing duplicate and irrelevant studies and evaluating the full text of eligible studies, 31 studies (4 case series and 27 case reports) were included in the systematic review (Figure).6-12,16-39 Quality assessment of the included studies is presented in Table 1.

Flowchart for a systematic review and meta-analysis using Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) criteria for articles published before December 2022.

Clinical Characteristics of Included Patients—Our systematic review included 36 patients with a mean age of 63 years and a male to female ratio of 2:1. The 2 most common causes for skin grafting were burn wounds and surgical excision of skin tumors. Most grafts were harvested from the thighs. The development of a solitary lesion on the donor area was reported in two-thirds of the patients, while more than 1 lesion developed in the remaining one-third of patients. The median time to tumor development was 6.5 weeks. In most cases, a split-thickness skin graft was used.

Cutaneous squamous cell carcinomas (CSCCs) were found in 23 patients, with well-differentiated CSCCs in 19 of these cases. Additionally, keratoacanthomas (KAs) were found in 10 patients. The majority of patients underwent surgical excision of the tumor. The median follow-up time was 12 months, during which recurrences were noted in a small percentage of cases. Clinical characteristics of included patients are presented in Table 2.

Comparison of Variables Between CSCC and KA Groups—The most common diagnoses among the included patients were CSCC and KA. There were no significant differences between the groups in clinical variables, including age, sex, reason for grafting, time to occurrence, and rate of recurrence (Table 3).

 

 

Comment

Reasons for Tumor Development on Skin Graft Donor Sites—The etiology behind epidermal tumor development on graft donor sites is unclear. According to one theory, iatrogenic contamination of the donor site during the removal of a primary epidermal tumor could be responsible. However, contemporary surgical procedures dictate the use of different sets of instruments for separate surgical sites. Moreover, this theory cannot explain the occurrence of epidermal tumors on donor sites in patients who have undergone skin grafting for the repair of burn wounds.37

Another theory suggests that hematogenous and/or lymphatic spread can occur from the site of the primary epidermal tumor to the donor site, which has increased vascularization.16,37 However, this theory also fails to provide an explanation for the development of epidermal tumors in patients who receive skin grafts for burn wounds.

A third theory states that the microenvironment of the donor site is key to tumor development. The donor site undergoes acute inflammation due to the trauma from harvesting the skin graft. According to this theory, acute inflammation could promote neoplastic growth and thus explain the development of epidermal tumors on the donor site.8,26 However, the relationship between acute inflammation and carcinogenesis remains unclear. What is known to date is that the development of CSCC has been documented primarily in chronically inflamed tissues, whereas the development of KA—a variant of CSCC with distinctive and more benign clinical characteristics—can be expected in the setting of acute trauma-related inflammation.13,40,41

Based on our systematic review, we propose that well-differentiated CSCC on graft donor sites might actually be misdiagnosed KA, given that the histopathologic differential diagnosis between CSCC and KA is extremely challenging.42 This hypothesis could explain the development of well-differentiated CSCC and KA on graft donor sites.

Conclusion

Development of CSCC and KA on graft donor sites can be listed among the postoperative complications of autologous skin grafting. Patients and physicians should be aware of this potential complication, and donor sites should be monitored for the occurrence of epidermal tumors.

References
  1. Adams DC, Ramsey ML. Grafts in dermatologic surgery: review and update on full- and split-thickness skin grafts, free cartilage grafts, and composite grafts. Dermatologic Surg. 2005;31(8, pt 2):1055-1067. doi:10.1111/j.1524-4725.2005.31831
  2. Shimizu R, Kishi K. Skin graft. Plast Surg Int. 2012;2012:563493. doi:10.1155/2012/563493
  3. Reddy S, El-Haddawi F, Fancourt M, et al. The incidence and risk factors for lower limb skin graft failure. Dermatol Res Pract. 2014;2014:582080. doi:10.1155/2014/582080
  4. Coughlin MJ, Dockery GD, Crawford ME, et al. Lower Extremity Soft Tissue & Cutaneous Plastic Surgery. 2nd ed. Saunders Ltd; 2012.
  5. Herskovitz I, Hughes OB, Macquhae F, et al. Epidermal skin grafting. Int Wound J. 2016;13(suppl 3):52-56. doi:10.1111/iwj.12631
  6. Wright H, McKinnell TH, Dunkin C. Recurrence of cutaneous squamous cell carcinoma at remote limb donor site. J Plast Reconstr Aesthet Surg. 2012;65:1265-1266. doi:10.1016/j.bjps.2012.01.022
  7. Thomas W, Rezzadeh K, Rossi K, et al. Squamous cell carcinoma arising at a skin graft donor site: case report and review of the literature. Plast Surg Case Stud. 2021;7:2513826X211008425. doi:10.1177/2513826X211008425
  8. Ponnuvelu G, Ng MFY, Connolly CM, et al. Inflammation to skin malignancy, time to rethink the link: SCC in skin graft donor sites. Surgeon. 2011;9:168-169. doi:10.1016/j.surge.2010.08.006
  9. Noori VJ, Trehan K, Savetamal A, et al. New onset squamous cell carcinoma in previous split-thickness skin graft donor site. Int J Surg. 2018;52:16-19. doi:10.1016/j.ijsu.2018.01.047
  10. Morritt DG, Khandwala AR. The development of squamous cell carcinomas in split-thickness skin graft donor sites. Eur J Plast Surg. 2013;36:377-380.
  11. McCormick M, Miotke S. Squamous cell carcinoma at split thickness skin graft donor site: a case report and review of the literature. J Burn Care Res. 2023;44:210-213. doi:10.1093/jbcr/irac137
  12. Haik J, Georgiou I, Farber N, et al. Squamous cell carcinoma arising in a split-thickness skin graft donor site. Burns. 2008;34:891-893. doi:10.1016/j.burns.2007.06.006
  13. Elder DE, Massi D, Scolyer RA WR. WHO Classification of Skin Tumours. 4th ed. IARC Press; 2018.
  14. Moher D, Liberati A, Tetzlaff J, et al. Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. Ann Intern Med. 2009;151:264-269, W64. doi:10.7326/0003-4819-151-4-200908180-00135
  15. Murad MH, Sultan S, Haffar S, et al. Methodological quality and synthesis of case series and case reports. BMJ. 2018;23:60-63. doi:10.1136/bmjebm-2017-110853
  16. de Moraes LPB, Burchett I, Nicholls S, et al. Large solitary distant metastasis of cutaneous squamous cell carcinoma to skin graft site with complete response following definitive radiotherapy. Int J Bioautomation. 2017;21:103-108.
  17. Nagase K, Suzuki Y, Misago N, et al. Acute development of keratoacanthoma at a full-thickness skin graft donor site shortly after surgery. J Dermatol. 2016;43:1232-1233. doi:10.1111/1346-8138.13368
  18. Taylor CD, Snelling CF, Nickerson D, et al. Acute development of invasive squamous cell carcinoma in a split-thickness skin graft donor site. J Burn Care Rehabil. 1998;19:382-385. doi:10.1097/00004630-199809000-00004
  19. de Delas J, Leache A, Vazquez Doval J, et al. Keratoacanthoma over the donor site of a laminar skin graft. Med Cutan Ibero Lat Am. 1989;17:225-228.
  20. Neilson D, Emerson DJ, Dunn L. Squamous cell carcinoma of skin developing in a skin graft donor site. Br J Plast Surg. 1988;41:417-419. doi:10.1016/0007-1226(88)90086-0
  21. May JT, Patil YJ. Keratoacanthoma-type squamous cell carcinoma developing in a skin graft donor site after tumor extirpation at a distant site. Ear Nose Throat J. 2010;89:E11-E13.
  22. Imbernón-Moya A, Vargas-Laguna E, Lobato-Berezo A, et al. Simultaneous onset of basal cell carcinoma over skin graft and donor site. JAAD Case Rep. 2015;1:244-246. doi:10.1016/j.jdcr.2015.05.004
  23. Lee S, Coutts I, Ryan A, et al. Keratoacanthoma formation after skin grafting: a brief report and pathophysiological hypothesis. Australas J Dermatol. 2017;58:e117-e119. doi:10.1111/ajd.12501
  24. Hammond JS, Thomsen S, Ward CG. Scar carcinoma arising acutelyin a skin graft donor site. J Trauma. 1987;27:681-683. doi:10.1097/00005373-198706000-00017
  25. Herard C, Arnaud D, Goga D, et al. Rapid onset of squamous cell carcinoma in a thin skin graft donor site. Ann Dermatol Venereol. 2016;143:457-461. doi:10.1016/j.annder.2015.03.027
  26. Ibrahim A, Moisidis E. Case series: rapidly growing squamous cell carcinoma after cutaneous surgical intervention. JPRAS Open. 2017;14:27-32. doi:10.1016/j.jpra.2017.08.004
  27. Kearney L, Dolan RT, Parfrey NA, et al. Squamous cell carcinoma arising in a skin graft donor site following melanoma extirpation at a distant site: a case report and review of the literature. JPRAS Open. 2015;3:35-38. doi:10.1016/j.jpra.2015.02.002
  28. Clark MA, Guitart J, Gerami P, et al. Eruptive keratoacanthomatous atypical squamous proliferations (KASPs) arising in skin graft sites. JAAD Case Rep. 2015;1:274-276. doi:10.1016/j.jdcr.2015.06.009
  29. Aloraifi F, Mulgrew S, James NK. Secondary Merkel cell carcinoma arising from a graft donor site. J Cutan Med Surg. 2017;21:167-169. doi:10.1177/1203475416676805
  30. Abadir R, Zurowski S. Case report: squamous cell carcinoma of the skin in both palms, axillary node, donor skin graft site and both soles—associated hyperkeratosis and porokeratosis. Br J Radiol. 1994;67:507-510. doi:10.1259/0007-1285-67-797-507
  31. Griffiths RW. Keratoacanthoma observed. Br J Plast Surg. 2004;57:485-501. doi:10.1016/j.bjps.2004.05.007
  32. Marous M, Brady K. Cutaneous squamous cell carcinoma arising in a split thickness skin graft donor site in a patient with systemic lupus erythematosus. Dermatologic Surg. 2021;47:1106-1107. doi:10.1097/DSS.0000000000002955
  33. Dibden FA, Fowler M. The multiple growth of molluscum sebaceum in donor and recipient sites of skin graft. Aust N Z J Surg. 1955;25:157-159. doi:10.1111/j.1445-2197.1955.tb05122.x
  34. Jeremiah BS. Squamous cell carcinoma development on donor area following removal of a split thickness skin graft. Plast Reconstr Surg. 1948;3:718-721.
  35. Tamir G, Morgenstern S, Ben-Amitay D, et al. Synchronous appearance of keratoacanthomas in burn scar and skin graft donor site shortly after injury. J Am Acad Dermatol. 1999;40(5, pt 2):870-871. doi:10.1053/jd.1999.v40.a94419
  36. Hamilton SA, Dickson WA, O’Brien CJ. Keratoacanthoma developing in a split skin graft donor site. Br J Plast Surg. 1997;50:560-561. doi:10.1016/s0007-1226(97)91308-4
  37. Hussain A, Ekwobi C, Watson S. Metastatic implantation squamous cell carcinoma in a split-thickness skin graft donor site. J Plast Reconstr Aesthet Surg. 2011;64:690-692. doi:10.1016/j.bjps.2010.06.004
  38. Wulsin JH. Keratoacanthoma: a benign cutaneous tumors arising in a skin graft donor site. Am Surg. 1958;24:689-692.
  39. Davis L, Butler D. Acute development of squamous cell carcinoma in a split-thickness skin graft donor site [abstract]. J Am Acad Dermatol. 2012;66:AB208. doi:10.1016/j.jaad.2011.11.874
  40. Shacter E, Weitzman SA. Chronic inflammation and cancer. Oncology (Williston Park). 2002;16:217-226, 229; discussion 230-232.
  41.  Piotrowski I, Kulcenty K, Suchorska W. Interplay between inflammation and cancer. Reports Pract Oncol Radiother. 2020;25:422-427. doi:10.1016/j.rpor.2020.04.004
  42. Carr RA, Houghton JP. Histopathologists’ approach to keratoacanthoma: a multisite survey of regional variation in Great Britain and Ireland. J Clin Pathol. 2014;67:637-638. doi:10.1136/jclinpath-2014-202255
Article PDF
Author and Disclosure Information

Dr. Chaitidis is from the Department of Dermatology and Venereology, 424 General Military Training Hospital, Thessaloniki, Greece. Dr. Papadopoulou is from the 3rd Department of Pediatrics, Hippokration General Hospital, Aristotle University of Thessaloniki. Dr. Paraschou is from the 2nd Department of Pulmonology, University General Hospital Attikon, National and Kapodistrian University of Athens, Haidari, Greece, and Hellenic Police Medical Center, Thessaloniki. Dr. Panagiotidis is from the 1st Department of Surgery, Papageorgiou General Hospital, Thessaloniki.

The authors report no conflict of interest.

Correspondence: Nikolaos Chaitidis, MD, Department of Dermatology and Venereology, 424 General Military Training Hospital, Thessaloniki, Greece, Perifereiaki Odos Neas Eukarpias 56429 (nchaitidauth@gmail.com).

Cutis. 2024 August;114(2):E6-E12. doi:10.12788/cutis.1079

Issue
Cutis - 114(2)
Publications
Topics
Page Number
E6-E12
Sections
Author and Disclosure Information

Dr. Chaitidis is from the Department of Dermatology and Venereology, 424 General Military Training Hospital, Thessaloniki, Greece. Dr. Papadopoulou is from the 3rd Department of Pediatrics, Hippokration General Hospital, Aristotle University of Thessaloniki. Dr. Paraschou is from the 2nd Department of Pulmonology, University General Hospital Attikon, National and Kapodistrian University of Athens, Haidari, Greece, and Hellenic Police Medical Center, Thessaloniki. Dr. Panagiotidis is from the 1st Department of Surgery, Papageorgiou General Hospital, Thessaloniki.

The authors report no conflict of interest.

Correspondence: Nikolaos Chaitidis, MD, Department of Dermatology and Venereology, 424 General Military Training Hospital, Thessaloniki, Greece, Perifereiaki Odos Neas Eukarpias 56429 (nchaitidauth@gmail.com).

Cutis. 2024 August;114(2):E6-E12. doi:10.12788/cutis.1079

Author and Disclosure Information

Dr. Chaitidis is from the Department of Dermatology and Venereology, 424 General Military Training Hospital, Thessaloniki, Greece. Dr. Papadopoulou is from the 3rd Department of Pediatrics, Hippokration General Hospital, Aristotle University of Thessaloniki. Dr. Paraschou is from the 2nd Department of Pulmonology, University General Hospital Attikon, National and Kapodistrian University of Athens, Haidari, Greece, and Hellenic Police Medical Center, Thessaloniki. Dr. Panagiotidis is from the 1st Department of Surgery, Papageorgiou General Hospital, Thessaloniki.

The authors report no conflict of interest.

Correspondence: Nikolaos Chaitidis, MD, Department of Dermatology and Venereology, 424 General Military Training Hospital, Thessaloniki, Greece, Perifereiaki Odos Neas Eukarpias 56429 (nchaitidauth@gmail.com).

Cutis. 2024 August;114(2):E6-E12. doi:10.12788/cutis.1079

Article PDF
Article PDF

Skin grafting is a surgical technique used to cover skin defects resulting from the removal of skin tumors, ulcers, or burn injuries.1-3 Complications can occur at both donor and recipient sites and may include bleeding, hematoma/seroma formation, postoperative pain, infection, scarring, paresthesia, skin pigmentation, graft contracture, and graft failure.1,2,4,5 The development of epidermal tumors is not commonly reported among the complications of skin grafting; however, cases of epidermal tumor development on skin graft donor sites during the postoperative period have been reported.6-12

We performed a systematic review of the literature for cases of epidermal tumor development on skin graft donor sites in patients undergoing autologous skin graft surgery. We present the clinical characteristics of these cases and discuss the nature of these tumors.

Methods

Search Strategy and Study Selection—A literature search was conducted by 2 independent researchers (Z.P. and V.P.) for articles published before December 2022 in the following databases: MEDLINE/PubMed, Web of Science, Scopus, Cochrane Library, OpenGrey, Google Scholar, and WorldCat. Search terms included all possible combinations of the following: keratoacanthoma, molluscum sebaceum, basal cell carcinoma, squamous cell carcinoma, acanthoma, wart, Merkel cell carcinoma, verruca, Bowen disease, keratosis, skin cancer, cutaneous cancer, skin neoplasia, cutaneous neoplasia, and skin tumor. The literature search terms were selected based on the World Health Organization classification of skin tumors.13 Manual bibliography checks were performed on all eligible search results for possible relevant studies. Discrepancies were resolved through discussion and, if needed, mediation by a third researcher (N.C.). To be included, a study had to report a case(s) of epidermal tumor(s) that was confirmed by histopathology and arose on a graft donor site in a patient receiving autologous skin grafts for any reason. No language, geographic, or report date restrictions were set.

Data Extraction, Quality Assessment, and Statistical Analysis—We adhered to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines.14 Two independent researchers (Z.P. and V.P.) retrieved the data from the included studies. We have used the terms case and patient interchangeably, and 1 month was measured as 4 weeks for simplicity. Disagreements were resolved by discussion and mediation by a third researcher (N.C.). The quality of the included studies was assessed by 2 researchers (M.P. and V.P.) using the tool proposed by Murad et al.15

We used descriptive statistical analysis to analyze clinical characteristics of the included cases. We performed separate descriptive analyses based on the most frequently reported types of epidermal tumors and compared the differences between different groups using the Mann-Whitney U test, χ2 test, and Fisher exact test. The level of significance was set at P<.05. All statistical analyses were conducted using SPSS (version 29).

 

 

Results

Literature Search and Characteristics of Included Studies—The initial literature search identified 1378 studies, which were screened based on title and abstract. After removing duplicate and irrelevant studies and evaluating the full text of eligible studies, 31 studies (4 case series and 27 case reports) were included in the systematic review (Figure).6-12,16-39 Quality assessment of the included studies is presented in Table 1.

Flowchart for a systematic review and meta-analysis using Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) criteria for articles published before December 2022.

Clinical Characteristics of Included Patients—Our systematic review included 36 patients with a mean age of 63 years and a male to female ratio of 2:1. The 2 most common causes for skin grafting were burn wounds and surgical excision of skin tumors. Most grafts were harvested from the thighs. The development of a solitary lesion on the donor area was reported in two-thirds of the patients, while more than 1 lesion developed in the remaining one-third of patients. The median time to tumor development was 6.5 weeks. In most cases, a split-thickness skin graft was used.

Cutaneous squamous cell carcinomas (CSCCs) were found in 23 patients, with well-differentiated CSCCs in 19 of these cases. Additionally, keratoacanthomas (KAs) were found in 10 patients. The majority of patients underwent surgical excision of the tumor. The median follow-up time was 12 months, during which recurrences were noted in a small percentage of cases. Clinical characteristics of included patients are presented in Table 2.

Comparison of Variables Between CSCC and KA Groups—The most common diagnoses among the included patients were CSCC and KA. There were no significant differences between the groups in clinical variables, including age, sex, reason for grafting, time to occurrence, and rate of recurrence (Table 3).

 

 

Comment

Reasons for Tumor Development on Skin Graft Donor Sites—The etiology behind epidermal tumor development on graft donor sites is unclear. According to one theory, iatrogenic contamination of the donor site during the removal of a primary epidermal tumor could be responsible. However, contemporary surgical procedures dictate the use of different sets of instruments for separate surgical sites. Moreover, this theory cannot explain the occurrence of epidermal tumors on donor sites in patients who have undergone skin grafting for the repair of burn wounds.37

Another theory suggests that hematogenous and/or lymphatic spread can occur from the site of the primary epidermal tumor to the donor site, which has increased vascularization.16,37 However, this theory also fails to provide an explanation for the development of epidermal tumors in patients who receive skin grafts for burn wounds.

A third theory states that the microenvironment of the donor site is key to tumor development. The donor site undergoes acute inflammation due to the trauma from harvesting the skin graft. According to this theory, acute inflammation could promote neoplastic growth and thus explain the development of epidermal tumors on the donor site.8,26 However, the relationship between acute inflammation and carcinogenesis remains unclear. What is known to date is that the development of CSCC has been documented primarily in chronically inflamed tissues, whereas the development of KA—a variant of CSCC with distinctive and more benign clinical characteristics—can be expected in the setting of acute trauma-related inflammation.13,40,41

Based on our systematic review, we propose that well-differentiated CSCC on graft donor sites might actually be misdiagnosed KA, given that the histopathologic differential diagnosis between CSCC and KA is extremely challenging.42 This hypothesis could explain the development of well-differentiated CSCC and KA on graft donor sites.

Conclusion

Development of CSCC and KA on graft donor sites can be listed among the postoperative complications of autologous skin grafting. Patients and physicians should be aware of this potential complication, and donor sites should be monitored for the occurrence of epidermal tumors.

Skin grafting is a surgical technique used to cover skin defects resulting from the removal of skin tumors, ulcers, or burn injuries.1-3 Complications can occur at both donor and recipient sites and may include bleeding, hematoma/seroma formation, postoperative pain, infection, scarring, paresthesia, skin pigmentation, graft contracture, and graft failure.1,2,4,5 The development of epidermal tumors is not commonly reported among the complications of skin grafting; however, cases of epidermal tumor development on skin graft donor sites during the postoperative period have been reported.6-12

We performed a systematic review of the literature for cases of epidermal tumor development on skin graft donor sites in patients undergoing autologous skin graft surgery. We present the clinical characteristics of these cases and discuss the nature of these tumors.

Methods

Search Strategy and Study Selection—A literature search was conducted by 2 independent researchers (Z.P. and V.P.) for articles published before December 2022 in the following databases: MEDLINE/PubMed, Web of Science, Scopus, Cochrane Library, OpenGrey, Google Scholar, and WorldCat. Search terms included all possible combinations of the following: keratoacanthoma, molluscum sebaceum, basal cell carcinoma, squamous cell carcinoma, acanthoma, wart, Merkel cell carcinoma, verruca, Bowen disease, keratosis, skin cancer, cutaneous cancer, skin neoplasia, cutaneous neoplasia, and skin tumor. The literature search terms were selected based on the World Health Organization classification of skin tumors.13 Manual bibliography checks were performed on all eligible search results for possible relevant studies. Discrepancies were resolved through discussion and, if needed, mediation by a third researcher (N.C.). To be included, a study had to report a case(s) of epidermal tumor(s) that was confirmed by histopathology and arose on a graft donor site in a patient receiving autologous skin grafts for any reason. No language, geographic, or report date restrictions were set.

