Purpuric Lesions on the Leg

Article Type
Changed
Tue, 10/01/2024 - 11:30
Display Headline
Purpuric Lesions on the Leg

THE DIAGNOSIS: Dengue Hemorrhagic Fever

The retiform purpura observed in our patient was suggestive of a vasculitic, thrombotic, or embolic etiology. Dengue IgM serologic testing performed based on her extensive travel history and recent return from a dengue-endemic area was positive, indicating acute infection. A clinical diagnosis of dengue hemorrhagic fever (DHF) was made based on the hemorrhagic appearance of the lesion. Histopathology revealed leukocytoclastic vasculitis (Figure). Anti–double-stranded DNA, antideoxyribonuclease, C3 and C4, CH50 (total hemolytic complement), antineutrophil cytoplasmic antibodies, HIV, and hepatitis B virus tests were normal. Direct immunofluorescence was negative.

A, Histopathology of a biopsy from the right medial leg showed early leukocytoclastic vasculitis with karyorrhexis and red cell extravasation (H&E, original magnification ×200). B, Extensive erythrocyte extravasation and expended vessel walls with fibrin deposition also were seen (H&E, original magnification ×100).

Dengue virus is a single-stranded RNA virus transmitted by Aedes aegypti and Aedes albopictus mosquitoes and is one of the most prevalent arthropod-borne viruses affecting humans today.1,2 Infection with the dengue virus generally is seen in travelers visiting tropical regions of Africa, Mexico, South America, South and Central Asia, Southeast Asia, and the Caribbean.1 The Table shows the global distribution of dengue serotypes from 2000 to 2014.3,4 There are 4 serotypes of the dengue virus: DENV-1 to DENV-4. Infection with 1 strain elicits longlasting immunity to that strain, but subsequent infection with another strain can result in severe DHF due to antibody cross-reaction.1

Dengue virus infection ranges from mildly symptomatic to a spectrum of increasingly severe conditions that comprise dengue fever (DF) and DHF, as well as dengue shock syndrome and brain stem hemorrhage, which may be fatal.2,5 Dengue fever manifests as severe myalgia, fever, headache (usually retro-orbital), arthralgia, erythema, and rubelliform exanthema.6 The frequency of skin eruptions in patients with DF varies with the virus strain and outbreaks.7 The lesions initially develop with the onset of fever and manifest as flushing or erythematous mottling of the face, neck, and chest areas.1,7 The morbilliform eruption develops 2 to 6 days after the onset of the fever, beginning on the trunk and spreading to the face and extremities.1,7 The rash may become confluent with characteristic sparing of small round areas of normal skin described as white islands in a sea of red.2 Verrucous papules on the ears also have been described and may resemble those seen in Cowden syndrome. In patients with prior infection with a different strain of the virus, hemorrhagic lesions may develop, including characteristic retiform purpura, a positive tourniquet test, and the appearance of petechiae on the lower legs. Pruritus and desquamation, especially on the palms and soles, may follow the termination of the eruption.7

The differential diagnosis of DF includes measles, rubella, enteroviruses, and influenza. Chikungunya and West Nile viruses in Asia and Africa and the O’nyong-nyong virus in Africa are also arboviruses that cause a clinical picture similar to DF but not DHF. Other diagnostic considerations include phases of scarlet fever, typhoid, malaria, leptospirosis, hepatitis A, and trypanosomal and rickettsial diseases.7 The differential diagnosis of DHF includes antineutrophil cytoplasmic antibody–associated vasculitis, rheumatoid vasculitis, and bacterial septic vasculitis.

Acute clinical diagnosis of DF can be challenging because of the nonspecific symptoms that can be seen in almost every infectious disease. Clinical presentation assessment should be confirmed with laboratory testing.6 Dengue virus infection usually is confirmed by the identification of viral genomic RNA, antigens, or the antibodies it elicits. Enzyme-linked immunosorbent assay–based serologic tests are cost-effective and easy to perform.5 IgM antibodies usually show cross-reactivity with platelets, but the antibody levels are not positively correlated with the severity of DF.8 Primary infection with the dengue virus is characterized by the elevation of specific IgM levels that usually occurs 3 to 5 days after symptom onset and persists during the postfebrile stage (up to 30 to 60 days). In secondary infections, the IgM levels usually rise more slowly and reach a lower level than in primary infections.9 For both primary and secondary infections, testing IgM levels after the febrile stage may be helpful with the laboratory diagnosis.

Currently, there is no antiviral drug available for dengue. Treatment of dengue infection is symptomatic and supportive.2

Dengue hemorrhagic fever is indicated by a rising hematocrit (≥20%) and a falling platelet count (>100,000/mm3) accompanying clinical signs of hemorrhage. Treatment includes intravenous fluid replacement and careful clinical monitoring of hematocrit levels, platelet count, vitals, urine output, and other signs of shock.5 For patients with a history of dengue infection, travel to areas with other serotypes is not recommended.

If any travel to a high-risk area is planned, countryspecific travel recommendations and warnings should be reviewed from the Centers for Disease Control and Prevention’s website (https://wwwnc.cdc.gov/travel/notices/level1/dengue-global). Use of an Environmental Protection Agency–registered insect repellent to avoid mosquito bites and acetaminophen for managing symptoms is advised. During travel, staying in places with window and door screens and using a bed net during sleep are suggested. Long-sleeved shirts and long pants also are preferred. Travelers should see a health care provider if they have symptoms of dengue.10

African tick bite fever (ATBF) is caused by Rickettsia africae transmitted by Amblyomma ticks. Skin findings in ATBF include erythematous, firm, tender papules with central eschars consistent with the feeding patterns of ticks.11 Histopathology of ATBF usually includes fibrinoid necrosis of vessels in the dermis with a perivascular inflammatory infiltrate and coagulation necrosis of the surrounding dermis consistent with eschar formation.12 The lack of an eschar weighs against this diagnosis.

African trypanosomiasis (also known as sleeping sickness) is caused by protozoa transmitted by the tsetse fly. A chancrelike, circumscribed, rubbery, indurated red or violaceous nodule measuring 2 to 5 cm in diameter often develops as the earliest cutaneous sign of the disease.13 Nonspecific histopathologic findings, such as infiltration of lymphocytes and macrophages and proliferation of endothelial cells and fibroblasts, may be observed.14 Extravascular parasites have been noted in skin biopsies.15 In later stages, skin lesions called trypanids may be observed as macular, papular, annular, targetoid, purpuric, and erythematous lesions, and histopathologic findings consistent with vasculitis also may be seen.13

Chikungunya virus infection is an acute-onset, mosquito-borne viral disease. Skin manifestations may start with nonspecific, generalized, morbilliform, maculopapular rashes coinciding with fever, which also may be seen initially with DHF. Skin hyperpigmentation, mostly centrofacial and involving the nose (chik sign); purpuric and ecchymotic lesions over the trunk and flexors of limbs in adults, often surmounted by subepidermal bullae and lesions resembling toxic epidermal necrolysis; and nonhealing ulcers in the genital and groin areas are common skin manifestations of chikungunya infection.16 Intraepithelial splitting with acantholysis and perivascular lymphohistiocytic infiltration may be observed in the histopathology of blistering lesions, which are not consistent with DHF.17

Zika virus infection is caused by an arbovirus within the Flaviviridae family, which also includes the dengue virus. Initial mucocutaneous findings of the Zika virus include nonspecific diffuse maculopapular eruptions. The eruption generally spares the palms and soles; however, various manifestations including involvement of the palms and soles have been reported.18 The morbilliform eruption begins on the face and extends to the trunk and extremities. Mild hemorrhagic manifestations, including petechiae and bleeding gums, may be observed. Distinguishing between dengue and Zika virus infection relies on the severity of symptoms and laboratory tests, including polymerase chain reaction or IgM antibody testing.19 The other conditions listed do not produce hemorrhagic fever.

References
  1. Pincus LB, Grossman ME, Fox LP. The exanthem of dengue fever: clinical features of two US tourists traveling abroad. J Am Acad Dermatol. 2008;58:308-316. doi:10.1016/j.jaad.2007.08.042
  2. Radakovic-Fijan S, Graninger W, Müller C, et al. Dengue hemorrhagic fever in a British travel guide. J Am Acad Dermatol. 2002;46:430-433. doi:10.1067/mjd.2002.111904
  3. Yamashita A, Sakamoto T, Sekizuka T, et al. DGV: dengue genographic viewer. Front Microbiol. 2016;7:875. doi:10.3389/fmicb.2016.00875
  4. Centers for Disease and Prevention. Dengue in the US states and territories. Updated October 7, 2020. Accessed September 30, 2024. https://www.cdc.gov/dengue/data-research/facts-stats/?CDC_AAref_Val=https://www.cdc.gov/dengue/areaswithrisk/in-the-us.html
  5. Khetarpal N, Khanna I. Dengue fever: causes, complications, and vaccine strategies. J Immunol Res. 2016;2016:6803098. doi:10.1155/2016/6803098
  6. Muller DA, Depelsenaire AC, Young PR. Clinical and laboratory diagnosis of dengue virus infection. J Infect Dis. 2017;215(suppl 2):S89-S95. doi:10.1093/infdis/jiw649
  7. Waterman SH, Gubler DJ. Dengue fever. Clin Dermatol. 1989;7:117-122. doi:10.1016/0738-081x(89)90034-5
  8. Lin CF, Lei HY, Liu CC, et al. Generation of IgM anti-platelet autoantibody in dengue patients. J Med Virol. 2001;63:143-149. doi:10.1002/1096- 9071(20000201)63:2<143::AID-JMV1009>3.0.CO;2-L
  9. Tripathi NK, Shrivastava A, Dash PK, et al. Detection of dengue virus. Methods Mol Biol. 2011;665:51-64. doi:10.1007/978-1-60761-817-1_4
  10. Centers for Disease Control and Prevention. Plan for travel. Accessed September 30, 2024. https://wwwnc.cdc.gov/travel
  11. Mack I, Ritz N. African tick-bite fever. N Engl J Med. 2019;380:960. doi:10.1056/NEJMicm1810093
  12. Lepidi H, Fournier PE, Raoult D. Histologic features and immunodetection of African tick-bite fever eschar. Emerg Infect Dis. 2006;12:1332- 1337. doi:10.3201/eid1209.051540
  13. McGovern TW, Williams W, Fitzpatrick JE, et al. Cutaneous manifestations of African trypanosomiasis. Arch Dermatol. 1995;131:1178-1182.
  14. Kristensson K, Bentivoglio M. Pathology of African trypanosomiasis. In: Dumas M, Bouteille B, Buguet A, eds. Progress in Human African Trypanosomiasis, Sleeping Sickness. Springer; 1999:157-181.
  15. Capewell P, Cren-Travaillé C, Marchesi F, et al. The skin is a significant but overlooked anatomical reservoir for vector-borne African trypanosomes. Elife. 2016;5:e17716. doi:10.7554/eLife.17716
  16. Singal A. Chikungunya and skin: current perspective. Indian Dermatol Online J. 2017;8:307-309. doi:10.4103/idoj.IDOJ_93_17
  17. Robin S, Ramful D, Zettor J, et al. Severe bullous skin lesions associated with chikungunya virus infection in small infants. Eur J Pediatr. 2009;169:67-72. doi:10.1007/s00431-009-0986-0
  18. Hussain A, Ali F, Latiwesh OB, et al. A comprehensive review of the manifestations and pathogenesis of Zika virus in neonates and adults. Cureus. 2018;10:E3290. doi:10.7759/cureus.3290
  19. Farahnik B, Beroukhim K, Blattner CM, et al. Cutaneous manifestations of the Zika virus. J Am Acad Dermatol. 2016;74:1286-1287. doi:10.1016/j.jaad.2016.02.1232
Article PDF
Author and Disclosure Information

Drs. Sagut and Elston are from the Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston. Dr. Gaster is from Avera Medical Group Dermatology Sioux Falls, South Dakota, and Physicians Laboratory, Sioux Falls.

The authors have no relevant financial disclosures to report.

The images are in the public domain.

Correspondence: Pelin Sagut, MD, 135 Rutledge Ave, MSC 578, Charleston, SC 29425 (pelin.sagut@gmail.com).

Cutis. 2024 September;114(3):E27-E30. doi:10.12788/cutis.1114

Issue
Cutis - 114(3)
Publications
Topics
Page Number
E27-E30
Sections
Author and Disclosure Information

Drs. Sagut and Elston are from the Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston. Dr. Gaster is from Avera Medical Group Dermatology Sioux Falls, South Dakota, and Physicians Laboratory, Sioux Falls.

The authors have no relevant financial disclosures to report.

The images are in the public domain.

Correspondence: Pelin Sagut, MD, 135 Rutledge Ave, MSC 578, Charleston, SC 29425 (pelin.sagut@gmail.com).

Cutis. 2024 September;114(3):E27-E30. doi:10.12788/cutis.1114

Author and Disclosure Information

Drs. Sagut and Elston are from the Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston. Dr. Gaster is from Avera Medical Group Dermatology Sioux Falls, South Dakota, and Physicians Laboratory, Sioux Falls.

The authors have no relevant financial disclosures to report.

The images are in the public domain.

Correspondence: Pelin Sagut, MD, 135 Rutledge Ave, MSC 578, Charleston, SC 29425 (pelin.sagut@gmail.com).

Cutis. 2024 September;114(3):E27-E30. doi:10.12788/cutis.1114

Article PDF
Article PDF
Related Articles

THE DIAGNOSIS: Dengue Hemorrhagic Fever

The retiform purpura observed in our patient was suggestive of a vasculitic, thrombotic, or embolic etiology. Dengue IgM serologic testing performed based on her extensive travel history and recent return from a dengue-endemic area was positive, indicating acute infection. A clinical diagnosis of dengue hemorrhagic fever (DHF) was made based on the hemorrhagic appearance of the lesion. Histopathology revealed leukocytoclastic vasculitis (Figure). Anti–double-stranded DNA, antideoxyribonuclease, C3 and C4, CH50 (total hemolytic complement), antineutrophil cytoplasmic antibodies, HIV, and hepatitis B virus tests were normal. Direct immunofluorescence was negative.

A, Histopathology of a biopsy from the right medial leg showed early leukocytoclastic vasculitis with karyorrhexis and red cell extravasation (H&E, original magnification ×200). B, Extensive erythrocyte extravasation and expended vessel walls with fibrin deposition also were seen (H&E, original magnification ×100).

Dengue virus is a single-stranded RNA virus transmitted by Aedes aegypti and Aedes albopictus mosquitoes and is one of the most prevalent arthropod-borne viruses affecting humans today.1,2 Infection with the dengue virus generally is seen in travelers visiting tropical regions of Africa, Mexico, South America, South and Central Asia, Southeast Asia, and the Caribbean.1 The Table shows the global distribution of dengue serotypes from 2000 to 2014.3,4 There are 4 serotypes of the dengue virus: DENV-1 to DENV-4. Infection with 1 strain elicits longlasting immunity to that strain, but subsequent infection with another strain can result in severe DHF due to antibody cross-reaction.1

Dengue virus infection ranges from mildly symptomatic to a spectrum of increasingly severe conditions that comprise dengue fever (DF) and DHF, as well as dengue shock syndrome and brain stem hemorrhage, which may be fatal.2,5 Dengue fever manifests as severe myalgia, fever, headache (usually retro-orbital), arthralgia, erythema, and rubelliform exanthema.6 The frequency of skin eruptions in patients with DF varies with the virus strain and outbreaks.7 The lesions initially develop with the onset of fever and manifest as flushing or erythematous mottling of the face, neck, and chest areas.1,7 The morbilliform eruption develops 2 to 6 days after the onset of the fever, beginning on the trunk and spreading to the face and extremities.1,7 The rash may become confluent with characteristic sparing of small round areas of normal skin described as white islands in a sea of red.2 Verrucous papules on the ears also have been described and may resemble those seen in Cowden syndrome. In patients with prior infection with a different strain of the virus, hemorrhagic lesions may develop, including characteristic retiform purpura, a positive tourniquet test, and the appearance of petechiae on the lower legs. Pruritus and desquamation, especially on the palms and soles, may follow the termination of the eruption.7

The differential diagnosis of DF includes measles, rubella, enteroviruses, and influenza. Chikungunya and West Nile viruses in Asia and Africa and the O’nyong-nyong virus in Africa are also arboviruses that cause a clinical picture similar to DF but not DHF. Other diagnostic considerations include phases of scarlet fever, typhoid, malaria, leptospirosis, hepatitis A, and trypanosomal and rickettsial diseases.7 The differential diagnosis of DHF includes antineutrophil cytoplasmic antibody–associated vasculitis, rheumatoid vasculitis, and bacterial septic vasculitis.

Acute clinical diagnosis of DF can be challenging because of the nonspecific symptoms that can be seen in almost every infectious disease. Clinical presentation assessment should be confirmed with laboratory testing.6 Dengue virus infection usually is confirmed by the identification of viral genomic RNA, antigens, or the antibodies it elicits. Enzyme-linked immunosorbent assay–based serologic tests are cost-effective and easy to perform.5 IgM antibodies usually show cross-reactivity with platelets, but the antibody levels are not positively correlated with the severity of DF.8 Primary infection with the dengue virus is characterized by the elevation of specific IgM levels that usually occurs 3 to 5 days after symptom onset and persists during the postfebrile stage (up to 30 to 60 days). In secondary infections, the IgM levels usually rise more slowly and reach a lower level than in primary infections.9 For both primary and secondary infections, testing IgM levels after the febrile stage may be helpful with the laboratory diagnosis.

Currently, there is no antiviral drug available for dengue. Treatment of dengue infection is symptomatic and supportive.2

Dengue hemorrhagic fever is indicated by a rising hematocrit (≥20%) and a falling platelet count (>100,000/mm3) accompanying clinical signs of hemorrhage. Treatment includes intravenous fluid replacement and careful clinical monitoring of hematocrit levels, platelet count, vitals, urine output, and other signs of shock.5 For patients with a history of dengue infection, travel to areas with other serotypes is not recommended.

If any travel to a high-risk area is planned, countryspecific travel recommendations and warnings should be reviewed from the Centers for Disease Control and Prevention’s website (https://wwwnc.cdc.gov/travel/notices/level1/dengue-global). Use of an Environmental Protection Agency–registered insect repellent to avoid mosquito bites and acetaminophen for managing symptoms is advised. During travel, staying in places with window and door screens and using a bed net during sleep are suggested. Long-sleeved shirts and long pants also are preferred. Travelers should see a health care provider if they have symptoms of dengue.10

African tick bite fever (ATBF) is caused by Rickettsia africae transmitted by Amblyomma ticks. Skin findings in ATBF include erythematous, firm, tender papules with central eschars consistent with the feeding patterns of ticks.11 Histopathology of ATBF usually includes fibrinoid necrosis of vessels in the dermis with a perivascular inflammatory infiltrate and coagulation necrosis of the surrounding dermis consistent with eschar formation.12 The lack of an eschar weighs against this diagnosis.

African trypanosomiasis (also known as sleeping sickness) is caused by protozoa transmitted by the tsetse fly. A chancrelike, circumscribed, rubbery, indurated red or violaceous nodule measuring 2 to 5 cm in diameter often develops as the earliest cutaneous sign of the disease.13 Nonspecific histopathologic findings, such as infiltration of lymphocytes and macrophages and proliferation of endothelial cells and fibroblasts, may be observed.14 Extravascular parasites have been noted in skin biopsies.15 In later stages, skin lesions called trypanids may be observed as macular, papular, annular, targetoid, purpuric, and erythematous lesions, and histopathologic findings consistent with vasculitis also may be seen.13

Chikungunya virus infection is an acute-onset, mosquito-borne viral disease. Skin manifestations may start with nonspecific, generalized, morbilliform, maculopapular rashes coinciding with fever, which also may be seen initially with DHF. Skin hyperpigmentation, mostly centrofacial and involving the nose (chik sign); purpuric and ecchymotic lesions over the trunk and flexors of limbs in adults, often surmounted by subepidermal bullae and lesions resembling toxic epidermal necrolysis; and nonhealing ulcers in the genital and groin areas are common skin manifestations of chikungunya infection.16 Intraepithelial splitting with acantholysis and perivascular lymphohistiocytic infiltration may be observed in the histopathology of blistering lesions, which are not consistent with DHF.17

Zika virus infection is caused by an arbovirus within the Flaviviridae family, which also includes the dengue virus. Initial mucocutaneous findings of the Zika virus include nonspecific diffuse maculopapular eruptions. The eruption generally spares the palms and soles; however, various manifestations including involvement of the palms and soles have been reported.18 The morbilliform eruption begins on the face and extends to the trunk and extremities. Mild hemorrhagic manifestations, including petechiae and bleeding gums, may be observed. Distinguishing between dengue and Zika virus infection relies on the severity of symptoms and laboratory tests, including polymerase chain reaction or IgM antibody testing.19 The other conditions listed do not produce hemorrhagic fever.

THE DIAGNOSIS: Dengue Hemorrhagic Fever

The retiform purpura observed in our patient was suggestive of a vasculitic, thrombotic, or embolic etiology. Dengue IgM serologic testing performed based on her extensive travel history and recent return from a dengue-endemic area was positive, indicating acute infection. A clinical diagnosis of dengue hemorrhagic fever (DHF) was made based on the hemorrhagic appearance of the lesion. Histopathology revealed leukocytoclastic vasculitis (Figure). Anti–double-stranded DNA, antideoxyribonuclease, C3 and C4, CH50 (total hemolytic complement), antineutrophil cytoplasmic antibodies, HIV, and hepatitis B virus tests were normal. Direct immunofluorescence was negative.

A, Histopathology of a biopsy from the right medial leg showed early leukocytoclastic vasculitis with karyorrhexis and red cell extravasation (H&E, original magnification ×200). B, Extensive erythrocyte extravasation and expended vessel walls with fibrin deposition also were seen (H&E, original magnification ×100).

Dengue virus is a single-stranded RNA virus transmitted by Aedes aegypti and Aedes albopictus mosquitoes and is one of the most prevalent arthropod-borne viruses affecting humans today.1,2 Infection with the dengue virus generally is seen in travelers visiting tropical regions of Africa, Mexico, South America, South and Central Asia, Southeast Asia, and the Caribbean.1 The Table shows the global distribution of dengue serotypes from 2000 to 2014.3,4 There are 4 serotypes of the dengue virus: DENV-1 to DENV-4. Infection with 1 strain elicits longlasting immunity to that strain, but subsequent infection with another strain can result in severe DHF due to antibody cross-reaction.1

Dengue virus infection ranges from mildly symptomatic to a spectrum of increasingly severe conditions that comprise dengue fever (DF) and DHF, as well as dengue shock syndrome and brain stem hemorrhage, which may be fatal.2,5 Dengue fever manifests as severe myalgia, fever, headache (usually retro-orbital), arthralgia, erythema, and rubelliform exanthema.6 The frequency of skin eruptions in patients with DF varies with the virus strain and outbreaks.7 The lesions initially develop with the onset of fever and manifest as flushing or erythematous mottling of the face, neck, and chest areas.1,7 The morbilliform eruption develops 2 to 6 days after the onset of the fever, beginning on the trunk and spreading to the face and extremities.1,7 The rash may become confluent with characteristic sparing of small round areas of normal skin described as white islands in a sea of red.2 Verrucous papules on the ears also have been described and may resemble those seen in Cowden syndrome. In patients with prior infection with a different strain of the virus, hemorrhagic lesions may develop, including characteristic retiform purpura, a positive tourniquet test, and the appearance of petechiae on the lower legs. Pruritus and desquamation, especially on the palms and soles, may follow the termination of the eruption.7

The differential diagnosis of DF includes measles, rubella, enteroviruses, and influenza. Chikungunya and West Nile viruses in Asia and Africa and the O’nyong-nyong virus in Africa are also arboviruses that cause a clinical picture similar to DF but not DHF. Other diagnostic considerations include phases of scarlet fever, typhoid, malaria, leptospirosis, hepatitis A, and trypanosomal and rickettsial diseases.7 The differential diagnosis of DHF includes antineutrophil cytoplasmic antibody–associated vasculitis, rheumatoid vasculitis, and bacterial septic vasculitis.

Acute clinical diagnosis of DF can be challenging because of the nonspecific symptoms that can be seen in almost every infectious disease. Clinical presentation assessment should be confirmed with laboratory testing.6 Dengue virus infection usually is confirmed by the identification of viral genomic RNA, antigens, or the antibodies it elicits. Enzyme-linked immunosorbent assay–based serologic tests are cost-effective and easy to perform.5 IgM antibodies usually show cross-reactivity with platelets, but the antibody levels are not positively correlated with the severity of DF.8 Primary infection with the dengue virus is characterized by the elevation of specific IgM levels that usually occurs 3 to 5 days after symptom onset and persists during the postfebrile stage (up to 30 to 60 days). In secondary infections, the IgM levels usually rise more slowly and reach a lower level than in primary infections.9 For both primary and secondary infections, testing IgM levels after the febrile stage may be helpful with the laboratory diagnosis.

Currently, there is no antiviral drug available for dengue. Treatment of dengue infection is symptomatic and supportive.2

Dengue hemorrhagic fever is indicated by a rising hematocrit (≥20%) and a falling platelet count (>100,000/mm3) accompanying clinical signs of hemorrhage. Treatment includes intravenous fluid replacement and careful clinical monitoring of hematocrit levels, platelet count, vitals, urine output, and other signs of shock.5 For patients with a history of dengue infection, travel to areas with other serotypes is not recommended.

If any travel to a high-risk area is planned, countryspecific travel recommendations and warnings should be reviewed from the Centers for Disease Control and Prevention’s website (https://wwwnc.cdc.gov/travel/notices/level1/dengue-global). Use of an Environmental Protection Agency–registered insect repellent to avoid mosquito bites and acetaminophen for managing symptoms is advised. During travel, staying in places with window and door screens and using a bed net during sleep are suggested. Long-sleeved shirts and long pants also are preferred. Travelers should see a health care provider if they have symptoms of dengue.10

African tick bite fever (ATBF) is caused by Rickettsia africae transmitted by Amblyomma ticks. Skin findings in ATBF include erythematous, firm, tender papules with central eschars consistent with the feeding patterns of ticks.11 Histopathology of ATBF usually includes fibrinoid necrosis of vessels in the dermis with a perivascular inflammatory infiltrate and coagulation necrosis of the surrounding dermis consistent with eschar formation.12 The lack of an eschar weighs against this diagnosis.

African trypanosomiasis (also known as sleeping sickness) is caused by protozoa transmitted by the tsetse fly. A chancrelike, circumscribed, rubbery, indurated red or violaceous nodule measuring 2 to 5 cm in diameter often develops as the earliest cutaneous sign of the disease.13 Nonspecific histopathologic findings, such as infiltration of lymphocytes and macrophages and proliferation of endothelial cells and fibroblasts, may be observed.14 Extravascular parasites have been noted in skin biopsies.15 In later stages, skin lesions called trypanids may be observed as macular, papular, annular, targetoid, purpuric, and erythematous lesions, and histopathologic findings consistent with vasculitis also may be seen.13

Chikungunya virus infection is an acute-onset, mosquito-borne viral disease. Skin manifestations may start with nonspecific, generalized, morbilliform, maculopapular rashes coinciding with fever, which also may be seen initially with DHF. Skin hyperpigmentation, mostly centrofacial and involving the nose (chik sign); purpuric and ecchymotic lesions over the trunk and flexors of limbs in adults, often surmounted by subepidermal bullae and lesions resembling toxic epidermal necrolysis; and nonhealing ulcers in the genital and groin areas are common skin manifestations of chikungunya infection.16 Intraepithelial splitting with acantholysis and perivascular lymphohistiocytic infiltration may be observed in the histopathology of blistering lesions, which are not consistent with DHF.17

Zika virus infection is caused by an arbovirus within the Flaviviridae family, which also includes the dengue virus. Initial mucocutaneous findings of the Zika virus include nonspecific diffuse maculopapular eruptions. The eruption generally spares the palms and soles; however, various manifestations including involvement of the palms and soles have been reported.18 The morbilliform eruption begins on the face and extends to the trunk and extremities. Mild hemorrhagic manifestations, including petechiae and bleeding gums, may be observed. Distinguishing between dengue and Zika virus infection relies on the severity of symptoms and laboratory tests, including polymerase chain reaction or IgM antibody testing.19 The other conditions listed do not produce hemorrhagic fever.

References
  1. Pincus LB, Grossman ME, Fox LP. The exanthem of dengue fever: clinical features of two US tourists traveling abroad. J Am Acad Dermatol. 2008;58:308-316. doi:10.1016/j.jaad.2007.08.042
  2. Radakovic-Fijan S, Graninger W, Müller C, et al. Dengue hemorrhagic fever in a British travel guide. J Am Acad Dermatol. 2002;46:430-433. doi:10.1067/mjd.2002.111904
  3. Yamashita A, Sakamoto T, Sekizuka T, et al. DGV: dengue genographic viewer. Front Microbiol. 2016;7:875. doi:10.3389/fmicb.2016.00875
  4. Centers for Disease and Prevention. Dengue in the US states and territories. Updated October 7, 2020. Accessed September 30, 2024. https://www.cdc.gov/dengue/data-research/facts-stats/?CDC_AAref_Val=https://www.cdc.gov/dengue/areaswithrisk/in-the-us.html
  5. Khetarpal N, Khanna I. Dengue fever: causes, complications, and vaccine strategies. J Immunol Res. 2016;2016:6803098. doi:10.1155/2016/6803098
  6. Muller DA, Depelsenaire AC, Young PR. Clinical and laboratory diagnosis of dengue virus infection. J Infect Dis. 2017;215(suppl 2):S89-S95. doi:10.1093/infdis/jiw649
  7. Waterman SH, Gubler DJ. Dengue fever. Clin Dermatol. 1989;7:117-122. doi:10.1016/0738-081x(89)90034-5
  8. Lin CF, Lei HY, Liu CC, et al. Generation of IgM anti-platelet autoantibody in dengue patients. J Med Virol. 2001;63:143-149. doi:10.1002/1096- 9071(20000201)63:2<143::AID-JMV1009>3.0.CO;2-L
  9. Tripathi NK, Shrivastava A, Dash PK, et al. Detection of dengue virus. Methods Mol Biol. 2011;665:51-64. doi:10.1007/978-1-60761-817-1_4
  10. Centers for Disease Control and Prevention. Plan for travel. Accessed September 30, 2024. https://wwwnc.cdc.gov/travel
  11. Mack I, Ritz N. African tick-bite fever. N Engl J Med. 2019;380:960. doi:10.1056/NEJMicm1810093
  12. Lepidi H, Fournier PE, Raoult D. Histologic features and immunodetection of African tick-bite fever eschar. Emerg Infect Dis. 2006;12:1332- 1337. doi:10.3201/eid1209.051540
  13. McGovern TW, Williams W, Fitzpatrick JE, et al. Cutaneous manifestations of African trypanosomiasis. Arch Dermatol. 1995;131:1178-1182.
  14. Kristensson K, Bentivoglio M. Pathology of African trypanosomiasis. In: Dumas M, Bouteille B, Buguet A, eds. Progress in Human African Trypanosomiasis, Sleeping Sickness. Springer; 1999:157-181.
  15. Capewell P, Cren-Travaillé C, Marchesi F, et al. The skin is a significant but overlooked anatomical reservoir for vector-borne African trypanosomes. Elife. 2016;5:e17716. doi:10.7554/eLife.17716
  16. Singal A. Chikungunya and skin: current perspective. Indian Dermatol Online J. 2017;8:307-309. doi:10.4103/idoj.IDOJ_93_17
  17. Robin S, Ramful D, Zettor J, et al. Severe bullous skin lesions associated with chikungunya virus infection in small infants. Eur J Pediatr. 2009;169:67-72. doi:10.1007/s00431-009-0986-0
  18. Hussain A, Ali F, Latiwesh OB, et al. A comprehensive review of the manifestations and pathogenesis of Zika virus in neonates and adults. Cureus. 2018;10:E3290. doi:10.7759/cureus.3290
  19. Farahnik B, Beroukhim K, Blattner CM, et al. Cutaneous manifestations of the Zika virus. J Am Acad Dermatol. 2016;74:1286-1287. doi:10.1016/j.jaad.2016.02.1232
References
  1. Pincus LB, Grossman ME, Fox LP. The exanthem of dengue fever: clinical features of two US tourists traveling abroad. J Am Acad Dermatol. 2008;58:308-316. doi:10.1016/j.jaad.2007.08.042
  2. Radakovic-Fijan S, Graninger W, Müller C, et al. Dengue hemorrhagic fever in a British travel guide. J Am Acad Dermatol. 2002;46:430-433. doi:10.1067/mjd.2002.111904
  3. Yamashita A, Sakamoto T, Sekizuka T, et al. DGV: dengue genographic viewer. Front Microbiol. 2016;7:875. doi:10.3389/fmicb.2016.00875
  4. Centers for Disease and Prevention. Dengue in the US states and territories. Updated October 7, 2020. Accessed September 30, 2024. https://www.cdc.gov/dengue/data-research/facts-stats/?CDC_AAref_Val=https://www.cdc.gov/dengue/areaswithrisk/in-the-us.html
  5. Khetarpal N, Khanna I. Dengue fever: causes, complications, and vaccine strategies. J Immunol Res. 2016;2016:6803098. doi:10.1155/2016/6803098
  6. Muller DA, Depelsenaire AC, Young PR. Clinical and laboratory diagnosis of dengue virus infection. J Infect Dis. 2017;215(suppl 2):S89-S95. doi:10.1093/infdis/jiw649
  7. Waterman SH, Gubler DJ. Dengue fever. Clin Dermatol. 1989;7:117-122. doi:10.1016/0738-081x(89)90034-5
  8. Lin CF, Lei HY, Liu CC, et al. Generation of IgM anti-platelet autoantibody in dengue patients. J Med Virol. 2001;63:143-149. doi:10.1002/1096- 9071(20000201)63:2<143::AID-JMV1009>3.0.CO;2-L
  9. Tripathi NK, Shrivastava A, Dash PK, et al. Detection of dengue virus. Methods Mol Biol. 2011;665:51-64. doi:10.1007/978-1-60761-817-1_4
  10. Centers for Disease Control and Prevention. Plan for travel. Accessed September 30, 2024. https://wwwnc.cdc.gov/travel
  11. Mack I, Ritz N. African tick-bite fever. N Engl J Med. 2019;380:960. doi:10.1056/NEJMicm1810093
  12. Lepidi H, Fournier PE, Raoult D. Histologic features and immunodetection of African tick-bite fever eschar. Emerg Infect Dis. 2006;12:1332- 1337. doi:10.3201/eid1209.051540
  13. McGovern TW, Williams W, Fitzpatrick JE, et al. Cutaneous manifestations of African trypanosomiasis. Arch Dermatol. 1995;131:1178-1182.
  14. Kristensson K, Bentivoglio M. Pathology of African trypanosomiasis. In: Dumas M, Bouteille B, Buguet A, eds. Progress in Human African Trypanosomiasis, Sleeping Sickness. Springer; 1999:157-181.
  15. Capewell P, Cren-Travaillé C, Marchesi F, et al. The skin is a significant but overlooked anatomical reservoir for vector-borne African trypanosomes. Elife. 2016;5:e17716. doi:10.7554/eLife.17716
  16. Singal A. Chikungunya and skin: current perspective. Indian Dermatol Online J. 2017;8:307-309. doi:10.4103/idoj.IDOJ_93_17
  17. Robin S, Ramful D, Zettor J, et al. Severe bullous skin lesions associated with chikungunya virus infection in small infants. Eur J Pediatr. 2009;169:67-72. doi:10.1007/s00431-009-0986-0
  18. Hussain A, Ali F, Latiwesh OB, et al. A comprehensive review of the manifestations and pathogenesis of Zika virus in neonates and adults. Cureus. 2018;10:E3290. doi:10.7759/cureus.3290
  19. Farahnik B, Beroukhim K, Blattner CM, et al. Cutaneous manifestations of the Zika virus. J Am Acad Dermatol. 2016;74:1286-1287. doi:10.1016/j.jaad.2016.02.1232
Issue
Cutis - 114(3)
Issue
Cutis - 114(3)
Page Number
E27-E30
Page Number
E27-E30
Publications
Publications
Topics
Article Type
Display Headline
Purpuric Lesions on the Leg
Display Headline
Purpuric Lesions on the Leg
Sections
Questionnaire Body

A 74-year-old woman who frequently traveled abroad presented to the dermatology department with retiform purpura of the lower leg along with gastrointestinal cramps, fatigue, and myalgia. The patient reported that the symptoms had started 10 days after returning from a recent trip to Africa.

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Tue, 10/01/2024 - 10:15
Un-Gate On Date
Tue, 10/01/2024 - 10:15
Use ProPublica
CFC Schedule Remove Status
Tue, 10/01/2024 - 10:15
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

Multiple Draining Sinus Tracts on the Thigh

Article Type
Changed
Fri, 09/06/2024 - 09:52
Display Headline
Multiple Draining Sinus Tracts on the Thigh

The Diagnosis: Mycobacterial Infection

An injury sustained in a wet environment that results in chronic indolent abscesses, nodules, or draining sinus tracts suggests a mycobacterial infection. In our patient, a culture revealed MycobacteriuM fortuitum, which is classified in the rapid grower nontuberculous mycobacteria (NTM) group, along with Mycobacterium chelonae and Mycobacterium abscessus.1 The patient’s history of skin injury while cutting wet grass and the common presence of M fortuitum in the environment suggested that the organism entered the wound. The patient healed completely following surgical excision and a 2-month course of clarithromycin 1 g daily and rifampin 600 mg daily.

MycobacteriuM fortuitum was first isolated from an amphibian source in 1905 and later identified in a human with cutaneous infection in 1938. It commonly is found in soil and water.2 Skin and soft-tissue infections with M fortuitum usually are acquired from direct entry of the organism through a damaged skin barrier from trauma, medical injection, surgery, or tattoo placement.2,3

Skin lesions caused by NTM often are nonspecific and can mimic a variety of other dermatologic conditions, making clinical diagnosis challenging. As such, cutaneous manifestations of M fortuitum infection can include recurrent cutaneous abscesses, nodular lesions, chronic discharging sinuses, cellulitis, and surgical site infections.4 Although cutaneous infection with M fortuitum classically manifests with a single subcutaneous nodule at the site of trauma or surgery,5 it also can manifest as multiple draining sinus tracts, as seen in our patient. Hence, the diagnosis and treatment of cutaneous NTM infection is challenging, especially when M fortuitum skin manifestations can take up to 4 to 6 weeks to develop after inoculation. Diagnosis often requires a detailed patient history, tissue cultures, and histopathology.5

In recent years, rapid detection with polymerase chain reaction (PCR) techniques has been employed more widely. Notably, a molecular system based on multiplex real-time PCR with high-resolution melting was shown to have a sensitivity of up to 54% for distinguishing M fortuitum from other NTM.6 More recently, a 2-step real-time PCR method has demonstrated diagnostic sensitivity and specificity for differentiating NTM from Mycobacterium tuberculosis infections and identifying the causative NTM agent.7

Compared to immunocompetent individuals, those who are immunocompromised are more susceptible to less pathogenic strains of NTM, which can cause dissemination and lead to tenosynovitis, myositis, osteomyelitis, and septic arthritis.8-12 Nonetheless, cases of infections with NTM—including M fortuitum—are becoming harder to treat. Several single nucleotide polymorphisms and point mutations have been demonstrated in the ribosomal RNA methylase gene erm(39) related to clarithromycin resistance and in the rrl gene related to linezolid resistance.13 Due to increasing inducible resistance to common classes of antibiotics, such as macrolides and linezolid, treatment of M fortuitum requires multidrug regimens.13,14 Drug susceptibility testing also may be required, as M fortuitum has shown low resistance to tigecycline, tetracycline, cefmetazole, imipenem, and aminoglycosides (eg, amikacin, tobramycin, neomycin, gentamycin). Surgery is an important adjunctive tool in treating M fortuitum infections; patients with a single lesion are more likely to undergo surgical treatment alone or in combination with antibiotic therapy.15 More recently, antimicrobial photodynamic therapy has been explored as an alternative to eliminate NTM, including M fortuitum.16

The differential diagnosis for skin lesions manifesting with draining fistulae and sinus tracts includes conditions with infectious (cellulitis and chromomycosis) and inflammatory (pyoderma gangrenosum [PG] and hidradenitis suppurativa [HS]) causes.

Cellulitis is a common infection of the skin and subcutaneous tissue that predominantly is caused by gram-positive organisms such as β-hemolytic streptococci.17 Clinical manifestations include acute skin erythema, swelling, tenderness, and warmth. The legs are the most common sites of infection, but any area of the skin can be involved.17 Cellulitis comprises 10% of all infectious disease hospitalizations and up to 11% of all dermatologic admissions.18,19 It frequently is misdiagnosed, perhaps due to the lack of a reliable confirmatory laboratory test or imaging study, in addition to the plethora of diseases that mimic cellulitis, such as stasis dermatitis, lipodermatosclerosis, contact dermatitis, lymphedema, eosinophilic cellulitis, and papular urticaria.20,21 The consequences of misdiagnosis include but are not limited to unnecessary hospitalizations, inappropriate antibiotic use, and delayed management of the disease; thus, there is an urgent need for a reliable standard test to confirm the diagnosis, especially among nonspecialist physicians. 20 Most patients with uncomplicated cellulitis can be treated with empiric oral antibiotics that target β-hemolytic streptococci (ie, penicillin V potassium, amoxicillin).17 Methicillin-resistant Staphylococcus aureus coverage generally is unnecessary for nonpurulent cellulitis, but clinicians can consider adding amoxicillin-clavulanate, dicloxacillin, and cephalexin to the regimen. For purulent cellulitis, incision and drainage should be performed. In severe cases that manifest with sepsis, altered mental status, or hemodynamic instability, inpatient management is required.17

Chromomycosis (also known as chromoblastomycosis) is a chronic, indolent, granulomatous, suppurative mycosis of the skin and subcutaneous tissue22 that is caused by traumatic inoculation of various fungi of the order Chaetothyriales and family Herpotrichiellaceae, which are present in soil, plants, and decomposing wood. Chromomycosis is prevalent in tropical and subtropical regions.23,24 Clinically, it manifests as oligosymptomatic or asymptomatic lesions around an infection site that can manifest as papules with centrifugal growth evolving into nodular, verrucous, plaque, tumoral, or atrophic forms.22 Diagnosis is made with direct microscopy using potassium hydroxide, which reveals muriform bodies. Fungal culture in Sabouraud agar also can be used to isolate the causative pathogen.22 Unfortunately, chromomycosis is difficult to treat, with low cure rates and high relapse rates. Antifungal agents combined with surgery, cryotherapy, or thermotherapy often are used, with cure rates ranging from 15% to 80%.22,25

Pyoderma gangrenosum is a reactive noninfectious inflammatory dermatosis associated with inflammatory bowel disease and rheumatoid arthritis. The exact etiology is not clearly understood, but it generally is considered an autoinflammatory disorder.26 The most common form—classical PG—occurs in approximately 85% of cases and manifests as a painful erythematous lesion that progresses to a blistered or necrotic ulcer. It primarily affects the lower legs but can occur in other body sites.27 The diagnosis is based on clinical symptoms after excluding other similar conditions; histopathology of biopsied wound tissues often are required for confirmation. Treatment of PG starts with fast-acting immunosuppressive drugs (corticosteroids and/or cyclosporine) followed by slowacting immunosuppressive drugs (biologics).26

Hidradenitis suppurativa is a chronic recurrent disease of the hair follicle unit that develops after puberty.28 Clinically, HS manifests with painful nodules, abscesses, chronically draining fistulas, and scarring in areas of the body rich in apocrine glands.29,30 Treatment of HS is challenging due to its diverse clinical manifestations and unclear etiology. Topical therapy, systemic treatments, biologic agents, surgery, and light therapy have shown variable results.28,31

References
  1. Franco-Paredes C, Marcos LA, Henao-Martínez AF, et al. Cutaneous mycobacterial infections. Clin Microbiol Rev. 2018;32: E00069-18. doi:10.1128/CMR.00069-18
  2. Brown TH. The rapidly growing mycobacteria—MycobacteriuM fortuitum and Mycobacterium chelonae. Infect Control. 1985;6:283-238. doi:10.1017/s0195941700061762
  3. Hooper J; Beltrami EJ; Santoro F; et al. Remember the fite: a case of cutaneous MycobacteriuM fortuitum infection. Am J Dermatopathol. 2023;45:214-215. doi:10.1097/DAD.0000000000002336
  4. Franco-Paredes C, Chastain DB, Allen L, et al. Overview of cutaneous mycobacterial infections. Curr Trop Med Rep. 2018;5:228-232. doi:10.1007/s40475-018-0161-7
  5. Gonzalez-Santiago TM, Drage LA. Nontuberculous mycobacteria: skin and soft tissue infections. Dermatol Clin. 2015;33:563-77. doi:10.1016/j.det.2015.03.017
  6. Peixoto ADS, Montenegro LML, Lima AS, et al. Identification of nontuberculous mycobacteria species by multiplex real-time PCR with high-resolution melting. Rev Soc Bras Med Trop. 2020;53:E20200211. doi:10.1590/0037-8682-0211-2020
  7. Park J, Kwak N, Chae JC, et al. A two-step real-time PCR method to identify Mycobacterium tuberculosis infections and six dominant nontuberculous mycobacterial infections from clinical specimens. Microbiol Spectr. 2023:E0160623. doi:10.1128/spectrum.01606-23
  8. Fowler J, Mahlen SD. Localized cutaneous infections in immunocompetent individuals due to rapidly growing mycobacteria. Arch Pathol Lab Med. 2014;138:1106-1109. doi:10.5858/arpa.2012-0203-RS
  9. Gardini G, Gregori N, Matteelli A, et al. Mycobacterial skin infection. Curr Opin Infect Dis. 2022;35:79-87. doi:10.1097/QCO.0000000000000820
  10. Wang SH, Pancholi P. Mycobacterial skin and soft tissue infection. Curr Infect Dis Rep. 2014;16:438. doi:10.1007/s11908-014-0438-5
  11. Griffith DE, Aksamit T, Brown-Elliott BA, et al; ATS Mycobacterial Diseases Subcommittee; American Thoracic Society; Infectious Disease Society of America. An official ATS/IDSA statement: diagnosis, treatment, and prevention of nontuberculous mycobacterial diseases. Am J Respir Crit Care Med. 2007;175:367-416. doi:10.1164/rccm.200604-571ST
  12. Mougari F, Guglielmetti L, Raskine L, et al. Infections caused by Mycobacterium abscessus: epidemiology, diagnostic tools and treatment. Expert Rev Anti Infect Ther. 2016;14:1139-1154. doi:10.1080/14787210.201 6.1238304
  13. Tu HZ, Lee HS, Chen YS, et al. High rates of antimicrobial resistance in rapidly growing mycobacterial infections in Taiwan. Pathogens. 2022;11:969. doi:10.3390/pathogens11090969
  14. Hashemzadeh M, Zadegan Dezfuli AA, Khosravi AD, et al. F requency of mutations in erm(39) related to clarithromycin resistance and in rrl related to linezolid resistance in clinical isolates of MycobacteriuM fortuitum in Iran. Acta Microbiol Immunol Hung. 2023;70:167-176. doi:10.1556/030.2023.02020
  15. Uslan DZ, Kowalski TJ, Wengenack NL, et al. Skin and soft tissue infections due to rapidly growing mycobacteria: comparison of clinical features, treatment, and susceptibility. Arch Dermatol. 2006;142:1287-1292. doi:10.1001/archderm.142.10.1287
  16. Miretti M, Juri L, Peralta A, et al. Photoinactivation of non-tuberculous mycobacteria using Zn-phthalocyanine loaded into liposomes. Tuberculosis (Edinb). 2022;136:102247. doi:10.1016/j.tube.2022.102247
  17. Bystritsky RJ. Cellulitis. Infect Dis Clin North Am. 2021;35:49-60. doi:10.1016/j.idc.2020.10.002
  18. Christensen K, Holman R, Steiner C, et al. Infectious disease hospitalizations in the United States. Clin Infect Dis. 2009;49:1025-1035. doi:10.1086/605562
  19. Yang JJ, Maloney NJ, Bach DQ, et al. Dermatology in the emergency department: prescriptions, rates of inpatient admission, and predictors of high utilization in the United States from 1996 to 2012. J Am Acad Dermatol. 2021;84:1480-1483. doi:10.1016/J.JAAD.2020.07.055
  20. Cutler TS, Jannat-Khah DP, Kam B, et al. Prevalence of misdiagnosis of cellulitis: a systematic review and meta-analysis. J Hosp Med. 2023;18:254-261. doi:10.1002/jhm.12977
  21. Keller EC, Tomecki KJ, Alraies MC. Distinguishing cellulitis from its mimics. Cleve Clin J Med. 2012;79:547-52. doi:10.3949/ccjm.79a.11121
  22. Brito AC, Bittencourt MJS. Chromoblastomycosis: an etiological, epidemiological, clinical, diagnostic, and treatment update. An Bras Dermatol. 2018;93:495-506. doi:10.1590/abd1806-4841.20187321
  23. McGinnis MR. Chromoblastomycosis and phaeohyphomycosis: new concepts, diagnosis, and mycology. J Am Acad Dermatol. 1983;8:1-16.
  24. Rubin HA, Bruce S, Rosen T, et al. Evidence for percutaneous inoculation as the mode of transmission for chromoblastomycosis. J Am Acad Dermatol. 1991;25:951-954.
  25. Bonifaz A, Paredes-Solís V, Saúl A. Treating chromoblastomycosis with systemic antifungals. Expert Opin Pharmacother. 2004;5:247-254.
  26. Maverakis E, Marzano AV, Le ST, et al. Pyoderma gangrenosum. Nat Rev Dis Primers. 2020;6:81. doi:10.1038/s41572-020-0213-x
  27. George C, Deroide F, Rustin M. Pyoderma gangrenosum—a guide to diagnosis and management. Clin Med (Lond). 2019;19:224-228. doi:10.7861/clinmedicine.19-3-224
  28. Narla S, Lyons AB, Hamzavi IH. The most recent advances in understanding and managing hidradenitis suppurativa. F1000Res. 2020;9:F1000 Faculty Rev-1049. doi:10.12688/f1000research.26083.1
  29. Garg A, Lavian J, Lin G, et al. Incidence of hidradenitis suppurativa in the United States: a sex- and age-adjusted population analysis. J Am Acad Dermatol. 2017;77:118-122. doi:10.1016/j.jaad.2017.02.005
  30. Daxhelet M, Suppa M, White J, et al. Proposed definitions of typical lesions in hidradenitis suppurativa. Dermatology. 2020;236:431-438. doi:10.1159/000507348
  31. Amat-Samaranch V, Agut-Busquet E, Vilarrasa E, et al. New perspectives on the treatment of hidradenitis suppurativa. Ther Adv Chronic Dis. 2021;12:20406223211055920. doi:10.1177/20406223211055920
Article PDF
Author and Disclosure Information

Toan S. Bui is from the University of Maryland School of Medicine, Baltimore. Dr. Elston is from the Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston.

The authors report no conflict of interest.

Correspondence: Toan S. Bui, BS, 655 W Baltimore St S, Baltimore, MD 21201 (toanbui@som.umaryland.edu).

Cutis. 2024 September;114(3):71, 77-78. doi:10.12788/cutis.1084

Issue
Cutis - 114(3)
Publications
Topics
Page Number
71,77-78
Sections
Author and Disclosure Information

Toan S. Bui is from the University of Maryland School of Medicine, Baltimore. Dr. Elston is from the Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston.

The authors report no conflict of interest.

Correspondence: Toan S. Bui, BS, 655 W Baltimore St S, Baltimore, MD 21201 (toanbui@som.umaryland.edu).

Cutis. 2024 September;114(3):71, 77-78. doi:10.12788/cutis.1084

Author and Disclosure Information

Toan S. Bui is from the University of Maryland School of Medicine, Baltimore. Dr. Elston is from the Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston.

The authors report no conflict of interest.

Correspondence: Toan S. Bui, BS, 655 W Baltimore St S, Baltimore, MD 21201 (toanbui@som.umaryland.edu).

Cutis. 2024 September;114(3):71, 77-78. doi:10.12788/cutis.1084

Article PDF
Article PDF
Related Articles

The Diagnosis: Mycobacterial Infection

An injury sustained in a wet environment that results in chronic indolent abscesses, nodules, or draining sinus tracts suggests a mycobacterial infection. In our patient, a culture revealed MycobacteriuM fortuitum, which is classified in the rapid grower nontuberculous mycobacteria (NTM) group, along with Mycobacterium chelonae and Mycobacterium abscessus.1 The patient’s history of skin injury while cutting wet grass and the common presence of M fortuitum in the environment suggested that the organism entered the wound. The patient healed completely following surgical excision and a 2-month course of clarithromycin 1 g daily and rifampin 600 mg daily.

MycobacteriuM fortuitum was first isolated from an amphibian source in 1905 and later identified in a human with cutaneous infection in 1938. It commonly is found in soil and water.2 Skin and soft-tissue infections with M fortuitum usually are acquired from direct entry of the organism through a damaged skin barrier from trauma, medical injection, surgery, or tattoo placement.2,3

Skin lesions caused by NTM often are nonspecific and can mimic a variety of other dermatologic conditions, making clinical diagnosis challenging. As such, cutaneous manifestations of M fortuitum infection can include recurrent cutaneous abscesses, nodular lesions, chronic discharging sinuses, cellulitis, and surgical site infections.4 Although cutaneous infection with M fortuitum classically manifests with a single subcutaneous nodule at the site of trauma or surgery,5 it also can manifest as multiple draining sinus tracts, as seen in our patient. Hence, the diagnosis and treatment of cutaneous NTM infection is challenging, especially when M fortuitum skin manifestations can take up to 4 to 6 weeks to develop after inoculation. Diagnosis often requires a detailed patient history, tissue cultures, and histopathology.5

In recent years, rapid detection with polymerase chain reaction (PCR) techniques has been employed more widely. Notably, a molecular system based on multiplex real-time PCR with high-resolution melting was shown to have a sensitivity of up to 54% for distinguishing M fortuitum from other NTM.6 More recently, a 2-step real-time PCR method has demonstrated diagnostic sensitivity and specificity for differentiating NTM from Mycobacterium tuberculosis infections and identifying the causative NTM agent.7

Compared to immunocompetent individuals, those who are immunocompromised are more susceptible to less pathogenic strains of NTM, which can cause dissemination and lead to tenosynovitis, myositis, osteomyelitis, and septic arthritis.8-12 Nonetheless, cases of infections with NTM—including M fortuitum—are becoming harder to treat. Several single nucleotide polymorphisms and point mutations have been demonstrated in the ribosomal RNA methylase gene erm(39) related to clarithromycin resistance and in the rrl gene related to linezolid resistance.13 Due to increasing inducible resistance to common classes of antibiotics, such as macrolides and linezolid, treatment of M fortuitum requires multidrug regimens.13,14 Drug susceptibility testing also may be required, as M fortuitum has shown low resistance to tigecycline, tetracycline, cefmetazole, imipenem, and aminoglycosides (eg, amikacin, tobramycin, neomycin, gentamycin). Surgery is an important adjunctive tool in treating M fortuitum infections; patients with a single lesion are more likely to undergo surgical treatment alone or in combination with antibiotic therapy.15 More recently, antimicrobial photodynamic therapy has been explored as an alternative to eliminate NTM, including M fortuitum.16

The differential diagnosis for skin lesions manifesting with draining fistulae and sinus tracts includes conditions with infectious (cellulitis and chromomycosis) and inflammatory (pyoderma gangrenosum [PG] and hidradenitis suppurativa [HS]) causes.

Cellulitis is a common infection of the skin and subcutaneous tissue that predominantly is caused by gram-positive organisms such as β-hemolytic streptococci.17 Clinical manifestations include acute skin erythema, swelling, tenderness, and warmth. The legs are the most common sites of infection, but any area of the skin can be involved.17 Cellulitis comprises 10% of all infectious disease hospitalizations and up to 11% of all dermatologic admissions.18,19 It frequently is misdiagnosed, perhaps due to the lack of a reliable confirmatory laboratory test or imaging study, in addition to the plethora of diseases that mimic cellulitis, such as stasis dermatitis, lipodermatosclerosis, contact dermatitis, lymphedema, eosinophilic cellulitis, and papular urticaria.20,21 The consequences of misdiagnosis include but are not limited to unnecessary hospitalizations, inappropriate antibiotic use, and delayed management of the disease; thus, there is an urgent need for a reliable standard test to confirm the diagnosis, especially among nonspecialist physicians. 20 Most patients with uncomplicated cellulitis can be treated with empiric oral antibiotics that target β-hemolytic streptococci (ie, penicillin V potassium, amoxicillin).17 Methicillin-resistant Staphylococcus aureus coverage generally is unnecessary for nonpurulent cellulitis, but clinicians can consider adding amoxicillin-clavulanate, dicloxacillin, and cephalexin to the regimen. For purulent cellulitis, incision and drainage should be performed. In severe cases that manifest with sepsis, altered mental status, or hemodynamic instability, inpatient management is required.17

Chromomycosis (also known as chromoblastomycosis) is a chronic, indolent, granulomatous, suppurative mycosis of the skin and subcutaneous tissue22 that is caused by traumatic inoculation of various fungi of the order Chaetothyriales and family Herpotrichiellaceae, which are present in soil, plants, and decomposing wood. Chromomycosis is prevalent in tropical and subtropical regions.23,24 Clinically, it manifests as oligosymptomatic or asymptomatic lesions around an infection site that can manifest as papules with centrifugal growth evolving into nodular, verrucous, plaque, tumoral, or atrophic forms.22 Diagnosis is made with direct microscopy using potassium hydroxide, which reveals muriform bodies. Fungal culture in Sabouraud agar also can be used to isolate the causative pathogen.22 Unfortunately, chromomycosis is difficult to treat, with low cure rates and high relapse rates. Antifungal agents combined with surgery, cryotherapy, or thermotherapy often are used, with cure rates ranging from 15% to 80%.22,25

Pyoderma gangrenosum is a reactive noninfectious inflammatory dermatosis associated with inflammatory bowel disease and rheumatoid arthritis. The exact etiology is not clearly understood, but it generally is considered an autoinflammatory disorder.26 The most common form—classical PG—occurs in approximately 85% of cases and manifests as a painful erythematous lesion that progresses to a blistered or necrotic ulcer. It primarily affects the lower legs but can occur in other body sites.27 The diagnosis is based on clinical symptoms after excluding other similar conditions; histopathology of biopsied wound tissues often are required for confirmation. Treatment of PG starts with fast-acting immunosuppressive drugs (corticosteroids and/or cyclosporine) followed by slowacting immunosuppressive drugs (biologics).26

Hidradenitis suppurativa is a chronic recurrent disease of the hair follicle unit that develops after puberty.28 Clinically, HS manifests with painful nodules, abscesses, chronically draining fistulas, and scarring in areas of the body rich in apocrine glands.29,30 Treatment of HS is challenging due to its diverse clinical manifestations and unclear etiology. Topical therapy, systemic treatments, biologic agents, surgery, and light therapy have shown variable results.28,31

The Diagnosis: Mycobacterial Infection

An injury sustained in a wet environment that results in chronic indolent abscesses, nodules, or draining sinus tracts suggests a mycobacterial infection. In our patient, a culture revealed MycobacteriuM fortuitum, which is classified in the rapid grower nontuberculous mycobacteria (NTM) group, along with Mycobacterium chelonae and Mycobacterium abscessus.1 The patient’s history of skin injury while cutting wet grass and the common presence of M fortuitum in the environment suggested that the organism entered the wound. The patient healed completely following surgical excision and a 2-month course of clarithromycin 1 g daily and rifampin 600 mg daily.

MycobacteriuM fortuitum was first isolated from an amphibian source in 1905 and later identified in a human with cutaneous infection in 1938. It commonly is found in soil and water.2 Skin and soft-tissue infections with M fortuitum usually are acquired from direct entry of the organism through a damaged skin barrier from trauma, medical injection, surgery, or tattoo placement.2,3

Skin lesions caused by NTM often are nonspecific and can mimic a variety of other dermatologic conditions, making clinical diagnosis challenging. As such, cutaneous manifestations of M fortuitum infection can include recurrent cutaneous abscesses, nodular lesions, chronic discharging sinuses, cellulitis, and surgical site infections.4 Although cutaneous infection with M fortuitum classically manifests with a single subcutaneous nodule at the site of trauma or surgery,5 it also can manifest as multiple draining sinus tracts, as seen in our patient. Hence, the diagnosis and treatment of cutaneous NTM infection is challenging, especially when M fortuitum skin manifestations can take up to 4 to 6 weeks to develop after inoculation. Diagnosis often requires a detailed patient history, tissue cultures, and histopathology.5

In recent years, rapid detection with polymerase chain reaction (PCR) techniques has been employed more widely. Notably, a molecular system based on multiplex real-time PCR with high-resolution melting was shown to have a sensitivity of up to 54% for distinguishing M fortuitum from other NTM.6 More recently, a 2-step real-time PCR method has demonstrated diagnostic sensitivity and specificity for differentiating NTM from Mycobacterium tuberculosis infections and identifying the causative NTM agent.7

Compared to immunocompetent individuals, those who are immunocompromised are more susceptible to less pathogenic strains of NTM, which can cause dissemination and lead to tenosynovitis, myositis, osteomyelitis, and septic arthritis.8-12 Nonetheless, cases of infections with NTM—including M fortuitum—are becoming harder to treat. Several single nucleotide polymorphisms and point mutations have been demonstrated in the ribosomal RNA methylase gene erm(39) related to clarithromycin resistance and in the rrl gene related to linezolid resistance.13 Due to increasing inducible resistance to common classes of antibiotics, such as macrolides and linezolid, treatment of M fortuitum requires multidrug regimens.13,14 Drug susceptibility testing also may be required, as M fortuitum has shown low resistance to tigecycline, tetracycline, cefmetazole, imipenem, and aminoglycosides (eg, amikacin, tobramycin, neomycin, gentamycin). Surgery is an important adjunctive tool in treating M fortuitum infections; patients with a single lesion are more likely to undergo surgical treatment alone or in combination with antibiotic therapy.15 More recently, antimicrobial photodynamic therapy has been explored as an alternative to eliminate NTM, including M fortuitum.16

The differential diagnosis for skin lesions manifesting with draining fistulae and sinus tracts includes conditions with infectious (cellulitis and chromomycosis) and inflammatory (pyoderma gangrenosum [PG] and hidradenitis suppurativa [HS]) causes.

Cellulitis is a common infection of the skin and subcutaneous tissue that predominantly is caused by gram-positive organisms such as β-hemolytic streptococci.17 Clinical manifestations include acute skin erythema, swelling, tenderness, and warmth. The legs are the most common sites of infection, but any area of the skin can be involved.17 Cellulitis comprises 10% of all infectious disease hospitalizations and up to 11% of all dermatologic admissions.18,19 It frequently is misdiagnosed, perhaps due to the lack of a reliable confirmatory laboratory test or imaging study, in addition to the plethora of diseases that mimic cellulitis, such as stasis dermatitis, lipodermatosclerosis, contact dermatitis, lymphedema, eosinophilic cellulitis, and papular urticaria.20,21 The consequences of misdiagnosis include but are not limited to unnecessary hospitalizations, inappropriate antibiotic use, and delayed management of the disease; thus, there is an urgent need for a reliable standard test to confirm the diagnosis, especially among nonspecialist physicians. 20 Most patients with uncomplicated cellulitis can be treated with empiric oral antibiotics that target β-hemolytic streptococci (ie, penicillin V potassium, amoxicillin).17 Methicillin-resistant Staphylococcus aureus coverage generally is unnecessary for nonpurulent cellulitis, but clinicians can consider adding amoxicillin-clavulanate, dicloxacillin, and cephalexin to the regimen. For purulent cellulitis, incision and drainage should be performed. In severe cases that manifest with sepsis, altered mental status, or hemodynamic instability, inpatient management is required.17

Chromomycosis (also known as chromoblastomycosis) is a chronic, indolent, granulomatous, suppurative mycosis of the skin and subcutaneous tissue22 that is caused by traumatic inoculation of various fungi of the order Chaetothyriales and family Herpotrichiellaceae, which are present in soil, plants, and decomposing wood. Chromomycosis is prevalent in tropical and subtropical regions.23,24 Clinically, it manifests as oligosymptomatic or asymptomatic lesions around an infection site that can manifest as papules with centrifugal growth evolving into nodular, verrucous, plaque, tumoral, or atrophic forms.22 Diagnosis is made with direct microscopy using potassium hydroxide, which reveals muriform bodies. Fungal culture in Sabouraud agar also can be used to isolate the causative pathogen.22 Unfortunately, chromomycosis is difficult to treat, with low cure rates and high relapse rates. Antifungal agents combined with surgery, cryotherapy, or thermotherapy often are used, with cure rates ranging from 15% to 80%.22,25

Pyoderma gangrenosum is a reactive noninfectious inflammatory dermatosis associated with inflammatory bowel disease and rheumatoid arthritis. The exact etiology is not clearly understood, but it generally is considered an autoinflammatory disorder.26 The most common form—classical PG—occurs in approximately 85% of cases and manifests as a painful erythematous lesion that progresses to a blistered or necrotic ulcer. It primarily affects the lower legs but can occur in other body sites.27 The diagnosis is based on clinical symptoms after excluding other similar conditions; histopathology of biopsied wound tissues often are required for confirmation. Treatment of PG starts with fast-acting immunosuppressive drugs (corticosteroids and/or cyclosporine) followed by slowacting immunosuppressive drugs (biologics).26

Hidradenitis suppurativa is a chronic recurrent disease of the hair follicle unit that develops after puberty.28 Clinically, HS manifests with painful nodules, abscesses, chronically draining fistulas, and scarring in areas of the body rich in apocrine glands.29,30 Treatment of HS is challenging due to its diverse clinical manifestations and unclear etiology. Topical therapy, systemic treatments, biologic agents, surgery, and light therapy have shown variable results.28,31

References
  1. Franco-Paredes C, Marcos LA, Henao-Martínez AF, et al. Cutaneous mycobacterial infections. Clin Microbiol Rev. 2018;32: E00069-18. doi:10.1128/CMR.00069-18
  2. Brown TH. The rapidly growing mycobacteria—MycobacteriuM fortuitum and Mycobacterium chelonae. Infect Control. 1985;6:283-238. doi:10.1017/s0195941700061762
  3. Hooper J; Beltrami EJ; Santoro F; et al. Remember the fite: a case of cutaneous MycobacteriuM fortuitum infection. Am J Dermatopathol. 2023;45:214-215. doi:10.1097/DAD.0000000000002336
  4. Franco-Paredes C, Chastain DB, Allen L, et al. Overview of cutaneous mycobacterial infections. Curr Trop Med Rep. 2018;5:228-232. doi:10.1007/s40475-018-0161-7
  5. Gonzalez-Santiago TM, Drage LA. Nontuberculous mycobacteria: skin and soft tissue infections. Dermatol Clin. 2015;33:563-77. doi:10.1016/j.det.2015.03.017
  6. Peixoto ADS, Montenegro LML, Lima AS, et al. Identification of nontuberculous mycobacteria species by multiplex real-time PCR with high-resolution melting. Rev Soc Bras Med Trop. 2020;53:E20200211. doi:10.1590/0037-8682-0211-2020
  7. Park J, Kwak N, Chae JC, et al. A two-step real-time PCR method to identify Mycobacterium tuberculosis infections and six dominant nontuberculous mycobacterial infections from clinical specimens. Microbiol Spectr. 2023:E0160623. doi:10.1128/spectrum.01606-23
  8. Fowler J, Mahlen SD. Localized cutaneous infections in immunocompetent individuals due to rapidly growing mycobacteria. Arch Pathol Lab Med. 2014;138:1106-1109. doi:10.5858/arpa.2012-0203-RS
  9. Gardini G, Gregori N, Matteelli A, et al. Mycobacterial skin infection. Curr Opin Infect Dis. 2022;35:79-87. doi:10.1097/QCO.0000000000000820
  10. Wang SH, Pancholi P. Mycobacterial skin and soft tissue infection. Curr Infect Dis Rep. 2014;16:438. doi:10.1007/s11908-014-0438-5
  11. Griffith DE, Aksamit T, Brown-Elliott BA, et al; ATS Mycobacterial Diseases Subcommittee; American Thoracic Society; Infectious Disease Society of America. An official ATS/IDSA statement: diagnosis, treatment, and prevention of nontuberculous mycobacterial diseases. Am J Respir Crit Care Med. 2007;175:367-416. doi:10.1164/rccm.200604-571ST
  12. Mougari F, Guglielmetti L, Raskine L, et al. Infections caused by Mycobacterium abscessus: epidemiology, diagnostic tools and treatment. Expert Rev Anti Infect Ther. 2016;14:1139-1154. doi:10.1080/14787210.201 6.1238304
  13. Tu HZ, Lee HS, Chen YS, et al. High rates of antimicrobial resistance in rapidly growing mycobacterial infections in Taiwan. Pathogens. 2022;11:969. doi:10.3390/pathogens11090969
  14. Hashemzadeh M, Zadegan Dezfuli AA, Khosravi AD, et al. F requency of mutations in erm(39) related to clarithromycin resistance and in rrl related to linezolid resistance in clinical isolates of MycobacteriuM fortuitum in Iran. Acta Microbiol Immunol Hung. 2023;70:167-176. doi:10.1556/030.2023.02020
  15. Uslan DZ, Kowalski TJ, Wengenack NL, et al. Skin and soft tissue infections due to rapidly growing mycobacteria: comparison of clinical features, treatment, and susceptibility. Arch Dermatol. 2006;142:1287-1292. doi:10.1001/archderm.142.10.1287
  16. Miretti M, Juri L, Peralta A, et al. Photoinactivation of non-tuberculous mycobacteria using Zn-phthalocyanine loaded into liposomes. Tuberculosis (Edinb). 2022;136:102247. doi:10.1016/j.tube.2022.102247
  17. Bystritsky RJ. Cellulitis. Infect Dis Clin North Am. 2021;35:49-60. doi:10.1016/j.idc.2020.10.002
  18. Christensen K, Holman R, Steiner C, et al. Infectious disease hospitalizations in the United States. Clin Infect Dis. 2009;49:1025-1035. doi:10.1086/605562
  19. Yang JJ, Maloney NJ, Bach DQ, et al. Dermatology in the emergency department: prescriptions, rates of inpatient admission, and predictors of high utilization in the United States from 1996 to 2012. J Am Acad Dermatol. 2021;84:1480-1483. doi:10.1016/J.JAAD.2020.07.055
  20. Cutler TS, Jannat-Khah DP, Kam B, et al. Prevalence of misdiagnosis of cellulitis: a systematic review and meta-analysis. J Hosp Med. 2023;18:254-261. doi:10.1002/jhm.12977
  21. Keller EC, Tomecki KJ, Alraies MC. Distinguishing cellulitis from its mimics. Cleve Clin J Med. 2012;79:547-52. doi:10.3949/ccjm.79a.11121
  22. Brito AC, Bittencourt MJS. Chromoblastomycosis: an etiological, epidemiological, clinical, diagnostic, and treatment update. An Bras Dermatol. 2018;93:495-506. doi:10.1590/abd1806-4841.20187321
  23. McGinnis MR. Chromoblastomycosis and phaeohyphomycosis: new concepts, diagnosis, and mycology. J Am Acad Dermatol. 1983;8:1-16.
  24. Rubin HA, Bruce S, Rosen T, et al. Evidence for percutaneous inoculation as the mode of transmission for chromoblastomycosis. J Am Acad Dermatol. 1991;25:951-954.
  25. Bonifaz A, Paredes-Solís V, Saúl A. Treating chromoblastomycosis with systemic antifungals. Expert Opin Pharmacother. 2004;5:247-254.
  26. Maverakis E, Marzano AV, Le ST, et al. Pyoderma gangrenosum. Nat Rev Dis Primers. 2020;6:81. doi:10.1038/s41572-020-0213-x
  27. George C, Deroide F, Rustin M. Pyoderma gangrenosum—a guide to diagnosis and management. Clin Med (Lond). 2019;19:224-228. doi:10.7861/clinmedicine.19-3-224
  28. Narla S, Lyons AB, Hamzavi IH. The most recent advances in understanding and managing hidradenitis suppurativa. F1000Res. 2020;9:F1000 Faculty Rev-1049. doi:10.12688/f1000research.26083.1
  29. Garg A, Lavian J, Lin G, et al. Incidence of hidradenitis suppurativa in the United States: a sex- and age-adjusted population analysis. J Am Acad Dermatol. 2017;77:118-122. doi:10.1016/j.jaad.2017.02.005
  30. Daxhelet M, Suppa M, White J, et al. Proposed definitions of typical lesions in hidradenitis suppurativa. Dermatology. 2020;236:431-438. doi:10.1159/000507348
  31. Amat-Samaranch V, Agut-Busquet E, Vilarrasa E, et al. New perspectives on the treatment of hidradenitis suppurativa. Ther Adv Chronic Dis. 2021;12:20406223211055920. doi:10.1177/20406223211055920
References
  1. Franco-Paredes C, Marcos LA, Henao-Martínez AF, et al. Cutaneous mycobacterial infections. Clin Microbiol Rev. 2018;32: E00069-18. doi:10.1128/CMR.00069-18
  2. Brown TH. The rapidly growing mycobacteria—MycobacteriuM fortuitum and Mycobacterium chelonae. Infect Control. 1985;6:283-238. doi:10.1017/s0195941700061762
  3. Hooper J; Beltrami EJ; Santoro F; et al. Remember the fite: a case of cutaneous MycobacteriuM fortuitum infection. Am J Dermatopathol. 2023;45:214-215. doi:10.1097/DAD.0000000000002336
  4. Franco-Paredes C, Chastain DB, Allen L, et al. Overview of cutaneous mycobacterial infections. Curr Trop Med Rep. 2018;5:228-232. doi:10.1007/s40475-018-0161-7
  5. Gonzalez-Santiago TM, Drage LA. Nontuberculous mycobacteria: skin and soft tissue infections. Dermatol Clin. 2015;33:563-77. doi:10.1016/j.det.2015.03.017
  6. Peixoto ADS, Montenegro LML, Lima AS, et al. Identification of nontuberculous mycobacteria species by multiplex real-time PCR with high-resolution melting. Rev Soc Bras Med Trop. 2020;53:E20200211. doi:10.1590/0037-8682-0211-2020
  7. Park J, Kwak N, Chae JC, et al. A two-step real-time PCR method to identify Mycobacterium tuberculosis infections and six dominant nontuberculous mycobacterial infections from clinical specimens. Microbiol Spectr. 2023:E0160623. doi:10.1128/spectrum.01606-23
  8. Fowler J, Mahlen SD. Localized cutaneous infections in immunocompetent individuals due to rapidly growing mycobacteria. Arch Pathol Lab Med. 2014;138:1106-1109. doi:10.5858/arpa.2012-0203-RS
  9. Gardini G, Gregori N, Matteelli A, et al. Mycobacterial skin infection. Curr Opin Infect Dis. 2022;35:79-87. doi:10.1097/QCO.0000000000000820
  10. Wang SH, Pancholi P. Mycobacterial skin and soft tissue infection. Curr Infect Dis Rep. 2014;16:438. doi:10.1007/s11908-014-0438-5
  11. Griffith DE, Aksamit T, Brown-Elliott BA, et al; ATS Mycobacterial Diseases Subcommittee; American Thoracic Society; Infectious Disease Society of America. An official ATS/IDSA statement: diagnosis, treatment, and prevention of nontuberculous mycobacterial diseases. Am J Respir Crit Care Med. 2007;175:367-416. doi:10.1164/rccm.200604-571ST
  12. Mougari F, Guglielmetti L, Raskine L, et al. Infections caused by Mycobacterium abscessus: epidemiology, diagnostic tools and treatment. Expert Rev Anti Infect Ther. 2016;14:1139-1154. doi:10.1080/14787210.201 6.1238304
  13. Tu HZ, Lee HS, Chen YS, et al. High rates of antimicrobial resistance in rapidly growing mycobacterial infections in Taiwan. Pathogens. 2022;11:969. doi:10.3390/pathogens11090969
  14. Hashemzadeh M, Zadegan Dezfuli AA, Khosravi AD, et al. F requency of mutations in erm(39) related to clarithromycin resistance and in rrl related to linezolid resistance in clinical isolates of MycobacteriuM fortuitum in Iran. Acta Microbiol Immunol Hung. 2023;70:167-176. doi:10.1556/030.2023.02020
  15. Uslan DZ, Kowalski TJ, Wengenack NL, et al. Skin and soft tissue infections due to rapidly growing mycobacteria: comparison of clinical features, treatment, and susceptibility. Arch Dermatol. 2006;142:1287-1292. doi:10.1001/archderm.142.10.1287
  16. Miretti M, Juri L, Peralta A, et al. Photoinactivation of non-tuberculous mycobacteria using Zn-phthalocyanine loaded into liposomes. Tuberculosis (Edinb). 2022;136:102247. doi:10.1016/j.tube.2022.102247
  17. Bystritsky RJ. Cellulitis. Infect Dis Clin North Am. 2021;35:49-60. doi:10.1016/j.idc.2020.10.002
  18. Christensen K, Holman R, Steiner C, et al. Infectious disease hospitalizations in the United States. Clin Infect Dis. 2009;49:1025-1035. doi:10.1086/605562
  19. Yang JJ, Maloney NJ, Bach DQ, et al. Dermatology in the emergency department: prescriptions, rates of inpatient admission, and predictors of high utilization in the United States from 1996 to 2012. J Am Acad Dermatol. 2021;84:1480-1483. doi:10.1016/J.JAAD.2020.07.055
  20. Cutler TS, Jannat-Khah DP, Kam B, et al. Prevalence of misdiagnosis of cellulitis: a systematic review and meta-analysis. J Hosp Med. 2023;18:254-261. doi:10.1002/jhm.12977
  21. Keller EC, Tomecki KJ, Alraies MC. Distinguishing cellulitis from its mimics. Cleve Clin J Med. 2012;79:547-52. doi:10.3949/ccjm.79a.11121
  22. Brito AC, Bittencourt MJS. Chromoblastomycosis: an etiological, epidemiological, clinical, diagnostic, and treatment update. An Bras Dermatol. 2018;93:495-506. doi:10.1590/abd1806-4841.20187321
  23. McGinnis MR. Chromoblastomycosis and phaeohyphomycosis: new concepts, diagnosis, and mycology. J Am Acad Dermatol. 1983;8:1-16.
  24. Rubin HA, Bruce S, Rosen T, et al. Evidence for percutaneous inoculation as the mode of transmission for chromoblastomycosis. J Am Acad Dermatol. 1991;25:951-954.
  25. Bonifaz A, Paredes-Solís V, Saúl A. Treating chromoblastomycosis with systemic antifungals. Expert Opin Pharmacother. 2004;5:247-254.
  26. Maverakis E, Marzano AV, Le ST, et al. Pyoderma gangrenosum. Nat Rev Dis Primers. 2020;6:81. doi:10.1038/s41572-020-0213-x
  27. George C, Deroide F, Rustin M. Pyoderma gangrenosum—a guide to diagnosis and management. Clin Med (Lond). 2019;19:224-228. doi:10.7861/clinmedicine.19-3-224
  28. Narla S, Lyons AB, Hamzavi IH. The most recent advances in understanding and managing hidradenitis suppurativa. F1000Res. 2020;9:F1000 Faculty Rev-1049. doi:10.12688/f1000research.26083.1
  29. Garg A, Lavian J, Lin G, et al. Incidence of hidradenitis suppurativa in the United States: a sex- and age-adjusted population analysis. J Am Acad Dermatol. 2017;77:118-122. doi:10.1016/j.jaad.2017.02.005
  30. Daxhelet M, Suppa M, White J, et al. Proposed definitions of typical lesions in hidradenitis suppurativa. Dermatology. 2020;236:431-438. doi:10.1159/000507348
  31. Amat-Samaranch V, Agut-Busquet E, Vilarrasa E, et al. New perspectives on the treatment of hidradenitis suppurativa. Ther Adv Chronic Dis. 2021;12:20406223211055920. doi:10.1177/20406223211055920
Issue
Cutis - 114(3)
Issue
Cutis - 114(3)
Page Number
71,77-78
Page Number
71,77-78
Publications
Publications
Topics
Article Type
Display Headline
Multiple Draining Sinus Tracts on the Thigh
Display Headline
Multiple Draining Sinus Tracts on the Thigh
Sections
Questionnaire Body