Data Extraction, Quality Assessment, and Statistical Analysis—We adhered to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines.14 Two independent researchers (Z.P. and V.P.) retrieved the data from the included studies. We have used the terms case and patient interchangeably, and 1 month was measured as 4 weeks for simplicity. Disagreements were resolved by discussion and mediation by a third researcher (N.C.). The quality of the included studies was assessed by 2 researchers (M.P. and V.P.) using the tool proposed by Murad et al.15

We used descriptive statistical analysis to analyze clinical characteristics of the included cases. We performed separate descriptive analyses based on the most frequently reported types of epidermal tumors and compared the differences between different groups using the Mann-Whitney U test, χ2 test, and Fisher exact test. The level of significance was set at P<.05. All statistical analyses were conducted using SPSS (version 29).

 

 

Results

Literature Search and Characteristics of Included Studies—The initial literature search identified 1378 studies, which were screened based on title and abstract. After removing duplicate and irrelevant studies and evaluating the full text of eligible studies, 31 studies (4 case series and 27 case reports) were included in the systematic review (Figure).6-12,16-39 Quality assessment of the included studies is presented in Table 1.

Flowchart for a systematic review and meta-analysis using Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) criteria for articles published before December 2022.

Clinical Characteristics of Included Patients—Our systematic review included 36 patients with a mean age of 63 years and a male to female ratio of 2:1. The 2 most common causes for skin grafting were burn wounds and surgical excision of skin tumors. Most grafts were harvested from the thighs. The development of a solitary lesion on the donor area was reported in two-thirds of the patients, while more than 1 lesion developed in the remaining one-third of patients. The median time to tumor development was 6.5 weeks. In most cases, a split-thickness skin graft was used.

Cutaneous squamous cell carcinomas (CSCCs) were found in 23 patients, with well-differentiated CSCCs in 19 of these cases. Additionally, keratoacanthomas (KAs) were found in 10 patients. The majority of patients underwent surgical excision of the tumor. The median follow-up time was 12 months, during which recurrences were noted in a small percentage of cases. Clinical characteristics of included patients are presented in Table 2.

Comparison of Variables Between CSCC and KA Groups—The most common diagnoses among the included patients were CSCC and KA. There were no significant differences between the groups in clinical variables, including age, sex, reason for grafting, time to occurrence, and rate of recurrence (Table 3).

 

 

Comment

Reasons for Tumor Development on Skin Graft Donor Sites—The etiology behind epidermal tumor development on graft donor sites is unclear. According to one theory, iatrogenic contamination of the donor site during the removal of a primary epidermal tumor could be responsible. However, contemporary surgical procedures dictate the use of different sets of instruments for separate surgical sites. Moreover, this theory cannot explain the occurrence of epidermal tumors on donor sites in patients who have undergone skin grafting for the repair of burn wounds.37

Another theory suggests that hematogenous and/or lymphatic spread can occur from the site of the primary epidermal tumor to the donor site, which has increased vascularization.16,37 However, this theory also fails to provide an explanation for the development of epidermal tumors in patients who receive skin grafts for burn wounds.

A third theory states that the microenvironment of the donor site is key to tumor development. The donor site undergoes acute inflammation due to the trauma from harvesting the skin graft. According to this theory, acute inflammation could promote neoplastic growth and thus explain the development of epidermal tumors on the donor site.8,26 However, the relationship between acute inflammation and carcinogenesis remains unclear. What is known to date is that the development of CSCC has been documented primarily in chronically inflamed tissues, whereas the development of KA—a variant of CSCC with distinctive and more benign clinical characteristics—can be expected in the setting of acute trauma-related inflammation.13,40,41

Based on our systematic review, we propose that well-differentiated CSCC on graft donor sites might actually be misdiagnosed KA, given that the histopathologic differential diagnosis between CSCC and KA is extremely challenging.42 This hypothesis could explain the development of well-differentiated CSCC and KA on graft donor sites.

Conclusion

Development of CSCC and KA on graft donor sites can be listed among the postoperative complications of autologous skin grafting. Patients and physicians should be aware of this potential complication, and donor sites should be monitored for the occurrence of epidermal tumors.

References
  1. Adams DC, Ramsey ML. Grafts in dermatologic surgery: review and update on full- and split-thickness skin grafts, free cartilage grafts, and composite grafts. Dermatologic Surg. 2005;31(8, pt 2):1055-1067. doi:10.1111/j.1524-4725.2005.31831
  2. Shimizu R, Kishi K. Skin graft. Plast Surg Int. 2012;2012:563493. doi:10.1155/2012/563493
  3. Reddy S, El-Haddawi F, Fancourt M, et al. The incidence and risk factors for lower limb skin graft failure. Dermatol Res Pract. 2014;2014:582080. doi:10.1155/2014/582080
  4. Coughlin MJ, Dockery GD, Crawford ME, et al. Lower Extremity Soft Tissue & Cutaneous Plastic Surgery. 2nd ed. Saunders Ltd; 2012.
  5. Herskovitz I, Hughes OB, Macquhae F, et al. Epidermal skin grafting. Int Wound J. 2016;13(suppl 3):52-56. doi:10.1111/iwj.12631
  6. Wright H, McKinnell TH, Dunkin C. Recurrence of cutaneous squamous cell carcinoma at remote limb donor site. J Plast Reconstr Aesthet Surg. 2012;65:1265-1266. doi:10.1016/j.bjps.2012.01.022
  7. Thomas W, Rezzadeh K, Rossi K, et al. Squamous cell carcinoma arising at a skin graft donor site: case report and review of the literature. Plast Surg Case Stud. 2021;7:2513826X211008425. doi:10.1177/2513826X211008425
  8. Ponnuvelu G, Ng MFY, Connolly CM, et al. Inflammation to skin malignancy, time to rethink the link: SCC in skin graft donor sites. Surgeon. 2011;9:168-169. doi:10.1016/j.surge.2010.08.006
  9. Noori VJ, Trehan K, Savetamal A, et al. New onset squamous cell carcinoma in previous split-thickness skin graft donor site. Int J Surg. 2018;52:16-19. doi:10.1016/j.ijsu.2018.01.047
  10. Morritt DG, Khandwala AR. The development of squamous cell carcinomas in split-thickness skin graft donor sites. Eur J Plast Surg. 2013;36:377-380.
  11. McCormick M, Miotke S. Squamous cell carcinoma at split thickness skin graft donor site: a case report and review of the literature. J Burn Care Res. 2023;44:210-213. doi:10.1093/jbcr/irac137
  12. Haik J, Georgiou I, Farber N, et al. Squamous cell carcinoma arising in a split-thickness skin graft donor site. Burns. 2008;34:891-893. doi:10.1016/j.burns.2007.06.006
  13. Elder DE, Massi D, Scolyer RA WR. WHO Classification of Skin Tumours. 4th ed. IARC Press; 2018.
  14. Moher D, Liberati A, Tetzlaff J, et al. Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. Ann Intern Med. 2009;151:264-269, W64. doi:10.7326/0003-4819-151-4-200908180-00135
  15. Murad MH, Sultan S, Haffar S, et al. Methodological quality and synthesis of case series and case reports. BMJ. 2018;23:60-63. doi:10.1136/bmjebm-2017-110853
  16. de Moraes LPB, Burchett I, Nicholls S, et al. Large solitary distant metastasis of cutaneous squamous cell carcinoma to skin graft site with complete response following definitive radiotherapy. Int J Bioautomation. 2017;21:103-108.
  17. Nagase K, Suzuki Y, Misago N, et al. Acute development of keratoacanthoma at a full-thickness skin graft donor site shortly after surgery. J Dermatol. 2016;43:1232-1233. doi:10.1111/1346-8138.13368
  18. Taylor CD, Snelling CF, Nickerson D, et al. Acute development of invasive squamous cell carcinoma in a split-thickness skin graft donor site. J Burn Care Rehabil. 1998;19:382-385. doi:10.1097/00004630-199809000-00004
  19. de Delas J, Leache A, Vazquez Doval J, et al. Keratoacanthoma over the donor site of a laminar skin graft. Med Cutan Ibero Lat Am. 1989;17:225-228.
  20. Neilson D, Emerson DJ, Dunn L. Squamous cell carcinoma of skin developing in a skin graft donor site. Br J Plast Surg. 1988;41:417-419. doi:10.1016/0007-1226(88)90086-0
  21. May JT, Patil YJ. Keratoacanthoma-type squamous cell carcinoma developing in a skin graft donor site after tumor extirpation at a distant site. Ear Nose Throat J. 2010;89:E11-E13.
  22. Imbernón-Moya A, Vargas-Laguna E, Lobato-Berezo A, et al. Simultaneous onset of basal cell carcinoma over skin graft and donor site. JAAD Case Rep. 2015;1:244-246. doi:10.1016/j.jdcr.2015.05.004
  23. Lee S, Coutts I, Ryan A, et al. Keratoacanthoma formation after skin grafting: a brief report and pathophysiological hypothesis. Australas J Dermatol. 2017;58:e117-e119. doi:10.1111/ajd.12501
  24. Hammond JS, Thomsen S, Ward CG. Scar carcinoma arising acutelyin a skin graft donor site. J Trauma. 1987;27:681-683. doi:10.1097/00005373-198706000-00017
  25. Herard C, Arnaud D, Goga D, et al. Rapid onset of squamous cell carcinoma in a thin skin graft donor site. Ann Dermatol Venereol. 2016;143:457-461. doi:10.1016/j.annder.2015.03.027
  26. Ibrahim A, Moisidis E. Case series: rapidly growing squamous cell carcinoma after cutaneous surgical intervention. JPRAS Open. 2017;14:27-32. doi:10.1016/j.jpra.2017.08.004
  27. Kearney L, Dolan RT, Parfrey NA, et al. Squamous cell carcinoma arising in a skin graft donor site following melanoma extirpation at a distant site: a case report and review of the literature. JPRAS Open. 2015;3:35-38. doi:10.1016/j.jpra.2015.02.002
  28. Clark MA, Guitart J, Gerami P, et al. Eruptive keratoacanthomatous atypical squamous proliferations (KASPs) arising in skin graft sites. JAAD Case Rep. 2015;1:274-276. doi:10.1016/j.jdcr.2015.06.009
  29. Aloraifi F, Mulgrew S, James NK. Secondary Merkel cell carcinoma arising from a graft donor site. J Cutan Med Surg. 2017;21:167-169. doi:10.1177/1203475416676805
  30. Abadir R, Zurowski S. Case report: squamous cell carcinoma of the skin in both palms, axillary node, donor skin graft site and both soles—associated hyperkeratosis and porokeratosis. Br J Radiol. 1994;67:507-510. doi:10.1259/0007-1285-67-797-507
  31. Griffiths RW. Keratoacanthoma observed. Br J Plast Surg. 2004;57:485-501. doi:10.1016/j.bjps.2004.05.007
  32. Marous M, Brady K. Cutaneous squamous cell carcinoma arising in a split thickness skin graft donor site in a patient with systemic lupus erythematosus. Dermatologic Surg. 2021;47:1106-1107. doi:10.1097/DSS.0000000000002955
  33. Dibden FA, Fowler M. The multiple growth of molluscum sebaceum in donor and recipient sites of skin graft. Aust N Z J Surg. 1955;25:157-159. doi:10.1111/j.1445-2197.1955.tb05122.x
  34. Jeremiah BS. Squamous cell carcinoma development on donor area following removal of a split thickness skin graft. Plast Reconstr Surg. 1948;3:718-721.
  35. Tamir G, Morgenstern S, Ben-Amitay D, et al. Synchronous appearance of keratoacanthomas in burn scar and skin graft donor site shortly after injury. J Am Acad Dermatol. 1999;40(5, pt 2):870-871. doi:10.1053/jd.1999.v40.a94419
  36. Hamilton SA, Dickson WA, O’Brien CJ. Keratoacanthoma developing in a split skin graft donor site. Br J Plast Surg. 1997;50:560-561. doi:10.1016/s0007-1226(97)91308-4
  37. Hussain A, Ekwobi C, Watson S. Metastatic implantation squamous cell carcinoma in a split-thickness skin graft donor site. J Plast Reconstr Aesthet Surg. 2011;64:690-692. doi:10.1016/j.bjps.2010.06.004
  38. Wulsin JH. Keratoacanthoma: a benign cutaneous tumors arising in a skin graft donor site. Am Surg. 1958;24:689-692.
  39. Davis L, Butler D. Acute development of squamous cell carcinoma in a split-thickness skin graft donor site [abstract]. J Am Acad Dermatol. 2012;66:AB208. doi:10.1016/j.jaad.2011.11.874
  40. Shacter E, Weitzman SA. Chronic inflammation and cancer. Oncology (Williston Park). 2002;16:217-226, 229; discussion 230-232.
  41.  Piotrowski I, Kulcenty K, Suchorska W. Interplay between inflammation and cancer. Reports Pract Oncol Radiother. 2020;25:422-427. doi:10.1016/j.rpor.2020.04.004
  42. Carr RA, Houghton JP. Histopathologists’ approach to keratoacanthoma: a multisite survey of regional variation in Great Britain and Ireland. J Clin Pathol. 2014;67:637-638. doi:10.1136/jclinpath-2014-202255
References
  1. Adams DC, Ramsey ML. Grafts in dermatologic surgery: review and update on full- and split-thickness skin grafts, free cartilage grafts, and composite grafts. Dermatologic Surg. 2005;31(8, pt 2):1055-1067. doi:10.1111/j.1524-4725.2005.31831
  2. Shimizu R, Kishi K. Skin graft. Plast Surg Int. 2012;2012:563493. doi:10.1155/2012/563493
  3. Reddy S, El-Haddawi F, Fancourt M, et al. The incidence and risk factors for lower limb skin graft failure. Dermatol Res Pract. 2014;2014:582080. doi:10.1155/2014/582080
  4. Coughlin MJ, Dockery GD, Crawford ME, et al. Lower Extremity Soft Tissue & Cutaneous Plastic Surgery. 2nd ed. Saunders Ltd; 2012.
  5. Herskovitz I, Hughes OB, Macquhae F, et al. Epidermal skin grafting. Int Wound J. 2016;13(suppl 3):52-56. doi:10.1111/iwj.12631
  6. Wright H, McKinnell TH, Dunkin C. Recurrence of cutaneous squamous cell carcinoma at remote limb donor site. J Plast Reconstr Aesthet Surg. 2012;65:1265-1266. doi:10.1016/j.bjps.2012.01.022
  7. Thomas W, Rezzadeh K, Rossi K, et al. Squamous cell carcinoma arising at a skin graft donor site: case report and review of the literature. Plast Surg Case Stud. 2021;7:2513826X211008425. doi:10.1177/2513826X211008425
  8. Ponnuvelu G, Ng MFY, Connolly CM, et al. Inflammation to skin malignancy, time to rethink the link: SCC in skin graft donor sites. Surgeon. 2011;9:168-169. doi:10.1016/j.surge.2010.08.006
  9. Noori VJ, Trehan K, Savetamal A, et al. New onset squamous cell carcinoma in previous split-thickness skin graft donor site. Int J Surg. 2018;52:16-19. doi:10.1016/j.ijsu.2018.01.047
  10. Morritt DG, Khandwala AR. The development of squamous cell carcinomas in split-thickness skin graft donor sites. Eur J Plast Surg. 2013;36:377-380.
  11. McCormick M, Miotke S. Squamous cell carcinoma at split thickness skin graft donor site: a case report and review of the literature. J Burn Care Res. 2023;44:210-213. doi:10.1093/jbcr/irac137
  12. Haik J, Georgiou I, Farber N, et al. Squamous cell carcinoma arising in a split-thickness skin graft donor site. Burns. 2008;34:891-893. doi:10.1016/j.burns.2007.06.006
  13. Elder DE, Massi D, Scolyer RA WR. WHO Classification of Skin Tumours. 4th ed. IARC Press; 2018.
  14. Moher D, Liberati A, Tetzlaff J, et al. Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. Ann Intern Med. 2009;151:264-269, W64. doi:10.7326/0003-4819-151-4-200908180-00135
  15. Murad MH, Sultan S, Haffar S, et al. Methodological quality and synthesis of case series and case reports. BMJ. 2018;23:60-63. doi:10.1136/bmjebm-2017-110853
  16. de Moraes LPB, Burchett I, Nicholls S, et al. Large solitary distant metastasis of cutaneous squamous cell carcinoma to skin graft site with complete response following definitive radiotherapy. Int J Bioautomation. 2017;21:103-108.
  17. Nagase K, Suzuki Y, Misago N, et al. Acute development of keratoacanthoma at a full-thickness skin graft donor site shortly after surgery. J Dermatol. 2016;43:1232-1233. doi:10.1111/1346-8138.13368
  18. Taylor CD, Snelling CF, Nickerson D, et al. Acute development of invasive squamous cell carcinoma in a split-thickness skin graft donor site. J Burn Care Rehabil. 1998;19:382-385. doi:10.1097/00004630-199809000-00004
  19. de Delas J, Leache A, Vazquez Doval J, et al. Keratoacanthoma over the donor site of a laminar skin graft. Med Cutan Ibero Lat Am. 1989;17:225-228.
  20. Neilson D, Emerson DJ, Dunn L. Squamous cell carcinoma of skin developing in a skin graft donor site. Br J Plast Surg. 1988;41:417-419. doi:10.1016/0007-1226(88)90086-0
  21. May JT, Patil YJ. Keratoacanthoma-type squamous cell carcinoma developing in a skin graft donor site after tumor extirpation at a distant site. Ear Nose Throat J. 2010;89:E11-E13.
  22. Imbernón-Moya A, Vargas-Laguna E, Lobato-Berezo A, et al. Simultaneous onset of basal cell carcinoma over skin graft and donor site. JAAD Case Rep. 2015;1:244-246. doi:10.1016/j.jdcr.2015.05.004
  23. Lee S, Coutts I, Ryan A, et al. Keratoacanthoma formation after skin grafting: a brief report and pathophysiological hypothesis. Australas J Dermatol. 2017;58:e117-e119. doi:10.1111/ajd.12501
  24. Hammond JS, Thomsen S, Ward CG. Scar carcinoma arising acutelyin a skin graft donor site. J Trauma. 1987;27:681-683. doi:10.1097/00005373-198706000-00017
  25. Herard C, Arnaud D, Goga D, et al. Rapid onset of squamous cell carcinoma in a thin skin graft donor site. Ann Dermatol Venereol. 2016;143:457-461. doi:10.1016/j.annder.2015.03.027
  26. Ibrahim A, Moisidis E. Case series: rapidly growing squamous cell carcinoma after cutaneous surgical intervention. JPRAS Open. 2017;14:27-32. doi:10.1016/j.jpra.2017.08.004
  27. Kearney L, Dolan RT, Parfrey NA, et al. Squamous cell carcinoma arising in a skin graft donor site following melanoma extirpation at a distant site: a case report and review of the literature. JPRAS Open. 2015;3:35-38. doi:10.1016/j.jpra.2015.02.002
  28. Clark MA, Guitart J, Gerami P, et al. Eruptive keratoacanthomatous atypical squamous proliferations (KASPs) arising in skin graft sites. JAAD Case Rep. 2015;1:274-276. doi:10.1016/j.jdcr.2015.06.009
  29. Aloraifi F, Mulgrew S, James NK. Secondary Merkel cell carcinoma arising from a graft donor site. J Cutan Med Surg. 2017;21:167-169. doi:10.1177/1203475416676805
  30. Abadir R, Zurowski S. Case report: squamous cell carcinoma of the skin in both palms, axillary node, donor skin graft site and both soles—associated hyperkeratosis and porokeratosis. Br J Radiol. 1994;67:507-510. doi:10.1259/0007-1285-67-797-507
  31. Griffiths RW. Keratoacanthoma observed. Br J Plast Surg. 2004;57:485-501. doi:10.1016/j.bjps.2004.05.007
  32. Marous M, Brady K. Cutaneous squamous cell carcinoma arising in a split thickness skin graft donor site in a patient with systemic lupus erythematosus. Dermatologic Surg. 2021;47:1106-1107. doi:10.1097/DSS.0000000000002955
  33. Dibden FA, Fowler M. The multiple growth of molluscum sebaceum in donor and recipient sites of skin graft. Aust N Z J Surg. 1955;25:157-159. doi:10.1111/j.1445-2197.1955.tb05122.x
  34. Jeremiah BS. Squamous cell carcinoma development on donor area following removal of a split thickness skin graft. Plast Reconstr Surg. 1948;3:718-721.
  35. Tamir G, Morgenstern S, Ben-Amitay D, et al. Synchronous appearance of keratoacanthomas in burn scar and skin graft donor site shortly after injury. J Am Acad Dermatol. 1999;40(5, pt 2):870-871. doi:10.1053/jd.1999.v40.a94419
  36. Hamilton SA, Dickson WA, O’Brien CJ. Keratoacanthoma developing in a split skin graft donor site. Br J Plast Surg. 1997;50:560-561. doi:10.1016/s0007-1226(97)91308-4
  37. Hussain A, Ekwobi C, Watson S. Metastatic implantation squamous cell carcinoma in a split-thickness skin graft donor site. J Plast Reconstr Aesthet Surg. 2011;64:690-692. doi:10.1016/j.bjps.2010.06.004
  38. Wulsin JH. Keratoacanthoma: a benign cutaneous tumors arising in a skin graft donor site. Am Surg. 1958;24:689-692.
  39. Davis L, Butler D. Acute development of squamous cell carcinoma in a split-thickness skin graft donor site [abstract]. J Am Acad Dermatol. 2012;66:AB208. doi:10.1016/j.jaad.2011.11.874
  40. Shacter E, Weitzman SA. Chronic inflammation and cancer. Oncology (Williston Park). 2002;16:217-226, 229; discussion 230-232.
  41.  Piotrowski I, Kulcenty K, Suchorska W. Interplay between inflammation and cancer. Reports Pract Oncol Radiother. 2020;25:422-427. doi:10.1016/j.rpor.2020.04.004
  42. Carr RA, Houghton JP. Histopathologists’ approach to keratoacanthoma: a multisite survey of regional variation in Great Britain and Ireland. J Clin Pathol. 2014;67:637-638. doi:10.1136/jclinpath-2014-202255
Issue
Cutis - 114(2)
Issue
Cutis - 114(2)
Page Number
E6-E12
Page Number
E6-E12
Publications
Publications
Topics
Article Type
Display Headline
Epidermal Tumors Arising on Donor Sites From Autologous Skin Grafts: A Systematic Review
Display Headline
Epidermal Tumors Arising on Donor Sites From Autologous Skin Grafts: A Systematic Review
Sections
Inside the Article

Practice Points

  • Donor site cutaneous squamous cell carcinoma (CSCC) and keratoacanthoma (KA) can be postoperative complications of autologous skin grafting.
  • Surgical excision of donor site CSCC and KA typically is curative.
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.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

‘Emerging Threat’ Xylazine Use Continues to Spread Across the United States

Article Type
Changed
Wed, 09/04/2024 - 10:19

 

Illicit use of the veterinary tranquilizer xylazine continues to spread across the United States. The drug, which is increasingly mixed with fentanyl, often fails to respond to the opioid overdose reversal medication naloxone and can cause severe necrotic lesions.

A report released by Millennium Health, a specialty lab that provides medication monitoring for pain management, drug treatment, and behavioral and substance use disorder treatment centers across the country, showed the number of urine specimens collected and tested at the US drug treatment centers were positive for xylazine in the most recent 6 months.

As previously reported by this news organization, in late 2022, the US Food and Drug Administration (FDA) issued a communication alerting clinicians about the special management required for opioid overdoses tainted with xylazine, which is also known as “tranq” or “tranq dope.”

Subsequently, in early 2023, The White House Office of National Drug Control Policy designated xylazine combined with fentanyl as an emerging threat to the United States.

Both the FDA and the Drug Enforcement Administration have taken steps to try to stop trafficking of the combination. However, despite these efforts, xylazine use has continued to spread.

The Millennium Health Signals report showed that the greatest increase in xylazine use was largely in the western United States. In the first 6 months of 2023, 3% of urine drug tests (UDTs) in Washington, Oregon, California, Hawaii, and Alaska were positive for xylazine. From November 2023 to April 2024, this rose to 8%, a 147% increase. In the Mountain West, xylazine-positive UDTs increased from 2% in 2023 to 4% in 2024, an increase of 94%. In addition to growth in the West, the report showed that xylazine use increased by more than 100% in New England — from 14% in 2023 to 28% in 2024.

Nationally, 16% of all urine specimens were positive for xylazine from late 2023 to April 2024, up slightly from 14% from April to October 2023.

Xylazine use was highest in the East and in the mid-Atlantic United States. Still, positivity rates in the mid-Atlantic dropped from 44% to 33%. The states included in that group were New York, Pennsylvania, Delaware, and New Jersey. East North Central states (Ohio, Michigan, Wisconsin, Indiana, and Illinois) also experienced a decline in positive tests from 32% to 30%.

The South Atlantic states, which include Maryland, Virginia, West Virginia, North and South Carolina, Georgia, and Florida, had a 17% increase in positivity — from 22% to 26%.

From April 2023 to April 2024 state-level UDT positivity rates were 40% in Pennsylvania, 37% in New York, and 35% in Ohio. But rates vary by locality. In Clermont and Hamilton counties in Ohio — both in the Cincinnati area — about 70% of specimens were positive for xylazine.

About one third of specimens in Maryland and South Carolina contained xylazine.

“Because xylazine exposure remains a significant challenge in the East and is a growing concern in the West, clinicians across the US need to be prepared to recognize and address the consequences of xylazine use — like diminished responses to naloxone and severe skin wounds that may lead to amputation — among people who use fentanyl,” Millennium Health Chief Clinical Officer Angela Huskey, PharmD, said in a press release.

The Health Signals Alert analyzed more than 50,000 fentanyl-positive UDT specimens collected between April 12, 2023, and April 11, 2024. Millennium Health researchers analyzed xylazine positivity rates in fentanyl-positive UDT specimens by the US Census Division and state.

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

Publications
Topics
Sections

 

Illicit use of the veterinary tranquilizer xylazine continues to spread across the United States. The drug, which is increasingly mixed with fentanyl, often fails to respond to the opioid overdose reversal medication naloxone and can cause severe necrotic lesions.

A report released by Millennium Health, a specialty lab that provides medication monitoring for pain management, drug treatment, and behavioral and substance use disorder treatment centers across the country, showed the number of urine specimens collected and tested at the US drug treatment centers were positive for xylazine in the most recent 6 months.

As previously reported by this news organization, in late 2022, the US Food and Drug Administration (FDA) issued a communication alerting clinicians about the special management required for opioid overdoses tainted with xylazine, which is also known as “tranq” or “tranq dope.”

Subsequently, in early 2023, The White House Office of National Drug Control Policy designated xylazine combined with fentanyl as an emerging threat to the United States.

Both the FDA and the Drug Enforcement Administration have taken steps to try to stop trafficking of the combination. However, despite these efforts, xylazine use has continued to spread.

The Millennium Health Signals report showed that the greatest increase in xylazine use was largely in the western United States. In the first 6 months of 2023, 3% of urine drug tests (UDTs) in Washington, Oregon, California, Hawaii, and Alaska were positive for xylazine. From November 2023 to April 2024, this rose to 8%, a 147% increase. In the Mountain West, xylazine-positive UDTs increased from 2% in 2023 to 4% in 2024, an increase of 94%. In addition to growth in the West, the report showed that xylazine use increased by more than 100% in New England — from 14% in 2023 to 28% in 2024.

Nationally, 16% of all urine specimens were positive for xylazine from late 2023 to April 2024, up slightly from 14% from April to October 2023.

Xylazine use was highest in the East and in the mid-Atlantic United States. Still, positivity rates in the mid-Atlantic dropped from 44% to 33%. The states included in that group were New York, Pennsylvania, Delaware, and New Jersey. East North Central states (Ohio, Michigan, Wisconsin, Indiana, and Illinois) also experienced a decline in positive tests from 32% to 30%.

The South Atlantic states, which include Maryland, Virginia, West Virginia, North and South Carolina, Georgia, and Florida, had a 17% increase in positivity — from 22% to 26%.

From April 2023 to April 2024 state-level UDT positivity rates were 40% in Pennsylvania, 37% in New York, and 35% in Ohio. But rates vary by locality. In Clermont and Hamilton counties in Ohio — both in the Cincinnati area — about 70% of specimens were positive for xylazine.

About one third of specimens in Maryland and South Carolina contained xylazine.

“Because xylazine exposure remains a significant challenge in the East and is a growing concern in the West, clinicians across the US need to be prepared to recognize and address the consequences of xylazine use — like diminished responses to naloxone and severe skin wounds that may lead to amputation — among people who use fentanyl,” Millennium Health Chief Clinical Officer Angela Huskey, PharmD, said in a press release.

The Health Signals Alert analyzed more than 50,000 fentanyl-positive UDT specimens collected between April 12, 2023, and April 11, 2024. Millennium Health researchers analyzed xylazine positivity rates in fentanyl-positive UDT specimens by the US Census Division and state.

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

 

Illicit use of the veterinary tranquilizer xylazine continues to spread across the United States. The drug, which is increasingly mixed with fentanyl, often fails to respond to the opioid overdose reversal medication naloxone and can cause severe necrotic lesions.

A report released by Millennium Health, a specialty lab that provides medication monitoring for pain management, drug treatment, and behavioral and substance use disorder treatment centers across the country, showed the number of urine specimens collected and tested at the US drug treatment centers were positive for xylazine in the most recent 6 months.

As previously reported by this news organization, in late 2022, the US Food and Drug Administration (FDA) issued a communication alerting clinicians about the special management required for opioid overdoses tainted with xylazine, which is also known as “tranq” or “tranq dope.”

Subsequently, in early 2023, The White House Office of National Drug Control Policy designated xylazine combined with fentanyl as an emerging threat to the United States.

Both the FDA and the Drug Enforcement Administration have taken steps to try to stop trafficking of the combination. However, despite these efforts, xylazine use has continued to spread.

The Millennium Health Signals report showed that the greatest increase in xylazine use was largely in the western United States. In the first 6 months of 2023, 3% of urine drug tests (UDTs) in Washington, Oregon, California, Hawaii, and Alaska were positive for xylazine. From November 2023 to April 2024, this rose to 8%, a 147% increase. In the Mountain West, xylazine-positive UDTs increased from 2% in 2023 to 4% in 2024, an increase of 94%. In addition to growth in the West, the report showed that xylazine use increased by more than 100% in New England — from 14% in 2023 to 28% in 2024.

Nationally, 16% of all urine specimens were positive for xylazine from late 2023 to April 2024, up slightly from 14% from April to October 2023.

Xylazine use was highest in the East and in the mid-Atlantic United States. Still, positivity rates in the mid-Atlantic dropped from 44% to 33%. The states included in that group were New York, Pennsylvania, Delaware, and New Jersey. East North Central states (Ohio, Michigan, Wisconsin, Indiana, and Illinois) also experienced a decline in positive tests from 32% to 30%.

The South Atlantic states, which include Maryland, Virginia, West Virginia, North and South Carolina, Georgia, and Florida, had a 17% increase in positivity — from 22% to 26%.

From April 2023 to April 2024 state-level UDT positivity rates were 40% in Pennsylvania, 37% in New York, and 35% in Ohio. But rates vary by locality. In Clermont and Hamilton counties in Ohio — both in the Cincinnati area — about 70% of specimens were positive for xylazine.

About one third of specimens in Maryland and South Carolina contained xylazine.

“Because xylazine exposure remains a significant challenge in the East and is a growing concern in the West, clinicians across the US need to be prepared to recognize and address the consequences of xylazine use — like diminished responses to naloxone and severe skin wounds that may lead to amputation — among people who use fentanyl,” Millennium Health Chief Clinical Officer Angela Huskey, PharmD, said in a press release.

The Health Signals Alert analyzed more than 50,000 fentanyl-positive UDT specimens collected between April 12, 2023, and April 11, 2024. Millennium Health researchers analyzed xylazine positivity rates in fentanyl-positive UDT specimens by the US Census Division and state.

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

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Tackling Inflammatory and Infectious Nail Disorders in Children

Article Type
Changed
Wed, 08/07/2024 - 11:57
Display Headline
Tackling Inflammatory and Infectious Nail Disorders in Children

Nail disorders are common among pediatric patients but often are underdiagnosed or misdiagnosed because of their unique disease manifestations. These conditions may severely impact quality of life. There are few nail disease clinical trials that include children. Consequently, most treatment recommendations are based on case series and expert consensus recommendations. We review inflammatory and infectious nail disorders in pediatric patients. By describing characteristics, clinical manifestations, and management approaches for these conditions, we aim to provide guidance to dermatologists in their diagnosis and treatment.

INFLAMMATORY NAIL DISORDERS

Nail Psoriasis

Nail involvement in children with psoriasis is common, with prevalence estimates ranging from 17% to 39.2%.1 Nail matrix psoriasis may manifest with pitting (large irregular pits) and leukonychia as well as chromonychia and nail plate crumbling. Onycholysis, oil drop spots (salmon patches), and subungual hyperkeratosis can be seen in nail bed psoriasis. Nail pitting is the most frequently observed clinical finding (Figure 1).2,3 In a cross-sectional multicenter study of 313 children with cutaneous psoriasis in France, nail findings were present in 101 patients (32.3%). There were associations between nail findings and presence of psoriatic arthritis (P=.03), palmoplantar psoriasis (P<.001), and severity of psoriatic disease, defined as use of systemic treatment with phototherapy (psoralen plus UVA, UVB), traditional systemic treatment (acitretin, methotrexate, cyclosporine), or a biologic (P=.003).4

Topical steroids and vitamin D analogues may be used with or without occlusion and may be efficacious.5 Several case reports describe systemic treatments for psoriasis in children, including methotrexate, acitretin, and apremilast (approved for children 6 years and older for plaque psoriasis by the US Food and Drug Administration [FDA]).2 There are 5 biologic drugs currently approved for the treatment of pediatric psoriasis—adalimumab, etanercept, ustekinumab, secukinumab, ixekizumab—and 6 drugs currently undergoing phase 3 studies—brodalumab, guselkumab, risankizumab, tildrakizumab, certolizumab pegol, and deucravacitinib (Table 1).6-15 Adalimumab is specifically approved for moderate to severe nail psoriasis in adults 18 years and older.

FIGURE 1. Nail psoriasis in a 9-year-old girl with onycholysis, nail bed hyperkeratosis, and pitting, as well as discoloration.

 

Intralesional steroid injections are sometimes useful in the management of nail matrix psoriasis; however, appropriate patient selection is critical due to the pain associated with the procedure. In a prospective study of 16 children (age range, 9–17 years) with nail psoriasis treated with intralesional triamcinolone (ILTAC) 2.5 to 5 mg/mL every 4 to 8 weeks for a minimum of 3 to 6 months, 9 patients achieved resolution and 6 had improvement of clinical findings.16 Local adverse events were mild, including injection-site pain (66%), subungual hematoma (n=1), Beau lines (n=1), proximal nail fold hypopigmentation (n=2), and proximal nail fold atrophy (n=2). Because the proximal nail fold in children is thinner than in adults, there may be an increased risk for nail fold hypopigmentation and atrophy in children. Therefore, a maximum ILTAC concentration of 2.5 mg/mL with 0.2 mL maximum volume per nail per session is recommended for children younger than 15 years.16

Nail Lichen Planus

Nail lichen planus (NLP) is uncommon in children, with few biopsy-proven cases documented in the literature.17 Common clinical findings are onychorrhexis, nail plate thinning, fissuring, splitting, and atrophy with koilonychia.5 Although pterygium development (irreversible nail matrix scarring) is uncommon in pediatric patients, NLP can be progressive and may cause irreversible destruction of the nail matrix and subsequent nail loss, warranting therapeutic intervention.18

Treatment of NLP may be difficult, as there are no options that work in all patients. Current literature supports the use of systemic corticosteroids or ILTAC for the treatment of NLP; however, recurrence rates can be high. According to an expert consensus paper on NLP treatment, ILTAC may be injected in a concentration of 2.5, 5, or 10 mg/mL according to disease severity.19 In severe or resistant cases, intramuscular (IM) triamcinolone may be considered, especially if more than 3 nails are affected. A dosage of 0.5 to 1 mg/kg/mo for at least 3 to 6 months is recommended for both children and adults, with 1 mg/kg/mo recommended in the active treatment phase (first 2–3 months).19 In a retrospective review of 5 pediatric patients with NLP treated with IM triamcinolone 0.5 mg/kg/mo, 3 patients had resolution and 2 improved with treatment.20 In a prospective study of 10 children with NLP, IM triamcinolone at a dosage of 0.5 to 1 mg/kg every 30 days for 3 to 6 months resulted in resolution of nail findings in 9 patients.17 In a prospective study of 14 pediatric patients with NLP treated with 2.5 to 5 mg/mL of ILTAC, 10 achieved resolution and 3 improved.16

Intralesional triamcinolone injections may be better suited for teenagers compared to younger children who may be more apprehensive of needles. To minimize pain, it is recommended to inject ILTAC slowly at room temperature, with use of “talkesthesia” and vibration devices, 1% lidocaine, or ethyl chloride spray.18

Trachyonychia

Trachyonychia is characterized by the presence of sandpaperlike nails. It manifests with brittle thin nails with longitudinal ridging, onychoschizia, and thickened hyperkeratotic cuticles. Trachyonychia typically involves multiple nails, with a peak age of onset between 3 and 12 years.21,22 There are 2 variants: the opaque type with rough longitudinal ridging, and the shiny variant with opalescent nails and pits that reflect light. The opaque variant is more common and is associated with psoriasis, whereas the shiny variant is less common and is associated with alopecia areata.23 Although most cases are idiopathic, some are associated with psoriasis and alopecia areata, as previously noted, as well as atopic dermatitis (AD) and lichen planus.22,24

Fortunately, trachyonychia does not lead to permanent nail damage or pterygium, making treatment primarily focused on addressing functional and cosmetic concerns.24 Spontaneous resolution occurs in approximately 50% of patients. In a prospective study of 11 patients with idiopathic trachyonychia, there was partial improvement in 5 of 9 patients treated with topical steroids, 1 with only petrolatum, and 1 with vitamin supplements. Complete resolution was reported in 1 patient treated with topical steroids.25 Because trachyonychia often is self-resolving, no treatment is required and a conservative approach is strongly recommended.26 Treatment options include topical corticosteroids, tazarotene, and 5-fluorouracil. Intralesional triamcinolone, systemic cyclosporine, retinoids, systemic corticosteroids, and tofacitinib have been described in case reports, though none of these have been shown to be 100% efficacious.24

Nail Lichen Striatus

Lichen striatus involving the nail is uncommon and is characterized by onycholysis, longitudinal ridging, ­splitting, and fraying, as well as what appears to be a subungual tumor. It can encompass the entire nail or may be isolated to a portion of the nail (Figure 2). Usually, a Blaschko-linear array of flesh-colored papules on the more proximal digit directly adjacent to the nail dystrophy will be seen, though nail findings can occur in ­isolation.27-29 The underlying pathophysiology is not clear; however, one hypothesis is that a triggering event, such as trauma, induces the expression of antigens that elicit a self-limiting immune-mediated response by CD8 T lymphocytes.30

 

FIGURE 2. Lichen striatus in a 6-year-old boy with multiple fleshcolored papules in a Blaschko-linear distribution (arrows) as well as onychodystrophy and subungual hyperkeratosis of the nail. Republished under the Creative Commons Attribution (CC BY 4.0).27

Generally, nail lichen striatus spontaneously resolves in 1 to 2 years without treatment. In a prospective study of 5 patients with nail lichen striatus, the median time to resolution was 22.6 months (range, 10–30 months).31 Topical steroids may be used for pruritus. In one case report, a 3-year-old boy with nail lichen striatus of 4 months’ duration was treated with tacrolimus ointment 0.03% daily for 3 months.28

Nail AD

Nail changes with AD may be more common in adults than children or are underreported. In a study of 777 adults with AD, nail dystrophy was present in 124 patients (16%), whereas in a study of 250 pediatric patients with AD (aged 0-2 years), nail dystrophy was present in only 4 patients.32,33

Periungual inflammation from AD causes the nail changes.34 In a cross-sectional study of 24 pediatric patients with nail dystrophy due to AD, transverse grooves (Beau lines) were present in 25% (6/24), nail pitting in 16.7% (4/24), koilonychia in 16.7% (4/24), trachyonychia in 12.5% (3/24), leukonychia in 12.5% (3/24), brachyonychia in 8.3% (2/24), melanonychia in 8.3% (2/24), onychomadesis in 8.3% (2/24), onychoschizia in 8.3% (2/24), and onycholysis in 8.3% (2/24). There was an association between disease severity and presence of toenail dystrophy (P=.03).35

Topical steroids with or without occlusion can be used to treat nail changes. Although there is limited literature describing the treatment of nail AD in children, a 61-year-old man with nail changes associated with AD achieved resolution with 3 months of treatment with dupilumab.36 Anecdotally, most patients will improve with usual cutaneous AD management.

 

 

INFECTIOUS NAIL DISORDERS

Viral Infections

Hand, Foot, and Mouth Disease—Hand, foot, and mouth disease (HFMD) is a common childhood viral infection caused by various enteroviruses, most commonly coxsackievirus A16, with the A6 variant causing more severe disease. Fever and painful vesicles involving the oral mucosa as well as palms and soles give the disease its name. Nail changes are common. In a prospective study involving 130 patients with laboratory-confirmed coxsackievirus CA6 serotype infection, 37% developed onychomadesis vs only 5% of 145 cases with non-CA6 enterovirus infection who developed nail findings. There was an association between CA6 infection and presence of nail changes (P<.001).37

Findings ranging from transverse grooves (Beau lines) to complete nail shedding (onychomadesis)(Figure 3) may be seen.38,39 Nail findings in HFMD are due to transient inhibition of nail growth and present approximately 3 to 6 weeks after infection.40 Onychomadesis is seen in 30% to 68% of patients with HFMD.37,41,42 Nail findings in HFMD spontaneously resolve with nail growth (2–3 mm per month for fingernails and 1 mm per month for toenails) and do not require specific treatment. Although the appearance of nail changes associated with HFMD can be disturbing, dermatologists can reassure children and their parents that the nails will resolve with the next cycle of growth.

Kawasaki Disease—Kawasaki disease (KD) is a vasculitis primarily affecting children and infants. Although the specific pathogen and pathophysiology is not entirely clear, clinical observations have suggested an infectious cause, most likely a virus.43 In Japan, more than 15,000 cases of KD are documented annually, while approximately 4200 cases are seen in the United States.44 In a prospective study from 1984 to 1990, 4 of 26 (15.4%) patients with KD presented with nail manifestations during the late acute phase or early convalescent phase of disease. There were no significant associations between nail dystrophy and severity of KD, such as coronary artery aneurysm.45

Nail changes reported in children with KD include onychomadesis, onycholysis, orange-brown chromonychia, splinter hemorrhages, Beau lines, and pincer nails. In a review of nail changes associated with KD from 1980 to 2021, orange-brown transverse chromonychia, which may evolve into transverse leukonychia, was the most common nail finding reported, occurring in 17 of 31 (54.8%) patients.44 It has been hypothesized that nail changes may result from blood flow disturbance due to the underlying vasculitis.46 Nail changes appear several weeks after the onset of fever and are self-limited. Resolution occurs with nail growth, with no treatment required.

FIGURE 3. Onychomadesis from hand, foot, and mouth disease with yellow-orange discoloration of the nail plate. Republished under the Creative Commons Attribution (CC BY-NC-SA).39

 

 

FUNGAL INFECTIONS

Onychomycosis

Onychomycosis is a fungal infection of the nails that occurs in 0.2% to 5.5% of pediatric patients, and its prevalence may be increasing, which may be due to environmental factors or increased rates of diabetes mellitus and obesity in the pediatric population.47 Onychomycosis represents 15.5% of nail dystrophies in pediatric patients.48 Some dermatologists treat presumptive onychomycosis without confirmation; however, we do not recommend that approach. Because the differential is broad and the duration of treatment is long, mycologic examination (potassium hydroxide preparation, fungal culture, polymerase chain reaction, and/or histopathology) should be obtained to confirm onychomycosis prior to initiation of antifungal management. Family members of affected individuals should be evaluated and treated, if indicated, for onychomycosis and tinea pedis, as household transmission is common.