A 40-year-old woman presented with multiple draining sinus tracts on the right thigh following an injury sustained weeks earlier while mowing wet grass.

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Wed, 09/04/2024 - 11:00
Un-Gate On Date
Wed, 09/04/2024 - 11:00
Use ProPublica
CFC Schedule Remove Status
Wed, 09/04/2024 - 11:00
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

Erythema Nodosum Triggered by a Bite From a Copperhead Snake

Article Type
Changed
Tue, 09/03/2024 - 15:55
Display Headline
Erythema Nodosum Triggered by a Bite From a Copperhead Snake

The clinical manifestations of snakebites vary based on the species of snake, bite location, and amount and strength of the venom injected. Locally acting toxins in snake venom predominantly consist of enzymes, such as phospholipase A2, that cause local tissue destruction and can result in pain, swelling, blistering, ecchymosis, and tissue necrosis at the site of the bite within hours to days after the bite.1 Systemically acting toxins can target a wide variety of tissues and cause severe systemic complications including paralysis, rhabdomyolysis secondary to muscle damage, coagulopathy, sepsis, and cardiorespiratory failure.2

Although pain and swelling following snakebites typically resolve by 1 month after envenomation, copperhead snakes—a type of pit viper—may cause residual symptoms of pain and swelling lasting for a year or more.3 Additional cutaneous manifestations of copperhead snakebites include wound infections at the bite site, such as cellulitis and necrotizing fasciitis. More devastating complications that have been described following snake envenomation include tissue injury of an entire extremity and development of compartment syndrome, which requires urgent fasciotomy to prevent potential loss of the affected limb.4

Physicians should be aware of the potential complications of snakebites to properly manage and counsel their patients. We describe a 42-year-old woman with tender, erythematous, subcutaneous nodules persisting for 4 months following a copperhead snakebite. A biopsy confirmed the diagnosis of snakebite-associated erythema nodosum (EN).

Case Report

A 42-year-old woman presented to our clinic with progressive tender, pruritic, deep-seated, erythematous nodules in multiple locations on the legs after sustaining a bite by a copperhead snake on the left foot 4 months prior. The lesions tended to fluctuate in intensity. In the days following the bite, she initially developed painful red bumps on the left foot just proximal to the bite site with associated pain and swelling extending up to just below the left knee. She reported no other notable symptoms such as fever, arthralgia, fatigue, or gastrointestinal tract symptoms. Physical examination revealed bilateral pitting edema, which was worse in the left leg, along with multiple deep, palpable, tender subcutaneous nodules with erythematous surface change (Figure 1).

FIGURE 1. Multiple palpable, erythematous, subcutaneous nodules scattered on the right leg in a patient with erythema nodosum following a bite from a copperhead snake.

Workup performed by an outside provider over the previous month included 2 venous duplex ultrasounds of the left leg, which showed no signs of deep vein thrombosis. Additionally, the patient underwent lateral and anteroposterior radiographs of the left foot, tibia, and fibula, which showed no evidence of fracture.

Given the morphology and distribution of the lesions (Figure 2), EN was strongly favored as the cause of the symptoms, and a biopsy confirmed the diagnosis. All immunohistochemical stains including auramine-­rhodamine for acid-fast bacilli, Grocott-Gomori methenamine silver for fungal organisms, and Brown and Brenn were negative. Given the waxing and waning course of the lesions, which suggested an active neutrophilic rather than purely chronic granulomatous phase of EN, the patient was treated with colchicine 0.6 mg twice daily for 1 month.

FIGURE 2. Punch biopsy site of an isolated erythematous plaque on the left upper thigh in a patient with erythema nodosum following a copperhead snake bite.

 

 

Causes of EN and Clinical Manifestations

Erythema nodosum is a common form of septal panniculitis that can be precipitated by inflammatory conditions, infection, or medications (commonly oral contraceptive pills) but often is idiopathic.5 The acute phase is neutrophilic, with evolution over time to a granulomatous phase. Common etiologies include sarcoidosis; inflammatory bowel disease; and bacterial or fungal infections such as Streptococcus (especially common in children), histoplasmosis, and coccidioidomycosis. The patient was otherwise healthy and was not taking any medications that are known triggers of EN. A PubMed search of articles indexed for MEDLINE in the English-language literature using the terms copperhead snake bite, erythema nodosum snake, and copperhead snake erythema nodosum revealed no reports of EN following a bite from a copperhead snake; however, in one case, an adder bite led to erysipelas, likely due to disturbed blood and lymphatic flow, which then triggered EN.6 Additionally, EN has been reported as a delayed reaction to jellyfish stings.7

Clinical features of EN include the development of tender, erythematous, subcutaneous nodules and plaques most frequently over the pretibial region. Lesions typically evolve from raised, deep-seated nodules into flat indurated plaques over a span of weeks. Occasionally, there is a slight prodromal phase marked by nonspecific symptoms such as fever and arthralgia lasting for 3 to 6 days. Erythema nodosum typically results in spontaneous resolution after 4 to 8 weeks, and management involves treatment of any underlying condition with symptomatic care. Interestingly, our patient experienced persistent symptoms over the course of 4 months, with development of new nodular lesions throughout this time period. The most frequently used drugs for the management of symptomatic EN include nonsteroidal anti-inflammatory drugs, colchicine, and potassium iodide.8 A characteristic histologic finding of the granulomatous phase is the Miescher radial granuloma, which is a septal collection of histiocytes surrounding a cleft.9

Snakebite Reactions

Snakebites can result in a wide range of local and systemic manifestations, as snake venom may contain 20 or more toxins.10 Local complications of pit viper bites include pain, swelling, and fang marks; when examining fang marks, the presence of 2 distinct puncture wounds often indicates envenomation with a poisonous snake, whereas nonvenomous snakebites often result in smaller puncture wounds arranged in an arc. Following bites, pain can develop immediately and spread proximally up the affected limb, which occurred in our patient in the days following the bite. Intense local reactions can occur, as bites often result in intense edema of the affected limb spreading to the trunk in the days to weeks after the bite, occasionally accompanied by regional lymphadenopathy. Some bites can result in local necrosis and secondary bacterial infection caused by organisms in the oral cavity of the culprit snake.

Although they were not present in our patient, snakebites can result in a wide range of systemic toxicities ranging from clotting defects and hemolysis to neurotoxicity, myotoxicity, and nephrotoxicity.10 In severe cases, snake venom can result in disseminated intravascular coagulation, sepsis, and cardiorespiratory collapse.

The eastern copperhead (Agkistrodon contortrix) is a species of venomous snake that is endemic to eastern North America. Copperheads are members of the subfamily Crotalinae in the family Viperidae.11 Reported reactions to copperhead bites include cellulitis, necrotizing fasciitis, compartment syndrome, and tissue necrosis of an entire affected extremity.12,13 Our patient displayed no systemic symptoms to suggest envenomation.

Management of Snakebites

Treatment of snakebites varies based on the constellation and severity of symptoms as well as how recently the envenomation occurred. In urgent cases, antivenom may be administered to prevent further toxicity. In cases of progressive compartment syndrome, emergent surgical procedures such as fasciotomy or amputation are required to prevent further complications. When a superimposed bacterial infection is suspected, broad-spectrum antibiotics are required. Because our patient presented 4 months following the initial bite with isolated cutaneous manifestations, she was treated symptomatically with colchicine for EN.1,2

Final Thoughts

Our patient presented with EN following a bite from a copperhead snake. Physicians should be aware of possible etiologies of EN to evaluate patients who present with new-onset tender subcutaneous nodules. Additionally, physicians should be aware of venomous snakes endemic to their region and also understand the various complications that can result following a snakebite, with the potential for lingering cutaneous manifestations weeks to months following the initial bite.

 

References
  1. Warrell DA. Snake bite. Lancet. 2010;375:77-88. doi:10.1016/S0140-6736(09)61754-2
  2. White J. Overview of venomous snakes of the world. In: Dart RC, eds. Medical Toxicology. 3rd ed. Lippincott, Williams, & Wilkins; 2004:1543
  3. Spiller HA, Bosse GM. Prospective study of morbidity associated with snakebite envenomation. J Toxicol Clin Toxicol. 2003;41:125-130. doi:10.1081/clt-120019127
  4. Scharman EJ, Noffsinger VD. Copperhead snakebites: clinical severity of local effects. Ann Emerg Med. 2001;38:55-61. doi:10.1067/mem.2001.116148
  5. Hafsi W, Badri T. Erythema nodosum. In: StatPearls. StatPearls Publishing; November 28, 2022. Accessed July 22, 2024. https://www.ncbi.nlm.nih.gov/books/NBK470369/
  6. Nowowiejska J, Baran A, Flisiak I. Rare coexistence of unilateral erythema nodosum with erysipelas in the area of previous adder bite. Przegl Epidemiol. 2020;74:355-361. doi:10.32394/pe.74.28
  7. Auerbach PS, Hays JT. Erythema nodosum following a jellyfish sting. J Emerg Med. 1987;5:487-491. doi:10.1016/0736-4679(87)90211-3
  8. Gilchrist H, Patterson JW. Erythema nodosum and erythema induratum (nodular vasculitis): diagnosis and management. Dermatol Ther. 2010;23:320-327. doi:10.1111/j.1529-8019.2010.01332.x
  9. Sánchez Yus E, Sanz Vico MD, de Diego V. Miescher’s radial granuloma. a characteristic marker of erythema nodosum. Am J Dermatopathol. 1989;11:434-442. doi:10.1097/00000372-198910000-00005
  10. Mehta SR, Sashindran VK. Clinical features and management of snake bite. Med J Armed Forces India. 2002;58:247-249. doi:10.1016/S0377-1237(02)80140-X
  11. Brys AK, Gandolfi BM, Levinson H, et al. Copperhead envenomation resulting in a rare case of hand compartment syndrome and subsequent fasciotomy. Plast Reconstr Surg Glob Open. 2015;3:E396. doi:10.1097/GOX.0000000000000367
  12. Clark RF, Selden BS, Furbee B. The incidence of wound infection following crotalid envenomation. J Emerg Med. 1993;11:583-586. doi:10.1016/0736-4679(93)90313-v
  13. Buchanan JT, Thurman J. Crotalidae envenomation. In: StatPearls. StatPearls Publishing; October 3, 2022. Accessed July 22, 2024. https://www.ncbi.nlm.nih.gov/books/NBK551615/
Article PDF
Author and Disclosure Information

Dr. Newcomer is from Mayo Clinic, Rochester, Minnesota. Drs. Jansen and Elston are from the Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston.

The authors report no conflict of interest.

Correspondence: Jack Newcomer, MD, 200 1st St NW, Rochester, MN 55901 (jacknewcomer23@gmail.com).

Cutis. 2024 August;114(2):51-53. doi:10.12788/cutis.1074

Issue
Cutis - 114(2)
Publications
Topics
Page Number
51-53
Sections
Author and Disclosure Information

Dr. Newcomer is from Mayo Clinic, Rochester, Minnesota. Drs. Jansen and Elston are from the Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston.

The authors report no conflict of interest.

Correspondence: Jack Newcomer, MD, 200 1st St NW, Rochester, MN 55901 (jacknewcomer23@gmail.com).

Cutis. 2024 August;114(2):51-53. doi:10.12788/cutis.1074

Author and Disclosure Information

Dr. Newcomer is from Mayo Clinic, Rochester, Minnesota. Drs. Jansen and Elston are from the Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston.

The authors report no conflict of interest.

Correspondence: Jack Newcomer, MD, 200 1st St NW, Rochester, MN 55901 (jacknewcomer23@gmail.com).

Cutis. 2024 August;114(2):51-53. doi:10.12788/cutis.1074

Article PDF
Article PDF

The clinical manifestations of snakebites vary based on the species of snake, bite location, and amount and strength of the venom injected. Locally acting toxins in snake venom predominantly consist of enzymes, such as phospholipase A2, that cause local tissue destruction and can result in pain, swelling, blistering, ecchymosis, and tissue necrosis at the site of the bite within hours to days after the bite.1 Systemically acting toxins can target a wide variety of tissues and cause severe systemic complications including paralysis, rhabdomyolysis secondary to muscle damage, coagulopathy, sepsis, and cardiorespiratory failure.2

Although pain and swelling following snakebites typically resolve by 1 month after envenomation, copperhead snakes—a type of pit viper—may cause residual symptoms of pain and swelling lasting for a year or more.3 Additional cutaneous manifestations of copperhead snakebites include wound infections at the bite site, such as cellulitis and necrotizing fasciitis. More devastating complications that have been described following snake envenomation include tissue injury of an entire extremity and development of compartment syndrome, which requires urgent fasciotomy to prevent potential loss of the affected limb.4

Physicians should be aware of the potential complications of snakebites to properly manage and counsel their patients. We describe a 42-year-old woman with tender, erythematous, subcutaneous nodules persisting for 4 months following a copperhead snakebite. A biopsy confirmed the diagnosis of snakebite-associated erythema nodosum (EN).

Case Report

A 42-year-old woman presented to our clinic with progressive tender, pruritic, deep-seated, erythematous nodules in multiple locations on the legs after sustaining a bite by a copperhead snake on the left foot 4 months prior. The lesions tended to fluctuate in intensity. In the days following the bite, she initially developed painful red bumps on the left foot just proximal to the bite site with associated pain and swelling extending up to just below the left knee. She reported no other notable symptoms such as fever, arthralgia, fatigue, or gastrointestinal tract symptoms. Physical examination revealed bilateral pitting edema, which was worse in the left leg, along with multiple deep, palpable, tender subcutaneous nodules with erythematous surface change (Figure 1).

FIGURE 1. Multiple palpable, erythematous, subcutaneous nodules scattered on the right leg in a patient with erythema nodosum following a bite from a copperhead snake.

Workup performed by an outside provider over the previous month included 2 venous duplex ultrasounds of the left leg, which showed no signs of deep vein thrombosis. Additionally, the patient underwent lateral and anteroposterior radiographs of the left foot, tibia, and fibula, which showed no evidence of fracture.

Given the morphology and distribution of the lesions (Figure 2), EN was strongly favored as the cause of the symptoms, and a biopsy confirmed the diagnosis. All immunohistochemical stains including auramine-­rhodamine for acid-fast bacilli, Grocott-Gomori methenamine silver for fungal organisms, and Brown and Brenn were negative. Given the waxing and waning course of the lesions, which suggested an active neutrophilic rather than purely chronic granulomatous phase of EN, the patient was treated with colchicine 0.6 mg twice daily for 1 month.

FIGURE 2. Punch biopsy site of an isolated erythematous plaque on the left upper thigh in a patient with erythema nodosum following a copperhead snake bite.

 

 

Causes of EN and Clinical Manifestations

Erythema nodosum is a common form of septal panniculitis that can be precipitated by inflammatory conditions, infection, or medications (commonly oral contraceptive pills) but often is idiopathic.5 The acute phase is neutrophilic, with evolution over time to a granulomatous phase. Common etiologies include sarcoidosis; inflammatory bowel disease; and bacterial or fungal infections such as Streptococcus (especially common in children), histoplasmosis, and coccidioidomycosis. The patient was otherwise healthy and was not taking any medications that are known triggers of EN. A PubMed search of articles indexed for MEDLINE in the English-language literature using the terms copperhead snake bite, erythema nodosum snake, and copperhead snake erythema nodosum revealed no reports of EN following a bite from a copperhead snake; however, in one case, an adder bite led to erysipelas, likely due to disturbed blood and lymphatic flow, which then triggered EN.6 Additionally, EN has been reported as a delayed reaction to jellyfish stings.7

Clinical features of EN include the development of tender, erythematous, subcutaneous nodules and plaques most frequently over the pretibial region. Lesions typically evolve from raised, deep-seated nodules into flat indurated plaques over a span of weeks. Occasionally, there is a slight prodromal phase marked by nonspecific symptoms such as fever and arthralgia lasting for 3 to 6 days. Erythema nodosum typically results in spontaneous resolution after 4 to 8 weeks, and management involves treatment of any underlying condition with symptomatic care. Interestingly, our patient experienced persistent symptoms over the course of 4 months, with development of new nodular lesions throughout this time period. The most frequently used drugs for the management of symptomatic EN include nonsteroidal anti-inflammatory drugs, colchicine, and potassium iodide.8 A characteristic histologic finding of the granulomatous phase is the Miescher radial granuloma, which is a septal collection of histiocytes surrounding a cleft.9

Snakebite Reactions

Snakebites can result in a wide range of local and systemic manifestations, as snake venom may contain 20 or more toxins.10 Local complications of pit viper bites include pain, swelling, and fang marks; when examining fang marks, the presence of 2 distinct puncture wounds often indicates envenomation with a poisonous snake, whereas nonvenomous snakebites often result in smaller puncture wounds arranged in an arc. Following bites, pain can develop immediately and spread proximally up the affected limb, which occurred in our patient in the days following the bite. Intense local reactions can occur, as bites often result in intense edema of the affected limb spreading to the trunk in the days to weeks after the bite, occasionally accompanied by regional lymphadenopathy. Some bites can result in local necrosis and secondary bacterial infection caused by organisms in the oral cavity of the culprit snake.

Although they were not present in our patient, snakebites can result in a wide range of systemic toxicities ranging from clotting defects and hemolysis to neurotoxicity, myotoxicity, and nephrotoxicity.10 In severe cases, snake venom can result in disseminated intravascular coagulation, sepsis, and cardiorespiratory collapse.

The eastern copperhead (Agkistrodon contortrix) is a species of venomous snake that is endemic to eastern North America. Copperheads are members of the subfamily Crotalinae in the family Viperidae.11 Reported reactions to copperhead bites include cellulitis, necrotizing fasciitis, compartment syndrome, and tissue necrosis of an entire affected extremity.12,13 Our patient displayed no systemic symptoms to suggest envenomation.

Management of Snakebites

Treatment of snakebites varies based on the constellation and severity of symptoms as well as how recently the envenomation occurred. In urgent cases, antivenom may be administered to prevent further toxicity. In cases of progressive compartment syndrome, emergent surgical procedures such as fasciotomy or amputation are required to prevent further complications. When a superimposed bacterial infection is suspected, broad-spectrum antibiotics are required. Because our patient presented 4 months following the initial bite with isolated cutaneous manifestations, she was treated symptomatically with colchicine for EN.1,2

Final Thoughts

Our patient presented with EN following a bite from a copperhead snake. Physicians should be aware of possible etiologies of EN to evaluate patients who present with new-onset tender subcutaneous nodules. Additionally, physicians should be aware of venomous snakes endemic to their region and also understand the various complications that can result following a snakebite, with the potential for lingering cutaneous manifestations weeks to months following the initial bite.

 

The clinical manifestations of snakebites vary based on the species of snake, bite location, and amount and strength of the venom injected. Locally acting toxins in snake venom predominantly consist of enzymes, such as phospholipase A2, that cause local tissue destruction and can result in pain, swelling, blistering, ecchymosis, and tissue necrosis at the site of the bite within hours to days after the bite.1 Systemically acting toxins can target a wide variety of tissues and cause severe systemic complications including paralysis, rhabdomyolysis secondary to muscle damage, coagulopathy, sepsis, and cardiorespiratory failure.2

Although pain and swelling following snakebites typically resolve by 1 month after envenomation, copperhead snakes—a type of pit viper—may cause residual symptoms of pain and swelling lasting for a year or more.3 Additional cutaneous manifestations of copperhead snakebites include wound infections at the bite site, such as cellulitis and necrotizing fasciitis. More devastating complications that have been described following snake envenomation include tissue injury of an entire extremity and development of compartment syndrome, which requires urgent fasciotomy to prevent potential loss of the affected limb.4

Physicians should be aware of the potential complications of snakebites to properly manage and counsel their patients. We describe a 42-year-old woman with tender, erythematous, subcutaneous nodules persisting for 4 months following a copperhead snakebite. A biopsy confirmed the diagnosis of snakebite-associated erythema nodosum (EN).

Case Report

A 42-year-old woman presented to our clinic with progressive tender, pruritic, deep-seated, erythematous nodules in multiple locations on the legs after sustaining a bite by a copperhead snake on the left foot 4 months prior. The lesions tended to fluctuate in intensity. In the days following the bite, she initially developed painful red bumps on the left foot just proximal to the bite site with associated pain and swelling extending up to just below the left knee. She reported no other notable symptoms such as fever, arthralgia, fatigue, or gastrointestinal tract symptoms. Physical examination revealed bilateral pitting edema, which was worse in the left leg, along with multiple deep, palpable, tender subcutaneous nodules with erythematous surface change (Figure 1).

FIGURE 1. Multiple palpable, erythematous, subcutaneous nodules scattered on the right leg in a patient with erythema nodosum following a bite from a copperhead snake.

Workup performed by an outside provider over the previous month included 2 venous duplex ultrasounds of the left leg, which showed no signs of deep vein thrombosis. Additionally, the patient underwent lateral and anteroposterior radiographs of the left foot, tibia, and fibula, which showed no evidence of fracture.

Given the morphology and distribution of the lesions (Figure 2), EN was strongly favored as the cause of the symptoms, and a biopsy confirmed the diagnosis. All immunohistochemical stains including auramine-­rhodamine for acid-fast bacilli, Grocott-Gomori methenamine silver for fungal organisms, and Brown and Brenn were negative. Given the waxing and waning course of the lesions, which suggested an active neutrophilic rather than purely chronic granulomatous phase of EN, the patient was treated with colchicine 0.6 mg twice daily for 1 month.

FIGURE 2. Punch biopsy site of an isolated erythematous plaque on the left upper thigh in a patient with erythema nodosum following a copperhead snake bite.

 

 

Causes of EN and Clinical Manifestations

Erythema nodosum is a common form of septal panniculitis that can be precipitated by inflammatory conditions, infection, or medications (commonly oral contraceptive pills) but often is idiopathic.5 The acute phase is neutrophilic, with evolution over time to a granulomatous phase. Common etiologies include sarcoidosis; inflammatory bowel disease; and bacterial or fungal infections such as Streptococcus (especially common in children), histoplasmosis, and coccidioidomycosis. The patient was otherwise healthy and was not taking any medications that are known triggers of EN. A PubMed search of articles indexed for MEDLINE in the English-language literature using the terms copperhead snake bite, erythema nodosum snake, and copperhead snake erythema nodosum revealed no reports of EN following a bite from a copperhead snake; however, in one case, an adder bite led to erysipelas, likely due to disturbed blood and lymphatic flow, which then triggered EN.6 Additionally, EN has been reported as a delayed reaction to jellyfish stings.7

Clinical features of EN include the development of tender, erythematous, subcutaneous nodules and plaques most frequently over the pretibial region. Lesions typically evolve from raised, deep-seated nodules into flat indurated plaques over a span of weeks. Occasionally, there is a slight prodromal phase marked by nonspecific symptoms such as fever and arthralgia lasting for 3 to 6 days. Erythema nodosum typically results in spontaneous resolution after 4 to 8 weeks, and management involves treatment of any underlying condition with symptomatic care. Interestingly, our patient experienced persistent symptoms over the course of 4 months, with development of new nodular lesions throughout this time period. The most frequently used drugs for the management of symptomatic EN include nonsteroidal anti-inflammatory drugs, colchicine, and potassium iodide.8 A characteristic histologic finding of the granulomatous phase is the Miescher radial granuloma, which is a septal collection of histiocytes surrounding a cleft.9

Snakebite Reactions

Snakebites can result in a wide range of local and systemic manifestations, as snake venom may contain 20 or more toxins.10 Local complications of pit viper bites include pain, swelling, and fang marks; when examining fang marks, the presence of 2 distinct puncture wounds often indicates envenomation with a poisonous snake, whereas nonvenomous snakebites often result in smaller puncture wounds arranged in an arc. Following bites, pain can develop immediately and spread proximally up the affected limb, which occurred in our patient in the days following the bite. Intense local reactions can occur, as bites often result in intense edema of the affected limb spreading to the trunk in the days to weeks after the bite, occasionally accompanied by regional lymphadenopathy. Some bites can result in local necrosis and secondary bacterial infection caused by organisms in the oral cavity of the culprit snake.

Although they were not present in our patient, snakebites can result in a wide range of systemic toxicities ranging from clotting defects and hemolysis to neurotoxicity, myotoxicity, and nephrotoxicity.10 In severe cases, snake venom can result in disseminated intravascular coagulation, sepsis, and cardiorespiratory collapse.

The eastern copperhead (Agkistrodon contortrix) is a species of venomous snake that is endemic to eastern North America. Copperheads are members of the subfamily Crotalinae in the family Viperidae.11 Reported reactions to copperhead bites include cellulitis, necrotizing fasciitis, compartment syndrome, and tissue necrosis of an entire affected extremity.12,13 Our patient displayed no systemic symptoms to suggest envenomation.

Management of Snakebites

Treatment of snakebites varies based on the constellation and severity of symptoms as well as how recently the envenomation occurred. In urgent cases, antivenom may be administered to prevent further toxicity. In cases of progressive compartment syndrome, emergent surgical procedures such as fasciotomy or amputation are required to prevent further complications. When a superimposed bacterial infection is suspected, broad-spectrum antibiotics are required. Because our patient presented 4 months following the initial bite with isolated cutaneous manifestations, she was treated symptomatically with colchicine for EN.1,2

Final Thoughts

Our patient presented with EN following a bite from a copperhead snake. Physicians should be aware of possible etiologies of EN to evaluate patients who present with new-onset tender subcutaneous nodules. Additionally, physicians should be aware of venomous snakes endemic to their region and also understand the various complications that can result following a snakebite, with the potential for lingering cutaneous manifestations weeks to months following the initial bite.

 

References
  1. Warrell DA. Snake bite. Lancet. 2010;375:77-88. doi:10.1016/S0140-6736(09)61754-2
  2. White J. Overview of venomous snakes of the world. In: Dart RC, eds. Medical Toxicology. 3rd ed. Lippincott, Williams, & Wilkins; 2004:1543
  3. Spiller HA, Bosse GM. Prospective study of morbidity associated with snakebite envenomation. J Toxicol Clin Toxicol. 2003;41:125-130. doi:10.1081/clt-120019127
  4. Scharman EJ, Noffsinger VD. Copperhead snakebites: clinical severity of local effects. Ann Emerg Med. 2001;38:55-61. doi:10.1067/mem.2001.116148
  5. Hafsi W, Badri T. Erythema nodosum. In: StatPearls. StatPearls Publishing; November 28, 2022. Accessed July 22, 2024. https://www.ncbi.nlm.nih.gov/books/NBK470369/
  6. Nowowiejska J, Baran A, Flisiak I. Rare coexistence of unilateral erythema nodosum with erysipelas in the area of previous adder bite. Przegl Epidemiol. 2020;74:355-361. doi:10.32394/pe.74.28
  7. Auerbach PS, Hays JT. Erythema nodosum following a jellyfish sting. J Emerg Med. 1987;5:487-491. doi:10.1016/0736-4679(87)90211-3
  8. Gilchrist H, Patterson JW. Erythema nodosum and erythema induratum (nodular vasculitis): diagnosis and management. Dermatol Ther. 2010;23:320-327. doi:10.1111/j.1529-8019.2010.01332.x
  9. Sánchez Yus E, Sanz Vico MD, de Diego V. Miescher’s radial granuloma. a characteristic marker of erythema nodosum. Am J Dermatopathol. 1989;11:434-442. doi:10.1097/00000372-198910000-00005
  10. Mehta SR, Sashindran VK. Clinical features and management of snake bite. Med J Armed Forces India. 2002;58:247-249. doi:10.1016/S0377-1237(02)80140-X
  11. Brys AK, Gandolfi BM, Levinson H, et al. Copperhead envenomation resulting in a rare case of hand compartment syndrome and subsequent fasciotomy. Plast Reconstr Surg Glob Open. 2015;3:E396. doi:10.1097/GOX.0000000000000367
  12. Clark RF, Selden BS, Furbee B. The incidence of wound infection following crotalid envenomation. J Emerg Med. 1993;11:583-586. doi:10.1016/0736-4679(93)90313-v
  13. Buchanan JT, Thurman J. Crotalidae envenomation. In: StatPearls. StatPearls Publishing; October 3, 2022. Accessed July 22, 2024. https://www.ncbi.nlm.nih.gov/books/NBK551615/
References
  1. Warrell DA. Snake bite. Lancet. 2010;375:77-88. doi:10.1016/S0140-6736(09)61754-2
  2. White J. Overview of venomous snakes of the world. In: Dart RC, eds. Medical Toxicology. 3rd ed. Lippincott, Williams, & Wilkins; 2004:1543
  3. Spiller HA, Bosse GM. Prospective study of morbidity associated with snakebite envenomation. J Toxicol Clin Toxicol. 2003;41:125-130. doi:10.1081/clt-120019127
  4. Scharman EJ, Noffsinger VD. Copperhead snakebites: clinical severity of local effects. Ann Emerg Med. 2001;38:55-61. doi:10.1067/mem.2001.116148
  5. Hafsi W, Badri T. Erythema nodosum. In: StatPearls. StatPearls Publishing; November 28, 2022. Accessed July 22, 2024. https://www.ncbi.nlm.nih.gov/books/NBK470369/
  6. Nowowiejska J, Baran A, Flisiak I. Rare coexistence of unilateral erythema nodosum with erysipelas in the area of previous adder bite. Przegl Epidemiol. 2020;74:355-361. doi:10.32394/pe.74.28
  7. Auerbach PS, Hays JT. Erythema nodosum following a jellyfish sting. J Emerg Med. 1987;5:487-491. doi:10.1016/0736-4679(87)90211-3
  8. Gilchrist H, Patterson JW. Erythema nodosum and erythema induratum (nodular vasculitis): diagnosis and management. Dermatol Ther. 2010;23:320-327. doi:10.1111/j.1529-8019.2010.01332.x
  9. Sánchez Yus E, Sanz Vico MD, de Diego V. Miescher’s radial granuloma. a characteristic marker of erythema nodosum. Am J Dermatopathol. 1989;11:434-442. doi:10.1097/00000372-198910000-00005
  10. Mehta SR, Sashindran VK. Clinical features and management of snake bite. Med J Armed Forces India. 2002;58:247-249. doi:10.1016/S0377-1237(02)80140-X
  11. Brys AK, Gandolfi BM, Levinson H, et al. Copperhead envenomation resulting in a rare case of hand compartment syndrome and subsequent fasciotomy. Plast Reconstr Surg Glob Open. 2015;3:E396. doi:10.1097/GOX.0000000000000367
  12. Clark RF, Selden BS, Furbee B. The incidence of wound infection following crotalid envenomation. J Emerg Med. 1993;11:583-586. doi:10.1016/0736-4679(93)90313-v
  13. Buchanan JT, Thurman J. Crotalidae envenomation. In: StatPearls. StatPearls Publishing; October 3, 2022. Accessed July 22, 2024. https://www.ncbi.nlm.nih.gov/books/NBK551615/
Issue
Cutis - 114(2)
Issue
Cutis - 114(2)
Page Number
51-53
Page Number
51-53
Publications
Publications
Topics
Article Type
Display Headline
Erythema Nodosum Triggered by a Bite From a Copperhead Snake
Display Headline
Erythema Nodosum Triggered by a Bite From a Copperhead Snake
Sections
Inside the Article

Practice Points

  • Erythema nodosum (EN) can occur following snakebites from pit vipers such as the eastern copperhead.
  • The acute phase of EN is neutrophilic and responds to colchicine. The chronic phase of EN is granulomatous and responds best to rest and elevation as well as nonsteroidal anti-inflammatory drugs and iodides.
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

Generalized Fixed Drug Eruptions Require Urgent Care: A Case Series

Article Type
Changed
Wed, 08/07/2024 - 11:51
Display Headline
Generalized Fixed Drug Eruptions Require Urgent Care: A Case Series

Recognizing cutaneous drug eruptions is important for treatment and prevention of recurrence. Fixed drug eruptions (FDEs) typically are harmless but can have major negative cosmetic consequences for patients. In its more severe forms, patients are at risk for widespread epithelial necrosis with accompanying complications. We report 1 patient with generalized FDE and 2 with generalized bullous FDE. We also discuss the recognition and treatment of the condition. Two patients previously had been diagnosed with systemic lupus erythematosus (SLE).