Currently, there are 2 topical FDA-approved treatments for pediatric onychomycosis in children 6 years and older (Table 2).49,50 There is a discussion of the need for confirmatory testing for onychomycosis in children, particularly when systemic treatment is prescribed. In a retrospective review of 269 pediatric patients with onychomycosis prescribed terbinafine, 53.5% (n=144) underwent laboratory monitoring of liver function and complete blood cell counts, and 12.5% had grade 1 laboratory abnormalities either prior to (12/144 [8.3%]) or during (6/144 [4.2%]) therapy.51 Baseline transaminase monitoring is recommended, though subsequent routine laboratory monitoring in healthy children may have limited utility with associated increased costs, incidental findings, and patient discomfort and likely is not needed.51

Pediatric onychomycosis responds better to topical therapy than adult disease, and pediatric patients do not always require systemic treatment.52 Ciclopirox is not FDA approved for the treatment of pediatric onychomycosis, but in a 32-week clinical trial of ciclopirox lacquer 8% use in 40 patients, 77% (27/35) of treated patients achieved mycologic cure. Overall, 71% of treated patients (25/35) vs 22% (2/9) of controls achieved efficacy (defined as investigator global assessment score of 2 or lower).52 In an open-label, single-arm clinical trial assessing tavaborole solution 5% applied once daily for 48 weeks for the treatment of toenail onychomycosis in pediatric patients (aged 6–17 years), 36.2% (20/55) of patients achieved mycologic cure, and 8.5% (5/55) achieved complete cure at week 52 with mild or minimal adverse effects.53 In an open-label, phase 4 study of the safety and efficacy of efinaconazole solution 10% applied once daily for 48 weeks in pediatric patients (aged 6 to 16 years) (n=60), 65% (35/60) achieved mycologic cure, 42% (25/60) achieved clinical cure, and 40% (24/60) achieved complete cure at 52 weeks. The most common adverse effects of efina­conazole were local and included ingrown toenail (1/60), application-site dermatitis (1/60), application-site vesicles (1/60), and application-site pain (1/60).54

In a systematic review of systemic antifungals for onychomycosis in 151 pediatric patients, itraconazole, fluconazole, griseofulvin, and terbinafine resulted in complete cure rates similar to those of the adult population, with excellent safety profiles.55 Depending on the situation, initiation of treatment with topical medications followed by addition of systemic antifungal agents only if needed may be an appropriate course of action.

BACTERIAL INFECTIONS

Acute Paronychia

Acute paronychia is a nail-fold infection that develops after the protective nail barrier has been compromised.56 In children, thumb-sucking, nail-biting, frequent oral manipulation of the digits, and poor skin hygiene are risk factors. Acute paronychia also may develop in association with congenital malalignment of the great toenails.57

Clinical manifestations include localized pain, erythema, and nail fold edema (Figure 4). Purulent material and abscess formation may ensue. Staphylococcus aureus as well as methicillin-resistant S aureus and Streptococcus pyogenes are classically the most common causes of acute paronychia. Treatment of paronychia is based on severity. In mild cases, warm soaks with topical antibiotics are indicated. Oral antibiotics should be prescribed for more severe presentations. If there is no improvement after 48 hours, surgical drainage is required to facilitate healing.56

FINAL THOUGHTS

Inflammatory and infectious nail disorders in children are relatively common and may impact the physical and emotional well-being of young patients. By understanding the distinctive features of these nail disorders in pediatric patients, dermatologists can provide anticipatory guidance and informed treatment options to children and their parents. Further research is needed to expand our understanding of pediatric nail disorders and create targeted therapeutic interventions, particularly for NLP and psoriasis.

FIGURE 4. Acute paronychia in a 9-year-old girl with erythema, tenderness, and fluctuance of the periungual skin.

 

 

References
  1. Uber M, Carvalho VO, Abagge KT, et al. Clinical features and nail clippings in 52 children with psoriasis. Pediatr Dermatol. 2018;35:202-207. doi:10.1111/pde.13402
  2. Plachouri KM, Mulita F, Georgiou S. Management of pediatric nail psoriasis. Cutis. 2021;108:292-294. doi:10.12788/cutis.0386
  3. Smith RJ, Rubin AI. Pediatric nail disorders: a review. Curr Opin Pediatr. 2020;32:506-515. doi:10.1097/mop.0000000000000921
  4. Pourchot D, Bodemer C, Phan A, et al. Nail psoriasis: a systematic evaluation in 313 children with psoriasis. Pediatr Dermatol. 2017;34:58-63. doi:10.1111/pde.13028
  5. Richert B, André J. Nail disorders in children: diagnosis and management. Am J Clin Dermatol. 2011;12:101-112. doi:10.2165/11537110-000000000-00000
  6. Lee JYY. Severe 20-nail psoriasis successfully treated by low dose methotrexate. Dermatol Online J. 2009;15:8.
  7. Nogueira M, Paller AS, Torres T. Targeted therapy for pediatric psoriasis. Paediatr Drugs. May 2021;23:203-212. doi:10.1007/s40272-021-00443-5
  8. Hanoodi M, Mittal M. Methotrexate. StatPearls [Internet]. Updated August 16, 2023. Accessed July 1, 2024. https://www.ncbi.nlm.nih.gov/books/NBK556114/
  9. Teran CG, Teran-Escalera CN, Balderrama C. A severe case of erythrodermic psoriasis associated with advanced nail and joint manifestations: a case report. J Med Case Rep. 2010;4:179. doi:10.1186/1752-1947-4-179
  10. Paller AS, Seyger MMB, Magariños GA, et al. Long-term efficacy and safety of up to 108 weeks of ixekizumab in pediatric patients with moderate to severe plaque psoriasis: the IXORA-PEDS randomized clinical trial. JAMA Dermatol. 2022;158:533-541. doi:10.1001/jamadermatol.2022.0655
  11.  Diotallevi F, Simonetti O, Rizzetto G, et al. Biological treatments for pediatric psoriasis: state of the art and future perspectives. Int J Mol Sci. 2022;23:11128. doi:10.3390/ijms231911128
  12. Nash P, Mease PJ, Kirkham B, et al. Secukinumab provides sustained improvement in nail psoriasis, signs and symptoms of psoriatic arthritis and low rate of radiographic progression in patients with concomitant nail involvement: 2-year results from the Phase III FUTURE 5 study. Clin Exp Rheumatol. 2022;40:952-959. doi:10.55563/clinexprheumatol/3nuz51
  13. Wells LE, Evans T, Hilton R, et al. Use of secukinumab in a pediatric patient leads to significant improvement in nail psoriasis and psoriatic arthritis. Pediatr Dermatol. 2019;36:384-385. doi:10.1111/pde.13767
  14. Watabe D, Endoh K, Maeda F, et al. Childhood-onset psoriaticonycho-pachydermo-periostitis treated successfully with infliximab. Eur J Dermatol. 2015;25:506-508. doi:10.1684/ejd.2015.2616
  15. Pereira TM, Vieira AP, Fernandes JC, et al. Anti-TNF-alpha therapy in childhood pustular psoriasis. Dermatology. 2006;213:350-352. doi:10.1159/000096202
  16. Iorizzo M, Gioia Di Chiacchio N, Di Chiacchio N, et al. Intralesional steroid injections for inflammatory nail dystrophies in the pediatric population. Pediatr Dermatol. 2023;40:759-761. doi:10.1111/pde.15295
  17. Tosti A, Piraccini BM, Cambiaghi S, et al. Nail lichen planus in children: clinical features, response to treatment, and long-term follow-up. Arch Dermatol. 2001;137:1027-1032.
  18. Lipner SR. Nail lichen planus: a true nail emergency. J Am Acad Dermatol. 2019;80:e177-e178. doi:10.1016/j.jaad.2018.11.065
  19.  Iorizzo M, Tosti A, Starace M, et al. Isolated nail lichen planus: an expert consensus on treatment of the classical form. J Am Acad Dermatol. 2020;83:1717-1723. doi:10.1016/j.jaad.2020.02.056
  20. Piraccini BM, Saccani E, Starace M, et al. Nail lichen planus: response to treatment and long term follow-up. Eur J Dermatol. 2010;20:489-496. doi:10.1684/ejd.2010.0952
  21. Mahajan R, Kaushik A, De D, et al. Pediatric trachyonychia- a retrospective study of 17 cases. Indian J Dermatol. 2021;66:689-690. doi:10.4103/ijd.ijd_42_21
  22. Leung AKC, Leong KF, Barankin B. Trachyonychia. J Pediatr. 2020;216:239-239.e1. doi:10.1016/j.jpeds.2019.08.034
  23. Haber JS, Chairatchaneeboon M, Rubin AI. Trachyonychia: review and update on clinical aspects, histology, and therapy. Skin Appendage Disord. 2017;2:109-115. doi:10.1159/000449063
  24. Jacobsen AA, Tosti A. Trachyonychia and twenty-nail dystrophy: a comprehensive review and discussion of diagnostic accuracy. Skin Appendage Disord. 2016;2:7-13. doi:10.1159/000445544
  25. Kumar MG, Ciliberto H, Bayliss SJ. Long-term follow-up of pediatric trachyonychia. Pediatr Dermatol. 2015;32:198-200. doi:10.1111/pde.12427
  26. Tosti A, Piraccini BM, Iorizzo M. Trachyonychia and related disorders: evaluation and treatment plans. Dermatolog Ther. 2002;15:121-125. doi:10.1046/j.1529-8019.2002.01511.x
  27.  Leung AKC, Leong KF, Barankin B. Lichen striatus with nail involvement in a 6-year-old boy. Case Rep Pediatr. 2020;2020:1494760. doi:10.1155/2020/1494760
  28. Kim GW, Kim SH, Seo SH, et al. Lichen striatus with nail abnormality successfully treated with tacrolimus ointment. J Dermatol. 2009;36:616-617. doi:10.1111/j.1346-8138.2009.00720.x
  29. Iorizzo M, Rubin AI, Starace M. Nail lichen striatus: is dermoscopy useful for the diagnosis? Pediatr Dermatol. 2019;36:859-863. doi:10.1111/pde.13916
  30. Karp DL, Cohen BA. Onychodystrophy in lichen striatus. Pediatr Dermatol. 1993;10:359-361. doi:10.1111/j.1525-1470.1993.tb00399.x
  31. Tosti A, Peluso AM, Misciali C, et al. Nail lichen striatus: clinical features and long-term follow-up of five patients. J Am Acad Dermatol. 1997;36(6, pt 1):908-913. doi:10.1016/s0190-9622(97)80270-8
  32. Simpson EL, Thompson MM, Hanifin JM. Prevalence and morphology of hand eczema in patients with atopic dermatitis. Dermatitis. 2006;17:123-127. doi:10.2310/6620.2006.06005
  33. Sarifakioglu E, Yilmaz AE, Gorpelioglu C. Nail alterations in 250 infant patients: a clinical study. J Eur Acad Dermatol Venereol. 2008;22:741-744. doi:10.1111/j.1468-3083.2008.02592.x
  34.  Milanesi N, D’Erme AM, Gola M. Nail improvement during alitretinoin treatment: three case reports and review of the literature. Clin Exp Dermatol. 2015;40:533-536. doi:10.1111/ced.12584
  35. Chung BY, Choi YW, Kim HO, et al. Nail dystrophy in patients with atopic dermatitis and its association with disease severity. Ann Dermatol. 2019;31:121-126. doi:10.5021/ad.2019.31.2.121
  36. Navarro-Triviño FJ, Vega-Castillo JJ, Ruiz-Villaverde R. Nail changes successfully treated with dupilumab in a patient with severe atopic dermatitis. Australas J Dermatol. 2021;62:e468-e469. doi:10.1111/ajd.13633
  37. Wei SH, Huang YP, Liu MC, et al. An outbreak of coxsackievirus A6 hand, foot, and mouth disease associated with onychomadesis in Taiwan, 2010. BMC Infect Dis. 2011;11:346. doi:10.1186/1471-2334-11-346
  38. Shin JY, Cho BK, Park HJ. A clinical study of nail changes occurring secondary to hand-foot-mouth disease: onychomadesis and Beau’s lines. Ann Dermatol. 2014;26:280-283. doi:10.5021/ad.2014.26.2.280
  39. Verma S, Singal A. Nail changes in hand-foot-and-mouth disease (HFMD). Indian Dermatol Online J. 2021;12:656-657. doi:10.4103 /idoj.IDOJ_271_20
  40. Giordano LMC, de la Fuente LA, Lorca JMB, et al. Onychomadesis secondary to hand-foot-mouth disease: a frequent manifestation and cause of concern for parents. Article in Spanish. Rev Chil Pediatr. 2018;89:380-383. doi:10.4067/s0370-41062018005000203
  41. Justino MCA, da SMD, Souza MF, et al. Atypical hand-foot-mouth disease in Belém, Amazon region, northern Brazil, with detection of coxsackievirus A6. J Clin Virol. 2020;126:104307. doi:10.1016/j.jcv.2020.104307
  42. Cheng FF, Zhang BB, Cao ML, et al. Clinical characteristics of 68 children with atypical hand, foot, and mouth disease caused by coxsackievirus A6: a single-center retrospective analysis. Transl Pediatr. 2022;11:1502-1509. doi:10.21037/tp-22-352
  43. Nagata S. Causes of Kawasaki disease-from past to present. Front Pediatr. 2019;7:18. doi:10.3389/fped.2019.00018
  44. Mitsuishi T, Miyata K, Ando A, et al. Characteristic nail lesions in Kawasaki disease: case series and literature review. J Dermatol. 2022;49:232-238. doi:10.1111/1346-8138.16276
  45. Lindsley CB. Nail-bed lines in Kawasaki disease. Am J Dis Child. 1992;146:659-660. doi:10.1001/archpedi.1992.02160180017005
  46. Matsumura O, Nakagishi Y. Pincer nails upon convalescence from Kawasaki disease. J Pediatr. 2022;246:279. doi:10.1016/j.jpeds.2022.03.002
  47. Solís-Arias MP, García-Romero MT. Onychomycosis in children. a review. Int J Dermatol. 2017;56:123-130. doi:10.1111/ijd.13392
  48. Gupta AK, Mays RR, Versteeg SG, et al. Onychomycosis in children: safety and efficacy of antifungal agents. Pediatr Dermatol. 2018;35:552-559. doi:10.1111/pde.13561
  49. 49. Gupta AK, Venkataraman M, Shear NH, et al. Labeled use of efinaconazole topical solution 10% in treating onychomycosis in children and a review of the management of pediatric onychomycosis. Dermatol Ther. 2020;33:e13613. doi:10.1111/dth.13613
  50. Falotico JM, Lipner SR. Updated perspectives on the diagnosis and management of onychomycosis. Clin Cosmet Investig Dermatol. 2022;15:1933-1957. doi:10.2147/ccid.S362635
  51. Patel D, Castelo-Soccio LA, Rubin AI, et al. Laboratory monitoring during systemic terbinafine therapy for pediatric onychomycosis. JAMA Dermatol. 2017;153:1326-1327. doi:10.1001/jamadermatol.2017.4483
  52. Friedlander SF, Chan YC, Chan YH, et al. Onychomycosis does not always require systemic treatment for cure: a trial using topical therapy. Pediatr Dermatol. 2013;30:316-322. doi:10.1111/pde.12064
  53. Rich P, Spellman M, Purohit V, et al. Tavaborole 5% topical solution for the treatment of toenail onychomycosis in pediatric patients: results from a phase 4 open-label study. J Drugs Dermatol. 2019;18:190-195.
  54. Gupta AK, Venkataraman M, Abramovits W, et al. JUBLIA (efinaconazole 10% solution) in the treatment of pediatric onychomycosis. Skinmed. 2021;19:206-210.
  55. Gupta AK, Paquet M. Systemic antifungals to treat onychomycosis in children: a systematic review. Pediatr Dermatol. 2013;30:294-302. doi:10.1111/pde.12048
  56. Leggit JC. Acute and chronic paronychia. Am Fam Physician. 2017;96:44-51.
  57. Lipner SR, Scher RK. Congenital malalignment of the great toenails with acute paronychia. Pediatr Dermatol. 2016;33:e288-e289.doi:10.1111/pde.12924
Article PDF
Author and Disclosure Information

 

Eden N. Axler and Dr. Lipner are from the Israel Englander Department of Dermatology, Weill Cornell Medicine, New York, New York. Dr. Bellet is from the Department of Dermatology and the Department of Pediatrics, Duke University School of Medicine, Durham, North Carolina.

Eden N. Axler and Dr. Bellet report no conflict of interest. Dr. Lipner has served as a consultant for BelleTorus Corporation, Hoth Therapeutics, Moberg Pharma, and Ortho Dermatologics.

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

Cutis. 2024 July;114(1):E9-E15. doi:10.12788/cutis.1041

Issue
Cutis - 114(1)
Publications
Topics
Page Number
E9-E15
Sections
Author and Disclosure Information

 

Eden N. Axler and Dr. Lipner are from the Israel Englander Department of Dermatology, Weill Cornell Medicine, New York, New York. Dr. Bellet is from the Department of Dermatology and the Department of Pediatrics, Duke University School of Medicine, Durham, North Carolina.

Eden N. Axler and Dr. Bellet report no conflict of interest. Dr. Lipner has served as a consultant for BelleTorus Corporation, Hoth Therapeutics, Moberg Pharma, and Ortho Dermatologics.

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

Cutis. 2024 July;114(1):E9-E15. doi:10.12788/cutis.1041

Author and Disclosure Information

 

Eden N. Axler and Dr. Lipner are from the Israel Englander Department of Dermatology, Weill Cornell Medicine, New York, New York. Dr. Bellet is from the Department of Dermatology and the Department of Pediatrics, Duke University School of Medicine, Durham, North Carolina.

Eden N. Axler and Dr. Bellet report no conflict of interest. Dr. Lipner has served as a consultant for BelleTorus Corporation, Hoth Therapeutics, Moberg Pharma, and Ortho Dermatologics.

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

Cutis. 2024 July;114(1):E9-E15. doi:10.12788/cutis.1041

Article PDF
Article PDF

Nail disorders are common among pediatric patients but often are underdiagnosed or misdiagnosed because of their unique disease manifestations. These conditions may severely impact quality of life. There are few nail disease clinical trials that include children. Consequently, most treatment recommendations are based on case series and expert consensus recommendations. We review inflammatory and infectious nail disorders in pediatric patients. By describing characteristics, clinical manifestations, and management approaches for these conditions, we aim to provide guidance to dermatologists in their diagnosis and treatment.

INFLAMMATORY NAIL DISORDERS

Nail Psoriasis

Nail involvement in children with psoriasis is common, with prevalence estimates ranging from 17% to 39.2%.1 Nail matrix psoriasis may manifest with pitting (large irregular pits) and leukonychia as well as chromonychia and nail plate crumbling. Onycholysis, oil drop spots (salmon patches), and subungual hyperkeratosis can be seen in nail bed psoriasis. Nail pitting is the most frequently observed clinical finding (Figure 1).2,3 In a cross-sectional multicenter study of 313 children with cutaneous psoriasis in France, nail findings were present in 101 patients (32.3%). There were associations between nail findings and presence of psoriatic arthritis (P=.03), palmoplantar psoriasis (P<.001), and severity of psoriatic disease, defined as use of systemic treatment with phototherapy (psoralen plus UVA, UVB), traditional systemic treatment (acitretin, methotrexate, cyclosporine), or a biologic (P=.003).4

Topical steroids and vitamin D analogues may be used with or without occlusion and may be efficacious.5 Several case reports describe systemic treatments for psoriasis in children, including methotrexate, acitretin, and apremilast (approved for children 6 years and older for plaque psoriasis by the US Food and Drug Administration [FDA]).2 There are 5 biologic drugs currently approved for the treatment of pediatric psoriasis—adalimumab, etanercept, ustekinumab, secukinumab, ixekizumab—and 6 drugs currently undergoing phase 3 studies—brodalumab, guselkumab, risankizumab, tildrakizumab, certolizumab pegol, and deucravacitinib (Table 1).6-15 Adalimumab is specifically approved for moderate to severe nail psoriasis in adults 18 years and older.

FIGURE 1. Nail psoriasis in a 9-year-old girl with onycholysis, nail bed hyperkeratosis, and pitting, as well as discoloration.

 

Intralesional steroid injections are sometimes useful in the management of nail matrix psoriasis; however, appropriate patient selection is critical due to the pain associated with the procedure. In a prospective study of 16 children (age range, 9–17 years) with nail psoriasis treated with intralesional triamcinolone (ILTAC) 2.5 to 5 mg/mL every 4 to 8 weeks for a minimum of 3 to 6 months, 9 patients achieved resolution and 6 had improvement of clinical findings.16 Local adverse events were mild, including injection-site pain (66%), subungual hematoma (n=1), Beau lines (n=1), proximal nail fold hypopigmentation (n=2), and proximal nail fold atrophy (n=2). Because the proximal nail fold in children is thinner than in adults, there may be an increased risk for nail fold hypopigmentation and atrophy in children. Therefore, a maximum ILTAC concentration of 2.5 mg/mL with 0.2 mL maximum volume per nail per session is recommended for children younger than 15 years.16

Nail Lichen Planus

Nail lichen planus (NLP) is uncommon in children, with few biopsy-proven cases documented in the literature.17 Common clinical findings are onychorrhexis, nail plate thinning, fissuring, splitting, and atrophy with koilonychia.5 Although pterygium development (irreversible nail matrix scarring) is uncommon in pediatric patients, NLP can be progressive and may cause irreversible destruction of the nail matrix and subsequent nail loss, warranting therapeutic intervention.18

Treatment of NLP may be difficult, as there are no options that work in all patients. Current literature supports the use of systemic corticosteroids or ILTAC for the treatment of NLP; however, recurrence rates can be high. According to an expert consensus paper on NLP treatment, ILTAC may be injected in a concentration of 2.5, 5, or 10 mg/mL according to disease severity.19 In severe or resistant cases, intramuscular (IM) triamcinolone may be considered, especially if more than 3 nails are affected. A dosage of 0.5 to 1 mg/kg/mo for at least 3 to 6 months is recommended for both children and adults, with 1 mg/kg/mo recommended in the active treatment phase (first 2–3 months).19 In a retrospective review of 5 pediatric patients with NLP treated with IM triamcinolone 0.5 mg/kg/mo, 3 patients had resolution and 2 improved with treatment.20 In a prospective study of 10 children with NLP, IM triamcinolone at a dosage of 0.5 to 1 mg/kg every 30 days for 3 to 6 months resulted in resolution of nail findings in 9 patients.17 In a prospective study of 14 pediatric patients with NLP treated with 2.5 to 5 mg/mL of ILTAC, 10 achieved resolution and 3 improved.16

Intralesional triamcinolone injections may be better suited for teenagers compared to younger children who may be more apprehensive of needles. To minimize pain, it is recommended to inject ILTAC slowly at room temperature, with use of “talkesthesia” and vibration devices, 1% lidocaine, or ethyl chloride spray.18

Trachyonychia

Trachyonychia is characterized by the presence of sandpaperlike nails. It manifests with brittle thin nails with longitudinal ridging, onychoschizia, and thickened hyperkeratotic cuticles. Trachyonychia typically involves multiple nails, with a peak age of onset between 3 and 12 years.21,22 There are 2 variants: the opaque type with rough longitudinal ridging, and the shiny variant with opalescent nails and pits that reflect light. The opaque variant is more common and is associated with psoriasis, whereas the shiny variant is less common and is associated with alopecia areata.23 Although most cases are idiopathic, some are associated with psoriasis and alopecia areata, as previously noted, as well as atopic dermatitis (AD) and lichen planus.22,24

Fortunately, trachyonychia does not lead to permanent nail damage or pterygium, making treatment primarily focused on addressing functional and cosmetic concerns.24 Spontaneous resolution occurs in approximately 50% of patients. In a prospective study of 11 patients with idiopathic trachyonychia, there was partial improvement in 5 of 9 patients treated with topical steroids, 1 with only petrolatum, and 1 with vitamin supplements. Complete resolution was reported in 1 patient treated with topical steroids.25 Because trachyonychia often is self-resolving, no treatment is required and a conservative approach is strongly recommended.26 Treatment options include topical corticosteroids, tazarotene, and 5-fluorouracil. Intralesional triamcinolone, systemic cyclosporine, retinoids, systemic corticosteroids, and tofacitinib have been described in case reports, though none of these have been shown to be 100% efficacious.24

Nail Lichen Striatus

Lichen striatus involving the nail is uncommon and is characterized by onycholysis, longitudinal ridging, ­splitting, and fraying, as well as what appears to be a subungual tumor. It can encompass the entire nail or may be isolated to a portion of the nail (Figure 2). Usually, a Blaschko-linear array of flesh-colored papules on the more proximal digit directly adjacent to the nail dystrophy will be seen, though nail findings can occur in ­isolation.27-29 The underlying pathophysiology is not clear; however, one hypothesis is that a triggering event, such as trauma, induces the expression of antigens that elicit a self-limiting immune-mediated response by CD8 T lymphocytes.30

 

FIGURE 2. Lichen striatus in a 6-year-old boy with multiple fleshcolored papules in a Blaschko-linear distribution (arrows) as well as onychodystrophy and subungual hyperkeratosis of the nail. Republished under the Creative Commons Attribution (CC BY 4.0).27

Generally, nail lichen striatus spontaneously resolves in 1 to 2 years without treatment. In a prospective study of 5 patients with nail lichen striatus, the median time to resolution was 22.6 months (range, 10–30 months).31 Topical steroids may be used for pruritus. In one case report, a 3-year-old boy with nail lichen striatus of 4 months’ duration was treated with tacrolimus ointment 0.03% daily for 3 months.28

Nail AD

Nail changes with AD may be more common in adults than children or are underreported. In a study of 777 adults with AD, nail dystrophy was present in 124 patients (16%), whereas in a study of 250 pediatric patients with AD (aged 0-2 years), nail dystrophy was present in only 4 patients.32,33

Periungual inflammation from AD causes the nail changes.34 In a cross-sectional study of 24 pediatric patients with nail dystrophy due to AD, transverse grooves (Beau lines) were present in 25% (6/24), nail pitting in 16.7% (4/24), koilonychia in 16.7% (4/24), trachyonychia in 12.5% (3/24), leukonychia in 12.5% (3/24), brachyonychia in 8.3% (2/24), melanonychia in 8.3% (2/24), onychomadesis in 8.3% (2/24), onychoschizia in 8.3% (2/24), and onycholysis in 8.3% (2/24). There was an association between disease severity and presence of toenail dystrophy (P=.03).35

Topical steroids with or without occlusion can be used to treat nail changes. Although there is limited literature describing the treatment of nail AD in children, a 61-year-old man with nail changes associated with AD achieved resolution with 3 months of treatment with dupilumab.36 Anecdotally, most patients will improve with usual cutaneous AD management.

 

 

INFECTIOUS NAIL DISORDERS

Viral Infections

Hand, Foot, and Mouth Disease—Hand, foot, and mouth disease (HFMD) is a common childhood viral infection caused by various enteroviruses, most commonly coxsackievirus A16, with the A6 variant causing more severe disease. Fever and painful vesicles involving the oral mucosa as well as palms and soles give the disease its name. Nail changes are common. In a prospective study involving 130 patients with laboratory-confirmed coxsackievirus CA6 serotype infection, 37% developed onychomadesis vs only 5% of 145 cases with non-CA6 enterovirus infection who developed nail findings. There was an association between CA6 infection and presence of nail changes (P<.001).37

Findings ranging from transverse grooves (Beau lines) to complete nail shedding (onychomadesis)(Figure 3) may be seen.38,39 Nail findings in HFMD are due to transient inhibition of nail growth and present approximately 3 to 6 weeks after infection.40 Onychomadesis is seen in 30% to 68% of patients with HFMD.37,41,42 Nail findings in HFMD spontaneously resolve with nail growth (2–3 mm per month for fingernails and 1 mm per month for toenails) and do not require specific treatment. Although the appearance of nail changes associated with HFMD can be disturbing, dermatologists can reassure children and their parents that the nails will resolve with the next cycle of growth.

Kawasaki Disease—Kawasaki disease (KD) is a vasculitis primarily affecting children and infants. Although the specific pathogen and pathophysiology is not entirely clear, clinical observations have suggested an infectious cause, most likely a virus.43 In Japan, more than 15,000 cases of KD are documented annually, while approximately 4200 cases are seen in the United States.44 In a prospective study from 1984 to 1990, 4 of 26 (15.4%) patients with KD presented with nail manifestations during the late acute phase or early convalescent phase of disease. There were no significant associations between nail dystrophy and severity of KD, such as coronary artery aneurysm.45

Nail changes reported in children with KD include onychomadesis, onycholysis, orange-brown chromonychia, splinter hemorrhages, Beau lines, and pincer nails. In a review of nail changes associated with KD from 1980 to 2021, orange-brown transverse chromonychia, which may evolve into transverse leukonychia, was the most common nail finding reported, occurring in 17 of 31 (54.8%) patients.44 It has been hypothesized that nail changes may result from blood flow disturbance due to the underlying vasculitis.46 Nail changes appear several weeks after the onset of fever and are self-limited. Resolution occurs with nail growth, with no treatment required.

FIGURE 3. Onychomadesis from hand, foot, and mouth disease with yellow-orange discoloration of the nail plate. Republished under the Creative Commons Attribution (CC BY-NC-SA).39

 

 

FUNGAL INFECTIONS

Onychomycosis

Onychomycosis is a fungal infection of the nails that occurs in 0.2% to 5.5% of pediatric patients, and its prevalence may be increasing, which may be due to environmental factors or increased rates of diabetes mellitus and obesity in the pediatric population.47 Onychomycosis represents 15.5% of nail dystrophies in pediatric patients.48 Some dermatologists treat presumptive onychomycosis without confirmation; however, we do not recommend that approach. Because the differential is broad and the duration of treatment is long, mycologic examination (potassium hydroxide preparation, fungal culture, polymerase chain reaction, and/or histopathology) should be obtained to confirm onychomycosis prior to initiation of antifungal management. Family members of affected individuals should be evaluated and treated, if indicated, for onychomycosis and tinea pedis, as household transmission is common.

Currently, there are 2 topical FDA-approved treatments for pediatric onychomycosis in children 6 years and older (Table 2).49,50 There is a discussion of the need for confirmatory testing for onychomycosis in children, particularly when systemic treatment is prescribed. In a retrospective review of 269 pediatric patients with onychomycosis prescribed terbinafine, 53.5% (n=144) underwent laboratory monitoring of liver function and complete blood cell counts, and 12.5% had grade 1 laboratory abnormalities either prior to (12/144 [8.3%]) or during (6/144 [4.2%]) therapy.51 Baseline transaminase monitoring is recommended, though subsequent routine laboratory monitoring in healthy children may have limited utility with associated increased costs, incidental findings, and patient discomfort and likely is not needed.51

Pediatric onychomycosis responds better to topical therapy than adult disease, and pediatric patients do not always require systemic treatment.52 Ciclopirox is not FDA approved for the treatment of pediatric onychomycosis, but in a 32-week clinical trial of ciclopirox lacquer 8% use in 40 patients, 77% (27/35) of treated patients achieved mycologic cure. Overall, 71% of treated patients (25/35) vs 22% (2/9) of controls achieved efficacy (defined as investigator global assessment score of 2 or lower).52 In an open-label, single-arm clinical trial assessing tavaborole solution 5% applied once daily for 48 weeks for the treatment of toenail onychomycosis in pediatric patients (aged 6–17 years), 36.2% (20/55) of patients achieved mycologic cure, and 8.5% (5/55) achieved complete cure at week 52 with mild or minimal adverse effects.53 In an open-label, phase 4 study of the safety and efficacy of efinaconazole solution 10% applied once daily for 48 weeks in pediatric patients (aged 6 to 16 years) (n=60), 65% (35/60) achieved mycologic cure, 42% (25/60) achieved clinical cure, and 40% (24/60) achieved complete cure at 52 weeks. The most common adverse effects of efina­conazole were local and included ingrown toenail (1/60), application-site dermatitis (1/60), application-site vesicles (1/60), and application-site pain (1/60).54

In a systematic review of systemic antifungals for onychomycosis in 151 pediatric patients, itraconazole, fluconazole, griseofulvin, and terbinafine resulted in complete cure rates similar to those of the adult population, with excellent safety profiles.55 Depending on the situation, initiation of treatment with topical medications followed by addition of systemic antifungal agents only if needed may be an appropriate course of action.

BACTERIAL INFECTIONS

Acute Paronychia

Acute paronychia is a nail-fold infection that develops after the protective nail barrier has been compromised.56 In children, thumb-sucking, nail-biting, frequent oral manipulation of the digits, and poor skin hygiene are risk factors. Acute paronychia also may develop in association with congenital malalignment of the great toenails.57

Clinical manifestations include localized pain, erythema, and nail fold edema (Figure 4). Purulent material and abscess formation may ensue. Staphylococcus aureus as well as methicillin-resistant S aureus and Streptococcus pyogenes are classically the most common causes of acute paronychia. Treatment of paronychia is based on severity. In mild cases, warm soaks with topical antibiotics are indicated. Oral antibiotics should be prescribed for more severe presentations. If there is no improvement after 48 hours, surgical drainage is required to facilitate healing.56

FINAL THOUGHTS

Inflammatory and infectious nail disorders in children are relatively common and may impact the physical and emotional well-being of young patients. By understanding the distinctive features of these nail disorders in pediatric patients, dermatologists can provide anticipatory guidance and informed treatment options to children and their parents. Further research is needed to expand our understanding of pediatric nail disorders and create targeted therapeutic interventions, particularly for NLP and psoriasis.

FIGURE 4. Acute paronychia in a 9-year-old girl with erythema, tenderness, and fluctuance of the periungual skin.

 

 

Nail disorders are common among pediatric patients but often are underdiagnosed or misdiagnosed because of their unique disease manifestations. These conditions may severely impact quality of life. There are few nail disease clinical trials that include children. Consequently, most treatment recommendations are based on case series and expert consensus recommendations. We review inflammatory and infectious nail disorders in pediatric patients. By describing characteristics, clinical manifestations, and management approaches for these conditions, we aim to provide guidance to dermatologists in their diagnosis and treatment.

INFLAMMATORY NAIL DISORDERS

Nail Psoriasis

Nail involvement in children with psoriasis is common, with prevalence estimates ranging from 17% to 39.2%.1 Nail matrix psoriasis may manifest with pitting (large irregular pits) and leukonychia as well as chromonychia and nail plate crumbling. Onycholysis, oil drop spots (salmon patches), and subungual hyperkeratosis can be seen in nail bed psoriasis. Nail pitting is the most frequently observed clinical finding (Figure 1).2,3 In a cross-sectional multicenter study of 313 children with cutaneous psoriasis in France, nail findings were present in 101 patients (32.3%). There were associations between nail findings and presence of psoriatic arthritis (P=.03), palmoplantar psoriasis (P<.001), and severity of psoriatic disease, defined as use of systemic treatment with phototherapy (psoralen plus UVA, UVB), traditional systemic treatment (acitretin, methotrexate, cyclosporine), or a biologic (P=.003).4

Topical steroids and vitamin D analogues may be used with or without occlusion and may be efficacious.5 Several case reports describe systemic treatments for psoriasis in children, including methotrexate, acitretin, and apremilast (approved for children 6 years and older for plaque psoriasis by the US Food and Drug Administration [FDA]).2 There are 5 biologic drugs currently approved for the treatment of pediatric psoriasis—adalimumab, etanercept, ustekinumab, secukinumab, ixekizumab—and 6 drugs currently undergoing phase 3 studies—brodalumab, guselkumab, risankizumab, tildrakizumab, certolizumab pegol, and deucravacitinib (Table 1).6-15 Adalimumab is specifically approved for moderate to severe nail psoriasis in adults 18 years and older.

FIGURE 1. Nail psoriasis in a 9-year-old girl with onycholysis, nail bed hyperkeratosis, and pitting, as well as discoloration.

 

Intralesional steroid injections are sometimes useful in the management of nail matrix psoriasis; however, appropriate patient selection is critical due to the pain associated with the procedure. In a prospective study of 16 children (age range, 9–17 years) with nail psoriasis treated with intralesional triamcinolone (ILTAC) 2.5 to 5 mg/mL every 4 to 8 weeks for a minimum of 3 to 6 months, 9 patients achieved resolution and 6 had improvement of clinical findings.16 Local adverse events were mild, including injection-site pain (66%), subungual hematoma (n=1), Beau lines (n=1), proximal nail fold hypopigmentation (n=2), and proximal nail fold atrophy (n=2). Because the proximal nail fold in children is thinner than in adults, there may be an increased risk for nail fold hypopigmentation and atrophy in children. Therefore, a maximum ILTAC concentration of 2.5 mg/mL with 0.2 mL maximum volume per nail per session is recommended for children younger than 15 years.16

Nail Lichen Planus

Nail lichen planus (NLP) is uncommon in children, with few biopsy-proven cases documented in the literature.17 Common clinical findings are onychorrhexis, nail plate thinning, fissuring, splitting, and atrophy with koilonychia.5 Although pterygium development (irreversible nail matrix scarring) is uncommon in pediatric patients, NLP can be progressive and may cause irreversible destruction of the nail matrix and subsequent nail loss, warranting therapeutic intervention.18

Treatment of NLP may be difficult, as there are no options that work in all patients. Current literature supports the use of systemic corticosteroids or ILTAC for the treatment of NLP; however, recurrence rates can be high. According to an expert consensus paper on NLP treatment, ILTAC may be injected in a concentration of 2.5, 5, or 10 mg/mL according to disease severity.19 In severe or resistant cases, intramuscular (IM) triamcinolone may be considered, especially if more than 3 nails are affected. A dosage of 0.5 to 1 mg/kg/mo for at least 3 to 6 months is recommended for both children and adults, with 1 mg/kg/mo recommended in the active treatment phase (first 2–3 months).19 In a retrospective review of 5 pediatric patients with NLP treated with IM triamcinolone 0.5 mg/kg/mo, 3 patients had resolution and 2 improved with treatment.20 In a prospective study of 10 children with NLP, IM triamcinolone at a dosage of 0.5 to 1 mg/kg every 30 days for 3 to 6 months resulted in resolution of nail findings in 9 patients.17 In a prospective study of 14 pediatric patients with NLP treated with 2.5 to 5 mg/mL of ILTAC, 10 achieved resolution and 3 improved.16

Intralesional triamcinolone injections may be better suited for teenagers compared to younger children who may be more apprehensive of needles. To minimize pain, it is recommended to inject ILTAC slowly at room temperature, with use of “talkesthesia” and vibration devices, 1% lidocaine, or ethyl chloride spray.18

Trachyonychia

Trachyonychia is characterized by the presence of sandpaperlike nails. It manifests with brittle thin nails with longitudinal ridging, onychoschizia, and thickened hyperkeratotic cuticles. Trachyonychia typically involves multiple nails, with a peak age of onset between 3 and 12 years.21,22 There are 2 variants: the opaque type with rough longitudinal ridging, and the shiny variant with opalescent nails and pits that reflect light. The opaque variant is more common and is associated with psoriasis, whereas the shiny variant is less common and is associated with alopecia areata.23 Although most cases are idiopathic, some are associated with psoriasis and alopecia areata, as previously noted, as well as atopic dermatitis (AD) and lichen planus.22,24

Fortunately, trachyonychia does not lead to permanent nail damage or pterygium, making treatment primarily focused on addressing functional and cosmetic concerns.24 Spontaneous resolution occurs in approximately 50% of patients. In a prospective study of 11 patients with idiopathic trachyonychia, there was partial improvement in 5 of 9 patients treated with topical steroids, 1 with only petrolatum, and 1 with vitamin supplements. Complete resolution was reported in 1 patient treated with topical steroids.25 Because trachyonychia often is self-resolving, no treatment is required and a conservative approach is strongly recommended.26 Treatment options include topical corticosteroids, tazarotene, and 5-fluorouracil. Intralesional triamcinolone, systemic cyclosporine, retinoids, systemic corticosteroids, and tofacitinib have been described in case reports, though none of these have been shown to be 100% efficacious.24

Nail Lichen Striatus

Lichen striatus involving the nail is uncommon and is characterized by onycholysis, longitudinal ridging, ­splitting, and fraying, as well as what appears to be a subungual tumor. It can encompass the entire nail or may be isolated to a portion of the nail (Figure 2). Usually, a Blaschko-linear array of flesh-colored papules on the more proximal digit directly adjacent to the nail dystrophy will be seen, though nail findings can occur in ­isolation.27-29 The underlying pathophysiology is not clear; however, one hypothesis is that a triggering event, such as trauma, induces the expression of antigens that elicit a self-limiting immune-mediated response by CD8 T lymphocytes.30

 

FIGURE 2. Lichen striatus in a 6-year-old boy with multiple fleshcolored papules in a Blaschko-linear distribution (arrows) as well as onychodystrophy and subungual hyperkeratosis of the nail. Republished under the Creative Commons Attribution (CC BY 4.0).27

Generally, nail lichen striatus spontaneously resolves in 1 to 2 years without treatment. In a prospective study of 5 patients with nail lichen striatus, the median time to resolution was 22.6 months (range, 10–30 months).31 Topical steroids may be used for pruritus. In one case report, a 3-year-old boy with nail lichen striatus of 4 months’ duration was treated with tacrolimus ointment 0.03% daily for 3 months.28

Nail AD

Nail changes with AD may be more common in adults than children or are underreported. In a study of 777 adults with AD, nail dystrophy was present in 124 patients (16%), whereas in a study of 250 pediatric patients with AD (aged 0-2 years), nail dystrophy was present in only 4 patients.32,33

Periungual inflammation from AD causes the nail changes.34 In a cross-sectional study of 24 pediatric patients with nail dystrophy due to AD, transverse grooves (Beau lines) were present in 25% (6/24), nail pitting in 16.7% (4/24), koilonychia in 16.7% (4/24), trachyonychia in 12.5% (3/24), leukonychia in 12.5% (3/24), brachyonychia in 8.3% (2/24), melanonychia in 8.3% (2/24), onychomadesis in 8.3% (2/24), onychoschizia in 8.3% (2/24), and onycholysis in 8.3% (2/24). There was an association between disease severity and presence of toenail dystrophy (P=.03).35

Topical steroids with or without occlusion can be used to treat nail changes. Although there is limited literature describing the treatment of nail AD in children, a 61-year-old man with nail changes associated with AD achieved resolution with 3 months of treatment with dupilumab.36 Anecdotally, most patients will improve with usual cutaneous AD management.

 

 

INFECTIOUS NAIL DISORDERS

Viral Infections

Hand, Foot, and Mouth Disease—Hand, foot, and mouth disease (HFMD) is a common childhood viral infection caused by various enteroviruses, most commonly coxsackievirus A16, with the A6 variant causing more severe disease. Fever and painful vesicles involving the oral mucosa as well as palms and soles give the disease its name. Nail changes are common. In a prospective study involving 130 patients with laboratory-confirmed coxsackievirus CA6 serotype infection, 37% developed onychomadesis vs only 5% of 145 cases with non-CA6 enterovirus infection who developed nail findings. There was an association between CA6 infection and presence of nail changes (P<.001).37

Findings ranging from transverse grooves (Beau lines) to complete nail shedding (onychomadesis)(Figure 3) may be seen.38,39 Nail findings in HFMD are due to transient inhibition of nail growth and present approximately 3 to 6 weeks after infection.40 Onychomadesis is seen in 30% to 68% of patients with HFMD.37,41,42 Nail findings in HFMD spontaneously resolve with nail growth (2–3 mm per month for fingernails and 1 mm per month for toenails) and do not require specific treatment. Although the appearance of nail changes associated with HFMD can be disturbing, dermatologists can reassure children and their parents that the nails will resolve with the next cycle of growth.

Kawasaki Disease—Kawasaki disease (KD) is a vasculitis primarily affecting children and infants. Although the specific pathogen and pathophysiology is not entirely clear, clinical observations have suggested an infectious cause, most likely a virus.43 In Japan, more than 15,000 cases of KD are documented annually, while approximately 4200 cases are seen in the United States.44 In a prospective study from 1984 to 1990, 4 of 26 (15.4%) patients with KD presented with nail manifestations during the late acute phase or early convalescent phase of disease. There were no significant associations between nail dystrophy and severity of KD, such as coronary artery aneurysm.45

Nail changes reported in children with KD include onychomadesis, onycholysis, orange-brown chromonychia, splinter hemorrhages, Beau lines, and pincer nails. In a review of nail changes associated with KD from 1980 to 2021, orange-brown transverse chromonychia, which may evolve into transverse leukonychia, was the most common nail finding reported, occurring in 17 of 31 (54.8%) patients.44 It has been hypothesized that nail changes may result from blood flow disturbance due to the underlying vasculitis.46 Nail changes appear several weeks after the onset of fever and are self-limited. Resolution occurs with nail growth, with no treatment required.