Case Series

Patient 1—A 60-year-old woman presented to dermatology with a rash on the trunk and groin folds of 4 days’ duration. She had a history of SLE and cutaneous lupus treated with hydroxychloroquine 200 mg twice daily and topical corticosteroids. She had started sulfamethoxazole-trimethoprim for a urinary tract infection with a rash appearing 1 day later. She reported burning skin pain with progression to blisters that “sloughed” off. She denied any known history of allergy to sulfa drugs. Prior to evaluation by dermatology, she visited an urgent care facility and was prescribed hydroxyzine and intramuscular corticosteroids. At presentation to dermatology 3 days after taking sulfamethoxazole-trimethoprim, she had annular flaccid bullae and superficial erosions with dusky borders on the right posterior thigh, right side of the chest, left inframammary fold, and right inguinal fold (Figure 1). She had no ocular, oral, or vaginal erosions. A diagnosis of generalized bullous FDE was favored over erythema multiforme or Stevens-Johnson syndrome (SJS)/toxic epidermal necrolysis (TEN). Shave biopsies from lesions on the right posterior thigh and right inguinal fold demonstrated interface dermatitis with epidermal necrosis, pigment incontinence, and numerous eosinophils. Direct immunofluorescence of the perilesional skin was negative for immunoprotein deposition. These findings were consistent with the clinical impression of generalized bullous FDE. Prior to receiving the histopathology report, the patient was initiated on a regimen of cyclosporine 5 mg/kg/d in the setting of normal renal function and followed until the eruption resolved completely. Cyclosporine was tapered at 2 weeks and discontinued at 3 weeks.

FIGURE 1. A and B, Eroded bullae on annular hyperpigmented plaques of the left inframammary fold and right side of the chest, respectively, in a patient with a generalized bullous fixed drug eruption (patient 1).

Patient 2—A 32-year-old woman presented for follow-up management of discoid lupus erythematosus. She had a history of systemic and cutaneous lupus, juvenile rheumatoid arthritis, and mixed connective tissue disease managed with prednisone, hydroxychloroquine, azathioprine, and belimumab. Physical examination revealed scarring alopecia with dyspigmentation and active inflammation consistent with uncontrolled cutaneous lupus. However, she also had oval-shaped hyperpigmented patches over the left breast, clavicle, and anterior chest consistent with a generalized FDE (Figure 2). The patient did not recall a history of similar lesions and could not identify a possible trigger. She was counseled on possible culprits and advised to avoid unnecessary medications. She had an unremarkable clinical course; therefore, no further intervention was necessary.

 

FIGURE 2. Hyperpigmented patches were noted on the left side of the chest in a patient with a generalized fixed drug eruption (patient 2).

Patient 3—A 33-year-old man presented to the emergency department with a painful rash on the chest and back of 2 days’ duration that began 1 hour after taking naproxen (dosage unknown) for back pain. He had no notable medical history. The patient stated that the rash had slowly worsened and started to develop blisters. He visited an urgent care facility 1 day prior to the current presentation and was started on a 5-day course of prednisone 40 mg daily; the first 2 doses did not help. He denied any mucosal involvement apart from a tender lesion on the penis. He reported a history of an allergic reaction to penicillin. Physical examination revealed extensive dusky violaceous annular plaques with erythematous borders across the anterior and posterior trunk (Figure 3). Multiple flaccid bullae developed within these plaques, involving 15% of the body surface area. He was diagnosed with generalized bullous FDE based on the clinical history and histopathology. He was admitted to the burn intensive care unit and treated with cyclosporine 3 mg/kg/d with subsequent resolution of the eruption.

FIGURE 3. A, Erythematous patches were scattered across the chest with focal, intact, flaccid bullae in a patient with a generalized bullous fixed drug eruption (patient 3). B, Large confluent annular hyperpigmented, dusky patches with erythematous rims and several bullae were scattered across the back.

 

 

Comment

Presentation of FDEs—A fixed drug eruption manifests with 1 or more well-demarcated, red or violaceous, annular patches that resolve with postinflammatory hyperpigmentation; it occasionally may manifest with bullae. Initial eruptions may occur up to 2 weeks following medication exposure, but recurrent eruptions usually happen within minutes to hours later. They often are in the same location as prior lesions. A fixed drug eruption can be solitary, scattered, or generalized; a generalized FDE typically demonstrates multiple bilateral lesions that may itch, burn, or cause no symptoms. Patients can experience an FDE at any age, though the median age is reported as 35 to 60 years of age.1 A fixed drug eruption usually occurs after ingestion of oral medications, though there have been a few reports with iodinated contrast.2 Well-known culprits include antibiotics (eg, sulfamethoxazole-trimethoprim, tetracyclines, penicillins/cephalosporins, quinolones, dapsone), nonsteroidal anti-inflammatory drugs, acetaminophen (eg, paracetamol), barbiturates, antimalarials, and anticonvulsants. It also can occur with vaccines or with certain foods (fixed food eruption).3,4 Clinicians may try an oral drug challenge to identify the cause of an FDE, but in patients with a history of a generalized FDE, the risk for developing an increasingly severe reaction with repeated exposure to the medication is too high.5

 

Histopathology—Patch testing at the site of prior eruption with suspected drug culprits may be useful.6 Histopathology of FDE typically demonstrates vacuolar changes at the dermoepidermal junction with a lichenoid lymphocytic infiltrate. Early lesions often show a predominance of eosinophils. Subepidermal clefting is a feature of the bullous variant. In an active lesion, there are large numbers of CD8+ T lymphocytes expressing natural killer cell–associated molecules.7 The pathologic mechanism is not well understood, though it has been hypothesized that memory CD8+ cells are maintained in specific regions of the epidermis by IL-15 produced in the microenvironment and are activated upon rechallenge.7Considerations in Generalized Bullous FDE—Generalized FDE is defined in the literature as an FDE with involvement of 3 of 6 body areas: head, neck, trunk, upper limbs, lower limbs, and genital area. It may cover more or less than 10% of the body surface area.8-10 Although an isolated FDE frequently is asymptomatic and may not be cause for alarm, recurring drug eruptions increase the risk for development of generalized bullous FDE. Generalized bullous FDE is a rare subset. It is frequently misdiagnosed, and data on its incidence are uncertain.11 Of note, several pathologies causing bullous lesions may be in the differential diagnosis, including bullous pemphigoid; pemphigus vulgaris; bullous SLE; or bullae from cutaneous lupus, staphylococcal scalded skin syndrome, erythema multiforme, or SJS/TEN.12 When matched for body surface area involvement with SJS/TEN, generalized bullous FDE shares nearly identical mortality rates10; therefore, these patients should be treated with the same level of urgency and admitted to a critical care or burn unit, as they are at serious risk for infection and other complications.13

Clinical history and presentation along with histopathologic findings help to narrow down the differential diagnosis. Clinically, generalized bullous FDE does not affect the surrounding skin and manifests sooner after drug exposure (1–24 hours) with less mucosal involvement than SJS/TEN.9 Additionally, SJS/TEN patients frequently have generalized malaise and/or fever, while generalized bullous FDE patients do not. Finally, patients with generalized bullous FDE may report a history of a cutaneous eruption similar in morphology or in the same location.

Histopathologically, generalized bullous FDE may be similar to FDE with the addition of a subepidermal blister. Generalized bullous FDE patients have greater eosinophil infiltration and dermal melanophages than patients with SJS/TEN.9 Cellular infiltrates in generalized bullous FDE include more dermal CD41 cells, such as Foxp31 regulatory T cells; fewer intraepidermal CD561 cells; and fewer intraepidermal cells with granulysin.9 Occasionally, generalized bullous FDE causes full-thickness necrosis. In those cases, generalized bullous FDE cannot reliably be distinguished from other conditions with epidermal necrolysis on histopathology.13

FDE Diagnostics—A cytotoxin produced by cytotoxic T lymphocytes, granulysin can be measured to aid in diagnosis of FDE, though this test may not be widely available. High levels of granulysin in the blister fluid and serum can be used to distinguish SJS/TEN, erythema multiforme, and localized and generalized bullous FDE from other non–cytotoxic T lymphocyte–mediated bullous skin disorders, such as bullous pemphigoid, pemphigus, and bullous SLE.14 Blister granulysin levels are notably lower in generalized bullous FDE than in SJS/TEN.9,14 Chen et al14 also found that granulysin levels can be used to gauge disease progression given that the levels sharply decrease after patients have reached maximal skin detachment.

Management—Avoidance of the inciting drug often is sufficient for patients with an FDE, as demonstrated in patient 2 in our case series. Clinicians also should counsel patients on avoidance of potential cross-reacting drugs. Symptomatic treatment for itch or pain is appropriate and may include antihistamines or topical steroids. Nonsteroidal anti-inflammatory drugs may exacerbate or be causative of FDE. For generalized bullous FDE, cyclosporine is favored in the literature15,16 and was used to successfully treat both patients 1 and 3 in our case series. A short course of systemic corticosteroids or intravenous immunoglobulin also may be considered. Mild cases of generalized bullous FDE may be treated with close outpatient follow-up (patient 1), while severe cases require inpatient or even critical care monitoring with aggressive medical management to prevent the progression of skin desquamation (patient 3). Patients with severe oral lesions may require inpatient support for fluid maintenance.

Lupus History—Two patients in our case series had a history of lupus. Lupus itself can cause primary bullous lesions. Similar to FDE, bullous SLE can involve sun-exposed and nonexposed areas of the skin as well as the mucous membranes with a predilection for the lower vermilion lip.17 In bullous SLE, tense subepidermal blisters with a neutrophil-rich infiltrate form due to circulating antibodies to type VII collagen. These blisters have an erythematous or urticated base, most commonly on the face, upper trunk, and proximal extremities.18 In both SLE with skin manifestations and lupus limited to the skin, bullae may form due to extensive vacuolar degeneration. Similar to TEN, they can form rapidly in a widespread distribution.17 However, there is limited mucosal involvement, no clear drug association, and a better prognosis. Bullae caused by lupus will frequently demonstrate deposition of immunoproteins IgG, IgM, IgA, and complement component 3 at the basement membrane zone in perilesional skin on direct immunofluorescence. However, negative direct immunofluorescence does not rule out lupus.12 At the same time, patients with lupus frequently have comorbidities requiring multiple medications; the need for these medications may predispose patients to higher rates of cutaneous drug eruptions.19 To our knowledge, there is no known association between FDE and lupus.

Conclusion

Patients with acute eruptions following the initiation of a new prescription or over-the-counter medication require urgent evaluation. Generalized bullous FDE requires timely diagnosis and intervention. Patients with lupus have an increased risk for cutaneous drug eruptions due to polypharmacy. Further investigation is necessary to determine if there is a pathophysiologic mechanism responsible for the development of FDE in lupus patients.

References
  1. Anderson HJ, Lee JB. A review of fixed drug eruption with a special focus on generalized bullous fixed drug eruption. Medicina (Kaunas). 2021;57:925.
  2. Gavin M, Sharp L, Walker K, et al. Contrast-induced generalized bullous fixed drug eruption resembling Stevens-Johnson syndrome. Proc (Bayl Univ Med Cent). 2019;32:601-602.
  3. Kabir S, Feit EJ, Heilman ER. Generalized fixed drug eruption following Pfizer-BioNtech COVID-19 vaccination. Clin Case Rep. 2022;10:E6684.
  4. Choi S, Kim SH, Hwang JH, et al. Rapidly progressing generalized bullous fixed drug eruption after the first dose of COVID-19 messenger RNA vaccination. J Dermatol. 2023;50:1190-1193.
  5. Mahboob A, Haroon TS. Drugs causing fixed eruptions: a study of 450 cases. Int J Dermatol. 1998;37:833-838.
  6. Shiohara T. Fixed drug eruption: pathogenesis and diagnostic tests. Curr Opin Allergy Clin Immunol. 2009;9:316-321.
  7. Mizukawa Y, Yamazaki Y, Shiohara T. In vivo dynamics of intraepidermal CD8+ T cells and CD4+ T cells during the evolution of fixed drug eruption. Br J Dermatol. 2008;158:1230-1238.
  8. Lee CH, Chen YC, Cho YT, et al. Fixed-drug eruption: a retrospective study in a single referral center in northern Taiwan. Dermatologica Sinica. 2012;30:11-15.
  9. Cho YT, Lin JW, Chen YC, et al. Generalized bullous fixed drug eruption is distinct from Stevens-Johnson syndrome/toxic epidermal necrolysis by immunohistopathological features. J Am Acad Dermatol. 2014;70:539-548.
  10. Lipowicz S, Sekula P, Ingen-Housz-Oro S, et al. Prognosis of generalized bullous fixed drug eruption: comparison with Stevens-Johnson syndrome and toxic epidermal necrolysis. Br J Dermatol. 2013;168:726-732.
  11. Patel S, John AM, Handler MZ, et al. Fixed drug eruptions: an update, emphasizing the potentially lethal generalized bullous fixed drug eruption. Am J Clin Dermatol. 2020;21:393-399.
  12. Ranario JS, Smith JL. Bullous lesions in a patient with systemic lupus erythematosus. J Clin Aesthet Dermatol. 2014;7:44-49.
  13. Perron E, Viarnaud A, Marciano L, et al. Clinical and histological features of fixed drug eruption: a single-centre series of 73 cases with comparison between bullous and non-bullous forms. Eur J Dermatol. 2021;31:372-380.
  14. Chen CB, Kuo KL, Wang CW, et al. Detecting lesional granulysin levels for rapid diagnosis of cytotoxic T lymphocyte-mediated bullous skin disorders. J Allergy Clin Immunol Pract. 2021;9:1327-1337.e3.
  15. Beniwal R, Gupta LK, Khare AK, et al. Cyclosporine in generalized bullous-fixed drug eruption. Indian J Dermatol. 2018;63:432-433.
  16. Vargas Mora P, García S, Valenzuela F, et al. Generalized bullous fixed drug eruption successfully treated with cyclosporine. Dermatol Ther. 2020;33:E13492.
  17. Montagnon CM, Tolkachjov SN, Murrell DF, et al. Subepithelial autoimmune blistering dermatoses: clinical features and diagnosis. J Am Acad Dermatol. 2021;85:1-14.
  18. Sebaratnam DF, Murrell DF. Bullous systemic lupus erythematosus. Dermatol Clin. 2011;29:649-653.
  19. Zonzits E, Aberer W, Tappeiner G. Drug eruptions from mesna. After cyclophosphamide treatment of patients with systemic lupus erythematosus and dermatomyositis. Arch Dermatol. 1992;128:80-82.
Article PDF
Author and Disclosure Information

 

From the Medical University of South Carolina, Charleston.   Dr. Barker is from the Department of Internal Medicine. Drs. Elston and Lee are from the Department of Dermatology and Dermatologic Surgery.

The authors report no conflict of interest.

Correspondence: Catherine Shirer Barker, MD, 96 Jonathan Lucas St, Ste 807 CSB, MSC 623, Charleston, SC 29425 (catherinesbarker@gmail.com). 

Cutis. 2024 July;114(1):E31-E34. doi:10.12788/cutis.1063

Issue
Cutis - 114(1)
Publications
Topics
Page Number
E31-E34
Sections
Author and Disclosure Information

 

From the Medical University of South Carolina, Charleston.   Dr. Barker is from the Department of Internal Medicine. Drs. Elston and Lee are from the Department of Dermatology and Dermatologic Surgery.

The authors report no conflict of interest.

Correspondence: Catherine Shirer Barker, MD, 96 Jonathan Lucas St, Ste 807 CSB, MSC 623, Charleston, SC 29425 (catherinesbarker@gmail.com). 

Cutis. 2024 July;114(1):E31-E34. doi:10.12788/cutis.1063

Author and Disclosure Information

 

From the Medical University of South Carolina, Charleston.   Dr. Barker is from the Department of Internal Medicine. Drs. Elston and Lee are from the Department of Dermatology and Dermatologic Surgery.

The authors report no conflict of interest.

Correspondence: Catherine Shirer Barker, MD, 96 Jonathan Lucas St, Ste 807 CSB, MSC 623, Charleston, SC 29425 (catherinesbarker@gmail.com). 

Cutis. 2024 July;114(1):E31-E34. doi:10.12788/cutis.1063

Article PDF
Article PDF

Recognizing cutaneous drug eruptions is important for treatment and prevention of recurrence. Fixed drug eruptions (FDEs) typically are harmless but can have major negative cosmetic consequences for patients. In its more severe forms, patients are at risk for widespread epithelial necrosis with accompanying complications. We report 1 patient with generalized FDE and 2 with generalized bullous FDE. We also discuss the recognition and treatment of the condition. Two patients previously had been diagnosed with systemic lupus erythematosus (SLE).

Case Series

Patient 1—A 60-year-old woman presented to dermatology with a rash on the trunk and groin folds of 4 days’ duration. She had a history of SLE and cutaneous lupus treated with hydroxychloroquine 200 mg twice daily and topical corticosteroids. She had started sulfamethoxazole-trimethoprim for a urinary tract infection with a rash appearing 1 day later. She reported burning skin pain with progression to blisters that “sloughed” off. She denied any known history of allergy to sulfa drugs. Prior to evaluation by dermatology, she visited an urgent care facility and was prescribed hydroxyzine and intramuscular corticosteroids. At presentation to dermatology 3 days after taking sulfamethoxazole-trimethoprim, she had annular flaccid bullae and superficial erosions with dusky borders on the right posterior thigh, right side of the chest, left inframammary fold, and right inguinal fold (Figure 1). She had no ocular, oral, or vaginal erosions. A diagnosis of generalized bullous FDE was favored over erythema multiforme or Stevens-Johnson syndrome (SJS)/toxic epidermal necrolysis (TEN). Shave biopsies from lesions on the right posterior thigh and right inguinal fold demonstrated interface dermatitis with epidermal necrosis, pigment incontinence, and numerous eosinophils. Direct immunofluorescence of the perilesional skin was negative for immunoprotein deposition. These findings were consistent with the clinical impression of generalized bullous FDE. Prior to receiving the histopathology report, the patient was initiated on a regimen of cyclosporine 5 mg/kg/d in the setting of normal renal function and followed until the eruption resolved completely. Cyclosporine was tapered at 2 weeks and discontinued at 3 weeks.

FIGURE 1. A and B, Eroded bullae on annular hyperpigmented plaques of the left inframammary fold and right side of the chest, respectively, in a patient with a generalized bullous fixed drug eruption (patient 1).

Patient 2—A 32-year-old woman presented for follow-up management of discoid lupus erythematosus. She had a history of systemic and cutaneous lupus, juvenile rheumatoid arthritis, and mixed connective tissue disease managed with prednisone, hydroxychloroquine, azathioprine, and belimumab. Physical examination revealed scarring alopecia with dyspigmentation and active inflammation consistent with uncontrolled cutaneous lupus. However, she also had oval-shaped hyperpigmented patches over the left breast, clavicle, and anterior chest consistent with a generalized FDE (Figure 2). The patient did not recall a history of similar lesions and could not identify a possible trigger. She was counseled on possible culprits and advised to avoid unnecessary medications. She had an unremarkable clinical course; therefore, no further intervention was necessary.

 

FIGURE 2. Hyperpigmented patches were noted on the left side of the chest in a patient with a generalized fixed drug eruption (patient 2).

Patient 3—A 33-year-old man presented to the emergency department with a painful rash on the chest and back of 2 days’ duration that began 1 hour after taking naproxen (dosage unknown) for back pain. He had no notable medical history. The patient stated that the rash had slowly worsened and started to develop blisters. He visited an urgent care facility 1 day prior to the current presentation and was started on a 5-day course of prednisone 40 mg daily; the first 2 doses did not help. He denied any mucosal involvement apart from a tender lesion on the penis. He reported a history of an allergic reaction to penicillin. Physical examination revealed extensive dusky violaceous annular plaques with erythematous borders across the anterior and posterior trunk (Figure 3). Multiple flaccid bullae developed within these plaques, involving 15% of the body surface area. He was diagnosed with generalized bullous FDE based on the clinical history and histopathology. He was admitted to the burn intensive care unit and treated with cyclosporine 3 mg/kg/d with subsequent resolution of the eruption.

FIGURE 3. A, Erythematous patches were scattered across the chest with focal, intact, flaccid bullae in a patient with a generalized bullous fixed drug eruption (patient 3). B, Large confluent annular hyperpigmented, dusky patches with erythematous rims and several bullae were scattered across the back.

 

 

Comment

Presentation of FDEs—A fixed drug eruption manifests with 1 or more well-demarcated, red or violaceous, annular patches that resolve with postinflammatory hyperpigmentation; it occasionally may manifest with bullae. Initial eruptions may occur up to 2 weeks following medication exposure, but recurrent eruptions usually happen within minutes to hours later. They often are in the same location as prior lesions. A fixed drug eruption can be solitary, scattered, or generalized; a generalized FDE typically demonstrates multiple bilateral lesions that may itch, burn, or cause no symptoms. Patients can experience an FDE at any age, though the median age is reported as 35 to 60 years of age.1 A fixed drug eruption usually occurs after ingestion of oral medications, though there have been a few reports with iodinated contrast.2 Well-known culprits include antibiotics (eg, sulfamethoxazole-trimethoprim, tetracyclines, penicillins/cephalosporins, quinolones, dapsone), nonsteroidal anti-inflammatory drugs, acetaminophen (eg, paracetamol), barbiturates, antimalarials, and anticonvulsants. It also can occur with vaccines or with certain foods (fixed food eruption).3,4 Clinicians may try an oral drug challenge to identify the cause of an FDE, but in patients with a history of a generalized FDE, the risk for developing an increasingly severe reaction with repeated exposure to the medication is too high.5

 

Histopathology—Patch testing at the site of prior eruption with suspected drug culprits may be useful.6 Histopathology of FDE typically demonstrates vacuolar changes at the dermoepidermal junction with a lichenoid lymphocytic infiltrate. Early lesions often show a predominance of eosinophils. Subepidermal clefting is a feature of the bullous variant. In an active lesion, there are large numbers of CD8+ T lymphocytes expressing natural killer cell–associated molecules.7 The pathologic mechanism is not well understood, though it has been hypothesized that memory CD8+ cells are maintained in specific regions of the epidermis by IL-15 produced in the microenvironment and are activated upon rechallenge.7Considerations in Generalized Bullous FDE—Generalized FDE is defined in the literature as an FDE with involvement of 3 of 6 body areas: head, neck, trunk, upper limbs, lower limbs, and genital area. It may cover more or less than 10% of the body surface area.8-10 Although an isolated FDE frequently is asymptomatic and may not be cause for alarm, recurring drug eruptions increase the risk for development of generalized bullous FDE. Generalized bullous FDE is a rare subset. It is frequently misdiagnosed, and data on its incidence are uncertain.11 Of note, several pathologies causing bullous lesions may be in the differential diagnosis, including bullous pemphigoid; pemphigus vulgaris; bullous SLE; or bullae from cutaneous lupus, staphylococcal scalded skin syndrome, erythema multiforme, or SJS/TEN.12 When matched for body surface area involvement with SJS/TEN, generalized bullous FDE shares nearly identical mortality rates10; therefore, these patients should be treated with the same level of urgency and admitted to a critical care or burn unit, as they are at serious risk for infection and other complications.13

Clinical history and presentation along with histopathologic findings help to narrow down the differential diagnosis. Clinically, generalized bullous FDE does not affect the surrounding skin and manifests sooner after drug exposure (1–24 hours) with less mucosal involvement than SJS/TEN.9 Additionally, SJS/TEN patients frequently have generalized malaise and/or fever, while generalized bullous FDE patients do not. Finally, patients with generalized bullous FDE may report a history of a cutaneous eruption similar in morphology or in the same location.

Histopathologically, generalized bullous FDE may be similar to FDE with the addition of a subepidermal blister. Generalized bullous FDE patients have greater eosinophil infiltration and dermal melanophages than patients with SJS/TEN.9 Cellular infiltrates in generalized bullous FDE include more dermal CD41 cells, such as Foxp31 regulatory T cells; fewer intraepidermal CD561 cells; and fewer intraepidermal cells with granulysin.9 Occasionally, generalized bullous FDE causes full-thickness necrosis. In those cases, generalized bullous FDE cannot reliably be distinguished from other conditions with epidermal necrolysis on histopathology.13

FDE Diagnostics—A cytotoxin produced by cytotoxic T lymphocytes, granulysin can be measured to aid in diagnosis of FDE, though this test may not be widely available. High levels of granulysin in the blister fluid and serum can be used to distinguish SJS/TEN, erythema multiforme, and localized and generalized bullous FDE from other non–cytotoxic T lymphocyte–mediated bullous skin disorders, such as bullous pemphigoid, pemphigus, and bullous SLE.14 Blister granulysin levels are notably lower in generalized bullous FDE than in SJS/TEN.9,14 Chen et al14 also found that granulysin levels can be used to gauge disease progression given that the levels sharply decrease after patients have reached maximal skin detachment.

Management—Avoidance of the inciting drug often is sufficient for patients with an FDE, as demonstrated in patient 2 in our case series. Clinicians also should counsel patients on avoidance of potential cross-reacting drugs. Symptomatic treatment for itch or pain is appropriate and may include antihistamines or topical steroids. Nonsteroidal anti-inflammatory drugs may exacerbate or be causative of FDE. For generalized bullous FDE, cyclosporine is favored in the literature15,16 and was used to successfully treat both patients 1 and 3 in our case series. A short course of systemic corticosteroids or intravenous immunoglobulin also may be considered. Mild cases of generalized bullous FDE may be treated with close outpatient follow-up (patient 1), while severe cases require inpatient or even critical care monitoring with aggressive medical management to prevent the progression of skin desquamation (patient 3). Patients with severe oral lesions may require inpatient support for fluid maintenance.

Lupus History—Two patients in our case series had a history of lupus. Lupus itself can cause primary bullous lesions. Similar to FDE, bullous SLE can involve sun-exposed and nonexposed areas of the skin as well as the mucous membranes with a predilection for the lower vermilion lip.17 In bullous SLE, tense subepidermal blisters with a neutrophil-rich infiltrate form due to circulating antibodies to type VII collagen. These blisters have an erythematous or urticated base, most commonly on the face, upper trunk, and proximal extremities.18 In both SLE with skin manifestations and lupus limited to the skin, bullae may form due to extensive vacuolar degeneration. Similar to TEN, they can form rapidly in a widespread distribution.17 However, there is limited mucosal involvement, no clear drug association, and a better prognosis. Bullae caused by lupus will frequently demonstrate deposition of immunoproteins IgG, IgM, IgA, and complement component 3 at the basement membrane zone in perilesional skin on direct immunofluorescence. However, negative direct immunofluorescence does not rule out lupus.12 At the same time, patients with lupus frequently have comorbidities requiring multiple medications; the need for these medications may predispose patients to higher rates of cutaneous drug eruptions.19 To our knowledge, there is no known association between FDE and lupus.

Conclusion

Patients with acute eruptions following the initiation of a new prescription or over-the-counter medication require urgent evaluation. Generalized bullous FDE requires timely diagnosis and intervention. Patients with lupus have an increased risk for cutaneous drug eruptions due to polypharmacy. Further investigation is necessary to determine if there is a pathophysiologic mechanism responsible for the development of FDE in lupus patients.

Recognizing cutaneous drug eruptions is important for treatment and prevention of recurrence. Fixed drug eruptions (FDEs) typically are harmless but can have major negative cosmetic consequences for patients. In its more severe forms, patients are at risk for widespread epithelial necrosis with accompanying complications. We report 1 patient with generalized FDE and 2 with generalized bullous FDE. We also discuss the recognition and treatment of the condition. Two patients previously had been diagnosed with systemic lupus erythematosus (SLE).

Case Series

Patient 1—A 60-year-old woman presented to dermatology with a rash on the trunk and groin folds of 4 days’ duration. She had a history of SLE and cutaneous lupus treated with hydroxychloroquine 200 mg twice daily and topical corticosteroids. She had started sulfamethoxazole-trimethoprim for a urinary tract infection with a rash appearing 1 day later. She reported burning skin pain with progression to blisters that “sloughed” off. She denied any known history of allergy to sulfa drugs. Prior to evaluation by dermatology, she visited an urgent care facility and was prescribed hydroxyzine and intramuscular corticosteroids. At presentation to dermatology 3 days after taking sulfamethoxazole-trimethoprim, she had annular flaccid bullae and superficial erosions with dusky borders on the right posterior thigh, right side of the chest, left inframammary fold, and right inguinal fold (Figure 1). She had no ocular, oral, or vaginal erosions. A diagnosis of generalized bullous FDE was favored over erythema multiforme or Stevens-Johnson syndrome (SJS)/toxic epidermal necrolysis (TEN). Shave biopsies from lesions on the right posterior thigh and right inguinal fold demonstrated interface dermatitis with epidermal necrosis, pigment incontinence, and numerous eosinophils. Direct immunofluorescence of the perilesional skin was negative for immunoprotein deposition. These findings were consistent with the clinical impression of generalized bullous FDE. Prior to receiving the histopathology report, the patient was initiated on a regimen of cyclosporine 5 mg/kg/d in the setting of normal renal function and followed until the eruption resolved completely. Cyclosporine was tapered at 2 weeks and discontinued at 3 weeks.

FIGURE 1. A and B, Eroded bullae on annular hyperpigmented plaques of the left inframammary fold and right side of the chest, respectively, in a patient with a generalized bullous fixed drug eruption (patient 1).

Patient 2—A 32-year-old woman presented for follow-up management of discoid lupus erythematosus. She had a history of systemic and cutaneous lupus, juvenile rheumatoid arthritis, and mixed connective tissue disease managed with prednisone, hydroxychloroquine, azathioprine, and belimumab. Physical examination revealed scarring alopecia with dyspigmentation and active inflammation consistent with uncontrolled cutaneous lupus. However, she also had oval-shaped hyperpigmented patches over the left breast, clavicle, and anterior chest consistent with a generalized FDE (Figure 2). The patient did not recall a history of similar lesions and could not identify a possible trigger. She was counseled on possible culprits and advised to avoid unnecessary medications. She had an unremarkable clinical course; therefore, no further intervention was necessary.

 

FIGURE 2. Hyperpigmented patches were noted on the left side of the chest in a patient with a generalized fixed drug eruption (patient 2).

Patient 3—A 33-year-old man presented to the emergency department with a painful rash on the chest and back of 2 days’ duration that began 1 hour after taking naproxen (dosage unknown) for back pain. He had no notable medical history. The patient stated that the rash had slowly worsened and started to develop blisters. He visited an urgent care facility 1 day prior to the current presentation and was started on a 5-day course of prednisone 40 mg daily; the first 2 doses did not help. He denied any mucosal involvement apart from a tender lesion on the penis. He reported a history of an allergic reaction to penicillin. Physical examination revealed extensive dusky violaceous annular plaques with erythematous borders across the anterior and posterior trunk (Figure 3). Multiple flaccid bullae developed within these plaques, involving 15% of the body surface area. He was diagnosed with generalized bullous FDE based on the clinical history and histopathology. He was admitted to the burn intensive care unit and treated with cyclosporine 3 mg/kg/d with subsequent resolution of the eruption.

FIGURE 3. A, Erythematous patches were scattered across the chest with focal, intact, flaccid bullae in a patient with a generalized bullous fixed drug eruption (patient 3). B, Large confluent annular hyperpigmented, dusky patches with erythematous rims and several bullae were scattered across the back.

 

 

Comment

Presentation of FDEs—A fixed drug eruption manifests with 1 or more well-demarcated, red or violaceous, annular patches that resolve with postinflammatory hyperpigmentation; it occasionally may manifest with bullae. Initial eruptions may occur up to 2 weeks following medication exposure, but recurrent eruptions usually happen within minutes to hours later. They often are in the same location as prior lesions. A fixed drug eruption can be solitary, scattered, or generalized; a generalized FDE typically demonstrates multiple bilateral lesions that may itch, burn, or cause no symptoms. Patients can experience an FDE at any age, though the median age is reported as 35 to 60 years of age.1 A fixed drug eruption usually occurs after ingestion of oral medications, though there have been a few reports with iodinated contrast.2 Well-known culprits include antibiotics (eg, sulfamethoxazole-trimethoprim, tetracyclines, penicillins/cephalosporins, quinolones, dapsone), nonsteroidal anti-inflammatory drugs, acetaminophen (eg, paracetamol), barbiturates, antimalarials, and anticonvulsants. It also can occur with vaccines or with certain foods (fixed food eruption).3,4 Clinicians may try an oral drug challenge to identify the cause of an FDE, but in patients with a history of a generalized FDE, the risk for developing an increasingly severe reaction with repeated exposure to the medication is too high.5

 

Histopathology—Patch testing at the site of prior eruption with suspected drug culprits may be useful.6 Histopathology of FDE typically demonstrates vacuolar changes at the dermoepidermal junction with a lichenoid lymphocytic infiltrate. Early lesions often show a predominance of eosinophils. Subepidermal clefting is a feature of the bullous variant. In an active lesion, there are large numbers of CD8+ T lymphocytes expressing natural killer cell–associated molecules.7 The pathologic mechanism is not well understood, though it has been hypothesized that memory CD8+ cells are maintained in specific regions of the epidermis by IL-15 produced in the microenvironment and are activated upon rechallenge.7Considerations in Generalized Bullous FDE—Generalized FDE is defined in the literature as an FDE with involvement of 3 of 6 body areas: head, neck, trunk, upper limbs, lower limbs, and genital area. It may cover more or less than 10% of the body surface area.8-10 Although an isolated FDE frequently is asymptomatic and may not be cause for alarm, recurring drug eruptions increase the risk for development of generalized bullous FDE. Generalized bullous FDE is a rare subset. It is frequently misdiagnosed, and data on its incidence are uncertain.11 Of note, several pathologies causing bullous lesions may be in the differential diagnosis, including bullous pemphigoid; pemphigus vulgaris; bullous SLE; or bullae from cutaneous lupus, staphylococcal scalded skin syndrome, erythema multiforme, or SJS/TEN.12 When matched for body surface area involvement with SJS/TEN, generalized bullous FDE shares nearly identical mortality rates10; therefore, these patients should be treated with the same level of urgency and admitted to a critical care or burn unit, as they are at serious risk for infection and other complications.13

Clinical history and presentation along with histopathologic findings help to narrow down the differential diagnosis. Clinically, generalized bullous FDE does not affect the surrounding skin and manifests sooner after drug exposure (1–24 hours) with less mucosal involvement than SJS/TEN.9 Additionally, SJS/TEN patients frequently have generalized malaise and/or fever, while generalized bullous FDE patients do not. Finally, patients with generalized bullous FDE may report a history of a cutaneous eruption similar in morphology or in the same location.

Histopathologically, generalized bullous FDE may be similar to FDE with the addition of a subepidermal blister. Generalized bullous FDE patients have greater eosinophil infiltration and dermal melanophages than patients with SJS/TEN.9 Cellular infiltrates in generalized bullous FDE include more dermal CD41 cells, such as Foxp31 regulatory T cells; fewer intraepidermal CD561 cells; and fewer intraepidermal cells with granulysin.9 Occasionally, generalized bullous FDE causes full-thickness necrosis. In those cases, generalized bullous FDE cannot reliably be distinguished from other conditions with epidermal necrolysis on histopathology.13

FDE Diagnostics—A cytotoxin produced by cytotoxic T lymphocytes, granulysin can be measured to aid in diagnosis of FDE, though this test may not be widely available. High levels of granulysin in the blister fluid and serum can be used to distinguish SJS/TEN, erythema multiforme, and localized and generalized bullous FDE from other non–cytotoxic T lymphocyte–mediated bullous skin disorders, such as bullous pemphigoid, pemphigus, and bullous SLE.14 Blister granulysin levels are notably lower in generalized bullous FDE than in SJS/TEN.9,14 Chen et al14 also found that granulysin levels can be used to gauge disease progression given that the levels sharply decrease after patients have reached maximal skin detachment.

Management—Avoidance of the inciting drug often is sufficient for patients with an FDE, as demonstrated in patient 2 in our case series. Clinicians also should counsel patients on avoidance of potential cross-reacting drugs. Symptomatic treatment for itch or pain is appropriate and may include antihistamines or topical steroids. Nonsteroidal anti-inflammatory drugs may exacerbate or be causative of FDE. For generalized bullous FDE, cyclosporine is favored in the literature15,16 and was used to successfully treat both patients 1 and 3 in our case series. A short course of systemic corticosteroids or intravenous immunoglobulin also may be considered. Mild cases of generalized bullous FDE may be treated with close outpatient follow-up (patient 1), while severe cases require inpatient or even critical care monitoring with aggressive medical management to prevent the progression of skin desquamation (patient 3). Patients with severe oral lesions may require inpatient support for fluid maintenance.

Lupus History—Two patients in our case series had a history of lupus. Lupus itself can cause primary bullous lesions. Similar to FDE, bullous SLE can involve sun-exposed and nonexposed areas of the skin as well as the mucous membranes with a predilection for the lower vermilion lip.17 In bullous SLE, tense subepidermal blisters with a neutrophil-rich infiltrate form due to circulating antibodies to type VII collagen. These blisters have an erythematous or urticated base, most commonly on the face, upper trunk, and proximal extremities.18 In both SLE with skin manifestations and lupus limited to the skin, bullae may form due to extensive vacuolar degeneration. Similar to TEN, they can form rapidly in a widespread distribution.17 However, there is limited mucosal involvement, no clear drug association, and a better prognosis. Bullae caused by lupus will frequently demonstrate deposition of immunoproteins IgG, IgM, IgA, and complement component 3 at the basement membrane zone in perilesional skin on direct immunofluorescence. However, negative direct immunofluorescence does not rule out lupus.12 At the same time, patients with lupus frequently have comorbidities requiring multiple medications; the need for these medications may predispose patients to higher rates of cutaneous drug eruptions.19 To our knowledge, there is no known association between FDE and lupus.

Conclusion

Patients with acute eruptions following the initiation of a new prescription or over-the-counter medication require urgent evaluation. Generalized bullous FDE requires timely diagnosis and intervention. Patients with lupus have an increased risk for cutaneous drug eruptions due to polypharmacy. Further investigation is necessary to determine if there is a pathophysiologic mechanism responsible for the development of FDE in lupus patients.

References
  1. Anderson HJ, Lee JB. A review of fixed drug eruption with a special focus on generalized bullous fixed drug eruption. Medicina (Kaunas). 2021;57:925.
  2. Gavin M, Sharp L, Walker K, et al. Contrast-induced generalized bullous fixed drug eruption resembling Stevens-Johnson syndrome. Proc (Bayl Univ Med Cent). 2019;32:601-602.
  3. Kabir S, Feit EJ, Heilman ER. Generalized fixed drug eruption following Pfizer-BioNtech COVID-19 vaccination. Clin Case Rep. 2022;10:E6684.
  4. Choi S, Kim SH, Hwang JH, et al. Rapidly progressing generalized bullous fixed drug eruption after the first dose of COVID-19 messenger RNA vaccination. J Dermatol. 2023;50:1190-1193.
  5. Mahboob A, Haroon TS. Drugs causing fixed eruptions: a study of 450 cases. Int J Dermatol. 1998;37:833-838.
  6. Shiohara T. Fixed drug eruption: pathogenesis and diagnostic tests. Curr Opin Allergy Clin Immunol. 2009;9:316-321.
  7. Mizukawa Y, Yamazaki Y, Shiohara T. In vivo dynamics of intraepidermal CD8+ T cells and CD4+ T cells during the evolution of fixed drug eruption. Br J Dermatol. 2008;158:1230-1238.
  8. Lee CH, Chen YC, Cho YT, et al. Fixed-drug eruption: a retrospective study in a single referral center in northern Taiwan. Dermatologica Sinica. 2012;30:11-15.
  9. Cho YT, Lin JW, Chen YC, et al. Generalized bullous fixed drug eruption is distinct from Stevens-Johnson syndrome/toxic epidermal necrolysis by immunohistopathological features. J Am Acad Dermatol. 2014;70:539-548.
  10. Lipowicz S, Sekula P, Ingen-Housz-Oro S, et al. Prognosis of generalized bullous fixed drug eruption: comparison with Stevens-Johnson syndrome and toxic epidermal necrolysis. Br J Dermatol. 2013;168:726-732.
  11. Patel S, John AM, Handler MZ, et al. Fixed drug eruptions: an update, emphasizing the potentially lethal generalized bullous fixed drug eruption. Am J Clin Dermatol. 2020;21:393-399.
  12. Ranario JS, Smith JL. Bullous lesions in a patient with systemic lupus erythematosus. J Clin Aesthet Dermatol. 2014;7:44-49.
  13. Perron E, Viarnaud A, Marciano L, et al. Clinical and histological features of fixed drug eruption: a single-centre series of 73 cases with comparison between bullous and non-bullous forms. Eur J Dermatol. 2021;31:372-380.
  14. Chen CB, Kuo KL, Wang CW, et al. Detecting lesional granulysin levels for rapid diagnosis of cytotoxic T lymphocyte-mediated bullous skin disorders. J Allergy Clin Immunol Pract. 2021;9:1327-1337.e3.
  15. Beniwal R, Gupta LK, Khare AK, et al. Cyclosporine in generalized bullous-fixed drug eruption. Indian J Dermatol. 2018;63:432-433.
  16. Vargas Mora P, García S, Valenzuela F, et al. Generalized bullous fixed drug eruption successfully treated with cyclosporine. Dermatol Ther. 2020;33:E13492.
  17. Montagnon CM, Tolkachjov SN, Murrell DF, et al. Subepithelial autoimmune blistering dermatoses: clinical features and diagnosis. J Am Acad Dermatol. 2021;85:1-14.
  18. Sebaratnam DF, Murrell DF. Bullous systemic lupus erythematosus. Dermatol Clin. 2011;29:649-653.
  19. Zonzits E, Aberer W, Tappeiner G. Drug eruptions from mesna. After cyclophosphamide treatment of patients with systemic lupus erythematosus and dermatomyositis. Arch Dermatol. 1992;128:80-82.
References
  1. Anderson HJ, Lee JB. A review of fixed drug eruption with a special focus on generalized bullous fixed drug eruption. Medicina (Kaunas). 2021;57:925.
  2. Gavin M, Sharp L, Walker K, et al. Contrast-induced generalized bullous fixed drug eruption resembling Stevens-Johnson syndrome. Proc (Bayl Univ Med Cent). 2019;32:601-602.
  3. Kabir S, Feit EJ, Heilman ER. Generalized fixed drug eruption following Pfizer-BioNtech COVID-19 vaccination. Clin Case Rep. 2022;10:E6684.
  4. Choi S, Kim SH, Hwang JH, et al. Rapidly progressing generalized bullous fixed drug eruption after the first dose of COVID-19 messenger RNA vaccination. J Dermatol. 2023;50:1190-1193.
  5. Mahboob A, Haroon TS. Drugs causing fixed eruptions: a study of 450 cases. Int J Dermatol. 1998;37:833-838.
  6. Shiohara T. Fixed drug eruption: pathogenesis and diagnostic tests. Curr Opin Allergy Clin Immunol. 2009;9:316-321.
  7. Mizukawa Y, Yamazaki Y, Shiohara T. In vivo dynamics of intraepidermal CD8+ T cells and CD4+ T cells during the evolution of fixed drug eruption. Br J Dermatol. 2008;158:1230-1238.
  8. Lee CH, Chen YC, Cho YT, et al. Fixed-drug eruption: a retrospective study in a single referral center in northern Taiwan. Dermatologica Sinica. 2012;30:11-15.
  9. Cho YT, Lin JW, Chen YC, et al. Generalized bullous fixed drug eruption is distinct from Stevens-Johnson syndrome/toxic epidermal necrolysis by immunohistopathological features. J Am Acad Dermatol. 2014;70:539-548.
  10. Lipowicz S, Sekula P, Ingen-Housz-Oro S, et al. Prognosis of generalized bullous fixed drug eruption: comparison with Stevens-Johnson syndrome and toxic epidermal necrolysis. Br J Dermatol. 2013;168:726-732.
  11. Patel S, John AM, Handler MZ, et al. Fixed drug eruptions: an update, emphasizing the potentially lethal generalized bullous fixed drug eruption. Am J Clin Dermatol. 2020;21:393-399.
  12. Ranario JS, Smith JL. Bullous lesions in a patient with systemic lupus erythematosus. J Clin Aesthet Dermatol. 2014;7:44-49.
  13. Perron E, Viarnaud A, Marciano L, et al. Clinical and histological features of fixed drug eruption: a single-centre series of 73 cases with comparison between bullous and non-bullous forms. Eur J Dermatol. 2021;31:372-380.
  14. Chen CB, Kuo KL, Wang CW, et al. Detecting lesional granulysin levels for rapid diagnosis of cytotoxic T lymphocyte-mediated bullous skin disorders. J Allergy Clin Immunol Pract. 2021;9:1327-1337.e3.
  15. Beniwal R, Gupta LK, Khare AK, et al. Cyclosporine in generalized bullous-fixed drug eruption. Indian J Dermatol. 2018;63:432-433.
  16. Vargas Mora P, García S, Valenzuela F, et al. Generalized bullous fixed drug eruption successfully treated with cyclosporine. Dermatol Ther. 2020;33:E13492.
  17. Montagnon CM, Tolkachjov SN, Murrell DF, et al. Subepithelial autoimmune blistering dermatoses: clinical features and diagnosis. J Am Acad Dermatol. 2021;85:1-14.
  18. Sebaratnam DF, Murrell DF. Bullous systemic lupus erythematosus. Dermatol Clin. 2011;29:649-653.
  19. Zonzits E, Aberer W, Tappeiner G. Drug eruptions from mesna. After cyclophosphamide treatment of patients with systemic lupus erythematosus and dermatomyositis. Arch Dermatol. 1992;128:80-82.
Issue
Cutis - 114(1)
Issue
Cutis - 114(1)
Page Number
E31-E34
Page Number
E31-E34
Publications
Publications
Topics
Article Type
Display Headline
Generalized Fixed Drug Eruptions Require Urgent Care: A Case Series
Display Headline
Generalized Fixed Drug Eruptions Require Urgent Care: A Case Series
Sections
Inside the Article

 

Practice Points

  • Although localized fixed drug eruption (FDE) is a relatively benign diagnosis, generalized bullous FDE requires urgent management and may necessitate intensive burn care.
  • Patients with lupus are at increased risk for drug eruptions due to polypharmacy, and there is a wide differential for bullous eruptions in these patients.
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

Reticulated Brownish Erythema on the Lower Back

Article Type
Changed
Tue, 06/18/2024 - 12:04
Display Headline
Reticulated Brownish Erythema on the Lower Back

The Diagnosis: Erythema Ab Igne

Based on the patient's long-standing history of back pain treated with heating pads as well as the normal laboratory findings and skin examination, a diagnosis of erythema ab igne (EAI) was made.

Erythema ab igne presents as reticulated brownish erythema or hyperpigmentation on sites exposed to prolonged use of heat sources such as heating pads, laptops, and space heaters. Erythema ab igne most commonly affects the lower back, thighs, or legs1-6; however, EAI can appear on atypical sites such as the forehead and eyebrows due to newer technology (eg, virtual reality headsets).7 The level of heat required for EAI to occur is below the threshold for thermal burns (<45 °C [113 °F]).1 Erythema ab igne can occur at any age, and woman are more commonly affected than men.8 The pathophysiology currently is unknown; however, recurrent and prolonged heat exposure may damage superficial vessels. As a result, hemosiderin accumulates in the skin, and hyperpigmentation subsequently occurs.9

The diagnosis of EAI is clinical, and early stages of the rash present as blanching reticulated erythema in areas associated with heat exposure. If the offending source of heat is not removed, EAI can progress to nonblanching, fixed, hyperpigmented plaques with skin atrophy, bullae, or hyperkeratosis. Patients often are asymptomatic; however, mild burning may occur.2 Histopathology reveals cellular atypia, epidermal atrophy, dilation of dermal blood vessels, a minute inflammatory infiltrate, and keratinocyte apoptosis.10 Skin biopsy may be necessary in cases of suspected malignancy due to chronic heat exposure. Lesions that ulcerate or evolve should raise suspicion for malignancy.11 Squamous cell carcinoma is the most common malignancy associated with EAI; other malignancies that may manifest include basal cell carcinoma, Merkel cell carcinoma, or cutaneous marginal zone lymphoma.2,12-14

Erythema ab igne often is mistaken for livedo reticularis, which appears more erythematous without hyperpigmentation or epidermal changes and may be associated with a pathologic state.15 The differential diagnosis in our patient, who was in her 40s with a history of fatigue and joint pain, included livedo reticularis associated with lupus; however, the history of heating pad use, normal laboratory findings, and presence of epidermal changes suggested EAI. Lupus typically affects the hand and knee joints.16 Additionally, livedo reticularis more commonly appears on the legs.15

Other differentials for EAI include livedo racemosa, cutaneous T-cell lymphoma, and cutis marmorata. Livedo racemosa presents with broken rings of erythema in young to middle-aged women and primarily affects the trunk and proximal limbs. It is associated with an underlying condition such as polyarteritis nodosa and less commonly with lupus erythematosus with antiphospholipid or Sneddon syndrome.15,17 Cutaneous T-cell lymphoma typically manifests with poikilodermatous patches larger than the palm, especially in covered areas of skin.18 Cutis marmorata is transient and temperature dependent.9

The key intervention for EAI is removal of the offending heat source.2 Patients should be counseled that the erythema and hyperpigmentation may take months to years to resolve. Topical hydroquinone or tretinoin may be used in cases of persistent hyperpigmentation.19 Patients who continue to use heating pads for long-standing pain should be advised to limit their use to short intervals without occlusion. If malignancy is a concern, a biopsy should be performed.20

References
  1. Wipf AJ, Brown MR. Malignant transformation of erythema ab igne. JAAD Case Rep. 2022;26:85-87. doi:10.1016/j.jdcr.2022.06.018
  2. Sigmon JR, Cantrell J, Teague D, et al. Poorly differentiated carcinoma arising in the setting of erythema ab igne. Am J Dermatopathol. 2013;35:676-678. doi:10.1097/DAD.0b013e3182871648
  3. Patel DP. The evolving nomenclature of erythema ab igne-redness from fire. JAMA Dermatol. 2017;153:685. doi:10.1001/jamadermatol.2017.2021
  4. Arnold AW, Itin PH. Laptop computer-induced erythema ab igne in a child and review of the literature. Pediatrics. 2010;126:E1227-E1230. doi:10.1542/peds.2010-1390
  5. Riahi RR, Cohen PR. Laptop-induced erythema ab igne: report and review of literature. Dermatol Online J. 2012;18:5.
  6. Haleem Z, Philip J, Muhammad S. Erythema ab igne: a rare presentation of toasted skin syndrome with the use of a space heater. Cureus. 2021;13:e13401. doi:10.7759/cureus.13401
  7. Moreau T, Benzaquen M, Gueissaz F. Erythema ab igne after using a virtual reality headset: a new phenomenon to know. J Eur Acad Dermatol Venereol. 2022;36:E932-E933. doi:10.1111/jdv.18371
  8. Ozturk M, An I. Clinical features and etiology of patients with erythema ab igne: a retrospective multicenter study. J Cosmet Dermatol. 2020;19:1774-1779. doi:10.1111/jocd.13210
  9. Gmuca S, Yu J, Weiss PF, et al. Erythema ab igne in an adolescent with chronic pain: an alarming cutaneous eruption from heat exposure. Pediatr Emerg Care. 2020;36:E236-E238. doi:10.1097 /PEC.0000000000001460
  10. Wells A, Desai A, Rudnick EW, et al. Erythema ab igne with features resembling keratosis lichenoides chronica. J Cutan Pathol. 2021;48:151-153. doi:10.1111/cup.13885
  11. Milchak M, Smucker J, Chung CG, et al. Erythema ab igne due to heating pad use: a case report and review of clinical presentation, prevention, and complications. Case Rep Med. 2016;2016:1862480. doi:10.1155/2016/1862480
  12. Daneshvar E, Seraji S, Kamyab-Hesari K, et al. Basal cell carcinoma associated with erythema ab igne. Dermatol Online J. 2020;26:13030 /qt3kz985b4.
  13. Jones CS, Tyring SK, Lee PC, et al. Development of neuroendocrine (Merkel cell) carcinoma mixed with squamous cell carcinoma in erythema ab igne. Arch Dermatol. 1988;124:110-113.
  14. Wharton J, Roffwarg D, Miller J, et al. Cutaneous marginal zone lymphoma arising in the setting of erythema ab igne. J Am Acad Dermatol. 2010;62:1080-1081. doi:10.1016/j.jaad.2009.08.005
  15. Sajjan VV, Lunge S, Swamy MB, et al. Livedo reticularis: a review of the literature. Indian Dermatol Online J. 2015;6:315-321. doi:10.4103 /2229-5178.164493
  16. Grossman JM. Lupus arthritis. Best Pract Res Clin Rheumatol. 2009;23:495-506. doi:10.1016/j.berh.2009.04.003
  17. Aria AB, Chen L, Silapunt S. Erythema ab igne from heating pad use: a report of three clinical cases and a differential diagnosis. Cureus. 2018;10:E2635. doi:10.7759/cureus.2635
  18. Wilcox RA. Cutaneous T-cell lymphoma: 2017 update on diagnosis, risk-stratification, and management. Am J Hematol. 2017;92:1085-1102. doi:10.1002/ajh.24876
  19. Pennitz A, Kinberger M, Avila Valle G, et al. Self-applied topical interventions for melasma: a systematic review and meta-analysis of data from randomized, investigator-blinded clinical trials. Br J Dermatol. 2022;187:309-317.
  20. Sahl WJ, Taira JW. Erythema ab igne: treatment with 5-fluorouracil cream. J Am Acad Dermatol. 1992;27:109-110.
Article PDF
Author and Disclosure Information

Dr. Dao is from the Virginia Commonwealth University School of Medicine, Richmond. Dr. Elston is from the Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston.

The authors report no conflict of interest.

Correspondence: Diem-Phuong D. Dao, MD, 1001 E Leigh St, 11th Floor, Richmond, VA 23219 (daopd@vcu.edu).

Issue
Cutis - 113(6)
Publications
Topics
Page Number
E8-E10
Sections
Author and Disclosure Information

Dr. Dao is from the Virginia Commonwealth University School of Medicine, Richmond. Dr. Elston is from the Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston.

The authors report no conflict of interest.

Correspondence: Diem-Phuong D. Dao, MD, 1001 E Leigh St, 11th Floor, Richmond, VA 23219 (daopd@vcu.edu).

Author and Disclosure Information

Dr. Dao is from the Virginia Commonwealth University School of Medicine, Richmond. Dr. Elston is from the Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston.

The authors report no conflict of interest.

Correspondence: Diem-Phuong D. Dao, MD, 1001 E Leigh St, 11th Floor, Richmond, VA 23219 (daopd@vcu.edu).

Article PDF
Article PDF
Related Articles

The Diagnosis: Erythema Ab Igne

Based on the patient's long-standing history of back pain treated with heating pads as well as the normal laboratory findings and skin examination, a diagnosis of erythema ab igne (EAI) was made.

Erythema ab igne presents as reticulated brownish erythema or hyperpigmentation on sites exposed to prolonged use of heat sources such as heating pads, laptops, and space heaters. Erythema ab igne most commonly affects the lower back, thighs, or legs1-6; however, EAI can appear on atypical sites such as the forehead and eyebrows due to newer technology (eg, virtual reality headsets).7 The level of heat required for EAI to occur is below the threshold for thermal burns (<45 °C [113 °F]).1 Erythema ab igne can occur at any age, and woman are more commonly affected than men.8 The pathophysiology currently is unknown; however, recurrent and prolonged heat exposure may damage superficial vessels. As a result, hemosiderin accumulates in the skin, and hyperpigmentation subsequently occurs.9

The diagnosis of EAI is clinical, and early stages of the rash present as blanching reticulated erythema in areas associated with heat exposure. If the offending source of heat is not removed, EAI can progress to nonblanching, fixed, hyperpigmented plaques with skin atrophy, bullae, or hyperkeratosis. Patients often are asymptomatic; however, mild burning may occur.2 Histopathology reveals cellular atypia, epidermal atrophy, dilation of dermal blood vessels, a minute inflammatory infiltrate, and keratinocyte apoptosis.10 Skin biopsy may be necessary in cases of suspected malignancy due to chronic heat exposure. Lesions that ulcerate or evolve should raise suspicion for malignancy.11 Squamous cell carcinoma is the most common malignancy associated with EAI; other malignancies that may manifest include basal cell carcinoma, Merkel cell carcinoma, or cutaneous marginal zone lymphoma.2,12-14

Erythema ab igne often is mistaken for livedo reticularis, which appears more erythematous without hyperpigmentation or epidermal changes and may be associated with a pathologic state.15 The differential diagnosis in our patient, who was in her 40s with a history of fatigue and joint pain, included livedo reticularis associated with lupus; however, the history of heating pad use, normal laboratory findings, and presence of epidermal changes suggested EAI. Lupus typically affects the hand and knee joints.16 Additionally, livedo reticularis more commonly appears on the legs.15

Other differentials for EAI include livedo racemosa, cutaneous T-cell lymphoma, and cutis marmorata. Livedo racemosa presents with broken rings of erythema in young to middle-aged women and primarily affects the trunk and proximal limbs. It is associated with an underlying condition such as polyarteritis nodosa and less commonly with lupus erythematosus with antiphospholipid or Sneddon syndrome.15,17 Cutaneous T-cell lymphoma typically manifests with poikilodermatous patches larger than the palm, especially in covered areas of skin.18 Cutis marmorata is transient and temperature dependent.9

The key intervention for EAI is removal of the offending heat source.2 Patients should be counseled that the erythema and hyperpigmentation may take months to years to resolve. Topical hydroquinone or tretinoin may be used in cases of persistent hyperpigmentation.19 Patients who continue to use heating pads for long-standing pain should be advised to limit their use to short intervals without occlusion. If malignancy is a concern, a biopsy should be performed.20

The Diagnosis: Erythema Ab Igne

Based on the patient's long-standing history of back pain treated with heating pads as well as the normal laboratory findings and skin examination, a diagnosis of erythema ab igne (EAI) was made.

Erythema ab igne presents as reticulated brownish erythema or hyperpigmentation on sites exposed to prolonged use of heat sources such as heating pads, laptops, and space heaters. Erythema ab igne most commonly affects the lower back, thighs, or legs1-6; however, EAI can appear on atypical sites such as the forehead and eyebrows due to newer technology (eg, virtual reality headsets).7 The level of heat required for EAI to occur is below the threshold for thermal burns (<45 °C [113 °F]).1 Erythema ab igne can occur at any age, and woman are more commonly affected than men.8 The pathophysiology currently is unknown; however, recurrent and prolonged heat exposure may damage superficial vessels. As a result, hemosiderin accumulates in the skin, and hyperpigmentation subsequently occurs.9

The diagnosis of EAI is clinical, and early stages of the rash present as blanching reticulated erythema in areas associated with heat exposure. If the offending source of heat is not removed, EAI can progress to nonblanching, fixed, hyperpigmented plaques with skin atrophy, bullae, or hyperkeratosis. Patients often are asymptomatic; however, mild burning may occur.2 Histopathology reveals cellular atypia, epidermal atrophy, dilation of dermal blood vessels, a minute inflammatory infiltrate, and keratinocyte apoptosis.10 Skin biopsy may be necessary in cases of suspected malignancy due to chronic heat exposure. Lesions that ulcerate or evolve should raise suspicion for malignancy.11 Squamous cell carcinoma is the most common malignancy associated with EAI; other malignancies that may manifest include basal cell carcinoma, Merkel cell carcinoma, or cutaneous marginal zone lymphoma.2,12-14

Erythema ab igne often is mistaken for livedo reticularis, which appears more erythematous without hyperpigmentation or epidermal changes and may be associated with a pathologic state.15 The differential diagnosis in our patient, who was in her 40s with a history of fatigue and joint pain, included livedo reticularis associated with lupus; however, the history of heating pad use, normal laboratory findings, and presence of epidermal changes suggested EAI. Lupus typically affects the hand and knee joints.16 Additionally, livedo reticularis more commonly appears on the legs.15

Other differentials for EAI include livedo racemosa, cutaneous T-cell lymphoma, and cutis marmorata. Livedo racemosa presents with broken rings of erythema in young to middle-aged women and primarily affects the trunk and proximal limbs. It is associated with an underlying condition such as polyarteritis nodosa and less commonly with lupus erythematosus with antiphospholipid or Sneddon syndrome.15,17 Cutaneous T-cell lymphoma typically manifests with poikilodermatous patches larger than the palm, especially in covered areas of skin.18 Cutis marmorata is transient and temperature dependent.9

The key intervention for EAI is removal of the offending heat source.2 Patients should be counseled that the erythema and hyperpigmentation may take months to years to resolve. Topical hydroquinone or tretinoin may be used in cases of persistent hyperpigmentation.19 Patients who continue to use heating pads for long-standing pain should be advised to limit their use to short intervals without occlusion. If malignancy is a concern, a biopsy should be performed.20

References
  1. Wipf AJ, Brown MR. Malignant transformation of erythema ab igne. JAAD Case Rep. 2022;26:85-87. doi:10.1016/j.jdcr.2022.06.018
  2. Sigmon JR, Cantrell J, Teague D, et al. Poorly differentiated carcinoma arising in the setting of erythema ab igne. Am J Dermatopathol. 2013;35:676-678. doi:10.1097/DAD.0b013e3182871648
  3. Patel DP. The evolving nomenclature of erythema ab igne-redness from fire. JAMA Dermatol. 2017;153:685. doi:10.1001/jamadermatol.2017.2021
  4. Arnold AW, Itin PH. Laptop computer-induced erythema ab igne in a child and review of the literature. Pediatrics. 2010;126:E1227-E1230. doi:10.1542/peds.2010-1390
  5. Riahi RR, Cohen PR. Laptop-induced erythema ab igne: report and review of literature. Dermatol Online J. 2012;18:5.
  6. Haleem Z, Philip J, Muhammad S. Erythema ab igne: a rare presentation of toasted skin syndrome with the use of a space heater. Cureus. 2021;13:e13401. doi:10.7759/cureus.13401
  7. Moreau T, Benzaquen M, Gueissaz F. Erythema ab igne after using a virtual reality headset: a new phenomenon to know. J Eur Acad Dermatol Venereol. 2022;36:E932-E933. doi:10.1111/jdv.18371
  8. Ozturk M, An I. Clinical features and etiology of patients with erythema ab igne: a retrospective multicenter study. J Cosmet Dermatol. 2020;19:1774-1779. doi:10.1111/jocd.13210
  9. Gmuca S, Yu J, Weiss PF, et al. Erythema ab igne in an adolescent with chronic pain: an alarming cutaneous eruption from heat exposure. Pediatr Emerg Care. 2020;36:E236-E238. doi:10.1097 /PEC.0000000000001460
  10. Wells A, Desai A, Rudnick EW, et al. Erythema ab igne with features resembling keratosis lichenoides chronica. J Cutan Pathol. 2021;48:151-153. doi:10.1111/cup.13885
  11. Milchak M, Smucker J, Chung CG, et al. Erythema ab igne due to heating pad use: a case report and review of clinical presentation, prevention, and complications. Case Rep Med. 2016;2016:1862480. doi:10.1155/2016/1862480
  12. Daneshvar E, Seraji S, Kamyab-Hesari K, et al. Basal cell carcinoma associated with erythema ab igne. Dermatol Online J. 2020;26:13030 /qt3kz985b4.
  13. Jones CS, Tyring SK, Lee PC, et al. Development of neuroendocrine (Merkel cell) carcinoma mixed with squamous cell carcinoma in erythema ab igne. Arch Dermatol. 1988;124:110-113.
  14. Wharton J, Roffwarg D, Miller J, et al. Cutaneous marginal zone lymphoma arising in the setting of erythema ab igne. J Am Acad Dermatol. 2010;62:1080-1081. doi:10.1016/j.jaad.2009.08.005
  15. Sajjan VV, Lunge S, Swamy MB, et al. Livedo reticularis: a review of the literature. Indian Dermatol Online J. 2015;6:315-321. doi:10.4103 /2229-5178.164493
  16. Grossman JM. Lupus arthritis. Best Pract Res Clin Rheumatol. 2009;23:495-506. doi:10.1016/j.berh.2009.04.003
  17. Aria AB, Chen L, Silapunt S. Erythema ab igne from heating pad use: a report of three clinical cases and a differential diagnosis. Cureus. 2018;10:E2635. doi:10.7759/cureus.2635
  18. Wilcox RA. Cutaneous T-cell lymphoma: 2017 update on diagnosis, risk-stratification, and management. Am J Hematol. 2017;92:1085-1102. doi:10.1002/ajh.24876
  19. Pennitz A, Kinberger M, Avila Valle G, et al. Self-applied topical interventions for melasma: a systematic review and meta-analysis of data from randomized, investigator-blinded clinical trials. Br J Dermatol. 2022;187:309-317.
  20. Sahl WJ, Taira JW. Erythema ab igne: treatment with 5-fluorouracil cream. J Am Acad Dermatol. 1992;27:109-110.
References
  1. Wipf AJ, Brown MR. Malignant transformation of erythema ab igne. JAAD Case Rep. 2022;26:85-87. doi:10.1016/j.jdcr.2022.06.018
  2. Sigmon JR, Cantrell J, Teague D, et al. Poorly differentiated carcinoma arising in the setting of erythema ab igne. Am J Dermatopathol. 2013;35:676-678. doi:10.1097/DAD.0b013e3182871648
  3. Patel DP. The evolving nomenclature of erythema ab igne-redness from fire. JAMA Dermatol. 2017;153:685. doi:10.1001/jamadermatol.2017.2021
  4. Arnold AW, Itin PH. Laptop computer-induced erythema ab igne in a child and review of the literature. Pediatrics. 2010;126:E1227-E1230. doi:10.1542/peds.2010-1390
  5. Riahi RR, Cohen PR. Laptop-induced erythema ab igne: report and review of literature. Dermatol Online J. 2012;18:5.
  6. Haleem Z, Philip J, Muhammad S. Erythema ab igne: a rare presentation of toasted skin syndrome with the use of a space heater. Cureus. 2021;13:e13401. doi:10.7759/cureus.13401
  7. Moreau T, Benzaquen M, Gueissaz F. Erythema ab igne after using a virtual reality headset: a new phenomenon to know. J Eur Acad Dermatol Venereol. 2022;36:E932-E933. doi:10.1111/jdv.18371
  8. Ozturk M, An I. Clinical features and etiology of patients with erythema ab igne: a retrospective multicenter study. J Cosmet Dermatol. 2020;19:1774-1779. doi:10.1111/jocd.13210
  9. Gmuca S, Yu J, Weiss PF, et al. Erythema ab igne in an adolescent with chronic pain: an alarming cutaneous eruption from heat exposure. Pediatr Emerg Care. 2020;36:E236-E238. doi:10.1097 /PEC.0000000000001460
  10. Wells A, Desai A, Rudnick EW, et al. Erythema ab igne with features resembling keratosis lichenoides chronica. J Cutan Pathol. 2021;48:151-153. doi:10.1111/cup.13885
  11. Milchak M, Smucker J, Chung CG, et al. Erythema ab igne due to heating pad use: a case report and review of clinical presentation, prevention, and complications. Case Rep Med. 2016;2016:1862480. doi:10.1155/2016/1862480
  12. Daneshvar E, Seraji S, Kamyab-Hesari K, et al. Basal cell carcinoma associated with erythema ab igne. Dermatol Online J. 2020;26:13030 /qt3kz985b4.
  13. Jones CS, Tyring SK, Lee PC, et al. Development of neuroendocrine (Merkel cell) carcinoma mixed with squamous cell carcinoma in erythema ab igne. Arch Dermatol. 1988;124:110-113.
  14. Wharton J, Roffwarg D, Miller J, et al. Cutaneous marginal zone lymphoma arising in the setting of erythema ab igne. J Am Acad Dermatol. 2010;62:1080-1081. doi:10.1016/j.jaad.2009.08.005
  15. Sajjan VV, Lunge S, Swamy MB, et al. Livedo reticularis: a review of the literature. Indian Dermatol Online J. 2015;6:315-321. doi:10.4103 /2229-5178.164493
  16. Grossman JM. Lupus arthritis. Best Pract Res Clin Rheumatol. 2009;23:495-506. doi:10.1016/j.berh.2009.04.003
  17. Aria AB, Chen L, Silapunt S. Erythema ab igne from heating pad use: a report of three clinical cases and a differential diagnosis. Cureus. 2018;10:E2635. doi:10.7759/cureus.2635
  18. Wilcox RA. Cutaneous T-cell lymphoma: 2017 update on diagnosis, risk-stratification, and management. Am J Hematol. 2017;92:1085-1102. doi:10.1002/ajh.24876
  19. Pennitz A, Kinberger M, Avila Valle G, et al. Self-applied topical interventions for melasma: a systematic review and meta-analysis of data from randomized, investigator-blinded clinical trials. Br J Dermatol. 2022;187:309-317.
  20. Sahl WJ, Taira JW. Erythema ab igne: treatment with 5-fluorouracil cream. J Am Acad Dermatol. 1992;27:109-110.
Issue
Cutis - 113(6)
Issue
Cutis - 113(6)
Page Number
E8-E10
Page Number
E8-E10
Publications
Publications
Topics
Article Type
Display Headline
Reticulated Brownish Erythema on the Lower Back
Display Headline
Reticulated Brownish Erythema on the Lower Back
Sections
Questionnaire Body

A 42-year-old woman presented with an asymptomatic, erythematous, lacelike rash on the lower back of 8 months’ duration that was first noticed by her husband. The patient had a long-standing history of chronic fatigue and lower back pain treated with acetaminophen, diclofenac gel, and heating pads. Physical examination revealed reticulated brownish erythema confined to the lower back. Laboratory findings were unremarkable.

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Fri, 06/14/2024 - 10:00
Un-Gate On Date
Fri, 06/14/2024 - 10:00
Use ProPublica
CFC Schedule Remove Status
Fri, 06/14/2024 - 10:00
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

Aquatic Antagonists: Dermatologic Injuries From Sea Urchins (Echinoidea)

Article Type
Changed
Fri, 06/14/2024 - 12:41
Display Headline
Aquatic Antagonists: Dermatologic Injuries From Sea Urchins (Echinoidea)

Sea urchins—members of the phylum Echinodermata and the class Echinoidea—are spiny marine invertebrates. Their consumption of fleshy algae makes them essential players in maintaining reef ecosystems.1,2 Echinoids, a class that includes heart urchins and sand dollars, are ubiquitous in benthic marine environments, both free floating and rock boring, and inhabit a wide range of latitudes spanning from polar oceans to warm seas.3 Despite their immobility and nonaggression, sea urchin puncture wounds are common among divers, snorkelers, swimmers, surfers, and fishers who accidentally come into contact with their sharp spines. Although the epidemiology of sea urchin exposure and injury is difficult to assess, the American Association of Poison Control Centers’ most recent annual report in 2022 documents approximately 1426 annual aquatic bites and/or envenomations.4

Sea Urchin Morphology and Toxicity

Echinoderms (a term of Greek origin meaning spiny skin) share a radially symmetric calcium carbonate skeleton (termed stereom) that is supported by collagenous ligaments.1 Sea urchins possess spines composed of calcite crystals, which radiate from their body and play a role in locomotion and defense against predators—namely sea otters, starfish/sea stars, wolf eels, and triggerfish, among others (Figure).5 These brittle spines can easily penetrate human skin and subsequently break off the sea urchin body. Most species of sea urchins possess solid spines, but a small percentage (80 of approximately 700 extant species) have hollow spines containing various toxic substances.6 Penetration and systemic absorption of the toxins within these spines can generate severe systemic responses.

The venomous flower urchin (Toxopneustes pileolus), found in the Indian and Pacific oceans, is one of the more common species known to produce a systemic reaction involving neuromuscular blockage.7-9 The most common species harvested off the Pacific coast of the United States—Strongylocentrotus purpuratus (purple sea urchin) and Strongylocentrotus franciscanus (red sea urchins)—are not inherently venomous.8

Purple sea urchin (Strongylocentrotus purpuratus). Photograph courtesy of the South Carolina Aquarium (Charleston, South Carolina).