FIGURE 3. Onychomadesis from hand, foot, and mouth disease with yellow-orange discoloration of the nail plate. Republished under the Creative Commons Attribution (CC BY-NC-SA).39

 

 

FUNGAL INFECTIONS

Onychomycosis

Onychomycosis is a fungal infection of the nails that occurs in 0.2% to 5.5% of pediatric patients, and its prevalence may be increasing, which may be due to environmental factors or increased rates of diabetes mellitus and obesity in the pediatric population.47 Onychomycosis represents 15.5% of nail dystrophies in pediatric patients.48 Some dermatologists treat presumptive onychomycosis without confirmation; however, we do not recommend that approach. Because the differential is broad and the duration of treatment is long, mycologic examination (potassium hydroxide preparation, fungal culture, polymerase chain reaction, and/or histopathology) should be obtained to confirm onychomycosis prior to initiation of antifungal management. Family members of affected individuals should be evaluated and treated, if indicated, for onychomycosis and tinea pedis, as household transmission is common.

Currently, there are 2 topical FDA-approved treatments for pediatric onychomycosis in children 6 years and older (Table 2).49,50 There is a discussion of the need for confirmatory testing for onychomycosis in children, particularly when systemic treatment is prescribed. In a retrospective review of 269 pediatric patients with onychomycosis prescribed terbinafine, 53.5% (n=144) underwent laboratory monitoring of liver function and complete blood cell counts, and 12.5% had grade 1 laboratory abnormalities either prior to (12/144 [8.3%]) or during (6/144 [4.2%]) therapy.51 Baseline transaminase monitoring is recommended, though subsequent routine laboratory monitoring in healthy children may have limited utility with associated increased costs, incidental findings, and patient discomfort and likely is not needed.51

Pediatric onychomycosis responds better to topical therapy than adult disease, and pediatric patients do not always require systemic treatment.52 Ciclopirox is not FDA approved for the treatment of pediatric onychomycosis, but in a 32-week clinical trial of ciclopirox lacquer 8% use in 40 patients, 77% (27/35) of treated patients achieved mycologic cure. Overall, 71% of treated patients (25/35) vs 22% (2/9) of controls achieved efficacy (defined as investigator global assessment score of 2 or lower).52 In an open-label, single-arm clinical trial assessing tavaborole solution 5% applied once daily for 48 weeks for the treatment of toenail onychomycosis in pediatric patients (aged 6–17 years), 36.2% (20/55) of patients achieved mycologic cure, and 8.5% (5/55) achieved complete cure at week 52 with mild or minimal adverse effects.53 In an open-label, phase 4 study of the safety and efficacy of efinaconazole solution 10% applied once daily for 48 weeks in pediatric patients (aged 6 to 16 years) (n=60), 65% (35/60) achieved mycologic cure, 42% (25/60) achieved clinical cure, and 40% (24/60) achieved complete cure at 52 weeks. The most common adverse effects of efina­conazole were local and included ingrown toenail (1/60), application-site dermatitis (1/60), application-site vesicles (1/60), and application-site pain (1/60).54

In a systematic review of systemic antifungals for onychomycosis in 151 pediatric patients, itraconazole, fluconazole, griseofulvin, and terbinafine resulted in complete cure rates similar to those of the adult population, with excellent safety profiles.55 Depending on the situation, initiation of treatment with topical medications followed by addition of systemic antifungal agents only if needed may be an appropriate course of action.

BACTERIAL INFECTIONS

Acute Paronychia

Acute paronychia is a nail-fold infection that develops after the protective nail barrier has been compromised.56 In children, thumb-sucking, nail-biting, frequent oral manipulation of the digits, and poor skin hygiene are risk factors. Acute paronychia also may develop in association with congenital malalignment of the great toenails.57

Clinical manifestations include localized pain, erythema, and nail fold edema (Figure 4). Purulent material and abscess formation may ensue. Staphylococcus aureus as well as methicillin-resistant S aureus and Streptococcus pyogenes are classically the most common causes of acute paronychia. Treatment of paronychia is based on severity. In mild cases, warm soaks with topical antibiotics are indicated. Oral antibiotics should be prescribed for more severe presentations. If there is no improvement after 48 hours, surgical drainage is required to facilitate healing.56

FINAL THOUGHTS

Inflammatory and infectious nail disorders in children are relatively common and may impact the physical and emotional well-being of young patients. By understanding the distinctive features of these nail disorders in pediatric patients, dermatologists can provide anticipatory guidance and informed treatment options to children and their parents. Further research is needed to expand our understanding of pediatric nail disorders and create targeted therapeutic interventions, particularly for NLP and psoriasis.

FIGURE 4. Acute paronychia in a 9-year-old girl with erythema, tenderness, and fluctuance of the periungual skin.

 

 

References
  1. Uber M, Carvalho VO, Abagge KT, et al. Clinical features and nail clippings in 52 children with psoriasis. Pediatr Dermatol. 2018;35:202-207. doi:10.1111/pde.13402
  2. Plachouri KM, Mulita F, Georgiou S. Management of pediatric nail psoriasis. Cutis. 2021;108:292-294. doi:10.12788/cutis.0386
  3. Smith RJ, Rubin AI. Pediatric nail disorders: a review. Curr Opin Pediatr. 2020;32:506-515. doi:10.1097/mop.0000000000000921
  4. Pourchot D, Bodemer C, Phan A, et al. Nail psoriasis: a systematic evaluation in 313 children with psoriasis. Pediatr Dermatol. 2017;34:58-63. doi:10.1111/pde.13028
  5. Richert B, André J. Nail disorders in children: diagnosis and management. Am J Clin Dermatol. 2011;12:101-112. doi:10.2165/11537110-000000000-00000
  6. Lee JYY. Severe 20-nail psoriasis successfully treated by low dose methotrexate. Dermatol Online J. 2009;15:8.
  7. Nogueira M, Paller AS, Torres T. Targeted therapy for pediatric psoriasis. Paediatr Drugs. May 2021;23:203-212. doi:10.1007/s40272-021-00443-5
  8. Hanoodi M, Mittal M. Methotrexate. StatPearls [Internet]. Updated August 16, 2023. Accessed July 1, 2024. https://www.ncbi.nlm.nih.gov/books/NBK556114/
  9. Teran CG, Teran-Escalera CN, Balderrama C. A severe case of erythrodermic psoriasis associated with advanced nail and joint manifestations: a case report. J Med Case Rep. 2010;4:179. doi:10.1186/1752-1947-4-179
  10. Paller AS, Seyger MMB, Magariños GA, et al. Long-term efficacy and safety of up to 108 weeks of ixekizumab in pediatric patients with moderate to severe plaque psoriasis: the IXORA-PEDS randomized clinical trial. JAMA Dermatol. 2022;158:533-541. doi:10.1001/jamadermatol.2022.0655
  11.  Diotallevi F, Simonetti O, Rizzetto G, et al. Biological treatments for pediatric psoriasis: state of the art and future perspectives. Int J Mol Sci. 2022;23:11128. doi:10.3390/ijms231911128
  12. Nash P, Mease PJ, Kirkham B, et al. Secukinumab provides sustained improvement in nail psoriasis, signs and symptoms of psoriatic arthritis and low rate of radiographic progression in patients with concomitant nail involvement: 2-year results from the Phase III FUTURE 5 study. Clin Exp Rheumatol. 2022;40:952-959. doi:10.55563/clinexprheumatol/3nuz51
  13. Wells LE, Evans T, Hilton R, et al. Use of secukinumab in a pediatric patient leads to significant improvement in nail psoriasis and psoriatic arthritis. Pediatr Dermatol. 2019;36:384-385. doi:10.1111/pde.13767
  14. Watabe D, Endoh K, Maeda F, et al. Childhood-onset psoriaticonycho-pachydermo-periostitis treated successfully with infliximab. Eur J Dermatol. 2015;25:506-508. doi:10.1684/ejd.2015.2616
  15. Pereira TM, Vieira AP, Fernandes JC, et al. Anti-TNF-alpha therapy in childhood pustular psoriasis. Dermatology. 2006;213:350-352. doi:10.1159/000096202
  16. Iorizzo M, Gioia Di Chiacchio N, Di Chiacchio N, et al. Intralesional steroid injections for inflammatory nail dystrophies in the pediatric population. Pediatr Dermatol. 2023;40:759-761. doi:10.1111/pde.15295
  17. Tosti A, Piraccini BM, Cambiaghi S, et al. Nail lichen planus in children: clinical features, response to treatment, and long-term follow-up. Arch Dermatol. 2001;137:1027-1032.
  18. Lipner SR. Nail lichen planus: a true nail emergency. J Am Acad Dermatol. 2019;80:e177-e178. doi:10.1016/j.jaad.2018.11.065
  19.  Iorizzo M, Tosti A, Starace M, et al. Isolated nail lichen planus: an expert consensus on treatment of the classical form. J Am Acad Dermatol. 2020;83:1717-1723. doi:10.1016/j.jaad.2020.02.056
  20. Piraccini BM, Saccani E, Starace M, et al. Nail lichen planus: response to treatment and long term follow-up. Eur J Dermatol. 2010;20:489-496. doi:10.1684/ejd.2010.0952
  21. Mahajan R, Kaushik A, De D, et al. Pediatric trachyonychia- a retrospective study of 17 cases. Indian J Dermatol. 2021;66:689-690. doi:10.4103/ijd.ijd_42_21
  22. Leung AKC, Leong KF, Barankin B. Trachyonychia. J Pediatr. 2020;216:239-239.e1. doi:10.1016/j.jpeds.2019.08.034
  23. Haber JS, Chairatchaneeboon M, Rubin AI. Trachyonychia: review and update on clinical aspects, histology, and therapy. Skin Appendage Disord. 2017;2:109-115. doi:10.1159/000449063
  24. Jacobsen AA, Tosti A. Trachyonychia and twenty-nail dystrophy: a comprehensive review and discussion of diagnostic accuracy. Skin Appendage Disord. 2016;2:7-13. doi:10.1159/000445544
  25. Kumar MG, Ciliberto H, Bayliss SJ. Long-term follow-up of pediatric trachyonychia. Pediatr Dermatol. 2015;32:198-200. doi:10.1111/pde.12427
  26. Tosti A, Piraccini BM, Iorizzo M. Trachyonychia and related disorders: evaluation and treatment plans. Dermatolog Ther. 2002;15:121-125. doi:10.1046/j.1529-8019.2002.01511.x
  27.  Leung AKC, Leong KF, Barankin B. Lichen striatus with nail involvement in a 6-year-old boy. Case Rep Pediatr. 2020;2020:1494760. doi:10.1155/2020/1494760
  28. Kim GW, Kim SH, Seo SH, et al. Lichen striatus with nail abnormality successfully treated with tacrolimus ointment. J Dermatol. 2009;36:616-617. doi:10.1111/j.1346-8138.2009.00720.x
  29. Iorizzo M, Rubin AI, Starace M. Nail lichen striatus: is dermoscopy useful for the diagnosis? Pediatr Dermatol. 2019;36:859-863. doi:10.1111/pde.13916
  30. Karp DL, Cohen BA. Onychodystrophy in lichen striatus. Pediatr Dermatol. 1993;10:359-361. doi:10.1111/j.1525-1470.1993.tb00399.x
  31. Tosti A, Peluso AM, Misciali C, et al. Nail lichen striatus: clinical features and long-term follow-up of five patients. J Am Acad Dermatol. 1997;36(6, pt 1):908-913. doi:10.1016/s0190-9622(97)80270-8
  32. Simpson EL, Thompson MM, Hanifin JM. Prevalence and morphology of hand eczema in patients with atopic dermatitis. Dermatitis. 2006;17:123-127. doi:10.2310/6620.2006.06005
  33. Sarifakioglu E, Yilmaz AE, Gorpelioglu C. Nail alterations in 250 infant patients: a clinical study. J Eur Acad Dermatol Venereol. 2008;22:741-744. doi:10.1111/j.1468-3083.2008.02592.x
  34.  Milanesi N, D’Erme AM, Gola M. Nail improvement during alitretinoin treatment: three case reports and review of the literature. Clin Exp Dermatol. 2015;40:533-536. doi:10.1111/ced.12584
  35. Chung BY, Choi YW, Kim HO, et al. Nail dystrophy in patients with atopic dermatitis and its association with disease severity. Ann Dermatol. 2019;31:121-126. doi:10.5021/ad.2019.31.2.121
  36. Navarro-Triviño FJ, Vega-Castillo JJ, Ruiz-Villaverde R. Nail changes successfully treated with dupilumab in a patient with severe atopic dermatitis. Australas J Dermatol. 2021;62:e468-e469. doi:10.1111/ajd.13633
  37. Wei SH, Huang YP, Liu MC, et al. An outbreak of coxsackievirus A6 hand, foot, and mouth disease associated with onychomadesis in Taiwan, 2010. BMC Infect Dis. 2011;11:346. doi:10.1186/1471-2334-11-346
  38. Shin JY, Cho BK, Park HJ. A clinical study of nail changes occurring secondary to hand-foot-mouth disease: onychomadesis and Beau’s lines. Ann Dermatol. 2014;26:280-283. doi:10.5021/ad.2014.26.2.280
  39. Verma S, Singal A. Nail changes in hand-foot-and-mouth disease (HFMD). Indian Dermatol Online J. 2021;12:656-657. doi:10.4103 /idoj.IDOJ_271_20
  40. Giordano LMC, de la Fuente LA, Lorca JMB, et al. Onychomadesis secondary to hand-foot-mouth disease: a frequent manifestation and cause of concern for parents. Article in Spanish. Rev Chil Pediatr. 2018;89:380-383. doi:10.4067/s0370-41062018005000203
  41. Justino MCA, da SMD, Souza MF, et al. Atypical hand-foot-mouth disease in Belém, Amazon region, northern Brazil, with detection of coxsackievirus A6. J Clin Virol. 2020;126:104307. doi:10.1016/j.jcv.2020.104307
  42. Cheng FF, Zhang BB, Cao ML, et al. Clinical characteristics of 68 children with atypical hand, foot, and mouth disease caused by coxsackievirus A6: a single-center retrospective analysis. Transl Pediatr. 2022;11:1502-1509. doi:10.21037/tp-22-352
  43. Nagata S. Causes of Kawasaki disease-from past to present. Front Pediatr. 2019;7:18. doi:10.3389/fped.2019.00018
  44. Mitsuishi T, Miyata K, Ando A, et al. Characteristic nail lesions in Kawasaki disease: case series and literature review. J Dermatol. 2022;49:232-238. doi:10.1111/1346-8138.16276
  45. Lindsley CB. Nail-bed lines in Kawasaki disease. Am J Dis Child. 1992;146:659-660. doi:10.1001/archpedi.1992.02160180017005
  46. Matsumura O, Nakagishi Y. Pincer nails upon convalescence from Kawasaki disease. J Pediatr. 2022;246:279. doi:10.1016/j.jpeds.2022.03.002
  47. Solís-Arias MP, García-Romero MT. Onychomycosis in children. a review. Int J Dermatol. 2017;56:123-130. doi:10.1111/ijd.13392
  48. Gupta AK, Mays RR, Versteeg SG, et al. Onychomycosis in children: safety and efficacy of antifungal agents. Pediatr Dermatol. 2018;35:552-559. doi:10.1111/pde.13561
  49. 49. Gupta AK, Venkataraman M, Shear NH, et al. Labeled use of efinaconazole topical solution 10% in treating onychomycosis in children and a review of the management of pediatric onychomycosis. Dermatol Ther. 2020;33:e13613. doi:10.1111/dth.13613
  50. Falotico JM, Lipner SR. Updated perspectives on the diagnosis and management of onychomycosis. Clin Cosmet Investig Dermatol. 2022;15:1933-1957. doi:10.2147/ccid.S362635
  51. Patel D, Castelo-Soccio LA, Rubin AI, et al. Laboratory monitoring during systemic terbinafine therapy for pediatric onychomycosis. JAMA Dermatol. 2017;153:1326-1327. doi:10.1001/jamadermatol.2017.4483
  52. Friedlander SF, Chan YC, Chan YH, et al. Onychomycosis does not always require systemic treatment for cure: a trial using topical therapy. Pediatr Dermatol. 2013;30:316-322. doi:10.1111/pde.12064
  53. Rich P, Spellman M, Purohit V, et al. Tavaborole 5% topical solution for the treatment of toenail onychomycosis in pediatric patients: results from a phase 4 open-label study. J Drugs Dermatol. 2019;18:190-195.
  54. Gupta AK, Venkataraman M, Abramovits W, et al. JUBLIA (efinaconazole 10% solution) in the treatment of pediatric onychomycosis. Skinmed. 2021;19:206-210.
  55. Gupta AK, Paquet M. Systemic antifungals to treat onychomycosis in children: a systematic review. Pediatr Dermatol. 2013;30:294-302. doi:10.1111/pde.12048
  56. Leggit JC. Acute and chronic paronychia. Am Fam Physician. 2017;96:44-51.
  57. Lipner SR, Scher RK. Congenital malalignment of the great toenails with acute paronychia. Pediatr Dermatol. 2016;33:e288-e289.doi:10.1111/pde.12924
References
  1. Uber M, Carvalho VO, Abagge KT, et al. Clinical features and nail clippings in 52 children with psoriasis. Pediatr Dermatol. 2018;35:202-207. doi:10.1111/pde.13402
  2. Plachouri KM, Mulita F, Georgiou S. Management of pediatric nail psoriasis. Cutis. 2021;108:292-294. doi:10.12788/cutis.0386
  3. Smith RJ, Rubin AI. Pediatric nail disorders: a review. Curr Opin Pediatr. 2020;32:506-515. doi:10.1097/mop.0000000000000921
  4. Pourchot D, Bodemer C, Phan A, et al. Nail psoriasis: a systematic evaluation in 313 children with psoriasis. Pediatr Dermatol. 2017;34:58-63. doi:10.1111/pde.13028
  5. Richert B, André J. Nail disorders in children: diagnosis and management. Am J Clin Dermatol. 2011;12:101-112. doi:10.2165/11537110-000000000-00000
  6. Lee JYY. Severe 20-nail psoriasis successfully treated by low dose methotrexate. Dermatol Online J. 2009;15:8.
  7. Nogueira M, Paller AS, Torres T. Targeted therapy for pediatric psoriasis. Paediatr Drugs. May 2021;23:203-212. doi:10.1007/s40272-021-00443-5
  8. Hanoodi M, Mittal M. Methotrexate. StatPearls [Internet]. Updated August 16, 2023. Accessed July 1, 2024. https://www.ncbi.nlm.nih.gov/books/NBK556114/
  9. Teran CG, Teran-Escalera CN, Balderrama C. A severe case of erythrodermic psoriasis associated with advanced nail and joint manifestations: a case report. J Med Case Rep. 2010;4:179. doi:10.1186/1752-1947-4-179
  10. Paller AS, Seyger MMB, Magariños GA, et al. Long-term efficacy and safety of up to 108 weeks of ixekizumab in pediatric patients with moderate to severe plaque psoriasis: the IXORA-PEDS randomized clinical trial. JAMA Dermatol. 2022;158:533-541. doi:10.1001/jamadermatol.2022.0655
  11.  Diotallevi F, Simonetti O, Rizzetto G, et al. Biological treatments for pediatric psoriasis: state of the art and future perspectives. Int J Mol Sci. 2022;23:11128. doi:10.3390/ijms231911128
  12. Nash P, Mease PJ, Kirkham B, et al. Secukinumab provides sustained improvement in nail psoriasis, signs and symptoms of psoriatic arthritis and low rate of radiographic progression in patients with concomitant nail involvement: 2-year results from the Phase III FUTURE 5 study. Clin Exp Rheumatol. 2022;40:952-959. doi:10.55563/clinexprheumatol/3nuz51
  13. Wells LE, Evans T, Hilton R, et al. Use of secukinumab in a pediatric patient leads to significant improvement in nail psoriasis and psoriatic arthritis. Pediatr Dermatol. 2019;36:384-385. doi:10.1111/pde.13767
  14. Watabe D, Endoh K, Maeda F, et al. Childhood-onset psoriaticonycho-pachydermo-periostitis treated successfully with infliximab. Eur J Dermatol. 2015;25:506-508. doi:10.1684/ejd.2015.2616
  15. Pereira TM, Vieira AP, Fernandes JC, et al. Anti-TNF-alpha therapy in childhood pustular psoriasis. Dermatology. 2006;213:350-352. doi:10.1159/000096202
  16. Iorizzo M, Gioia Di Chiacchio N, Di Chiacchio N, et al. Intralesional steroid injections for inflammatory nail dystrophies in the pediatric population. Pediatr Dermatol. 2023;40:759-761. doi:10.1111/pde.15295
  17. Tosti A, Piraccini BM, Cambiaghi S, et al. Nail lichen planus in children: clinical features, response to treatment, and long-term follow-up. Arch Dermatol. 2001;137:1027-1032.
  18. Lipner SR. Nail lichen planus: a true nail emergency. J Am Acad Dermatol. 2019;80:e177-e178. doi:10.1016/j.jaad.2018.11.065
  19.  Iorizzo M, Tosti A, Starace M, et al. Isolated nail lichen planus: an expert consensus on treatment of the classical form. J Am Acad Dermatol. 2020;83:1717-1723. doi:10.1016/j.jaad.2020.02.056
  20. Piraccini BM, Saccani E, Starace M, et al. Nail lichen planus: response to treatment and long term follow-up. Eur J Dermatol. 2010;20:489-496. doi:10.1684/ejd.2010.0952
  21. Mahajan R, Kaushik A, De D, et al. Pediatric trachyonychia- a retrospective study of 17 cases. Indian J Dermatol. 2021;66:689-690. doi:10.4103/ijd.ijd_42_21
  22. Leung AKC, Leong KF, Barankin B. Trachyonychia. J Pediatr. 2020;216:239-239.e1. doi:10.1016/j.jpeds.2019.08.034
  23. Haber JS, Chairatchaneeboon M, Rubin AI. Trachyonychia: review and update on clinical aspects, histology, and therapy. Skin Appendage Disord. 2017;2:109-115. doi:10.1159/000449063
  24. Jacobsen AA, Tosti A. Trachyonychia and twenty-nail dystrophy: a comprehensive review and discussion of diagnostic accuracy. Skin Appendage Disord. 2016;2:7-13. doi:10.1159/000445544
  25. Kumar MG, Ciliberto H, Bayliss SJ. Long-term follow-up of pediatric trachyonychia. Pediatr Dermatol. 2015;32:198-200. doi:10.1111/pde.12427
  26. Tosti A, Piraccini BM, Iorizzo M. Trachyonychia and related disorders: evaluation and treatment plans. Dermatolog Ther. 2002;15:121-125. doi:10.1046/j.1529-8019.2002.01511.x
  27.  Leung AKC, Leong KF, Barankin B. Lichen striatus with nail involvement in a 6-year-old boy. Case Rep Pediatr. 2020;2020:1494760. doi:10.1155/2020/1494760
  28. Kim GW, Kim SH, Seo SH, et al. Lichen striatus with nail abnormality successfully treated with tacrolimus ointment. J Dermatol. 2009;36:616-617. doi:10.1111/j.1346-8138.2009.00720.x
  29. Iorizzo M, Rubin AI, Starace M. Nail lichen striatus: is dermoscopy useful for the diagnosis? Pediatr Dermatol. 2019;36:859-863. doi:10.1111/pde.13916
  30. Karp DL, Cohen BA. Onychodystrophy in lichen striatus. Pediatr Dermatol. 1993;10:359-361. doi:10.1111/j.1525-1470.1993.tb00399.x
  31. Tosti A, Peluso AM, Misciali C, et al. Nail lichen striatus: clinical features and long-term follow-up of five patients. J Am Acad Dermatol. 1997;36(6, pt 1):908-913. doi:10.1016/s0190-9622(97)80270-8
  32. Simpson EL, Thompson MM, Hanifin JM. Prevalence and morphology of hand eczema in patients with atopic dermatitis. Dermatitis. 2006;17:123-127. doi:10.2310/6620.2006.06005
  33. Sarifakioglu E, Yilmaz AE, Gorpelioglu C. Nail alterations in 250 infant patients: a clinical study. J Eur Acad Dermatol Venereol. 2008;22:741-744. doi:10.1111/j.1468-3083.2008.02592.x
  34.  Milanesi N, D’Erme AM, Gola M. Nail improvement during alitretinoin treatment: three case reports and review of the literature. Clin Exp Dermatol. 2015;40:533-536. doi:10.1111/ced.12584
  35. Chung BY, Choi YW, Kim HO, et al. Nail dystrophy in patients with atopic dermatitis and its association with disease severity. Ann Dermatol. 2019;31:121-126. doi:10.5021/ad.2019.31.2.121
  36. Navarro-Triviño FJ, Vega-Castillo JJ, Ruiz-Villaverde R. Nail changes successfully treated with dupilumab in a patient with severe atopic dermatitis. Australas J Dermatol. 2021;62:e468-e469. doi:10.1111/ajd.13633
  37. Wei SH, Huang YP, Liu MC, et al. An outbreak of coxsackievirus A6 hand, foot, and mouth disease associated with onychomadesis in Taiwan, 2010. BMC Infect Dis. 2011;11:346. doi:10.1186/1471-2334-11-346
  38. Shin JY, Cho BK, Park HJ. A clinical study of nail changes occurring secondary to hand-foot-mouth disease: onychomadesis and Beau’s lines. Ann Dermatol. 2014;26:280-283. doi:10.5021/ad.2014.26.2.280
  39. Verma S, Singal A. Nail changes in hand-foot-and-mouth disease (HFMD). Indian Dermatol Online J. 2021;12:656-657. doi:10.4103 /idoj.IDOJ_271_20
  40. Giordano LMC, de la Fuente LA, Lorca JMB, et al. Onychomadesis secondary to hand-foot-mouth disease: a frequent manifestation and cause of concern for parents. Article in Spanish. Rev Chil Pediatr. 2018;89:380-383. doi:10.4067/s0370-41062018005000203
  41. Justino MCA, da SMD, Souza MF, et al. Atypical hand-foot-mouth disease in Belém, Amazon region, northern Brazil, with detection of coxsackievirus A6. J Clin Virol. 2020;126:104307. doi:10.1016/j.jcv.2020.104307
  42. Cheng FF, Zhang BB, Cao ML, et al. Clinical characteristics of 68 children with atypical hand, foot, and mouth disease caused by coxsackievirus A6: a single-center retrospective analysis. Transl Pediatr. 2022;11:1502-1509. doi:10.21037/tp-22-352
  43. Nagata S. Causes of Kawasaki disease-from past to present. Front Pediatr. 2019;7:18. doi:10.3389/fped.2019.00018
  44. Mitsuishi T, Miyata K, Ando A, et al. Characteristic nail lesions in Kawasaki disease: case series and literature review. J Dermatol. 2022;49:232-238. doi:10.1111/1346-8138.16276
  45. Lindsley CB. Nail-bed lines in Kawasaki disease. Am J Dis Child. 1992;146:659-660. doi:10.1001/archpedi.1992.02160180017005
  46. Matsumura O, Nakagishi Y. Pincer nails upon convalescence from Kawasaki disease. J Pediatr. 2022;246:279. doi:10.1016/j.jpeds.2022.03.002
  47. Solís-Arias MP, García-Romero MT. Onychomycosis in children. a review. Int J Dermatol. 2017;56:123-130. doi:10.1111/ijd.13392
  48. Gupta AK, Mays RR, Versteeg SG, et al. Onychomycosis in children: safety and efficacy of antifungal agents. Pediatr Dermatol. 2018;35:552-559. doi:10.1111/pde.13561
  49. 49. Gupta AK, Venkataraman M, Shear NH, et al. Labeled use of efinaconazole topical solution 10% in treating onychomycosis in children and a review of the management of pediatric onychomycosis. Dermatol Ther. 2020;33:e13613. doi:10.1111/dth.13613
  50. Falotico JM, Lipner SR. Updated perspectives on the diagnosis and management of onychomycosis. Clin Cosmet Investig Dermatol. 2022;15:1933-1957. doi:10.2147/ccid.S362635
  51. Patel D, Castelo-Soccio LA, Rubin AI, et al. Laboratory monitoring during systemic terbinafine therapy for pediatric onychomycosis. JAMA Dermatol. 2017;153:1326-1327. doi:10.1001/jamadermatol.2017.4483
  52. Friedlander SF, Chan YC, Chan YH, et al. Onychomycosis does not always require systemic treatment for cure: a trial using topical therapy. Pediatr Dermatol. 2013;30:316-322. doi:10.1111/pde.12064
  53. Rich P, Spellman M, Purohit V, et al. Tavaborole 5% topical solution for the treatment of toenail onychomycosis in pediatric patients: results from a phase 4 open-label study. J Drugs Dermatol. 2019;18:190-195.
  54. Gupta AK, Venkataraman M, Abramovits W, et al. JUBLIA (efinaconazole 10% solution) in the treatment of pediatric onychomycosis. Skinmed. 2021;19:206-210.
  55. Gupta AK, Paquet M. Systemic antifungals to treat onychomycosis in children: a systematic review. Pediatr Dermatol. 2013;30:294-302. doi:10.1111/pde.12048
  56. Leggit JC. Acute and chronic paronychia. Am Fam Physician. 2017;96:44-51.
  57. Lipner SR, Scher RK. Congenital malalignment of the great toenails with acute paronychia. Pediatr Dermatol. 2016;33:e288-e289.doi:10.1111/pde.12924
Issue
Cutis - 114(1)
Issue
Cutis - 114(1)
Page Number
E9-E15
Page Number
E9-E15
Publications
Publications
Topics
Article Type
Display Headline
Tackling Inflammatory and Infectious Nail Disorders in Children
Display Headline
Tackling Inflammatory and Infectious Nail Disorders in Children
Sections
Inside the Article