Both the sea urchin body and spines are covered in a unique epithelium thought to be responsible for the majority of their proinflammatory and pronociceptive properties. Epithelial compounds identified include serotonin, histamines, steroids, glycosides, hemolysins, proteases, and bradykininlike and cholinergic substances.5,7 Additionally, certain sea urchin species possess 3-pronged pincerlike organs at the base of spines called pedicellariae, which are used in feeding.10 Skin penetration by the pedicellariae is especially dangerous, as they tightly adhere to wounds and contain venom-producing organs that allow them to continue injecting toxins after their detachment from the sea urchin body.11

Presentation and Diagnosis of Sea Urchin Injuries

Sea urchin injuries have a wide range of manifestations depending on the number of spines involved, the presence of venom, the depth and location of spine penetration, the duration of spine retention in the skin, and the time before treatment initiation. The most common site of sea urchin injury unsurprisingly is the lower extremities and feet, often in the context of divers and swimmers walking across the sea floor. The hands are another frequently injured site, along with the legs, arms, back, scalp, and even oral mucosa.11

Although clinical history and presentation frequently reveal the mechanism of aquatic injury, patients often are unsure of the agent to which they were exposed and may be unaware of retained foreign bodies. Dermoscopy can distinguish the distinct lines radiating from the core of sea urchin spines from other foreign bodies lodged within the skin.6 It also can be used to locate spines for removal or for their analysis following punch biopsy.6,12 The radiopaque nature of sea urchin spines makes radiography and magnetic resonance imaging useful tools in assessment of periarticular soft-tissue damage and spine removal.8,11,13 Ultrasonography can reveal spines that no longer appear on radiography due to absorption by human tissue.14

Immediate Dermatologic Effects

Sea urchin injuries can be broadly categorized into immediate and delayed reactions. Immediate manifestations of contact with sea urchin spines include localized pain, bleeding, erythema, myalgia, and edema at the site of injury that can last from a few hours to 1 week without proper wound care and spine removal.5 Systemic symptoms ranging from dizziness, lightheadedness, paresthesia, aphonia, paralysis, coma, and death generally are only seen following injuries from venomous species, attachment of pedicellariae, injuries involving neurovascular structures, or penetration by more than 15 spines.7,11

Initial treatment includes soaking the wound in hot water (113 °F [45 °C]) for 30 to 90 minutes and subsequently removing spines and pedicellariae to prevent development of delayed reactions.5,15,16 The compounds in the sea urchin epithelium are heat labile and will be inactivated upon soaking in hot water.16 Extraction of spines can be difficult, as they are brittle and easily break in the skin. Successful removal has been reported using forceps and a hypodermic needle as well as excision; both approaches may require local anesthesia.8,17 Another technique involves freezing the localized area with liquid nitrogen to allow easier removal upon skin blistering.18 Punch biopsy also has been utilized as an effective means of ensuring all spiny fragments are removed.9,19,20 These spines often cause black or purple tattoolike staining at the puncture site, which can persist for a few days after spine extraction.8 Ablation using the erbium-doped:YAG laser may be helpful for removal of associated pigment.21,22

Delayed Dermatologic Effects

Delayed reactions to sea urchin injuries often are attributable to prolonged retention of spines in the skin. Granulomatous reactions typically manifest 2 weeks after injury as firm nonsuppurative nodules with central umbilication and a hyperkeratotic surface.7 These nodules may or may not be painful. Histopathology most often reveals foreign body and sarcoidal-type granulomatous reactions. However, tuberculoid, necrobiotic, and suppurative granulomas also may develop.13 Other microscopic features include inflammatory reactions, suppurative dermatitis, focal necrosis, and microabscesses.23 Wounds with progression to granulomatous disease often require surgical debridement.

Other more serious sequalae can result from involvement of joint capsules, especially in the hands and feet. Sea urchin injury involving joint spaces should be treated aggressively, as progression to inflammatory or infectious synovitis and tenosynovitis can cause irreversible loss of joint function. Inflammatory synovitis occurs 1 to 2 months on average after injury following a period of minimal symptoms and begins as a gradual increase in joint swelling and decrease in range of motion.8 Infectious tenosynovitis manifests quite similarly. Although suppurative etiologies generally progress with a more acute onset, certain infectious organisms (eg, Mycobacterium) take on an indolent course and should not be overlooked as a cause of delayed symptoms.8 The Kavanel cardinal signs are a sensitive tool used in the diagnosis of infectious flexor sheath tenosynovitis.8,24 If suspicion for joint infection is high, emergency referral should be made for debridement and culture-guided antibiotic therapy. Left untreated, infectious tenosynovitis can result in tendon necrosis or rupture, digit necrosis, and systemic infection.24 Patients with joint involvement should be referred to specialty care (eg, hand surgeon), as they often require synovectomy and surgical removal of foreign material.8

From 1 month to 1 year after injury, prolonged granulomatous synovitis of the hand may eventually lead to joint destruction known as “sea urchin arthritis.” These patients present with decreased range of motion and numerous nodules on the hand with a hyperkeratotic surface. Radiography reveals joint space narrowing, osteolysis, subchondral sclerosis, and periosteal reaction. Synovectomy and debridement are necessary to prevent irreversible joint damage or the need for arthrodesis and bone grafting.24

Other Treatment Considerations

Other important considerations in the care of sea urchin spine injuries include assessment of tetanus immunization status and administration of necessary prophylaxis as soon as possible, even in delayed presentations (Table).16,25 Cultures should be taken only if infection is suspected. Prophylactic antibiotics are not recommended unless the patient is immunocompromised or otherwise has impaired wound healing. If a patient presents with systemic symptoms, they should be referred to an emergency care facility for further management.

Final Thoughts

Sea urchin injuries can lead to serious complications if not diagnosed quickly and treated properly. Retention of sea urchin spines in the deep tissues and joint spaces may lead to granulomas, inflammatory and infectious tenosynovitis (including mycobacterial infection), and sea urchin arthritis requiring surgical debridement and possible irreversible joint damage, up to a year after initial injury. Patients should be educated on the possibility of developing these delayed reactions and instructed to seek immediate care. Joint deformities, range-of-motion deficits, and involvement of neurovascular structures should be considered emergent and referred for proper management. Shoes and diving gear offer some protection but are easily penetrable by sharp sea urchin spines. Preventive focus should be aimed at educating patients and providers on the importance of prompt spine removal upon injury. Although dermatologic and systemic manifestations vary widely, a thorough history, physical examination, and appropriate use of imaging modalities can facilitate accurate diagnosis and guide treatment.

References
  1. Amemiya CT, Miyake T, Rast JP. Echinoderms. Curr Biol. 2005;15:R944-R946. doi:10.1016/j.cub.2005.11.026
  2. Koch NM, Coppard SE, Lessios HA, et al. A phylogenomic resolution of the sea urchin tree of life. BMC Evol Biol. 2018;18:189. doi:10.1186/s12862-018-1300-4
  3. Amir Y, Insler M, Giller A, et al. Senescence and longevity of sea urchins. Genes (Basel). 2020;11:573. doi:10.3390/genes11050573
  4. Gummin DD, Mowry JB, Beuhler MC, et al. 2022 Annual Report of the National Poison Data System® (NPDS) from America's Poison Centers®: 40th annual report. Clin Toxicol (Phila). 2023;61:717-939. doi:10.1080/15563650.2023.2268981
  5. Gelman Y, Kong EL, Murphy-Lavoie HM. Sea urchin toxicity. In: StatPearls [Internet]. StatPearls Publishing; 2021.
  6. Suarez-Conde MF, Vallone MG, González VM, et al. Sea urchin skin lesions: a case report. Dermatol Pract Concept. 2021;11:E2021009. doi:10.5826/dpc.1102a09
  7. Al-Kathiri L, Al-Najjar T, Sulaiman I. Sea urchin granuloma of the hands: a case report. Oman Med J. 2019;34:350-353. doi:10.5001/omj.2019.68
  8. Dahl WJ, Jebson P, Louis DS. Sea urchin injuries to the hand: a case report and review of the literature. Iowa Orthop J. 2010;30:153-156.
  9. Hatakeyama T, Ichise A, Unno H, et al. Carbohydrate recognition by the rhamnose-binding lectin SUL-I with a novel three-domain structure isolated from the venom of globiferous pedicellariae of the flower sea urchin Toxopneustes pileolus. Protein Sci. 2017;26:1574-1583. doi:10.1002/pro.3185
  10. Balhara KS, Stolbach A. Marine envenomations. Emerg Med Clin North Am. 2014;32:223-243. doi:10.1016/j.emc.2013.09.009
  11. Schwartz Z, Cohen M, Lipner SR. Sea urchin injuries: a review and clinical approach algorithm. J Dermatolog Treat. 2021;32:150-156. doi:10.1080/09546634.2019.1638884
  12. Park SJ, Park JW, Choi SY, et al. Use of dermoscopy after punch removal of a veiled sea urchin spine. Dermatol Ther. 2021;34:E14947. doi:10.1111/dth.14947
  13. Wada T, Soma T, Gaman K, et al. Sea urchin spine arthritis of the hand. J Hand Surg Am. 2008;33:398-401. doi:10.1016/j.jhsa.2007.11.016
  14. Groleau S, Chhem RK, Younge D, et al. Ultrasonography of foreign-body tenosynovitis. Can Assoc Radiol J. 1992;43:454-456. 
  15. Hornbeak KB, Auerbach PS. Marine envenomation. Emerg Med Clin North Am. 2017;35:321-337. doi:10.1016/j.emc.2016.12.004
  16. Noonburg GE. Management of extremity trauma and related infections occurring in the aquatic environment. J Am Acad Orthop Surg. 2005;13:243-253. doi:10.5435/00124635-200507000-00004
  17. Haddad Junior V. Observation of initial clinical manifestations and repercussions from the treatment of 314 human injuries caused by black sea urchins (Echinometra lucunter) on the southeastern Brazilian coast. Rev Soc Bras Med Trop. 2012;45:390-392. doi:10.1590/s0037-86822012000300021
  18. Gargus MD, Morohashi DK. A sea-urchin spine chilling remedy. N Engl J Med. 2012;367:1867-1868. doi:10.1056/NEJMc1209382
  19. Sjøberg T, de Weerd L. The usefulness of a skin biopsy punch to remove sea urchin spines. ANZ J Surg. 2010;80:383. doi:10.1111/j.1445-2197.2010.05296.x
  20. Cardenas-de la Garza JA, Cuellar-Barboza A, Ancer-Arellano J, et al. Classic dermatological tools: foreign body removal with punch biopsy.J Am Acad Dermatol. 2019;81:E93-E94. doi:10.1016/j.jaad.2018.10.038
  21. Gungor S, Tarikçi N, Gokdemir G. Removal of sea urchin spines using erbium-doped yttrium aluminum garnet ablation. Dermatol Surg. 2012;38:508-510. doi:10.1111/j.1524-4725.2011.02259.x
  22. Böer A, Ochsendorf FR, Beier C, et al. Effective removal of sea-urchin spines by erbium:YAG laser ablation. Br J Dermatol. 2001;145:169-170. doi:10.1046/j.1365-2133.2001.04306.x
  23. De La Torre C, Toribio J. Sea-urchin granuloma: histologic profile. a pathologic study of 50 biopsies. J Cutan Pathol. 2001;28:223-228. doi:10.1034/j.1600-0560.2001.028005223.x
  24. Yi A, Kennedy C, Chia B, et al. Radiographic soft tissue thickness differentiating pyogenic flexor tenosynovitis from other finger infections. J Hand Surg Am. 2019;44:394-399. doi:10.1016/j.jhsa.2019.01.013
  25. Callison C, Nguyen H. Tetanus prophylaxis. In: StatPearls [Internet]. StatPearls Publishing; 2022.
Article PDF
Author and Disclosure Information

 

From the Medical University of South Carolina, Charleston. Dr. Brailsford is from the College of Medicine, and Dr. Elston is from the Department of Dermatology and Dermatologic Surgery.

The authors report no conflict of interest.

Correspondence: Caroline J. Brailsford, MD, Medical University of South Carolina, 135 Rutledge Ave, 11th Floor, Charleston, SC 29425-5780 (cjbrailsford@gmail.com).

Cutis. 2024 June;113(6):255-257. doi:10.12788/cutis.1034

Issue
Cutis - 113(6)
Publications
Topics
Page Number
255-257
Sections
Author and Disclosure Information

 

From the Medical University of South Carolina, Charleston. Dr. Brailsford is from the College of Medicine, and Dr. Elston is from the Department of Dermatology and Dermatologic Surgery.

The authors report no conflict of interest.

Correspondence: Caroline J. Brailsford, MD, Medical University of South Carolina, 135 Rutledge Ave, 11th Floor, Charleston, SC 29425-5780 (cjbrailsford@gmail.com).

Cutis. 2024 June;113(6):255-257. doi:10.12788/cutis.1034

Author and Disclosure Information

 

From the Medical University of South Carolina, Charleston. Dr. Brailsford is from the College of Medicine, and Dr. Elston is from the Department of Dermatology and Dermatologic Surgery.

The authors report no conflict of interest.

Correspondence: Caroline J. Brailsford, MD, Medical University of South Carolina, 135 Rutledge Ave, 11th Floor, Charleston, SC 29425-5780 (cjbrailsford@gmail.com).

Cutis. 2024 June;113(6):255-257. doi:10.12788/cutis.1034

Article PDF
Article PDF

Sea urchins—members of the phylum Echinodermata and the class Echinoidea—are spiny marine invertebrates. Their consumption of fleshy algae makes them essential players in maintaining reef ecosystems.1,2 Echinoids, a class that includes heart urchins and sand dollars, are ubiquitous in benthic marine environments, both free floating and rock boring, and inhabit a wide range of latitudes spanning from polar oceans to warm seas.3 Despite their immobility and nonaggression, sea urchin puncture wounds are common among divers, snorkelers, swimmers, surfers, and fishers who accidentally come into contact with their sharp spines. Although the epidemiology of sea urchin exposure and injury is difficult to assess, the American Association of Poison Control Centers’ most recent annual report in 2022 documents approximately 1426 annual aquatic bites and/or envenomations.4

Sea Urchin Morphology and Toxicity

Echinoderms (a term of Greek origin meaning spiny skin) share a radially symmetric calcium carbonate skeleton (termed stereom) that is supported by collagenous ligaments.1 Sea urchins possess spines composed of calcite crystals, which radiate from their body and play a role in locomotion and defense against predators—namely sea otters, starfish/sea stars, wolf eels, and triggerfish, among others (Figure).5 These brittle spines can easily penetrate human skin and subsequently break off the sea urchin body. Most species of sea urchins possess solid spines, but a small percentage (80 of approximately 700 extant species) have hollow spines containing various toxic substances.6 Penetration and systemic absorption of the toxins within these spines can generate severe systemic responses.

The venomous flower urchin (Toxopneustes pileolus), found in the Indian and Pacific oceans, is one of the more common species known to produce a systemic reaction involving neuromuscular blockage.7-9 The most common species harvested off the Pacific coast of the United States—Strongylocentrotus purpuratus (purple sea urchin) and Strongylocentrotus franciscanus (red sea urchins)—are not inherently venomous.8

Purple sea urchin (Strongylocentrotus purpuratus). Photograph courtesy of the South Carolina Aquarium (Charleston, South Carolina).


Both the sea urchin body and spines are covered in a unique epithelium thought to be responsible for the majority of their proinflammatory and pronociceptive properties. Epithelial compounds identified include serotonin, histamines, steroids, glycosides, hemolysins, proteases, and bradykininlike and cholinergic substances.5,7 Additionally, certain sea urchin species possess 3-pronged pincerlike organs at the base of spines called pedicellariae, which are used in feeding.10 Skin penetration by the pedicellariae is especially dangerous, as they tightly adhere to wounds and contain venom-producing organs that allow them to continue injecting toxins after their detachment from the sea urchin body.11

Presentation and Diagnosis of Sea Urchin Injuries

Sea urchin injuries have a wide range of manifestations depending on the number of spines involved, the presence of venom, the depth and location of spine penetration, the duration of spine retention in the skin, and the time before treatment initiation. The most common site of sea urchin injury unsurprisingly is the lower extremities and feet, often in the context of divers and swimmers walking across the sea floor. The hands are another frequently injured site, along with the legs, arms, back, scalp, and even oral mucosa.11

Although clinical history and presentation frequently reveal the mechanism of aquatic injury, patients often are unsure of the agent to which they were exposed and may be unaware of retained foreign bodies. Dermoscopy can distinguish the distinct lines radiating from the core of sea urchin spines from other foreign bodies lodged within the skin.6 It also can be used to locate spines for removal or for their analysis following punch biopsy.6,12 The radiopaque nature of sea urchin spines makes radiography and magnetic resonance imaging useful tools in assessment of periarticular soft-tissue damage and spine removal.8,11,13 Ultrasonography can reveal spines that no longer appear on radiography due to absorption by human tissue.14

Immediate Dermatologic Effects

Sea urchin injuries can be broadly categorized into immediate and delayed reactions. Immediate manifestations of contact with sea urchin spines include localized pain, bleeding, erythema, myalgia, and edema at the site of injury that can last from a few hours to 1 week without proper wound care and spine removal.5 Systemic symptoms ranging from dizziness, lightheadedness, paresthesia, aphonia, paralysis, coma, and death generally are only seen following injuries from venomous species, attachment of pedicellariae, injuries involving neurovascular structures, or penetration by more than 15 spines.7,11

Initial treatment includes soaking the wound in hot water (113 °F [45 °C]) for 30 to 90 minutes and subsequently removing spines and pedicellariae to prevent development of delayed reactions.5,15,16 The compounds in the sea urchin epithelium are heat labile and will be inactivated upon soaking in hot water.16 Extraction of spines can be difficult, as they are brittle and easily break in the skin. Successful removal has been reported using forceps and a hypodermic needle as well as excision; both approaches may require local anesthesia.8,17 Another technique involves freezing the localized area with liquid nitrogen to allow easier removal upon skin blistering.18 Punch biopsy also has been utilized as an effective means of ensuring all spiny fragments are removed.9,19,20 These spines often cause black or purple tattoolike staining at the puncture site, which can persist for a few days after spine extraction.8 Ablation using the erbium-doped:YAG laser may be helpful for removal of associated pigment.21,22

Delayed Dermatologic Effects

Delayed reactions to sea urchin injuries often are attributable to prolonged retention of spines in the skin. Granulomatous reactions typically manifest 2 weeks after injury as firm nonsuppurative nodules with central umbilication and a hyperkeratotic surface.7 These nodules may or may not be painful. Histopathology most often reveals foreign body and sarcoidal-type granulomatous reactions. However, tuberculoid, necrobiotic, and suppurative granulomas also may develop.13 Other microscopic features include inflammatory reactions, suppurative dermatitis, focal necrosis, and microabscesses.23 Wounds with progression to granulomatous disease often require surgical debridement.

Other more serious sequalae can result from involvement of joint capsules, especially in the hands and feet. Sea urchin injury involving joint spaces should be treated aggressively, as progression to inflammatory or infectious synovitis and tenosynovitis can cause irreversible loss of joint function. Inflammatory synovitis occurs 1 to 2 months on average after injury following a period of minimal symptoms and begins as a gradual increase in joint swelling and decrease in range of motion.8 Infectious tenosynovitis manifests quite similarly. Although suppurative etiologies generally progress with a more acute onset, certain infectious organisms (eg, Mycobacterium) take on an indolent course and should not be overlooked as a cause of delayed symptoms.8 The Kavanel cardinal signs are a sensitive tool used in the diagnosis of infectious flexor sheath tenosynovitis.8,24 If suspicion for joint infection is high, emergency referral should be made for debridement and culture-guided antibiotic therapy. Left untreated, infectious tenosynovitis can result in tendon necrosis or rupture, digit necrosis, and systemic infection.24 Patients with joint involvement should be referred to specialty care (eg, hand surgeon), as they often require synovectomy and surgical removal of foreign material.8

From 1 month to 1 year after injury, prolonged granulomatous synovitis of the hand may eventually lead to joint destruction known as “sea urchin arthritis.” These patients present with decreased range of motion and numerous nodules on the hand with a hyperkeratotic surface. Radiography reveals joint space narrowing, osteolysis, subchondral sclerosis, and periosteal reaction. Synovectomy and debridement are necessary to prevent irreversible joint damage or the need for arthrodesis and bone grafting.24

Other Treatment Considerations

Other important considerations in the care of sea urchin spine injuries include assessment of tetanus immunization status and administration of necessary prophylaxis as soon as possible, even in delayed presentations (Table).16,25 Cultures should be taken only if infection is suspected. Prophylactic antibiotics are not recommended unless the patient is immunocompromised or otherwise has impaired wound healing. If a patient presents with systemic symptoms, they should be referred to an emergency care facility for further management.

Final Thoughts

Sea urchin injuries can lead to serious complications if not diagnosed quickly and treated properly. Retention of sea urchin spines in the deep tissues and joint spaces may lead to granulomas, inflammatory and infectious tenosynovitis (including mycobacterial infection), and sea urchin arthritis requiring surgical debridement and possible irreversible joint damage, up to a year after initial injury. Patients should be educated on the possibility of developing these delayed reactions and instructed to seek immediate care. Joint deformities, range-of-motion deficits, and involvement of neurovascular structures should be considered emergent and referred for proper management. Shoes and diving gear offer some protection but are easily penetrable by sharp sea urchin spines. Preventive focus should be aimed at educating patients and providers on the importance of prompt spine removal upon injury. Although dermatologic and systemic manifestations vary widely, a thorough history, physical examination, and appropriate use of imaging modalities can facilitate accurate diagnosis and guide treatment.

Sea urchins—members of the phylum Echinodermata and the class Echinoidea—are spiny marine invertebrates. Their consumption of fleshy algae makes them essential players in maintaining reef ecosystems.1,2 Echinoids, a class that includes heart urchins and sand dollars, are ubiquitous in benthic marine environments, both free floating and rock boring, and inhabit a wide range of latitudes spanning from polar oceans to warm seas.3 Despite their immobility and nonaggression, sea urchin puncture wounds are common among divers, snorkelers, swimmers, surfers, and fishers who accidentally come into contact with their sharp spines. Although the epidemiology of sea urchin exposure and injury is difficult to assess, the American Association of Poison Control Centers’ most recent annual report in 2022 documents approximately 1426 annual aquatic bites and/or envenomations.4

Sea Urchin Morphology and Toxicity

Echinoderms (a term of Greek origin meaning spiny skin) share a radially symmetric calcium carbonate skeleton (termed stereom) that is supported by collagenous ligaments.1 Sea urchins possess spines composed of calcite crystals, which radiate from their body and play a role in locomotion and defense against predators—namely sea otters, starfish/sea stars, wolf eels, and triggerfish, among others (Figure).5 These brittle spines can easily penetrate human skin and subsequently break off the sea urchin body. Most species of sea urchins possess solid spines, but a small percentage (80 of approximately 700 extant species) have hollow spines containing various toxic substances.6 Penetration and systemic absorption of the toxins within these spines can generate severe systemic responses.

The venomous flower urchin (Toxopneustes pileolus), found in the Indian and Pacific oceans, is one of the more common species known to produce a systemic reaction involving neuromuscular blockage.7-9 The most common species harvested off the Pacific coast of the United States—Strongylocentrotus purpuratus (purple sea urchin) and Strongylocentrotus franciscanus (red sea urchins)—are not inherently venomous.8

Purple sea urchin (Strongylocentrotus purpuratus). Photograph courtesy of the South Carolina Aquarium (Charleston, South Carolina).


Both the sea urchin body and spines are covered in a unique epithelium thought to be responsible for the majority of their proinflammatory and pronociceptive properties. Epithelial compounds identified include serotonin, histamines, steroids, glycosides, hemolysins, proteases, and bradykininlike and cholinergic substances.5,7 Additionally, certain sea urchin species possess 3-pronged pincerlike organs at the base of spines called pedicellariae, which are used in feeding.10 Skin penetration by the pedicellariae is especially dangerous, as they tightly adhere to wounds and contain venom-producing organs that allow them to continue injecting toxins after their detachment from the sea urchin body.11

Presentation and Diagnosis of Sea Urchin Injuries

Sea urchin injuries have a wide range of manifestations depending on the number of spines involved, the presence of venom, the depth and location of spine penetration, the duration of spine retention in the skin, and the time before treatment initiation. The most common site of sea urchin injury unsurprisingly is the lower extremities and feet, often in the context of divers and swimmers walking across the sea floor. The hands are another frequently injured site, along with the legs, arms, back, scalp, and even oral mucosa.11

Although clinical history and presentation frequently reveal the mechanism of aquatic injury, patients often are unsure of the agent to which they were exposed and may be unaware of retained foreign bodies. Dermoscopy can distinguish the distinct lines radiating from the core of sea urchin spines from other foreign bodies lodged within the skin.6 It also can be used to locate spines for removal or for their analysis following punch biopsy.6,12 The radiopaque nature of sea urchin spines makes radiography and magnetic resonance imaging useful tools in assessment of periarticular soft-tissue damage and spine removal.8,11,13 Ultrasonography can reveal spines that no longer appear on radiography due to absorption by human tissue.14

Immediate Dermatologic Effects

Sea urchin injuries can be broadly categorized into immediate and delayed reactions. Immediate manifestations of contact with sea urchin spines include localized pain, bleeding, erythema, myalgia, and edema at the site of injury that can last from a few hours to 1 week without proper wound care and spine removal.5 Systemic symptoms ranging from dizziness, lightheadedness, paresthesia, aphonia, paralysis, coma, and death generally are only seen following injuries from venomous species, attachment of pedicellariae, injuries involving neurovascular structures, or penetration by more than 15 spines.7,11

Initial treatment includes soaking the wound in hot water (113 °F [45 °C]) for 30 to 90 minutes and subsequently removing spines and pedicellariae to prevent development of delayed reactions.5,15,16 The compounds in the sea urchin epithelium are heat labile and will be inactivated upon soaking in hot water.16 Extraction of spines can be difficult, as they are brittle and easily break in the skin. Successful removal has been reported using forceps and a hypodermic needle as well as excision; both approaches may require local anesthesia.8,17 Another technique involves freezing the localized area with liquid nitrogen to allow easier removal upon skin blistering.18 Punch biopsy also has been utilized as an effective means of ensuring all spiny fragments are removed.9,19,20 These spines often cause black or purple tattoolike staining at the puncture site, which can persist for a few days after spine extraction.8 Ablation using the erbium-doped:YAG laser may be helpful for removal of associated pigment.21,22

Delayed Dermatologic Effects

Delayed reactions to sea urchin injuries often are attributable to prolonged retention of spines in the skin. Granulomatous reactions typically manifest 2 weeks after injury as firm nonsuppurative nodules with central umbilication and a hyperkeratotic surface.7 These nodules may or may not be painful. Histopathology most often reveals foreign body and sarcoidal-type granulomatous reactions. However, tuberculoid, necrobiotic, and suppurative granulomas also may develop.13 Other microscopic features include inflammatory reactions, suppurative dermatitis, focal necrosis, and microabscesses.23 Wounds with progression to granulomatous disease often require surgical debridement.

Other more serious sequalae can result from involvement of joint capsules, especially in the hands and feet. Sea urchin injury involving joint spaces should be treated aggressively, as progression to inflammatory or infectious synovitis and tenosynovitis can cause irreversible loss of joint function. Inflammatory synovitis occurs 1 to 2 months on average after injury following a period of minimal symptoms and begins as a gradual increase in joint swelling and decrease in range of motion.8 Infectious tenosynovitis manifests quite similarly. Although suppurative etiologies generally progress with a more acute onset, certain infectious organisms (eg, Mycobacterium) take on an indolent course and should not be overlooked as a cause of delayed symptoms.8 The Kavanel cardinal signs are a sensitive tool used in the diagnosis of infectious flexor sheath tenosynovitis.8,24 If suspicion for joint infection is high, emergency referral should be made for debridement and culture-guided antibiotic therapy. Left untreated, infectious tenosynovitis can result in tendon necrosis or rupture, digit necrosis, and systemic infection.24 Patients with joint involvement should be referred to specialty care (eg, hand surgeon), as they often require synovectomy and surgical removal of foreign material.8

From 1 month to 1 year after injury, prolonged granulomatous synovitis of the hand may eventually lead to joint destruction known as “sea urchin arthritis.” These patients present with decreased range of motion and numerous nodules on the hand with a hyperkeratotic surface. Radiography reveals joint space narrowing, osteolysis, subchondral sclerosis, and periosteal reaction. Synovectomy and debridement are necessary to prevent irreversible joint damage or the need for arthrodesis and bone grafting.24

Other Treatment Considerations

Other important considerations in the care of sea urchin spine injuries include assessment of tetanus immunization status and administration of necessary prophylaxis as soon as possible, even in delayed presentations (Table).16,25 Cultures should be taken only if infection is suspected. Prophylactic antibiotics are not recommended unless the patient is immunocompromised or otherwise has impaired wound healing. If a patient presents with systemic symptoms, they should be referred to an emergency care facility for further management.

Final Thoughts

Sea urchin injuries can lead to serious complications if not diagnosed quickly and treated properly. Retention of sea urchin spines in the deep tissues and joint spaces may lead to granulomas, inflammatory and infectious tenosynovitis (including mycobacterial infection), and sea urchin arthritis requiring surgical debridement and possible irreversible joint damage, up to a year after initial injury. Patients should be educated on the possibility of developing these delayed reactions and instructed to seek immediate care. Joint deformities, range-of-motion deficits, and involvement of neurovascular structures should be considered emergent and referred for proper management. Shoes and diving gear offer some protection but are easily penetrable by sharp sea urchin spines. Preventive focus should be aimed at educating patients and providers on the importance of prompt spine removal upon injury. Although dermatologic and systemic manifestations vary widely, a thorough history, physical examination, and appropriate use of imaging modalities can facilitate accurate diagnosis and guide treatment.

References
  1. Amemiya CT, Miyake T, Rast JP. Echinoderms. Curr Biol. 2005;15:R944-R946. doi:10.1016/j.cub.2005.11.026
  2. Koch NM, Coppard SE, Lessios HA, et al. A phylogenomic resolution of the sea urchin tree of life. BMC Evol Biol. 2018;18:189. doi:10.1186/s12862-018-1300-4
  3. Amir Y, Insler M, Giller A, et al. Senescence and longevity of sea urchins. Genes (Basel). 2020;11:573. doi:10.3390/genes11050573
  4. Gummin DD, Mowry JB, Beuhler MC, et al. 2022 Annual Report of the National Poison Data System® (NPDS) from America's Poison Centers®: 40th annual report. Clin Toxicol (Phila). 2023;61:717-939. doi:10.1080/15563650.2023.2268981
  5. Gelman Y, Kong EL, Murphy-Lavoie HM. Sea urchin toxicity. In: StatPearls [Internet]. StatPearls Publishing; 2021.
  6. Suarez-Conde MF, Vallone MG, González VM, et al. Sea urchin skin lesions: a case report. Dermatol Pract Concept. 2021;11:E2021009. doi:10.5826/dpc.1102a09
  7. Al-Kathiri L, Al-Najjar T, Sulaiman I. Sea urchin granuloma of the hands: a case report. Oman Med J. 2019;34:350-353. doi:10.5001/omj.2019.68
  8. Dahl WJ, Jebson P, Louis DS. Sea urchin injuries to the hand: a case report and review of the literature. Iowa Orthop J. 2010;30:153-156.
  9. Hatakeyama T, Ichise A, Unno H, et al. Carbohydrate recognition by the rhamnose-binding lectin SUL-I with a novel three-domain structure isolated from the venom of globiferous pedicellariae of the flower sea urchin Toxopneustes pileolus. Protein Sci. 2017;26:1574-1583. doi:10.1002/pro.3185
  10. Balhara KS, Stolbach A. Marine envenomations. Emerg Med Clin North Am. 2014;32:223-243. doi:10.1016/j.emc.2013.09.009
  11. Schwartz Z, Cohen M, Lipner SR. Sea urchin injuries: a review and clinical approach algorithm. J Dermatolog Treat. 2021;32:150-156. doi:10.1080/09546634.2019.1638884
  12. Park SJ, Park JW, Choi SY, et al. Use of dermoscopy after punch removal of a veiled sea urchin spine. Dermatol Ther. 2021;34:E14947. doi:10.1111/dth.14947
  13. Wada T, Soma T, Gaman K, et al. Sea urchin spine arthritis of the hand. J Hand Surg Am. 2008;33:398-401. doi:10.1016/j.jhsa.2007.11.016
  14. Groleau S, Chhem RK, Younge D, et al. Ultrasonography of foreign-body tenosynovitis. Can Assoc Radiol J. 1992;43:454-456. 
  15. Hornbeak KB, Auerbach PS. Marine envenomation. Emerg Med Clin North Am. 2017;35:321-337. doi:10.1016/j.emc.2016.12.004
  16. Noonburg GE. Management of extremity trauma and related infections occurring in the aquatic environment. J Am Acad Orthop Surg. 2005;13:243-253. doi:10.5435/00124635-200507000-00004
  17. Haddad Junior V. Observation of initial clinical manifestations and repercussions from the treatment of 314 human injuries caused by black sea urchins (Echinometra lucunter) on the southeastern Brazilian coast. Rev Soc Bras Med Trop. 2012;45:390-392. doi:10.1590/s0037-86822012000300021
  18. Gargus MD, Morohashi DK. A sea-urchin spine chilling remedy. N Engl J Med. 2012;367:1867-1868. doi:10.1056/NEJMc1209382
  19. Sjøberg T, de Weerd L. The usefulness of a skin biopsy punch to remove sea urchin spines. ANZ J Surg. 2010;80:383. doi:10.1111/j.1445-2197.2010.05296.x
  20. Cardenas-de la Garza JA, Cuellar-Barboza A, Ancer-Arellano J, et al. Classic dermatological tools: foreign body removal with punch biopsy.J Am Acad Dermatol. 2019;81:E93-E94. doi:10.1016/j.jaad.2018.10.038
  21. Gungor S, Tarikçi N, Gokdemir G. Removal of sea urchin spines using erbium-doped yttrium aluminum garnet ablation. Dermatol Surg. 2012;38:508-510. doi:10.1111/j.1524-4725.2011.02259.x
  22. Böer A, Ochsendorf FR, Beier C, et al. Effective removal of sea-urchin spines by erbium:YAG laser ablation. Br J Dermatol. 2001;145:169-170. doi:10.1046/j.1365-2133.2001.04306.x
  23. De La Torre C, Toribio J. Sea-urchin granuloma: histologic profile. a pathologic study of 50 biopsies. J Cutan Pathol. 2001;28:223-228. doi:10.1034/j.1600-0560.2001.028005223.x
  24. Yi A, Kennedy C, Chia B, et al. Radiographic soft tissue thickness differentiating pyogenic flexor tenosynovitis from other finger infections. J Hand Surg Am. 2019;44:394-399. doi:10.1016/j.jhsa.2019.01.013
  25. Callison C, Nguyen H. Tetanus prophylaxis. In: StatPearls [Internet]. StatPearls Publishing; 2022.
References
  1. Amemiya CT, Miyake T, Rast JP. Echinoderms. Curr Biol. 2005;15:R944-R946. doi:10.1016/j.cub.2005.11.026
  2. Koch NM, Coppard SE, Lessios HA, et al. A phylogenomic resolution of the sea urchin tree of life. BMC Evol Biol. 2018;18:189. doi:10.1186/s12862-018-1300-4
  3. Amir Y, Insler M, Giller A, et al. Senescence and longevity of sea urchins. Genes (Basel). 2020;11:573. doi:10.3390/genes11050573
  4. Gummin DD, Mowry JB, Beuhler MC, et al. 2022 Annual Report of the National Poison Data System® (NPDS) from America's Poison Centers®: 40th annual report. Clin Toxicol (Phila). 2023;61:717-939. doi:10.1080/15563650.2023.2268981
  5. Gelman Y, Kong EL, Murphy-Lavoie HM. Sea urchin toxicity. In: StatPearls [Internet]. StatPearls Publishing; 2021.
  6. Suarez-Conde MF, Vallone MG, González VM, et al. Sea urchin skin lesions: a case report. Dermatol Pract Concept. 2021;11:E2021009. doi:10.5826/dpc.1102a09
  7. Al-Kathiri L, Al-Najjar T, Sulaiman I. Sea urchin granuloma of the hands: a case report. Oman Med J. 2019;34:350-353. doi:10.5001/omj.2019.68
  8. Dahl WJ, Jebson P, Louis DS. Sea urchin injuries to the hand: a case report and review of the literature. Iowa Orthop J. 2010;30:153-156.
  9. Hatakeyama T, Ichise A, Unno H, et al. Carbohydrate recognition by the rhamnose-binding lectin SUL-I with a novel three-domain structure isolated from the venom of globiferous pedicellariae of the flower sea urchin Toxopneustes pileolus. Protein Sci. 2017;26:1574-1583. doi:10.1002/pro.3185
  10. Balhara KS, Stolbach A. Marine envenomations. Emerg Med Clin North Am. 2014;32:223-243. doi:10.1016/j.emc.2013.09.009
  11. Schwartz Z, Cohen M, Lipner SR. Sea urchin injuries: a review and clinical approach algorithm. J Dermatolog Treat. 2021;32:150-156. doi:10.1080/09546634.2019.1638884
  12. Park SJ, Park JW, Choi SY, et al. Use of dermoscopy after punch removal of a veiled sea urchin spine. Dermatol Ther. 2021;34:E14947. doi:10.1111/dth.14947
  13. Wada T, Soma T, Gaman K, et al. Sea urchin spine arthritis of the hand. J Hand Surg Am. 2008;33:398-401. doi:10.1016/j.jhsa.2007.11.016
  14. Groleau S, Chhem RK, Younge D, et al. Ultrasonography of foreign-body tenosynovitis. Can Assoc Radiol J. 1992;43:454-456. 
  15. Hornbeak KB, Auerbach PS. Marine envenomation. Emerg Med Clin North Am. 2017;35:321-337. doi:10.1016/j.emc.2016.12.004
  16. Noonburg GE. Management of extremity trauma and related infections occurring in the aquatic environment. J Am Acad Orthop Surg. 2005;13:243-253. doi:10.5435/00124635-200507000-00004
  17. Haddad Junior V. Observation of initial clinical manifestations and repercussions from the treatment of 314 human injuries caused by black sea urchins (Echinometra lucunter) on the southeastern Brazilian coast. Rev Soc Bras Med Trop. 2012;45:390-392. doi:10.1590/s0037-86822012000300021
  18. Gargus MD, Morohashi DK. A sea-urchin spine chilling remedy. N Engl J Med. 2012;367:1867-1868. doi:10.1056/NEJMc1209382
  19. Sjøberg T, de Weerd L. The usefulness of a skin biopsy punch to remove sea urchin spines. ANZ J Surg. 2010;80:383. doi:10.1111/j.1445-2197.2010.05296.x
  20. Cardenas-de la Garza JA, Cuellar-Barboza A, Ancer-Arellano J, et al. Classic dermatological tools: foreign body removal with punch biopsy.J Am Acad Dermatol. 2019;81:E93-E94. doi:10.1016/j.jaad.2018.10.038
  21. Gungor S, Tarikçi N, Gokdemir G. Removal of sea urchin spines using erbium-doped yttrium aluminum garnet ablation. Dermatol Surg. 2012;38:508-510. doi:10.1111/j.1524-4725.2011.02259.x
  22. Böer A, Ochsendorf FR, Beier C, et al. Effective removal of sea-urchin spines by erbium:YAG laser ablation. Br J Dermatol. 2001;145:169-170. doi:10.1046/j.1365-2133.2001.04306.x
  23. De La Torre C, Toribio J. Sea-urchin granuloma: histologic profile. a pathologic study of 50 biopsies. J Cutan Pathol. 2001;28:223-228. doi:10.1034/j.1600-0560.2001.028005223.x
  24. Yi A, Kennedy C, Chia B, et al. Radiographic soft tissue thickness differentiating pyogenic flexor tenosynovitis from other finger infections. J Hand Surg Am. 2019;44:394-399. doi:10.1016/j.jhsa.2019.01.013
  25. Callison C, Nguyen H. Tetanus prophylaxis. In: StatPearls [Internet]. StatPearls Publishing; 2022.
Issue
Cutis - 113(6)
Issue
Cutis - 113(6)
Page Number
255-257
Page Number
255-257
Publications
Publications
Topics
Article Type
Display Headline
Aquatic Antagonists: Dermatologic Injuries From Sea Urchins (Echinoidea)
Display Headline
Aquatic Antagonists: Dermatologic Injuries From Sea Urchins (Echinoidea)
Sections
Inside the Article

 

Practice Points

  • Sea urchin spines easily become embedded in human skin upon contact and cause localized pain, edema, and black or purple pinpoint markings.
  • Immediate treatment includes soaking in hot water (113 12°F [45 12°C]) for 30 to 90 minutes to inactivate proinflammatory compounds, followed by extraction of the spines.
  • Successful methods of spine removal include the use of forceps and a hypodermic needle, as well as excision, liquid nitrogen, and punch biopsy.
  • Prompt removal of the spines can reduce the incidence of delayed granulomatous reactions, synovitis, and sea urchin arthritis.
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

Dermatologic Care for Refugees: Effective Management of Scabies and Pediculosis

Article Type
Changed
Fri, 04/26/2024 - 09:13
Display Headline
Dermatologic Care for Refugees: Effective Management of Scabies and Pediculosis

Approximately 108 million individuals have been forcibly displaced across the globe as of 2022, 35 million of whom are formally designated as refugees.1,2 The United States has coordinated resettlement of more refugee populations than any other country; the most common countries of origin are the Democratic Republic of the Congo, Syria, Afghanistan, and Myanmar.3 In 2021, policy to increase the number of refugees resettled in the United States by more than 700% (from 15,000 up to 125,000) was established; since enactment, the United States has seen more than double the refugee arrivals in 2023 than the prior year, making medical care for this population increasingly relevant for the dermatologist.4

Understanding how to care for this population begins with an accurate understanding of the term refugee. The United Nations defines a refugee as a person who is unwilling or unable to return to their country of nationality because of persecution or well-founded fear of persecution due to race, religion, nationality, membership in a particular social group, or political opinion. This term grants a protected status under international law and encompasses access to travel assistance, housing, cultural orientation, and medical evaluation upon resettlement.5,6

The burden of treatable dermatologic conditions in refugee populations ranges from 19% to 96% in the literature7,8 and varies from inflammatory disorders to infectious and parasitic diseases.9 In one study of 6899 displaced individuals in Greece, the prevalence of dermatologic conditions was higher than traumatic injury, cardiac disease, psychological conditions, and dental disease.10

When outlining differential diagnoses for parasitic infestations of the skin that affect refugee populations, helpful considerations include the individual’s country of origin, route traveled, and method of travel.11 Parasitic infestations specifically are more common in refugee populations when there are barriers to basic hygiene, crowded living or travel conditions, or lack of access to health care, which they may experience at any point in their home country, during travel, or in resettlement housing.8

Even with limited examination and diagnostic resources, the skin is the most accessible first indication of patients’ overall well-being and often provides simple diagnostic clues—in combination with contextualization of the patient’s unique circumstances—necessary for successful diagnosis and treatment of scabies and pediculosis.12 The dermatologist working with refugee populations may be the first set of eyes available and trained to discern skin infestations and therefore has the potential to improve overall outcomes.