 

Practice Points

  • Nail plate pitting is the most common clinical sign of nail psoriasis in children.
  • Nail changes are common in hand, foot, and mouth disease, with the most frequent being onychomadesis.
  • Because onychomycosis may resemble other nail disorders, mycologic confirmation is recommended to avoid misdiagnosis.
  • Many nail conditions in children self-resolve but recognizing these manifestations is important in providing anticipatory guidance to patients and caregivers.
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.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

Gut Biomarkers Accurately Flag Autism Spectrum Disorder

Article Type
Changed
Thu, 07/11/2024 - 10:28

Bacterial and nonbacterial components of the gut microbiome and their function can accurately differentiate children with autism spectrum disorder (ASD) from neurotypical children, new research shows. 

The findings could form the basis for development of a noninvasive diagnostic test for ASD and also provide novel therapeutic targets, wrote investigators, led by Siew C. Ng, MBBS, PhD, with the Microbiota I-Center (MagIC), the Chinese University of Hong Kong.

Their study was published online in Nature Microbiology
 

Beyond Bacteria

The gut microbiome has been shown to play a central role in modulating the gut-brain axis, potentially influencing the development of ASD. 

However, most studies in ASD have focused on the bacterial component of the microbiome. Whether nonbacterial microorganisms (such as gut archaea, fungi, and viruses) or function of the gut microbiome are altered in ASD remains unclear. 

To investigate, the researchers performed metagenomic sequencing on fecal samples from 1627 boys and girls aged 1-13 years with and without ASD from five cohorts in China. 

After controlling for diet, medication, and comorbidity, they identified 14 archaea, 51 bacteria, 7 fungi, 18 viruses, 27 microbial genes, and 12 metabolic pathways that were altered in children with ASD. 

Machine-learning models using single-kingdom panels (archaea, bacteria, fungi, viruses) achieved area under the curve (AUC) values ranging from 0.68 to 0.87 in differentiating children with ASD from neurotypical control children. 

A model based on a panel of 31 multikingdom and functional markers achieved “high predictive value” for ASD, achieving an AUC of 0.91, with comparable performance among boys and girls. 

“The reproducible performance of the models across ages, sexes, and cohorts highlights their potential as promising diagnostic tools for ASD,” the investigators wrote. 

They also noted that the accuracy of the model was largely driven by the biosynthesis pathways of ubiquinol-7 and thiamine diphosphate, which were less abundant in children with ASD, and may serve as therapeutic targets. 
 

‘Exciting’ Possibilities 

“This study broadens our understanding by including fungi, archaea, and viruses, where previous studies have largely focused on the role of gut bacteria in autism,” Bhismadev Chakrabarti, PhD, research director of the Centre for Autism at the University of Reading, United Kingdom, said in a statement from the nonprofit UK Science Media Centre. 

“The results are broadly in line with previous studies that show reduced microbial diversity in autistic individuals. It also examines one of the largest samples seen in a study like this, which further strengthens the results,” Dr. Chakrabarti added. 

He said this research may provide “new ways of detecting autism, if microbial markers turn out to strengthen the ability of genetic and behavioral tests to detect autism. A future platform that can combine genetic, microbial, and simple behavioral assessments could help address the detection gap.

“One limitation of this data is that it cannot assess any causal role for the microbiota in the development of autism,” Dr. Chakrabarti noted. 

This study was supported by InnoHK, the Government of Hong Kong, Special Administrative Region of the People’s Republic of China, The D. H. Chen Foundation, and the New Cornerstone Science Foundation through the New Cornerstone Investigator Program. Dr. Ng has served as an advisory board member for Pfizer, Ferring, Janssen, and AbbVie; has received honoraria as a speaker for Ferring, Tillotts, Menarini, Janssen, AbbVie, and Takeda; is a scientific cofounder and shareholder of GenieBiome; receives patent royalties through her affiliated institutions; and is named as a co-inventor of patent applications that cover the therapeutic and diagnostic use of microbiome. Dr. Chakrabarti has no relevant conflicts of interest.
 

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

Publications
Topics
Sections

Bacterial and nonbacterial components of the gut microbiome and their function can accurately differentiate children with autism spectrum disorder (ASD) from neurotypical children, new research shows. 

The findings could form the basis for development of a noninvasive diagnostic test for ASD and also provide novel therapeutic targets, wrote investigators, led by Siew C. Ng, MBBS, PhD, with the Microbiota I-Center (MagIC), the Chinese University of Hong Kong.

Their study was published online in Nature Microbiology
 

Beyond Bacteria

The gut microbiome has been shown to play a central role in modulating the gut-brain axis, potentially influencing the development of ASD. 

However, most studies in ASD have focused on the bacterial component of the microbiome. Whether nonbacterial microorganisms (such as gut archaea, fungi, and viruses) or function of the gut microbiome are altered in ASD remains unclear. 

To investigate, the researchers performed metagenomic sequencing on fecal samples from 1627 boys and girls aged 1-13 years with and without ASD from five cohorts in China. 

After controlling for diet, medication, and comorbidity, they identified 14 archaea, 51 bacteria, 7 fungi, 18 viruses, 27 microbial genes, and 12 metabolic pathways that were altered in children with ASD. 

Machine-learning models using single-kingdom panels (archaea, bacteria, fungi, viruses) achieved area under the curve (AUC) values ranging from 0.68 to 0.87 in differentiating children with ASD from neurotypical control children. 

A model based on a panel of 31 multikingdom and functional markers achieved “high predictive value” for ASD, achieving an AUC of 0.91, with comparable performance among boys and girls. 

“The reproducible performance of the models across ages, sexes, and cohorts highlights their potential as promising diagnostic tools for ASD,” the investigators wrote. 

They also noted that the accuracy of the model was largely driven by the biosynthesis pathways of ubiquinol-7 and thiamine diphosphate, which were less abundant in children with ASD, and may serve as therapeutic targets. 
 

‘Exciting’ Possibilities 

“This study broadens our understanding by including fungi, archaea, and viruses, where previous studies have largely focused on the role of gut bacteria in autism,” Bhismadev Chakrabarti, PhD, research director of the Centre for Autism at the University of Reading, United Kingdom, said in a statement from the nonprofit UK Science Media Centre. 

“The results are broadly in line with previous studies that show reduced microbial diversity in autistic individuals. It also examines one of the largest samples seen in a study like this, which further strengthens the results,” Dr. Chakrabarti added. 

He said this research may provide “new ways of detecting autism, if microbial markers turn out to strengthen the ability of genetic and behavioral tests to detect autism. A future platform that can combine genetic, microbial, and simple behavioral assessments could help address the detection gap.

“One limitation of this data is that it cannot assess any causal role for the microbiota in the development of autism,” Dr. Chakrabarti noted. 

This study was supported by InnoHK, the Government of Hong Kong, Special Administrative Region of the People’s Republic of China, The D. H. Chen Foundation, and the New Cornerstone Science Foundation through the New Cornerstone Investigator Program. Dr. Ng has served as an advisory board member for Pfizer, Ferring, Janssen, and AbbVie; has received honoraria as a speaker for Ferring, Tillotts, Menarini, Janssen, AbbVie, and Takeda; is a scientific cofounder and shareholder of GenieBiome; receives patent royalties through her affiliated institutions; and is named as a co-inventor of patent applications that cover the therapeutic and diagnostic use of microbiome. Dr. Chakrabarti has no relevant conflicts of interest.
 

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

Bacterial and nonbacterial components of the gut microbiome and their function can accurately differentiate children with autism spectrum disorder (ASD) from neurotypical children, new research shows. 

The findings could form the basis for development of a noninvasive diagnostic test for ASD and also provide novel therapeutic targets, wrote investigators, led by Siew C. Ng, MBBS, PhD, with the Microbiota I-Center (MagIC), the Chinese University of Hong Kong.

Their study was published online in Nature Microbiology
 

Beyond Bacteria

The gut microbiome has been shown to play a central role in modulating the gut-brain axis, potentially influencing the development of ASD. 

However, most studies in ASD have focused on the bacterial component of the microbiome. Whether nonbacterial microorganisms (such as gut archaea, fungi, and viruses) or function of the gut microbiome are altered in ASD remains unclear. 

To investigate, the researchers performed metagenomic sequencing on fecal samples from 1627 boys and girls aged 1-13 years with and without ASD from five cohorts in China. 

After controlling for diet, medication, and comorbidity, they identified 14 archaea, 51 bacteria, 7 fungi, 18 viruses, 27 microbial genes, and 12 metabolic pathways that were altered in children with ASD. 

Machine-learning models using single-kingdom panels (archaea, bacteria, fungi, viruses) achieved area under the curve (AUC) values ranging from 0.68 to 0.87 in differentiating children with ASD from neurotypical control children. 

A model based on a panel of 31 multikingdom and functional markers achieved “high predictive value” for ASD, achieving an AUC of 0.91, with comparable performance among boys and girls. 

“The reproducible performance of the models across ages, sexes, and cohorts highlights their potential as promising diagnostic tools for ASD,” the investigators wrote. 

They also noted that the accuracy of the model was largely driven by the biosynthesis pathways of ubiquinol-7 and thiamine diphosphate, which were less abundant in children with ASD, and may serve as therapeutic targets. 
 

‘Exciting’ Possibilities 

“This study broadens our understanding by including fungi, archaea, and viruses, where previous studies have largely focused on the role of gut bacteria in autism,” Bhismadev Chakrabarti, PhD, research director of the Centre for Autism at the University of Reading, United Kingdom, said in a statement from the nonprofit UK Science Media Centre. 

“The results are broadly in line with previous studies that show reduced microbial diversity in autistic individuals. It also examines one of the largest samples seen in a study like this, which further strengthens the results,” Dr. Chakrabarti added. 

He said this research may provide “new ways of detecting autism, if microbial markers turn out to strengthen the ability of genetic and behavioral tests to detect autism. A future platform that can combine genetic, microbial, and simple behavioral assessments could help address the detection gap.

“One limitation of this data is that it cannot assess any causal role for the microbiota in the development of autism,” Dr. Chakrabarti noted. 

This study was supported by InnoHK, the Government of Hong Kong, Special Administrative Region of the People’s Republic of China, The D. H. Chen Foundation, and the New Cornerstone Science Foundation through the New Cornerstone Investigator Program. Dr. Ng has served as an advisory board member for Pfizer, Ferring, Janssen, and AbbVie; has received honoraria as a speaker for Ferring, Tillotts, Menarini, Janssen, AbbVie, and Takeda; is a scientific cofounder and shareholder of GenieBiome; receives patent royalties through her affiliated institutions; and is named as a co-inventor of patent applications that cover the therapeutic and diagnostic use of microbiome. Dr. Chakrabarti has no relevant conflicts of interest.
 

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

Publications
Publications
Topics
Article Type
Sections
Article Source

FROM NATURE MICROBIOLOGY

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.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Combat Exposure Increases Chronic Pain Among Women in the US Military

Article Type
Changed
Thu, 07/11/2024 - 10:27

 

TOPLINE:

Combat exposure is strongly associated with chronic pain in active-duty servicewomen and female civilian dependents of military personnel on active duty; a lower socioeconomic status and mental health conditions further increased the likelihood of chronic pain.

METHODOLOGY:

  • Researchers analyzed claims data from the Military Health System to identify chronic pain diagnoses among active-duty servicewomen and civilian dependents of individuals on active duty.
  • A total of 3,473,401 individuals (median age, 29 years) were included in the study, with 644,478 active-duty servicewomen and 2,828,923 civilian dependents.
  • The study compared the incidence of chronic pain during 2006-2013, a period of heightened deployment intensity, with 2014-2020, a period of reduced deployment intensity.
  • The primary outcome was the diagnosis of chronic pain.

TAKEAWAY:

  • Active-duty servicewomen in the years 2006-2013 had a 53% increase in the odds of reporting chronic pain compared with those in the period between 2014 and 2020 (odds ratio [OR], 1.53; 95% CI, 1.48-1.58).
  • Civilian dependents in the years 2006-2013 had a 96% increase in the odds of chronic pain compared with those in the later interval (OR, 1.96; 95% CI, 1.93-1.99).
  • In 2006-2013, junior enlisted active-duty servicewomen had nearly a twofold increase in the odds of chronic pain (OR, 1.95; 95% CI, 1.83-2.09), while junior enlisted dependents had more than a threefold increase in the odds of chronic pain (OR, 3.05; 95% CI, 2.87-3.25) compared with senior officers.
  • Comorbid mental conditions also were associated with an increased odds of reporting chronic pain (OR, 1.67; 95% CI, 1.65-1.69).

IN PRACTICE:

“The potential for higher rates of chronic pain in women veterans has been theorized to result from differences in support structures, family conflict, coping strategies, stress regulation, and exposure to military sexual trauma,” the authors wrote. “Our results suggest that these contributing factors may carry over to the women dependents of combat veterans in addition, indicating a line of research that requires urgent further exploration.”

SOURCE:

The study was led by Andrew J. Schoenfeld, MD, MSc, of the Center for Surgery and Public Health, Department of Orthopaedic Surgery at Brigham and Women’s Hospital and Harvard Medical School, in Boston. It was published online on July 5, 2024, in JAMA Network Open.

LIMITATIONS:

This study relied on claims-based data, which may have issues with coding accuracy and limited clinical granularity. The population size reduced over time owing to military downsizing, which could impact the findings. The prevalence of chronic pain in the population was likely underestimated because individuals who did not report symptoms or were diagnosed after separation from service were not identified.

DISCLOSURES:

This study was funded by the US Department of Defense. The lead author reported receiving grants and personal fees, serving as the editor-in-chief for Spine, acting as a consultant, and having other ties with various sources outside the submitted work.
 

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

Publications
Topics
Sections

 

TOPLINE:

Combat exposure is strongly associated with chronic pain in active-duty servicewomen and female civilian dependents of military personnel on active duty; a lower socioeconomic status and mental health conditions further increased the likelihood of chronic pain.

METHODOLOGY:

  • Researchers analyzed claims data from the Military Health System to identify chronic pain diagnoses among active-duty servicewomen and civilian dependents of individuals on active duty.
  • A total of 3,473,401 individuals (median age, 29 years) were included in the study, with 644,478 active-duty servicewomen and 2,828,923 civilian dependents.
  • The study compared the incidence of chronic pain during 2006-2013, a period of heightened deployment intensity, with 2014-2020, a period of reduced deployment intensity.
  • The primary outcome was the diagnosis of chronic pain.