Some parasitic infestations in refugee populations may fall under the category of neglected tropical diseases, including scabies, ascariasis, trypanosomiasis, leishmaniasis, and schistosomiasis; they affect an estimated 1 billion individuals across the globe but historically have been underrepresented in the literature and in health policy due in part to limited access to care.13 This review will focus on infestations by the scabies mite (Sarcoptes scabiei var hominis) and the human louse, as these frequently are encountered, easily diagnosed, and treatable by trained clinicians, even in resource-limited settings.

Scabies

Scabies is a parasitic skin infestation caused by the 8-legged mite Sarcoptes scabiei var hominis. The female mite begins the infestation process via penetration of the epidermis, particularly the stratum corneum, and commences laying eggs (Figure 1). The subsequent larvae emerge 48 to 72 hours later and remain burrowed in the epidermis. The larvae mature over the next 10 to 14 days and continue the reproductive cycle.14,15 Symptoms of infestation occurs due to a hypersensitivity reaction to the mite and its by-products.16 Transmission of the mite primarily occurs via direct (skin-to-skin) contact with infected individuals or environmental surfaces for 24 to36 hours in specific conditions, though the latter source has been debated in the literature.

Sarcoptes scabiei mite (A), ova (B), and scybala (C) on microscopic evaluation.
FIGURE 1. Sarcoptes scabiei mite (A), ova (B), and scybala (C) on microscopic evaluation.

 

 

The method of transmission is particularly important when considering care for refugee populations. Scabies is found most often in those living in or traveling from tropical regions including East Asia, Southeast Asia, Oceania, and Latin America.17 In displaced or refugee populations, a lack of access to basic hygiene, extended travel in close quarters, and suboptimal health care access all may lead to an increased incidence of untreated scabies infestations.18 Scabies is more prevalent in children, with increased potential for secondary bacterial infections with Streptococcus and Staphylococcus species due to excoriation in unsanitary conditions. Secondary infection with Streptococcus pyogenes can lead to acute poststreptococcal glomerulonephritis, which accounts for a large burden of chronic kidney disease in affected populations.19 However, scabies may be found in any population, regardless of hygiene or health care access. Treating health care providers should keep a broad differential.

Presentation—The latency of scabies symptoms is 2 to 6 weeks in a primary outbreak and may be as short as 1 to 3 days with re-infestation, following the course of delayed-type hypersensitivity.20 The initial hallmark symptom is pruritus with increased severity in the evening. Visible lesions, excoriations, and burrows associated with scattered vesicles or pustules may be seen over the web spaces of the hands and feet, volar surfaces of the wrists, axillae, waist, genitalia, inner thighs, or buttocks.19 Chronic infestation often manifests with genital nodules. In populations with limited access to health care, there are reports of a sensitization phenomenon in which the individual may become less symptomatic after 4 to 6 weeks and yet be a potential carrier of the mite.21

Those with compromised immune function, such as individuals living with HIV or severe malnutrition, may present with crusted scabies, a variant that manifests as widespread hyperkeratotic scaling with more pronounced involvement of the head, neck, and acral areas. In contrast to classic scabies, crusted scabies is associated with minimal pruritus.22

Diagnosis—The diagnosis of scabies is largely clinical with confirmation through skin scrapings. The International Alliance for Control of Scabies has established diagnostic criteria that include a combination of clinical findings, history, and visualization of mites.23 A dermatologist working with refugee populations may employ any combination of history (eg, nocturnal itch, exposure to an affected individual) or clinical findings along with a high degree of suspicion in those with elevated risk. Visualization of mites is helpful to confirm the diagnosis and may be completed with the application of mineral oil at the terminal end of a burrow, skin scraping with a surgical blade or needle, and examination under light microscopy.

Treatment—First-line treatment for scabies consists of application of permethrin cream 5% on the skin of the neck to the soles of the feet, which is to be left on for 8 to 14 hours followed by rinsing. Re-application is recommended in 1 to 2 weeks. Oral ivermectin is a reasonable alternative to permethrin cream due to its low cost and easy administration in large affected groups. It is not labeled for use in pregnant women or children weighing less than 15 kg but has no selective fetal toxicity. Treatment of scabies with ivermectin has the benefit of treating many other parasitic infections. Both medications are on the World Health Organization Model List of Essential Medications and are widely available for treating providers, even in resource-limited settings.24

Much of the world still uses benzyl benzoate or precipitated sulfur ointment to treat scabies, and some botanicals used in folk medicine have genuine antiscabetic properties. Pruritus may persist for 1 to 4 weeks following treatment and does not indicate treatment failure. Topical camphor and menthol preparations, low-potency topical corticosteroids, or emollients all may be employed for relief.25Sarna is a Spanish term for scabies and has become the proprietary name for topical antipruritic agents. Additional methods of treatment and prevention include washing clothes and linens in hot water and drying on high heat. If machine washing is not available, clothing and linens may be sealed in a plastic bag for 72 hours.

Pediculosis

Pediculosis is an infestation caused by the ectoparasite Pediculus humanus, an obligate, sesame seed–sized louse that feeds exclusively on the blood of its host (Figure 2).26 Of the lice species, 2 require humans as hosts; one is P humanus and the other is Pthirus pubis (pubic lice). Pediculus humanus may be further classified into morphologies based largely on the affected area: body (P humanus corporis) or head (P humanus capitis), both of which will be discussed.27

Pediculus humanus (louse), adult form.
FIGURE 2. Pediculus humanus (louse), adult form.

 

 

Lice primarily attach to clothing and hair shafts, then transfer to the skin for blood feeds. Females lay eggs that hatch 6 to 10 days later, subsequently maturing into adults. The lifespan of these parasites with regular access to a host is 1 to 3 months for head lice and 18 days for body lice vs only 3 to 5 days without a host.28 Transmission of P humanus capitis primarily occurs via direct contact with affected individuals, either head-to-head contact or sharing of items such as brushes and headscarves; P humanus corporis also may be transmitted via direct contact with affected individuals or clothing.

Pediculosis is an important infestation to consider when providing care for refugee populations. Risk factors include lack of access to basic hygiene, including regular bathing or laundering of clothing, and crowded conditions that make direct person-to-person contact with affected individuals more likely.29 Body lice are associated more often with domestic turbulence and displaced populations30 in comparison to head lice, which have broad demographic variables, most often affecting females and children.28 Fatty acids in adult male sebum make the scalp less hospitable to lice.

Presentation—The most common clinical manifestation of pediculosis is pruritus. Cutaneous findings can include papules, wheals, or hemorrhagic puncta secondary to the louse bite. Due to the Tyndall effect of deep hemosiderin pigment, blue-grey macules termed maculae ceruleae (Figure 3) also may be present in chronic infestations of pediculosis pubis, in contrast to pediculosis capitis or corporis.31 Body louse infestation is associated with a general pruritus concentrated on the neck, shoulders, and waist—areas where clothing makes the most direct contact. Lesions may be visible and include eczematous patches with excoriation and possible secondary bacterial infection. Chronic infestation may exhibit lichenification or hyperpigmentation in associated areas. Head lice most often manifest with localized scalp pruritus and associated excoriation and cervical or occipital lymphadenopathy.32

Maculae ceruleae—blue-grey macules—may be present on the skin secondary to Pediculosis infestation.
FIGURE 3. Maculae ceruleae—blue-grey macules—may be present on the skin secondary to Pediculosis infestation.

Diagnosis—The diagnosis of pediculosis is clinical, with confirmation requiring direct examination of the insect or nits (the egg case of the parasite)(Figure 4). Body lice and associated nits can be visualized on clothing seams near areas of highest body temperature, particularly the waistband. Head lice may be visualized crawling on hair shafts or on a louse comb. Nits are firmly attached to hair shafts and are visible to the naked eye, whereas pseudonits slide freely along the hair shaft and are not a manifestation of louse infestation (Figure 5).31

Pediculosis nits—the egg cases of the parasite—may firmly attach to the hair shaft.
FIGURE 4. Pediculosis nits—the egg cases of the parasite—may firmly attach to the hair shaft.

Treatment—Treatment varies by affected area. Pediculosis corporis may be treated with permethrin cream 5% applied to the entire body and left on for 8 to 10 hours, but this may not be necessary if facilities are available to wash and dry clothing.33 The use of oral ivermectin and permethrin-impregnated underwear both have been proposed.34,35 Treatment of pediculosis capitis may be accomplished with a variety of topical pediculicides including permethrin, pyrethrum with piperonyl butoxide, dimethicone, malathion, benzyl alcohol, spinosad, and topical ivermectin.22 Topical corticosteroids or emollients may be employed for residual pruritus.

The pseudonit closely mimics pediculosis nits but consists of keratinized cell casts that are freely dislodged.
FIGURE 5. The pseudonit closely mimics pediculosis nits but consists of keratinized cell casts that are freely dislodged.

Equally important is environmental elimination of infestation. Clothing should be discarded if possible or washed and dried using high heat. If neither approach is possible or appropriate, clothing may be sealed in a plastic bag for 2 weeks or treated with a pediculicide. Nit combing is an important adjunct in the treatment of pediculosis capitis.36 It is important to encourage return to work and/or school immediately after treatment. “No nit” policies are more harmful to education than helpful for prevention of investation.37

Pediculosis corporis may transmit infectious agents including Bartonella quintana, (trench fever, endocarditis, bacillary angiomatosis), Borrelia recurrentis (louse-borne relapsing fever), and Rickettsia prowazekii (epidemic typhus).31,38,39 Additionally, severe pediculosis infestations have the potential to cause chronic blood loss in affected populations. In a study of patients with active pediculosis infestation, mean hemoglobin values were found to be 2.5 g/dL lower than a matched population without infestation.40 It is important to consider pediculosis as a risk for iron-deficiency anemia in populations who are known to lack access to regular medical evaluation.41

 

 

Future Considerations

Increased access to tools and education for clinicians treating refugee populations is key to reducing the burden of parasitic skin disease and related morbidity and mortality in vulnerable groups both domestically and globally. One such tool, the Skin NTDs App, was launched by the World Health Organization in 2020. It is available for free for Android and iOS devices to assist clinicians in the field with the diagnosis and treatment of neglected tropical diseases—including scabies—that may affect refugee populations.42

Additionally, to both improve access and limit preventable sequelae, future investigations into appropriate models of community-based care are paramount. The model of community-based care is centered on the idea of care provision that prioritizes safety, accessibility, affordability, and acceptability in an environment closest to vulnerable populations. The largest dermatologic society, the International League of Dermatological Societies, formed a Migrant Health Dermatology Working Group that prioritizes understanding and improving care for refugee and migrant populations; this group hosted a summit in 2022, bringing together international subject matter leaders to discuss such models of care and set goals for the creation of tool kits for patients, frontline health care workers, and dermatologists.43

Conclusion

Improvement in dermatologic care of refugee populations includes provision of culturally and linguistically appropriate care by trained clinicians, adequate access to the most essential medications, and basic physical or legal access to health care systems in general.8,11,44 Parasitic infestations have the potential to remain asymptomatic for extended periods of time and result in spread to potentially nonendemic regions of resettlement.45 Additionally, the psychosocial well-being of refugee populations upon resettlement may be negatively affected by stigma of disease processes such as scabies and pediculosis, leading to additional barriers to successful re-entry into the patient’s new environment.46 Therefore, proper screening, diagnosis, and treatment of the most common parasitic infestations in this population have great potential to improve outcomes for large groups across the globe.

References
  1. Monin K, Batalova J, Lai T. Refugees and Asylees in the United States. Migration Information Source. Published May 13, 2021. Accessed April 4, 2024. https://www.migrationpolicy.org/article/refugees-and-asylees-united-states-2021
  2. UNHCR. Figures at a Glance. UNHCR USA. Update June 14, 2023. Accessed April 4, 2024. https://www.unhcr.org/en-us/figures-at-a-glance.html
  3. UNHCR. Refugee resettlement facts. Published October 2023. Accessed April 8, 2024. https://www.unhcr.org/us/media/refugee-resettlement-facts
  4. US Department of State. Report to Congress on Proposed Refugee Admissions for Fiscal Year 2024. Published November 3, 2023. Accessed April 8, 2024. https://www.state.gov/report-to-congress-on-proposed-refugee-admissions-for-fiscal-year-2024/
  5. UNHCR. Compact for Migration: Definitions. United Nations. Accessed April 4, 2024. https://refugeesmigrants.un.org/definitions
  6. United Nations High Commissioner for Refugees (UNHCR). Convention and Protocol Relating to the Status of Refugees. Published December 2010. Accessed January 11, 2024. https://www.unhcr.org/us/media/convention-and-protocol-relating-status-refugees
  7. Kibar Öztürk M. Skin diseases in rural Nyala, Sudan (in a rural hospital, in 12 orphanages, and in two refugee camps). Int J Dermatol. 2019;58:1341-1349. doi:10.1111/ijd.14619
  8. Padovese V, Knapp A. Challenges of managing skin diseases in refugees and migrants. Dermatol Clin. 2021;39:101-115. doi:10.1016/j.det.2020.08.010
  9. Saikal SL, Ge L, Mir A, et al. Skin disease profile of Syrian refugees in Jordan: a field-mission assessment. J Eur Acad Dermatol Venereol. 2020;34:419-425. doi:10.1111/jdv.15909
  10. Eonomopoulou A, Pavli A, Stasinopoulou P, et al. Migrant screening: lessons learned from the migrant holding level at the Greek-Turkish borders. J Infect Public Health. 2017;10:177-184. doi:10.1016/j.jiph.2016.04.012
  11. Marano N, Angelo KM, Merrill RD, et al. Expanding travel medicine in the 21st century to address the health needs of the world’s migrants.J Travel Med. 2018;25. doi:10.1093/jtm/tay067
  12. Hay RJ, Asiedu K. Skin-related neglected tropical diseases (skin NTDs)—a new challenge. Trop Med Infect Dis. 2018;4. doi:10.3390/tropicalmed4010004
  13. NIAID. Neglected tropical diseases. Updated July 11, 2016. Accessed April 4, 2024. https://www.niaid.nih.gov/research/neglected-tropical-diseases
  14. Arlian LG, Morgan MS. A review of Sarcoptes scabiei: past, present and future. Parasit Vectors. 2017;10:297. doi:10.1186/s13071-017-2234-1
  15. Arlian LG, Runyan RA, Achar S, et al. Survival and infectivity of Sarcoptes scabiei var. canis and var. hominis. J Am Acad Dermatol. 1984;11(2 pt 1):210-215. doi:10.1016/s0190-9622(84)70151-4
  16. Chandler DJ, Fuller LC. A review of scabies: an infestation more than skin deep. Dermatology. 2019;235:79-90. doi:10.1159/000495290
  17. Karimkhani C, Colombara DV, Drucker AM, et al. The global burden of scabies: a cross-sectional analysis from the Global Burden of Disease Study 2015. Lancet Infect Dis. 2017;17:1247-1254. doi:10.1016/S1473-3099(17)30483-8
  18. Romani L, Steer AC, Whitfeld MJ, et al. Prevalence of scabies and impetigo worldwide: a systematic review. Lancet Infect Dis. 2015;15:960-967. doi:10.1016/S1473-3099(15)00132-2
  19. Thomas C, Coates SJ, Engelman D, et al. Ectoparasites: scabies. J Am Acad Dermatol. 2020;82:533-548. doi:10.1016/j.jaad.2019.05.109
  20. Mellanby K, Johnson CG, Bartley WC. Treatment of scabies. Br Med J. 1942;2:1-4. doi:10.1136/bmj.2.4252.1
  21. Walton SF. The immunology of susceptibility and resistance to scabies. Parasit Immunol. 2010;32:532-540. doi:10.1111/j.1365-3024.2010.01218.x
  22. Coates SJ, Thomas C, Chosidow O, et al. Ectoparasites: pediculosis and tungiasis. J Am Acad Dermatol. 2020;82:551-569. doi:10.1016/j.jaad.2019.05.110
  23. Engelman D, Fuller LC, Steer AC; International Alliance for the Control of Scabies Delphi p. Consensus criteria for the diagnosis of scabies: a Delphi study of international experts. PLoS Negl Trop Dis. 2018;12:E0006549. doi:10.1371/journal.pntd.0006549
  24. World Health Organization. WHO Model Lists of Essential Medicines—23rd list, 2023. Updated July 26, 2023. Accessed April 8, 2024. https://www.who.int/publications/i/item/WHO-MHP-HPS-EML-2023.02
  25. Salavastru CM, Chosidow O, Boffa MJ, et al. European guideline for the management of scabies. J Eur Acad Dermatol Venereol. 2017;31:1248-1253. doi:10.1111/jdv.14351
  26. Badiaga S, Brouqui P. Human louse-transmitted infectious diseases. Clin Microbiol Infect. 2012;18:332-337. doi:10.1111/j.1469-0691.2012.03778.x
  27. Leo NP, Campbell NJH, Yang X, et al. Evidence from mitochondrial DNA that head lice and body lice of humans (Phthiraptera: Pediculidae) are conspecific. J Med Entomol. 2002;39:662-666. doi:10.1603/0022-2585-39.4.662
  28. Chosidow O. Scabies and pediculosis. Lancet. 2000;355:819-826. doi:10.1016/S0140-6736(99)09458-1
  29. Arnaud A, Chosidow O, Détrez M-A, et al. Prevalences of scabies and pediculosis corporis among homeless people in the Paris region: results from two randomized cross-sectional surveys (HYTPEAC study). Br J Dermatol. 2016;174:104-112. doi:10.1111/bjd.14226
  30. Brouqui P. Arthropod-borne diseases associated with political and social disorder. Annu Rev Entomol. 2011;56:357-374. doi:10.1146/annurev-ento-120709-144739
  31. Ko CJ, Elston DM. Pediculosis. J Am Acad Dermatol. 2004;50:1-12. doi:10.1016/S0190-9622(03)02729-4
  32. Bloomfield D. Head lice. Pediatr Rev. 2002;23:34-35; discussion 34-35. doi:10.1542/pir.23-1-34
  33. Stone SP GJ, Bacelieri RE. Scabies, other mites, and pediculosis. In: Wolf K GL, Katz SI, et al (eds). Fitzpatrick’s Dermatology in General Medicine. McGraw Hill; 2008:2029.
  34. Foucault C, Ranque S, Badiaga S, et al. Oral ivermectin in the treatment of body lice. J Infect Dis. 2006;193:474-476. doi:10.1086/499279
  35. Benkouiten S, Drali R, Badiaga S, et al. Effect of permethrin-impregnated underwear on body lice in sheltered homeless persons: a randomized controlled trial. JAMA Dermatol. 2014;150:273-279. doi:10.1001/jamadermatol.2013.6398
  36. CDC. Parasites: Treatment. Updated October 15, 2019. Accessed April 4, 2024. https://www.cdc.gov/parasites/lice/head/treatment.html
  37. Devore CD, Schutze GE; Council on School Health and Committee on Infectious Diseases, American Academy of Pediatrics. Head lice. Pediatrics. 2015;135:e1355-e1365. doi:10.1542/peds.2015-0746
  38. Ohl ME, Spach DH. Bartonella quintana and urban trench fever. Clin Infect Dis. 2000;31:131-135. doi:10.1086/313890
  39. Drali R, Sangaré AK, Boutellis A, et al. Bartonella quintana in body lice from scalp hair of homeless persons, France. Emerg Infect Dis. 2014;20:907-908. doi:10.3201/eid2005.131242
  40. Rudd N, Zakaria A, Kohn MA, et al. Association of body lice infestation with hemoglobin values in hospitalized dermatology patients. JAMA Dermatol. 2022;158:691-693. doi:10.1001/jamadermatol.2022.0818
  41. Guss DA, Koenig M, Castillo EM. Severe iron deficiency anemia and lice infestation. J Emergency Med. 2011;41:362-365. doi:10.1016/j.jemermed.2010.05.030
  42. Neglected tropical diseases of the skin: WHO launches mobile application to facilitate diagnosis. News release. World Health Organization; July 16, 2020. Accessed April 4, 2024. https://www.who.int/news/item/16-07-2020-neglected-tropical-diseases-of-the-skin-who-launches-mobile-application-to-facilitate-diagnosis
  43. Padovese V, Fuller LC, Griffiths CEM, et al; Migrant Health Dermatology Working Group of the International Foundation for Dermatology. Migrant skin health: perspectives from the Migrant Health Summit, Malta, 2022. Br J Dermatology. 2023;188:553-554. doi:10.1093/bjd/ljad001
  44. Knapp AP, Rehmus W, Chang AY. Skin diseases in displaced populations: a review of contributing factors, challenges, and approaches to care. Int J Dermatol. 2020;59:1299-1311. doi:10.1111/ijd.15063
  45. Norman FF, Comeche B, Chamorro S, et al. Overcoming challenges in the diagnosis and treatment of parasitic infectious diseases in migrants. Expert Rev Anti-infective Therapy. 2020;18:127-143. doi:10.1080/14787210.2020.1713099
  46. Skin NTDs: prioritizing integrated approaches to reduce suffering, psychosocial impact and stigmatization. News release. World Health Organization; October 29, 2020. Accessed April 4, 2024. https://www.who.int/news/item/29-10-2020-skin-ntds-prioritizing-integrated-approaches-to-reduce-suffering-psychosocial-impact-and-stigmatization
Article PDF
Author and Disclosure Information

Alexis G. Strahan is from the Mercer University School of Medicine, Savannah, Georgia. Dr. Elston is from the Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston.

The authors report no conflict of interest.

All images are in the public domain.

Correspondence: Alexis G. Strahan, MD, MSN, 55 Fruit St, Bartlett Hall 6R, Boston, MA 02114 (alexis.grabow.strahan@live.mercer.edu).

Issue
Cutis - 113(4)
Publications
Topics
Page Number
E16-E21
Sections
Author and Disclosure Information

Alexis G. Strahan is from the Mercer University School of Medicine, Savannah, Georgia. Dr. Elston is from the Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston.

The authors report no conflict of interest.

All images are in the public domain.

Correspondence: Alexis G. Strahan, MD, MSN, 55 Fruit St, Bartlett Hall 6R, Boston, MA 02114 (alexis.grabow.strahan@live.mercer.edu).

Author and Disclosure Information

Alexis G. Strahan is from the Mercer University School of Medicine, Savannah, Georgia. Dr. Elston is from the Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston.

The authors report no conflict of interest.

All images are in the public domain.

Correspondence: Alexis G. Strahan, MD, MSN, 55 Fruit St, Bartlett Hall 6R, Boston, MA 02114 (alexis.grabow.strahan@live.mercer.edu).

Article PDF
Article PDF

Approximately 108 million individuals have been forcibly displaced across the globe as of 2022, 35 million of whom are formally designated as refugees.1,2 The United States has coordinated resettlement of more refugee populations than any other country; the most common countries of origin are the Democratic Republic of the Congo, Syria, Afghanistan, and Myanmar.3 In 2021, policy to increase the number of refugees resettled in the United States by more than 700% (from 15,000 up to 125,000) was established; since enactment, the United States has seen more than double the refugee arrivals in 2023 than the prior year, making medical care for this population increasingly relevant for the dermatologist.4

Understanding how to care for this population begins with an accurate understanding of the term refugee. The United Nations defines a refugee as a person who is unwilling or unable to return to their country of nationality because of persecution or well-founded fear of persecution due to race, religion, nationality, membership in a particular social group, or political opinion. This term grants a protected status under international law and encompasses access to travel assistance, housing, cultural orientation, and medical evaluation upon resettlement.5,6

The burden of treatable dermatologic conditions in refugee populations ranges from 19% to 96% in the literature7,8 and varies from inflammatory disorders to infectious and parasitic diseases.9 In one study of 6899 displaced individuals in Greece, the prevalence of dermatologic conditions was higher than traumatic injury, cardiac disease, psychological conditions, and dental disease.10

When outlining differential diagnoses for parasitic infestations of the skin that affect refugee populations, helpful considerations include the individual’s country of origin, route traveled, and method of travel.11 Parasitic infestations specifically are more common in refugee populations when there are barriers to basic hygiene, crowded living or travel conditions, or lack of access to health care, which they may experience at any point in their home country, during travel, or in resettlement housing.8

Even with limited examination and diagnostic resources, the skin is the most accessible first indication of patients’ overall well-being and often provides simple diagnostic clues—in combination with contextualization of the patient’s unique circumstances—necessary for successful diagnosis and treatment of scabies and pediculosis.12 The dermatologist working with refugee populations may be the first set of eyes available and trained to discern skin infestations and therefore has the potential to improve overall outcomes.

Some parasitic infestations in refugee populations may fall under the category of neglected tropical diseases, including scabies, ascariasis, trypanosomiasis, leishmaniasis, and schistosomiasis; they affect an estimated 1 billion individuals across the globe but historically have been underrepresented in the literature and in health policy due in part to limited access to care.13 This review will focus on infestations by the scabies mite (Sarcoptes scabiei var hominis) and the human louse, as these frequently are encountered, easily diagnosed, and treatable by trained clinicians, even in resource-limited settings.

Scabies

Scabies is a parasitic skin infestation caused by the 8-legged mite Sarcoptes scabiei var hominis. The female mite begins the infestation process via penetration of the epidermis, particularly the stratum corneum, and commences laying eggs (Figure 1). The subsequent larvae emerge 48 to 72 hours later and remain burrowed in the epidermis. The larvae mature over the next 10 to 14 days and continue the reproductive cycle.14,15 Symptoms of infestation occurs due to a hypersensitivity reaction to the mite and its by-products.16 Transmission of the mite primarily occurs via direct (skin-to-skin) contact with infected individuals or environmental surfaces for 24 to36 hours in specific conditions, though the latter source has been debated in the literature.

Sarcoptes scabiei mite (A), ova (B), and scybala (C) on microscopic evaluation.
FIGURE 1. Sarcoptes scabiei mite (A), ova (B), and scybala (C) on microscopic evaluation.

 

 

The method of transmission is particularly important when considering care for refugee populations. Scabies is found most often in those living in or traveling from tropical regions including East Asia, Southeast Asia, Oceania, and Latin America.17 In displaced or refugee populations, a lack of access to basic hygiene, extended travel in close quarters, and suboptimal health care access all may lead to an increased incidence of untreated scabies infestations.18 Scabies is more prevalent in children, with increased potential for secondary bacterial infections with Streptococcus and Staphylococcus species due to excoriation in unsanitary conditions. Secondary infection with Streptococcus pyogenes can lead to acute poststreptococcal glomerulonephritis, which accounts for a large burden of chronic kidney disease in affected populations.19 However, scabies may be found in any population, regardless of hygiene or health care access. Treating health care providers should keep a broad differential.

Presentation—The latency of scabies symptoms is 2 to 6 weeks in a primary outbreak and may be as short as 1 to 3 days with re-infestation, following the course of delayed-type hypersensitivity.20 The initial hallmark symptom is pruritus with increased severity in the evening. Visible lesions, excoriations, and burrows associated with scattered vesicles or pustules may be seen over the web spaces of the hands and feet, volar surfaces of the wrists, axillae, waist, genitalia, inner thighs, or buttocks.19 Chronic infestation often manifests with genital nodules. In populations with limited access to health care, there are reports of a sensitization phenomenon in which the individual may become less symptomatic after 4 to 6 weeks and yet be a potential carrier of the mite.21

Those with compromised immune function, such as individuals living with HIV or severe malnutrition, may present with crusted scabies, a variant that manifests as widespread hyperkeratotic scaling with more pronounced involvement of the head, neck, and acral areas. In contrast to classic scabies, crusted scabies is associated with minimal pruritus.22

Diagnosis—The diagnosis of scabies is largely clinical with confirmation through skin scrapings. The International Alliance for Control of Scabies has established diagnostic criteria that include a combination of clinical findings, history, and visualization of mites.23 A dermatologist working with refugee populations may employ any combination of history (eg, nocturnal itch, exposure to an affected individual) or clinical findings along with a high degree of suspicion in those with elevated risk. Visualization of mites is helpful to confirm the diagnosis and may be completed with the application of mineral oil at the terminal end of a burrow, skin scraping with a surgical blade or needle, and examination under light microscopy.

Treatment—First-line treatment for scabies consists of application of permethrin cream 5% on the skin of the neck to the soles of the feet, which is to be left on for 8 to 14 hours followed by rinsing. Re-application is recommended in 1 to 2 weeks. Oral ivermectin is a reasonable alternative to permethrin cream due to its low cost and easy administration in large affected groups. It is not labeled for use in pregnant women or children weighing less than 15 kg but has no selective fetal toxicity. Treatment of scabies with ivermectin has the benefit of treating many other parasitic infections. Both medications are on the World Health Organization Model List of Essential Medications and are widely available for treating providers, even in resource-limited settings.24

Much of the world still uses benzyl benzoate or precipitated sulfur ointment to treat scabies, and some botanicals used in folk medicine have genuine antiscabetic properties. Pruritus may persist for 1 to 4 weeks following treatment and does not indicate treatment failure. Topical camphor and menthol preparations, low-potency topical corticosteroids, or emollients all may be employed for relief.25Sarna is a Spanish term for scabies and has become the proprietary name for topical antipruritic agents. Additional methods of treatment and prevention include washing clothes and linens in hot water and drying on high heat. If machine washing is not available, clothing and linens may be sealed in a plastic bag for 72 hours.

Pediculosis

Pediculosis is an infestation caused by the ectoparasite Pediculus humanus, an obligate, sesame seed–sized louse that feeds exclusively on the blood of its host (Figure 2).26 Of the lice species, 2 require humans as hosts; one is P humanus and the other is Pthirus pubis (pubic lice). Pediculus humanus may be further classified into morphologies based largely on the affected area: body (P humanus corporis) or head (P humanus capitis), both of which will be discussed.27

Pediculus humanus (louse), adult form.
FIGURE 2. Pediculus humanus (louse), adult form.

 

 

Lice primarily attach to clothing and hair shafts, then transfer to the skin for blood feeds. Females lay eggs that hatch 6 to 10 days later, subsequently maturing into adults. The lifespan of these parasites with regular access to a host is 1 to 3 months for head lice and 18 days for body lice vs only 3 to 5 days without a host.28 Transmission of P humanus capitis primarily occurs via direct contact with affected individuals, either head-to-head contact or sharing of items such as brushes and headscarves; P humanus corporis also may be transmitted via direct contact with affected individuals or clothing.

Pediculosis is an important infestation to consider when providing care for refugee populations. Risk factors include lack of access to basic hygiene, including regular bathing or laundering of clothing, and crowded conditions that make direct person-to-person contact with affected individuals more likely.29 Body lice are associated more often with domestic turbulence and displaced populations30 in comparison to head lice, which have broad demographic variables, most often affecting females and children.28 Fatty acids in adult male sebum make the scalp less hospitable to lice.

Presentation—The most common clinical manifestation of pediculosis is pruritus. Cutaneous findings can include papules, wheals, or hemorrhagic puncta secondary to the louse bite. Due to the Tyndall effect of deep hemosiderin pigment, blue-grey macules termed maculae ceruleae (Figure 3) also may be present in chronic infestations of pediculosis pubis, in contrast to pediculosis capitis or corporis.31 Body louse infestation is associated with a general pruritus concentrated on the neck, shoulders, and waist—areas where clothing makes the most direct contact. Lesions may be visible and include eczematous patches with excoriation and possible secondary bacterial infection. Chronic infestation may exhibit lichenification or hyperpigmentation in associated areas. Head lice most often manifest with localized scalp pruritus and associated excoriation and cervical or occipital lymphadenopathy.32

Maculae ceruleae—blue-grey macules—may be present on the skin secondary to Pediculosis infestation.
FIGURE 3. Maculae ceruleae—blue-grey macules—may be present on the skin secondary to Pediculosis infestation.