TAKEAWAY:

  • Active-duty servicewomen in the years 2006-2013 had a 53% increase in the odds of reporting chronic pain compared with those in the period between 2014 and 2020 (odds ratio [OR], 1.53; 95% CI, 1.48-1.58).
  • Civilian dependents in the years 2006-2013 had a 96% increase in the odds of chronic pain compared with those in the later interval (OR, 1.96; 95% CI, 1.93-1.99).
  • In 2006-2013, junior enlisted active-duty servicewomen had nearly a twofold increase in the odds of chronic pain (OR, 1.95; 95% CI, 1.83-2.09), while junior enlisted dependents had more than a threefold increase in the odds of chronic pain (OR, 3.05; 95% CI, 2.87-3.25) compared with senior officers.
  • Comorbid mental conditions also were associated with an increased odds of reporting chronic pain (OR, 1.67; 95% CI, 1.65-1.69).

IN PRACTICE:

“The potential for higher rates of chronic pain in women veterans has been theorized to result from differences in support structures, family conflict, coping strategies, stress regulation, and exposure to military sexual trauma,” the authors wrote. “Our results suggest that these contributing factors may carry over to the women dependents of combat veterans in addition, indicating a line of research that requires urgent further exploration.”

SOURCE:

The study was led by Andrew J. Schoenfeld, MD, MSc, of the Center for Surgery and Public Health, Department of Orthopaedic Surgery at Brigham and Women’s Hospital and Harvard Medical School, in Boston. It was published online on July 5, 2024, in JAMA Network Open.

LIMITATIONS:

This study relied on claims-based data, which may have issues with coding accuracy and limited clinical granularity. The population size reduced over time owing to military downsizing, which could impact the findings. The prevalence of chronic pain in the population was likely underestimated because individuals who did not report symptoms or were diagnosed after separation from service were not identified.

DISCLOSURES:

This study was funded by the US Department of Defense. The lead author reported receiving grants and personal fees, serving as the editor-in-chief for Spine, acting as a consultant, and having other ties with various sources outside the submitted work.
 

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

 

TOPLINE:

Combat exposure is strongly associated with chronic pain in active-duty servicewomen and female civilian dependents of military personnel on active duty; a lower socioeconomic status and mental health conditions further increased the likelihood of chronic pain.

METHODOLOGY:

  • Researchers analyzed claims data from the Military Health System to identify chronic pain diagnoses among active-duty servicewomen and civilian dependents of individuals on active duty.
  • A total of 3,473,401 individuals (median age, 29 years) were included in the study, with 644,478 active-duty servicewomen and 2,828,923 civilian dependents.
  • The study compared the incidence of chronic pain during 2006-2013, a period of heightened deployment intensity, with 2014-2020, a period of reduced deployment intensity.
  • The primary outcome was the diagnosis of chronic pain.

TAKEAWAY:

  • Active-duty servicewomen in the years 2006-2013 had a 53% increase in the odds of reporting chronic pain compared with those in the period between 2014 and 2020 (odds ratio [OR], 1.53; 95% CI, 1.48-1.58).
  • Civilian dependents in the years 2006-2013 had a 96% increase in the odds of chronic pain compared with those in the later interval (OR, 1.96; 95% CI, 1.93-1.99).
  • In 2006-2013, junior enlisted active-duty servicewomen had nearly a twofold increase in the odds of chronic pain (OR, 1.95; 95% CI, 1.83-2.09), while junior enlisted dependents had more than a threefold increase in the odds of chronic pain (OR, 3.05; 95% CI, 2.87-3.25) compared with senior officers.
  • Comorbid mental conditions also were associated with an increased odds of reporting chronic pain (OR, 1.67; 95% CI, 1.65-1.69).

IN PRACTICE:

“The potential for higher rates of chronic pain in women veterans has been theorized to result from differences in support structures, family conflict, coping strategies, stress regulation, and exposure to military sexual trauma,” the authors wrote. “Our results suggest that these contributing factors may carry over to the women dependents of combat veterans in addition, indicating a line of research that requires urgent further exploration.”

SOURCE:

The study was led by Andrew J. Schoenfeld, MD, MSc, of the Center for Surgery and Public Health, Department of Orthopaedic Surgery at Brigham and Women’s Hospital and Harvard Medical School, in Boston. It was published online on July 5, 2024, in JAMA Network Open.

LIMITATIONS:

This study relied on claims-based data, which may have issues with coding accuracy and limited clinical granularity. The population size reduced over time owing to military downsizing, which could impact the findings. The prevalence of chronic pain in the population was likely underestimated because individuals who did not report symptoms or were diagnosed after separation from service were not identified.

DISCLOSURES:

This study was funded by the US Department of Defense. The lead author reported receiving grants and personal fees, serving as the editor-in-chief for Spine, acting as a consultant, and having other ties with various sources outside the submitted work.
 

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

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Women’s Risk for Lupus Rises With Greater Intake of Ultraprocessed Foods

Article Type
Changed
Thu, 07/11/2024 - 10:28

 

TOPLINE:

A higher intake of ultraprocessed foods increases the risk for systemic lupus erythematosus (SLE) by over 50% in women. The risk doubled in those with anti–double-stranded DNA antibodies.

METHODOLOGY:

  • Researchers assessed 204,175 women from two Nurses’ Health Study cohorts from 1984 to 2016.
  • Participants completed semiquantitative food frequency questionnaires every 4 years for the assessment of dietary intake.
  • Incident SLE cases were self-reported and confirmed using medical records, with 212 cases identified.

TAKEAWAY:

  • A higher cumulative average daily intake of ultraprocessed foods was associated with a 56% increased risk for SLE (95% confidence interval [CI], 1.04-2.32).
  • The risk for anti–double-stranded DNA antibody-positive SLE was more than doubled (hazard ratio, 2.05; 95% CI, 1.15-3.65).
  • Sugar or artificially sweetened beverages were associated with a 45% increased risk for SLE (95% CI, 1.01-2.09).
  • No significant interactions with body mass index were observed in the association between ultraprocessed food intake and SLE.

IN PRACTICE:

This study is too preliminary to have practical application.

SOURCE:

The study was led by Sinara Rossato, PhD, Harvard T.H. Chan School of Public Health, Boston. It was published online in Arthritis Care & Research.

LIMITATIONS:

The study’s generalizability is limited due to the predominantly White female population of registered nurses. The relatively high baseline age of participants may not fully capture the peak incidence age range for SLE. The observational nature of the study cannot establish causality between ultraprocessed food intake and SLE risk.

DISCLOSURES:

The study was supported by the National Institutes of Health. The authors did not declare any competing interests.

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

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

Publications
Topics
Sections

 

TOPLINE:

A higher intake of ultraprocessed foods increases the risk for systemic lupus erythematosus (SLE) by over 50% in women. The risk doubled in those with anti–double-stranded DNA antibodies.

METHODOLOGY:

  • Researchers assessed 204,175 women from two Nurses’ Health Study cohorts from 1984 to 2016.
  • Participants completed semiquantitative food frequency questionnaires every 4 years for the assessment of dietary intake.
  • Incident SLE cases were self-reported and confirmed using medical records, with 212 cases identified.

TAKEAWAY:

  • A higher cumulative average daily intake of ultraprocessed foods was associated with a 56% increased risk for SLE (95% confidence interval [CI], 1.04-2.32).
  • The risk for anti–double-stranded DNA antibody-positive SLE was more than doubled (hazard ratio, 2.05; 95% CI, 1.15-3.65).
  • Sugar or artificially sweetened beverages were associated with a 45% increased risk for SLE (95% CI, 1.01-2.09).
  • No significant interactions with body mass index were observed in the association between ultraprocessed food intake and SLE.

IN PRACTICE:

This study is too preliminary to have practical application.

SOURCE:

The study was led by Sinara Rossato, PhD, Harvard T.H. Chan School of Public Health, Boston. It was published online in Arthritis Care & Research.

LIMITATIONS:

The study’s generalizability is limited due to the predominantly White female population of registered nurses. The relatively high baseline age of participants may not fully capture the peak incidence age range for SLE. The observational nature of the study cannot establish causality between ultraprocessed food intake and SLE risk.

DISCLOSURES:

The study was supported by the National Institutes of Health. The authors did not declare any competing interests.

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

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

 

TOPLINE:

A higher intake of ultraprocessed foods increases the risk for systemic lupus erythematosus (SLE) by over 50% in women. The risk doubled in those with anti–double-stranded DNA antibodies.

METHODOLOGY:

  • Researchers assessed 204,175 women from two Nurses’ Health Study cohorts from 1984 to 2016.
  • Participants completed semiquantitative food frequency questionnaires every 4 years for the assessment of dietary intake.
  • Incident SLE cases were self-reported and confirmed using medical records, with 212 cases identified.

TAKEAWAY:

  • A higher cumulative average daily intake of ultraprocessed foods was associated with a 56% increased risk for SLE (95% confidence interval [CI], 1.04-2.32).
  • The risk for anti–double-stranded DNA antibody-positive SLE was more than doubled (hazard ratio, 2.05; 95% CI, 1.15-3.65).
  • Sugar or artificially sweetened beverages were associated with a 45% increased risk for SLE (95% CI, 1.01-2.09).
  • No significant interactions with body mass index were observed in the association between ultraprocessed food intake and SLE.

IN PRACTICE:

This study is too preliminary to have practical application.

SOURCE:

The study was led by Sinara Rossato, PhD, Harvard T.H. Chan School of Public Health, Boston. It was published online in Arthritis Care & Research.

LIMITATIONS:

The study’s generalizability is limited due to the predominantly White female population of registered nurses. The relatively high baseline age of participants may not fully capture the peak incidence age range for SLE. The observational nature of the study cannot establish causality between ultraprocessed food intake and SLE risk.

DISCLOSURES:

The study was supported by the National Institutes of Health. The authors did not declare any competing interests.

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

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

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Plastic Surgeon to Pay $5 Million for Restriction of Negative Reviews, Directing Fake Reviews

Article Type
Changed
Thu, 07/11/2024 - 10:27

A Seattle plastic surgeon who illegally restricted patients from posting negative reviews about his practice and directed his staff to post fake positive reviews will pay $5 million for violating Washington state’s consumer protection law.

According to a July 1 consent decree, Javad Sajan, MD, and his practice Allure Esthetic must pay $1.5 million in restitution to 21,000 patients and $3.5 million to the state for manipulation of patient ratings.

The settlement resolves a federal lawsuit brought by Washington State Attorney General Bob Ferguson that accused the doctor of illegally suppressing patients’ negative reviews by “forcing” them to sign nondisclosure agreements (NDAs) before they received care. In an April decision, US District Judge Ricardo S. Martinez sided with the state, ruling that Allure Esthetic’s actions violated the federal Consumer Review Fairness Act (CRFA).

“Writing a truthful review about a business should not subject you to threats or intimidation,” Mr. Ferguson said in a July 2 statement. “Consumers rely on reviews when determining who to trust, especially services that affect their health and safety. This resolution holds Allure accountable for brazenly violating that trust — and the law — and ensures the clinic stops its harmful conduct.”

In court documents, Dr. Sajan’s attorneys had argued that the agreements did not violate CRFA because patients had the opportunity to modify the language or decline signing them.

The surgeon’s practice is “pleased to have resolved its case with the Attorney General’s Office,” according to a statement provided by Dr. Sajan’s attorney. “The cooperative settlement, while not admitting fault and resolving claims asserted by both sides, allows Allure Esthetic to continue to focus on its core mission of providing compassionate care to patients and serving the community. The decision to settle was not an easy one, but it was necessary to allocate time and resources where they matter most — the patients.” 

The dispute stemmed from NDAs that Dr. Sajan’s practice required patients to sign starting in 2017, according to Mr. Ferguson’s complaint. The terms instructed patients to contact the business directly if they had concerns rather than post a negative review.

If patients posted negative reviews, the clinic, in some cases, threatened litigation, according to the lawsuit. In other cases, patients were allegedly offered money and free services in exchange for taking the reviews down. Patients who accepted cash or services were required to sign a second agreement forbidding them from posting future negative reviews and imposing a $250,000 penalty for failure to comply, according to court documents.

In addition, Mr. Ferguson accused Dr. Sajan of creating fake positive accounts of patient experiences and buying fake followers on social media. State investigators found Dr. Sajan directed Allure Esthetic’s employees to create fake Gmail accounts to post the false reviews, many of which are still online today, according to the state’s complaint.

Mr. Ferguson also claimed Dr. Sajan and his practice manipulated social media to appear more popular by purchasing followers through an online vendor. The practice also allegedly used a social media bot tool to buy thousands of fake likes on Instagram, YouTube, and other social media.

After filing the lawsuit, Mr. Ferguson’s office uncovered further evidence of Dr. Sajan’s efforts to influence his professional reputation through fabrication, according to the July 2 release. Allure Esthetic “rigged” “best doctor” competitions hosted by local media outlets by paying staff and contractors to vote for Dr. Sajan as “best plastic surgeon” in the region, according to the release. The staff cast as many votes as websites allowed, despite not being patients of Allure Esthetic.

The practice also allegedly edited before-and-after photos of patients to make their results appear better and kept tens of thousands of dollars in rebates intended for its patients.

In addition to paying $5 million, the consent decree requires Dr. Sajan and his practice also:

  • Stop posting or influencing consumer reviews; perform a full audit of all public reviews on Google, Yelp, and other third-party review platforms; and request removal of every review Allure Esthetic was involved in creating, posting, or shaping in any manner.
  • Remove all misleading “before-and-after” photographs of plastic surgery procedures from its website and social media and stop altering photographs of future procedures.
  • Cease use of and attempts to enforce all illegal NDAs and notify patients who previously signed them that they are released from the terms of those NDAs.
  • Pay a third-party forensic accounting firm to perform a full, independent audit of Allure Esthetic’s consumer rebate program to identify consumers who are owed rebates that were unlawfully claimed by Allure Esthetic.

Additionally, the attorney general’s office will continue to monitor Allure Esthetic, and upon request, the practice must provide information that demonstrates its compliance with the consent decree for the next 10 years.

The practice must also develop internal policies and implement a training program to educate staff about nondeceptive advertising and compliance with consumer protection laws.

Dr. Sajan and his practice agreed to the terms of the consent decree, and the settlement is not considered an admission of liability.

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

Publications
Topics
Sections

A Seattle plastic surgeon who illegally restricted patients from posting negative reviews about his practice and directed his staff to post fake positive reviews will pay $5 million for violating Washington state’s consumer protection law.

According to a July 1 consent decree, Javad Sajan, MD, and his practice Allure Esthetic must pay $1.5 million in restitution to 21,000 patients and $3.5 million to the state for manipulation of patient ratings.

The settlement resolves a federal lawsuit brought by Washington State Attorney General Bob Ferguson that accused the doctor of illegally suppressing patients’ negative reviews by “forcing” them to sign nondisclosure agreements (NDAs) before they received care. In an April decision, US District Judge Ricardo S. Martinez sided with the state, ruling that Allure Esthetic’s actions violated the federal Consumer Review Fairness Act (CRFA).

“Writing a truthful review about a business should not subject you to threats or intimidation,” Mr. Ferguson said in a July 2 statement. “Consumers rely on reviews when determining who to trust, especially services that affect their health and safety. This resolution holds Allure accountable for brazenly violating that trust — and the law — and ensures the clinic stops its harmful conduct.”

In court documents, Dr. Sajan’s attorneys had argued that the agreements did not violate CRFA because patients had the opportunity to modify the language or decline signing them.

The surgeon’s practice is “pleased to have resolved its case with the Attorney General’s Office,” according to a statement provided by Dr. Sajan’s attorney. “The cooperative settlement, while not admitting fault and resolving claims asserted by both sides, allows Allure Esthetic to continue to focus on its core mission of providing compassionate care to patients and serving the community. The decision to settle was not an easy one, but it was necessary to allocate time and resources where they matter most — the patients.” 

The dispute stemmed from NDAs that Dr. Sajan’s practice required patients to sign starting in 2017, according to Mr. Ferguson’s complaint. The terms instructed patients to contact the business directly if they had concerns rather than post a negative review.

If patients posted negative reviews, the clinic, in some cases, threatened litigation, according to the lawsuit. In other cases, patients were allegedly offered money and free services in exchange for taking the reviews down. Patients who accepted cash or services were required to sign a second agreement forbidding them from posting future negative reviews and imposing a $250,000 penalty for failure to comply, according to court documents.

In addition, Mr. Ferguson accused Dr. Sajan of creating fake positive accounts of patient experiences and buying fake followers on social media. State investigators found Dr. Sajan directed Allure Esthetic’s employees to create fake Gmail accounts to post the false reviews, many of which are still online today, according to the state’s complaint.

Mr. Ferguson also claimed Dr. Sajan and his practice manipulated social media to appear more popular by purchasing followers through an online vendor. The practice also allegedly used a social media bot tool to buy thousands of fake likes on Instagram, YouTube, and other social media.

After filing the lawsuit, Mr. Ferguson’s office uncovered further evidence of Dr. Sajan’s efforts to influence his professional reputation through fabrication, according to the July 2 release. Allure Esthetic “rigged” “best doctor” competitions hosted by local media outlets by paying staff and contractors to vote for Dr. Sajan as “best plastic surgeon” in the region, according to the release. The staff cast as many votes as websites allowed, despite not being patients of Allure Esthetic.

The practice also allegedly edited before-and-after photos of patients to make their results appear better and kept tens of thousands of dollars in rebates intended for its patients.

In addition to paying $5 million, the consent decree requires Dr. Sajan and his practice also:

  • Stop posting or influencing consumer reviews; perform a full audit of all public reviews on Google, Yelp, and other third-party review platforms; and request removal of every review Allure Esthetic was involved in creating, posting, or shaping in any manner.
  • Remove all misleading “before-and-after” photographs of plastic surgery procedures from its website and social media and stop altering photographs of future procedures.
  • Cease use of and attempts to enforce all illegal NDAs and notify patients who previously signed them that they are released from the terms of those NDAs.
  • Pay a third-party forensic accounting firm to perform a full, independent audit of Allure Esthetic’s consumer rebate program to identify consumers who are owed rebates that were unlawfully claimed by Allure Esthetic.

Additionally, the attorney general’s office will continue to monitor Allure Esthetic, and upon request, the practice must provide information that demonstrates its compliance with the consent decree for the next 10 years.

The practice must also develop internal policies and implement a training program to educate staff about nondeceptive advertising and compliance with consumer protection laws.

Dr. Sajan and his practice agreed to the terms of the consent decree, and the settlement is not considered an admission of liability.

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

A Seattle plastic surgeon who illegally restricted patients from posting negative reviews about his practice and directed his staff to post fake positive reviews will pay $5 million for violating Washington state’s consumer protection law.

According to a July 1 consent decree, Javad Sajan, MD, and his practice Allure Esthetic must pay $1.5 million in restitution to 21,000 patients and $3.5 million to the state for manipulation of patient ratings.

The settlement resolves a federal lawsuit brought by Washington State Attorney General Bob Ferguson that accused the doctor of illegally suppressing patients’ negative reviews by “forcing” them to sign nondisclosure agreements (NDAs) before they received care. In an April decision, US District Judge Ricardo S. Martinez sided with the state, ruling that Allure Esthetic’s actions violated the federal Consumer Review Fairness Act (CRFA).

“Writing a truthful review about a business should not subject you to threats or intimidation,” Mr. Ferguson said in a July 2 statement. “Consumers rely on reviews when determining who to trust, especially services that affect their health and safety. This resolution holds Allure accountable for brazenly violating that trust — and the law — and ensures the clinic stops its harmful conduct.”

In court documents, Dr. Sajan’s attorneys had argued that the agreements did not violate CRFA because patients had the opportunity to modify the language or decline signing them.

The surgeon’s practice is “pleased to have resolved its case with the Attorney General’s Office,” according to a statement provided by Dr. Sajan’s attorney. “The cooperative settlement, while not admitting fault and resolving claims asserted by both sides, allows Allure Esthetic to continue to focus on its core mission of providing compassionate care to patients and serving the community. The decision to settle was not an easy one, but it was necessary to allocate time and resources where they matter most — the patients.” 

The dispute stemmed from NDAs that Dr. Sajan’s practice required patients to sign starting in 2017, according to Mr. Ferguson’s complaint. The terms instructed patients to contact the business directly if they had concerns rather than post a negative review.

If patients posted negative reviews, the clinic, in some cases, threatened litigation, according to the lawsuit. In other cases, patients were allegedly offered money and free services in exchange for taking the reviews down. Patients who accepted cash or services were required to sign a second agreement forbidding them from posting future negative reviews and imposing a $250,000 penalty for failure to comply, according to court documents.

In addition, Mr. Ferguson accused Dr. Sajan of creating fake positive accounts of patient experiences and buying fake followers on social media. State investigators found Dr. Sajan directed Allure Esthetic’s employees to create fake Gmail accounts to post the false reviews, many of which are still online today, according to the state’s complaint.

Mr. Ferguson also claimed Dr. Sajan and his practice manipulated social media to appear more popular by purchasing followers through an online vendor. The practice also allegedly used a social media bot tool to buy thousands of fake likes on Instagram, YouTube, and other social media.

After filing the lawsuit, Mr. Ferguson’s office uncovered further evidence of Dr. Sajan’s efforts to influence his professional reputation through fabrication, according to the July 2 release. Allure Esthetic “rigged” “best doctor” competitions hosted by local media outlets by paying staff and contractors to vote for Dr. Sajan as “best plastic surgeon” in the region, according to the release. The staff cast as many votes as websites allowed, despite not being patients of Allure Esthetic.

The practice also allegedly edited before-and-after photos of patients to make their results appear better and kept tens of thousands of dollars in rebates intended for its patients.

In addition to paying $5 million, the consent decree requires Dr. Sajan and his practice also:

  • Stop posting or influencing consumer reviews; perform a full audit of all public reviews on Google, Yelp, and other third-party review platforms; and request removal of every review Allure Esthetic was involved in creating, posting, or shaping in any manner.
  • Remove all misleading “before-and-after” photographs of plastic surgery procedures from its website and social media and stop altering photographs of future procedures.
  • Cease use of and attempts to enforce all illegal NDAs and notify patients who previously signed them that they are released from the terms of those NDAs.
  • Pay a third-party forensic accounting firm to perform a full, independent audit of Allure Esthetic’s consumer rebate program to identify consumers who are owed rebates that were unlawfully claimed by Allure Esthetic.

Additionally, the attorney general’s office will continue to monitor Allure Esthetic, and upon request, the practice must provide information that demonstrates its compliance with the consent decree for the next 10 years.

The practice must also develop internal policies and implement a training program to educate staff about nondeceptive advertising and compliance with consumer protection laws.

Dr. Sajan and his practice agreed to the terms of the consent decree, and the settlement is not considered an admission of liability.

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

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article