Diagnosis—The diagnosis of pediculosis is clinical, with confirmation requiring direct examination of the insect or nits (the egg case of the parasite)(Figure 4). Body lice and associated nits can be visualized on clothing seams near areas of highest body temperature, particularly the waistband. Head lice may be visualized crawling on hair shafts or on a louse comb. Nits are firmly attached to hair shafts and are visible to the naked eye, whereas pseudonits slide freely along the hair shaft and are not a manifestation of louse infestation (Figure 5).31

Pediculosis nits—the egg cases of the parasite—may firmly attach to the hair shaft.
FIGURE 4. Pediculosis nits—the egg cases of the parasite—may firmly attach to the hair shaft.

Treatment—Treatment varies by affected area. Pediculosis corporis may be treated with permethrin cream 5% applied to the entire body and left on for 8 to 10 hours, but this may not be necessary if facilities are available to wash and dry clothing.33 The use of oral ivermectin and permethrin-impregnated underwear both have been proposed.34,35 Treatment of pediculosis capitis may be accomplished with a variety of topical pediculicides including permethrin, pyrethrum with piperonyl butoxide, dimethicone, malathion, benzyl alcohol, spinosad, and topical ivermectin.22 Topical corticosteroids or emollients may be employed for residual pruritus.

The pseudonit closely mimics pediculosis nits but consists of keratinized cell casts that are freely dislodged.
FIGURE 5. The pseudonit closely mimics pediculosis nits but consists of keratinized cell casts that are freely dislodged.

Equally important is environmental elimination of infestation. Clothing should be discarded if possible or washed and dried using high heat. If neither approach is possible or appropriate, clothing may be sealed in a plastic bag for 2 weeks or treated with a pediculicide. Nit combing is an important adjunct in the treatment of pediculosis capitis.36 It is important to encourage return to work and/or school immediately after treatment. “No nit” policies are more harmful to education than helpful for prevention of investation.37

Pediculosis corporis may transmit infectious agents including Bartonella quintana, (trench fever, endocarditis, bacillary angiomatosis), Borrelia recurrentis (louse-borne relapsing fever), and Rickettsia prowazekii (epidemic typhus).31,38,39 Additionally, severe pediculosis infestations have the potential to cause chronic blood loss in affected populations. In a study of patients with active pediculosis infestation, mean hemoglobin values were found to be 2.5 g/dL lower than a matched population without infestation.40 It is important to consider pediculosis as a risk for iron-deficiency anemia in populations who are known to lack access to regular medical evaluation.41

 

 

Future Considerations

Increased access to tools and education for clinicians treating refugee populations is key to reducing the burden of parasitic skin disease and related morbidity and mortality in vulnerable groups both domestically and globally. One such tool, the Skin NTDs App, was launched by the World Health Organization in 2020. It is available for free for Android and iOS devices to assist clinicians in the field with the diagnosis and treatment of neglected tropical diseases—including scabies—that may affect refugee populations.42

Additionally, to both improve access and limit preventable sequelae, future investigations into appropriate models of community-based care are paramount. The model of community-based care is centered on the idea of care provision that prioritizes safety, accessibility, affordability, and acceptability in an environment closest to vulnerable populations. The largest dermatologic society, the International League of Dermatological Societies, formed a Migrant Health Dermatology Working Group that prioritizes understanding and improving care for refugee and migrant populations; this group hosted a summit in 2022, bringing together international subject matter leaders to discuss such models of care and set goals for the creation of tool kits for patients, frontline health care workers, and dermatologists.43

Conclusion

Improvement in dermatologic care of refugee populations includes provision of culturally and linguistically appropriate care by trained clinicians, adequate access to the most essential medications, and basic physical or legal access to health care systems in general.8,11,44 Parasitic infestations have the potential to remain asymptomatic for extended periods of time and result in spread to potentially nonendemic regions of resettlement.45 Additionally, the psychosocial well-being of refugee populations upon resettlement may be negatively affected by stigma of disease processes such as scabies and pediculosis, leading to additional barriers to successful re-entry into the patient’s new environment.46 Therefore, proper screening, diagnosis, and treatment of the most common parasitic infestations in this population have great potential to improve outcomes for large groups across the globe.

Approximately 108 million individuals have been forcibly displaced across the globe as of 2022, 35 million of whom are formally designated as refugees.1,2 The United States has coordinated resettlement of more refugee populations than any other country; the most common countries of origin are the Democratic Republic of the Congo, Syria, Afghanistan, and Myanmar.3 In 2021, policy to increase the number of refugees resettled in the United States by more than 700% (from 15,000 up to 125,000) was established; since enactment, the United States has seen more than double the refugee arrivals in 2023 than the prior year, making medical care for this population increasingly relevant for the dermatologist.4

Understanding how to care for this population begins with an accurate understanding of the term refugee. The United Nations defines a refugee as a person who is unwilling or unable to return to their country of nationality because of persecution or well-founded fear of persecution due to race, religion, nationality, membership in a particular social group, or political opinion. This term grants a protected status under international law and encompasses access to travel assistance, housing, cultural orientation, and medical evaluation upon resettlement.5,6

The burden of treatable dermatologic conditions in refugee populations ranges from 19% to 96% in the literature7,8 and varies from inflammatory disorders to infectious and parasitic diseases.9 In one study of 6899 displaced individuals in Greece, the prevalence of dermatologic conditions was higher than traumatic injury, cardiac disease, psychological conditions, and dental disease.10

When outlining differential diagnoses for parasitic infestations of the skin that affect refugee populations, helpful considerations include the individual’s country of origin, route traveled, and method of travel.11 Parasitic infestations specifically are more common in refugee populations when there are barriers to basic hygiene, crowded living or travel conditions, or lack of access to health care, which they may experience at any point in their home country, during travel, or in resettlement housing.8

Even with limited examination and diagnostic resources, the skin is the most accessible first indication of patients’ overall well-being and often provides simple diagnostic clues—in combination with contextualization of the patient’s unique circumstances—necessary for successful diagnosis and treatment of scabies and pediculosis.12 The dermatologist working with refugee populations may be the first set of eyes available and trained to discern skin infestations and therefore has the potential to improve overall outcomes.

Some parasitic infestations in refugee populations may fall under the category of neglected tropical diseases, including scabies, ascariasis, trypanosomiasis, leishmaniasis, and schistosomiasis; they affect an estimated 1 billion individuals across the globe but historically have been underrepresented in the literature and in health policy due in part to limited access to care.13 This review will focus on infestations by the scabies mite (Sarcoptes scabiei var hominis) and the human louse, as these frequently are encountered, easily diagnosed, and treatable by trained clinicians, even in resource-limited settings.

Scabies

Scabies is a parasitic skin infestation caused by the 8-legged mite Sarcoptes scabiei var hominis. The female mite begins the infestation process via penetration of the epidermis, particularly the stratum corneum, and commences laying eggs (Figure 1). The subsequent larvae emerge 48 to 72 hours later and remain burrowed in the epidermis. The larvae mature over the next 10 to 14 days and continue the reproductive cycle.14,15 Symptoms of infestation occurs due to a hypersensitivity reaction to the mite and its by-products.16 Transmission of the mite primarily occurs via direct (skin-to-skin) contact with infected individuals or environmental surfaces for 24 to36 hours in specific conditions, though the latter source has been debated in the literature.

Sarcoptes scabiei mite (A), ova (B), and scybala (C) on microscopic evaluation.
FIGURE 1. Sarcoptes scabiei mite (A), ova (B), and scybala (C) on microscopic evaluation.

 

 

The method of transmission is particularly important when considering care for refugee populations. Scabies is found most often in those living in or traveling from tropical regions including East Asia, Southeast Asia, Oceania, and Latin America.17 In displaced or refugee populations, a lack of access to basic hygiene, extended travel in close quarters, and suboptimal health care access all may lead to an increased incidence of untreated scabies infestations.18 Scabies is more prevalent in children, with increased potential for secondary bacterial infections with Streptococcus and Staphylococcus species due to excoriation in unsanitary conditions. Secondary infection with Streptococcus pyogenes can lead to acute poststreptococcal glomerulonephritis, which accounts for a large burden of chronic kidney disease in affected populations.19 However, scabies may be found in any population, regardless of hygiene or health care access. Treating health care providers should keep a broad differential.

Presentation—The latency of scabies symptoms is 2 to 6 weeks in a primary outbreak and may be as short as 1 to 3 days with re-infestation, following the course of delayed-type hypersensitivity.20 The initial hallmark symptom is pruritus with increased severity in the evening. Visible lesions, excoriations, and burrows associated with scattered vesicles or pustules may be seen over the web spaces of the hands and feet, volar surfaces of the wrists, axillae, waist, genitalia, inner thighs, or buttocks.19 Chronic infestation often manifests with genital nodules. In populations with limited access to health care, there are reports of a sensitization phenomenon in which the individual may become less symptomatic after 4 to 6 weeks and yet be a potential carrier of the mite.21

Those with compromised immune function, such as individuals living with HIV or severe malnutrition, may present with crusted scabies, a variant that manifests as widespread hyperkeratotic scaling with more pronounced involvement of the head, neck, and acral areas. In contrast to classic scabies, crusted scabies is associated with minimal pruritus.22

Diagnosis—The diagnosis of scabies is largely clinical with confirmation through skin scrapings. The International Alliance for Control of Scabies has established diagnostic criteria that include a combination of clinical findings, history, and visualization of mites.23 A dermatologist working with refugee populations may employ any combination of history (eg, nocturnal itch, exposure to an affected individual) or clinical findings along with a high degree of suspicion in those with elevated risk. Visualization of mites is helpful to confirm the diagnosis and may be completed with the application of mineral oil at the terminal end of a burrow, skin scraping with a surgical blade or needle, and examination under light microscopy.

Treatment—First-line treatment for scabies consists of application of permethrin cream 5% on the skin of the neck to the soles of the feet, which is to be left on for 8 to 14 hours followed by rinsing. Re-application is recommended in 1 to 2 weeks. Oral ivermectin is a reasonable alternative to permethrin cream due to its low cost and easy administration in large affected groups. It is not labeled for use in pregnant women or children weighing less than 15 kg but has no selective fetal toxicity. Treatment of scabies with ivermectin has the benefit of treating many other parasitic infections. Both medications are on the World Health Organization Model List of Essential Medications and are widely available for treating providers, even in resource-limited settings.24

Much of the world still uses benzyl benzoate or precipitated sulfur ointment to treat scabies, and some botanicals used in folk medicine have genuine antiscabetic properties. Pruritus may persist for 1 to 4 weeks following treatment and does not indicate treatment failure. Topical camphor and menthol preparations, low-potency topical corticosteroids, or emollients all may be employed for relief.25Sarna is a Spanish term for scabies and has become the proprietary name for topical antipruritic agents. Additional methods of treatment and prevention include washing clothes and linens in hot water and drying on high heat. If machine washing is not available, clothing and linens may be sealed in a plastic bag for 72 hours.

Pediculosis

Pediculosis is an infestation caused by the ectoparasite Pediculus humanus, an obligate, sesame seed–sized louse that feeds exclusively on the blood of its host (Figure 2).26 Of the lice species, 2 require humans as hosts; one is P humanus and the other is Pthirus pubis (pubic lice). Pediculus humanus may be further classified into morphologies based largely on the affected area: body (P humanus corporis) or head (P humanus capitis), both of which will be discussed.27

Pediculus humanus (louse), adult form.
FIGURE 2. Pediculus humanus (louse), adult form.

 

 

Lice primarily attach to clothing and hair shafts, then transfer to the skin for blood feeds. Females lay eggs that hatch 6 to 10 days later, subsequently maturing into adults. The lifespan of these parasites with regular access to a host is 1 to 3 months for head lice and 18 days for body lice vs only 3 to 5 days without a host.28 Transmission of P humanus capitis primarily occurs via direct contact with affected individuals, either head-to-head contact or sharing of items such as brushes and headscarves; P humanus corporis also may be transmitted via direct contact with affected individuals or clothing.

Pediculosis is an important infestation to consider when providing care for refugee populations. Risk factors include lack of access to basic hygiene, including regular bathing or laundering of clothing, and crowded conditions that make direct person-to-person contact with affected individuals more likely.29 Body lice are associated more often with domestic turbulence and displaced populations30 in comparison to head lice, which have broad demographic variables, most often affecting females and children.28 Fatty acids in adult male sebum make the scalp less hospitable to lice.

Presentation—The most common clinical manifestation of pediculosis is pruritus. Cutaneous findings can include papules, wheals, or hemorrhagic puncta secondary to the louse bite. Due to the Tyndall effect of deep hemosiderin pigment, blue-grey macules termed maculae ceruleae (Figure 3) also may be present in chronic infestations of pediculosis pubis, in contrast to pediculosis capitis or corporis.31 Body louse infestation is associated with a general pruritus concentrated on the neck, shoulders, and waist—areas where clothing makes the most direct contact. Lesions may be visible and include eczematous patches with excoriation and possible secondary bacterial infection. Chronic infestation may exhibit lichenification or hyperpigmentation in associated areas. Head lice most often manifest with localized scalp pruritus and associated excoriation and cervical or occipital lymphadenopathy.32

Maculae ceruleae—blue-grey macules—may be present on the skin secondary to Pediculosis infestation.
FIGURE 3. Maculae ceruleae—blue-grey macules—may be present on the skin secondary to Pediculosis infestation.

Diagnosis—The diagnosis of pediculosis is clinical, with confirmation requiring direct examination of the insect or nits (the egg case of the parasite)(Figure 4). Body lice and associated nits can be visualized on clothing seams near areas of highest body temperature, particularly the waistband. Head lice may be visualized crawling on hair shafts or on a louse comb. Nits are firmly attached to hair shafts and are visible to the naked eye, whereas pseudonits slide freely along the hair shaft and are not a manifestation of louse infestation (Figure 5).31

Pediculosis nits—the egg cases of the parasite—may firmly attach to the hair shaft.
FIGURE 4. Pediculosis nits—the egg cases of the parasite—may firmly attach to the hair shaft.

Treatment—Treatment varies by affected area. Pediculosis corporis may be treated with permethrin cream 5% applied to the entire body and left on for 8 to 10 hours, but this may not be necessary if facilities are available to wash and dry clothing.33 The use of oral ivermectin and permethrin-impregnated underwear both have been proposed.34,35 Treatment of pediculosis capitis may be accomplished with a variety of topical pediculicides including permethrin, pyrethrum with piperonyl butoxide, dimethicone, malathion, benzyl alcohol, spinosad, and topical ivermectin.22 Topical corticosteroids or emollients may be employed for residual pruritus.

The pseudonit closely mimics pediculosis nits but consists of keratinized cell casts that are freely dislodged.
FIGURE 5. The pseudonit closely mimics pediculosis nits but consists of keratinized cell casts that are freely dislodged.

Equally important is environmental elimination of infestation. Clothing should be discarded if possible or washed and dried using high heat. If neither approach is possible or appropriate, clothing may be sealed in a plastic bag for 2 weeks or treated with a pediculicide. Nit combing is an important adjunct in the treatment of pediculosis capitis.36 It is important to encourage return to work and/or school immediately after treatment. “No nit” policies are more harmful to education than helpful for prevention of investation.37

Pediculosis corporis may transmit infectious agents including Bartonella quintana, (trench fever, endocarditis, bacillary angiomatosis), Borrelia recurrentis (louse-borne relapsing fever), and Rickettsia prowazekii (epidemic typhus).31,38,39 Additionally, severe pediculosis infestations have the potential to cause chronic blood loss in affected populations. In a study of patients with active pediculosis infestation, mean hemoglobin values were found to be 2.5 g/dL lower than a matched population without infestation.40 It is important to consider pediculosis as a risk for iron-deficiency anemia in populations who are known to lack access to regular medical evaluation.41

 

 

Future Considerations

Increased access to tools and education for clinicians treating refugee populations is key to reducing the burden of parasitic skin disease and related morbidity and mortality in vulnerable groups both domestically and globally. One such tool, the Skin NTDs App, was launched by the World Health Organization in 2020. It is available for free for Android and iOS devices to assist clinicians in the field with the diagnosis and treatment of neglected tropical diseases—including scabies—that may affect refugee populations.42

Additionally, to both improve access and limit preventable sequelae, future investigations into appropriate models of community-based care are paramount. The model of community-based care is centered on the idea of care provision that prioritizes safety, accessibility, affordability, and acceptability in an environment closest to vulnerable populations. The largest dermatologic society, the International League of Dermatological Societies, formed a Migrant Health Dermatology Working Group that prioritizes understanding and improving care for refugee and migrant populations; this group hosted a summit in 2022, bringing together international subject matter leaders to discuss such models of care and set goals for the creation of tool kits for patients, frontline health care workers, and dermatologists.43

Conclusion

Improvement in dermatologic care of refugee populations includes provision of culturally and linguistically appropriate care by trained clinicians, adequate access to the most essential medications, and basic physical or legal access to health care systems in general.8,11,44 Parasitic infestations have the potential to remain asymptomatic for extended periods of time and result in spread to potentially nonendemic regions of resettlement.45 Additionally, the psychosocial well-being of refugee populations upon resettlement may be negatively affected by stigma of disease processes such as scabies and pediculosis, leading to additional barriers to successful re-entry into the patient’s new environment.46 Therefore, proper screening, diagnosis, and treatment of the most common parasitic infestations in this population have great potential to improve outcomes for large groups across the globe.

References
  1. Monin K, Batalova J, Lai T. Refugees and Asylees in the United States. Migration Information Source. Published May 13, 2021. Accessed April 4, 2024. https://www.migrationpolicy.org/article/refugees-and-asylees-united-states-2021
  2. UNHCR. Figures at a Glance. UNHCR USA. Update June 14, 2023. Accessed April 4, 2024. https://www.unhcr.org/en-us/figures-at-a-glance.html
  3. UNHCR. Refugee resettlement facts. Published October 2023. Accessed April 8, 2024. https://www.unhcr.org/us/media/refugee-resettlement-facts
  4. US Department of State. Report to Congress on Proposed Refugee Admissions for Fiscal Year 2024. Published November 3, 2023. Accessed April 8, 2024. https://www.state.gov/report-to-congress-on-proposed-refugee-admissions-for-fiscal-year-2024/
  5. UNHCR. Compact for Migration: Definitions. United Nations. Accessed April 4, 2024. https://refugeesmigrants.un.org/definitions
  6. United Nations High Commissioner for Refugees (UNHCR). Convention and Protocol Relating to the Status of Refugees. Published December 2010. Accessed January 11, 2024. https://www.unhcr.org/us/media/convention-and-protocol-relating-status-refugees
  7. Kibar Öztürk M. Skin diseases in rural Nyala, Sudan (in a rural hospital, in 12 orphanages, and in two refugee camps). Int J Dermatol. 2019;58:1341-1349. doi:10.1111/ijd.14619
  8. Padovese V, Knapp A. Challenges of managing skin diseases in refugees and migrants. Dermatol Clin. 2021;39:101-115. doi:10.1016/j.det.2020.08.010
  9. Saikal SL, Ge L, Mir A, et al. Skin disease profile of Syrian refugees in Jordan: a field-mission assessment. J Eur Acad Dermatol Venereol. 2020;34:419-425. doi:10.1111/jdv.15909
  10. Eonomopoulou A, Pavli A, Stasinopoulou P, et al. Migrant screening: lessons learned from the migrant holding level at the Greek-Turkish borders. J Infect Public Health. 2017;10:177-184. doi:10.1016/j.jiph.2016.04.012
  11. Marano N, Angelo KM, Merrill RD, et al. Expanding travel medicine in the 21st century to address the health needs of the world’s migrants.J Travel Med. 2018;25. doi:10.1093/jtm/tay067
  12. Hay RJ, Asiedu K. Skin-related neglected tropical diseases (skin NTDs)—a new challenge. Trop Med Infect Dis. 2018;4. doi:10.3390/tropicalmed4010004
  13. NIAID. Neglected tropical diseases. Updated July 11, 2016. Accessed April 4, 2024. https://www.niaid.nih.gov/research/neglected-tropical-diseases
  14. Arlian LG, Morgan MS. A review of Sarcoptes scabiei: past, present and future. Parasit Vectors. 2017;10:297. doi:10.1186/s13071-017-2234-1
  15. Arlian LG, Runyan RA, Achar S, et al. Survival and infectivity of Sarcoptes scabiei var. canis and var. hominis. J Am Acad Dermatol. 1984;11(2 pt 1):210-215. doi:10.1016/s0190-9622(84)70151-4
  16. Chandler DJ, Fuller LC. A review of scabies: an infestation more than skin deep. Dermatology. 2019;235:79-90. doi:10.1159/000495290
  17. Karimkhani C, Colombara DV, Drucker AM, et al. The global burden of scabies: a cross-sectional analysis from the Global Burden of Disease Study 2015. Lancet Infect Dis. 2017;17:1247-1254. doi:10.1016/S1473-3099(17)30483-8
  18. Romani L, Steer AC, Whitfeld MJ, et al. Prevalence of scabies and impetigo worldwide: a systematic review. Lancet Infect Dis. 2015;15:960-967. doi:10.1016/S1473-3099(15)00132-2
  19. Thomas C, Coates SJ, Engelman D, et al. Ectoparasites: scabies. J Am Acad Dermatol. 2020;82:533-548. doi:10.1016/j.jaad.2019.05.109
  20. Mellanby K, Johnson CG, Bartley WC. Treatment of scabies. Br Med J. 1942;2:1-4. doi:10.1136/bmj.2.4252.1
  21. Walton SF. The immunology of susceptibility and resistance to scabies. Parasit Immunol. 2010;32:532-540. doi:10.1111/j.1365-3024.2010.01218.x
  22. Coates SJ, Thomas C, Chosidow O, et al. Ectoparasites: pediculosis and tungiasis. J Am Acad Dermatol. 2020;82:551-569. doi:10.1016/j.jaad.2019.05.110
  23. Engelman D, Fuller LC, Steer AC; International Alliance for the Control of Scabies Delphi p. Consensus criteria for the diagnosis of scabies: a Delphi study of international experts. PLoS Negl Trop Dis. 2018;12:E0006549. doi:10.1371/journal.pntd.0006549
  24. World Health Organization. WHO Model Lists of Essential Medicines—23rd list, 2023. Updated July 26, 2023. Accessed April 8, 2024. https://www.who.int/publications/i/item/WHO-MHP-HPS-EML-2023.02
  25. Salavastru CM, Chosidow O, Boffa MJ, et al. European guideline for the management of scabies. J Eur Acad Dermatol Venereol. 2017;31:1248-1253. doi:10.1111/jdv.14351
  26. Badiaga S, Brouqui P. Human louse-transmitted infectious diseases. Clin Microbiol Infect. 2012;18:332-337. doi:10.1111/j.1469-0691.2012.03778.x
  27. Leo NP, Campbell NJH, Yang X, et al. Evidence from mitochondrial DNA that head lice and body lice of humans (Phthiraptera: Pediculidae) are conspecific. J Med Entomol. 2002;39:662-666. doi:10.1603/0022-2585-39.4.662
  28. Chosidow O. Scabies and pediculosis. Lancet. 2000;355:819-826. doi:10.1016/S0140-6736(99)09458-1
  29. Arnaud A, Chosidow O, Détrez M-A, et al. Prevalences of scabies and pediculosis corporis among homeless people in the Paris region: results from two randomized cross-sectional surveys (HYTPEAC study). Br J Dermatol. 2016;174:104-112. doi:10.1111/bjd.14226
  30. Brouqui P. Arthropod-borne diseases associated with political and social disorder. Annu Rev Entomol. 2011;56:357-374. doi:10.1146/annurev-ento-120709-144739
  31. Ko CJ, Elston DM. Pediculosis. J Am Acad Dermatol. 2004;50:1-12. doi:10.1016/S0190-9622(03)02729-4
  32. Bloomfield D. Head lice. Pediatr Rev. 2002;23:34-35; discussion 34-35. doi:10.1542/pir.23-1-34
  33. Stone SP GJ, Bacelieri RE. Scabies, other mites, and pediculosis. In: Wolf K GL, Katz SI, et al (eds). Fitzpatrick’s Dermatology in General Medicine. McGraw Hill; 2008:2029.
  34. Foucault C, Ranque S, Badiaga S, et al. Oral ivermectin in the treatment of body lice. J Infect Dis. 2006;193:474-476. doi:10.1086/499279
  35. Benkouiten S, Drali R, Badiaga S, et al. Effect of permethrin-impregnated underwear on body lice in sheltered homeless persons: a randomized controlled trial. JAMA Dermatol. 2014;150:273-279. doi:10.1001/jamadermatol.2013.6398
  36. CDC. Parasites: Treatment. Updated October 15, 2019. Accessed April 4, 2024. https://www.cdc.gov/parasites/lice/head/treatment.html
  37. Devore CD, Schutze GE; Council on School Health and Committee on Infectious Diseases, American Academy of Pediatrics. Head lice. Pediatrics. 2015;135:e1355-e1365. doi:10.1542/peds.2015-0746
  38. Ohl ME, Spach DH. Bartonella quintana and urban trench fever. Clin Infect Dis. 2000;31:131-135. doi:10.1086/313890
  39. Drali R, Sangaré AK, Boutellis A, et al. Bartonella quintana in body lice from scalp hair of homeless persons, France. Emerg Infect Dis. 2014;20:907-908. doi:10.3201/eid2005.131242
  40. Rudd N, Zakaria A, Kohn MA, et al. Association of body lice infestation with hemoglobin values in hospitalized dermatology patients. JAMA Dermatol. 2022;158:691-693. doi:10.1001/jamadermatol.2022.0818
  41. Guss DA, Koenig M, Castillo EM. Severe iron deficiency anemia and lice infestation. J Emergency Med. 2011;41:362-365. doi:10.1016/j.jemermed.2010.05.030
  42. Neglected tropical diseases of the skin: WHO launches mobile application to facilitate diagnosis. News release. World Health Organization; July 16, 2020. Accessed April 4, 2024. https://www.who.int/news/item/16-07-2020-neglected-tropical-diseases-of-the-skin-who-launches-mobile-application-to-facilitate-diagnosis
  43. Padovese V, Fuller LC, Griffiths CEM, et al; Migrant Health Dermatology Working Group of the International Foundation for Dermatology. Migrant skin health: perspectives from the Migrant Health Summit, Malta, 2022. Br J Dermatology. 2023;188:553-554. doi:10.1093/bjd/ljad001
  44. Knapp AP, Rehmus W, Chang AY. Skin diseases in displaced populations: a review of contributing factors, challenges, and approaches to care. Int J Dermatol. 2020;59:1299-1311. doi:10.1111/ijd.15063
  45. Norman FF, Comeche B, Chamorro S, et al. Overcoming challenges in the diagnosis and treatment of parasitic infectious diseases in migrants. Expert Rev Anti-infective Therapy. 2020;18:127-143. doi:10.1080/14787210.2020.1713099
  46. Skin NTDs: prioritizing integrated approaches to reduce suffering, psychosocial impact and stigmatization. News release. World Health Organization; October 29, 2020. Accessed April 4, 2024. https://www.who.int/news/item/29-10-2020-skin-ntds-prioritizing-integrated-approaches-to-reduce-suffering-psychosocial-impact-and-stigmatization
References
  1. Monin K, Batalova J, Lai T. Refugees and Asylees in the United States. Migration Information Source. Published May 13, 2021. Accessed April 4, 2024. https://www.migrationpolicy.org/article/refugees-and-asylees-united-states-2021
  2. UNHCR. Figures at a Glance. UNHCR USA. Update June 14, 2023. Accessed April 4, 2024. https://www.unhcr.org/en-us/figures-at-a-glance.html
  3. UNHCR. Refugee resettlement facts. Published October 2023. Accessed April 8, 2024. https://www.unhcr.org/us/media/refugee-resettlement-facts
  4. US Department of State. Report to Congress on Proposed Refugee Admissions for Fiscal Year 2024. Published November 3, 2023. Accessed April 8, 2024. https://www.state.gov/report-to-congress-on-proposed-refugee-admissions-for-fiscal-year-2024/
  5. UNHCR. Compact for Migration: Definitions. United Nations. Accessed April 4, 2024. https://refugeesmigrants.un.org/definitions
  6. United Nations High Commissioner for Refugees (UNHCR). Convention and Protocol Relating to the Status of Refugees. Published December 2010. Accessed January 11, 2024. https://www.unhcr.org/us/media/convention-and-protocol-relating-status-refugees
  7. Kibar Öztürk M. Skin diseases in rural Nyala, Sudan (in a rural hospital, in 12 orphanages, and in two refugee camps). Int J Dermatol. 2019;58:1341-1349. doi:10.1111/ijd.14619
  8. Padovese V, Knapp A. Challenges of managing skin diseases in refugees and migrants. Dermatol Clin. 2021;39:101-115. doi:10.1016/j.det.2020.08.010
  9. Saikal SL, Ge L, Mir A, et al. Skin disease profile of Syrian refugees in Jordan: a field-mission assessment. J Eur Acad Dermatol Venereol. 2020;34:419-425. doi:10.1111/jdv.15909
  10. Eonomopoulou A, Pavli A, Stasinopoulou P, et al. Migrant screening: lessons learned from the migrant holding level at the Greek-Turkish borders. J Infect Public Health. 2017;10:177-184. doi:10.1016/j.jiph.2016.04.012
  11. Marano N, Angelo KM, Merrill RD, et al. Expanding travel medicine in the 21st century to address the health needs of the world’s migrants.J Travel Med. 2018;25. doi:10.1093/jtm/tay067
  12. Hay RJ, Asiedu K. Skin-related neglected tropical diseases (skin NTDs)—a new challenge. Trop Med Infect Dis. 2018;4. doi:10.3390/tropicalmed4010004
  13. NIAID. Neglected tropical diseases. Updated July 11, 2016. Accessed April 4, 2024. https://www.niaid.nih.gov/research/neglected-tropical-diseases
  14. Arlian LG, Morgan MS. A review of Sarcoptes scabiei: past, present and future. Parasit Vectors. 2017;10:297. doi:10.1186/s13071-017-2234-1
  15. Arlian LG, Runyan RA, Achar S, et al. Survival and infectivity of Sarcoptes scabiei var. canis and var. hominis. J Am Acad Dermatol. 1984;11(2 pt 1):210-215. doi:10.1016/s0190-9622(84)70151-4
  16. Chandler DJ, Fuller LC. A review of scabies: an infestation more than skin deep. Dermatology. 2019;235:79-90. doi:10.1159/000495290
  17. Karimkhani C, Colombara DV, Drucker AM, et al. The global burden of scabies: a cross-sectional analysis from the Global Burden of Disease Study 2015. Lancet Infect Dis. 2017;17:1247-1254. doi:10.1016/S1473-3099(17)30483-8
  18. Romani L, Steer AC, Whitfeld MJ, et al. Prevalence of scabies and impetigo worldwide: a systematic review. Lancet Infect Dis. 2015;15:960-967. doi:10.1016/S1473-3099(15)00132-2
  19. Thomas C, Coates SJ, Engelman D, et al. Ectoparasites: scabies. J Am Acad Dermatol. 2020;82:533-548. doi:10.1016/j.jaad.2019.05.109
  20. Mellanby K, Johnson CG, Bartley WC. Treatment of scabies. Br Med J. 1942;2:1-4. doi:10.1136/bmj.2.4252.1
  21. Walton SF. The immunology of susceptibility and resistance to scabies. Parasit Immunol. 2010;32:532-540. doi:10.1111/j.1365-3024.2010.01218.x
  22. Coates SJ, Thomas C, Chosidow O, et al. Ectoparasites: pediculosis and tungiasis. J Am Acad Dermatol. 2020;82:551-569. doi:10.1016/j.jaad.2019.05.110
  23. Engelman D, Fuller LC, Steer AC; International Alliance for the Control of Scabies Delphi p. Consensus criteria for the diagnosis of scabies: a Delphi study of international experts. PLoS Negl Trop Dis. 2018;12:E0006549. doi:10.1371/journal.pntd.0006549
  24. World Health Organization. WHO Model Lists of Essential Medicines—23rd list, 2023. Updated July 26, 2023. Accessed April 8, 2024. https://www.who.int/publications/i/item/WHO-MHP-HPS-EML-2023.02
  25. Salavastru CM, Chosidow O, Boffa MJ, et al. European guideline for the management of scabies. J Eur Acad Dermatol Venereol. 2017;31:1248-1253. doi:10.1111/jdv.14351
  26. Badiaga S, Brouqui P. Human louse-transmitted infectious diseases. Clin Microbiol Infect. 2012;18:332-337. doi:10.1111/j.1469-0691.2012.03778.x
  27. Leo NP, Campbell NJH, Yang X, et al. Evidence from mitochondrial DNA that head lice and body lice of humans (Phthiraptera: Pediculidae) are conspecific. J Med Entomol. 2002;39:662-666. doi:10.1603/0022-2585-39.4.662
  28. Chosidow O. Scabies and pediculosis. Lancet. 2000;355:819-826. doi:10.1016/S0140-6736(99)09458-1
  29. Arnaud A, Chosidow O, Détrez M-A, et al. Prevalences of scabies and pediculosis corporis among homeless people in the Paris region: results from two randomized cross-sectional surveys (HYTPEAC study). Br J Dermatol. 2016;174:104-112. doi:10.1111/bjd.14226
  30. Brouqui P. Arthropod-borne diseases associated with political and social disorder. Annu Rev Entomol. 2011;56:357-374. doi:10.1146/annurev-ento-120709-144739
  31. Ko CJ, Elston DM. Pediculosis. J Am Acad Dermatol. 2004;50:1-12. doi:10.1016/S0190-9622(03)02729-4
  32. Bloomfield D. Head lice. Pediatr Rev. 2002;23:34-35; discussion 34-35. doi:10.1542/pir.23-1-34
  33. Stone SP GJ, Bacelieri RE. Scabies, other mites, and pediculosis. In: Wolf K GL, Katz SI, et al (eds). Fitzpatrick’s Dermatology in General Medicine. McGraw Hill; 2008:2029.
  34. Foucault C, Ranque S, Badiaga S, et al. Oral ivermectin in the treatment of body lice. J Infect Dis. 2006;193:474-476. doi:10.1086/499279
  35. Benkouiten S, Drali R, Badiaga S, et al. Effect of permethrin-impregnated underwear on body lice in sheltered homeless persons: a randomized controlled trial. JAMA Dermatol. 2014;150:273-279. doi:10.1001/jamadermatol.2013.6398
  36. CDC. Parasites: Treatment. Updated October 15, 2019. Accessed April 4, 2024. https://www.cdc.gov/parasites/lice/head/treatment.html
  37. Devore CD, Schutze GE; Council on School Health and Committee on Infectious Diseases, American Academy of Pediatrics. Head lice. Pediatrics. 2015;135:e1355-e1365. doi:10.1542/peds.2015-0746
  38. Ohl ME, Spach DH. Bartonella quintana and urban trench fever. Clin Infect Dis. 2000;31:131-135. doi:10.1086/313890
  39. Drali R, Sangaré AK, Boutellis A, et al. Bartonella quintana in body lice from scalp hair of homeless persons, France. Emerg Infect Dis. 2014;20:907-908. doi:10.3201/eid2005.131242
  40. Rudd N, Zakaria A, Kohn MA, et al. Association of body lice infestation with hemoglobin values in hospitalized dermatology patients. JAMA Dermatol. 2022;158:691-693. doi:10.1001/jamadermatol.2022.0818
  41. Guss DA, Koenig M, Castillo EM. Severe iron deficiency anemia and lice infestation. J Emergency Med. 2011;41:362-365. doi:10.1016/j.jemermed.2010.05.030
  42. Neglected tropical diseases of the skin: WHO launches mobile application to facilitate diagnosis. News release. World Health Organization; July 16, 2020. Accessed April 4, 2024. https://www.who.int/news/item/16-07-2020-neglected-tropical-diseases-of-the-skin-who-launches-mobile-application-to-facilitate-diagnosis
  43. Padovese V, Fuller LC, Griffiths CEM, et al; Migrant Health Dermatology Working Group of the International Foundation for Dermatology. Migrant skin health: perspectives from the Migrant Health Summit, Malta, 2022. Br J Dermatology. 2023;188:553-554. doi:10.1093/bjd/ljad001
  44. Knapp AP, Rehmus W, Chang AY. Skin diseases in displaced populations: a review of contributing factors, challenges, and approaches to care. Int J Dermatol. 2020;59:1299-1311. doi:10.1111/ijd.15063
  45. Norman FF, Comeche B, Chamorro S, et al. Overcoming challenges in the diagnosis and treatment of parasitic infectious diseases in migrants. Expert Rev Anti-infective Therapy. 2020;18:127-143. doi:10.1080/14787210.2020.1713099
  46. Skin NTDs: prioritizing integrated approaches to reduce suffering, psychosocial impact and stigmatization. News release. World Health Organization; October 29, 2020. Accessed April 4, 2024. https://www.who.int/news/item/29-10-2020-skin-ntds-prioritizing-integrated-approaches-to-reduce-suffering-psychosocial-impact-and-stigmatization
Issue
Cutis - 113(4)
Issue
Cutis - 113(4)
Page Number
E16-E21
Page Number
E16-E21
Publications
Publications
Topics
Article Type
Display Headline
Dermatologic Care for Refugees: Effective Management of Scabies and Pediculosis
Display Headline
Dermatologic Care for Refugees: Effective Management of Scabies and Pediculosis
Sections
Inside the Article

Practice Points

  • War and natural disasters displace populations and disrupt infrastructure and access to medical care.
  • Infestations and cutaneous infections are common among refugee populations, and impetigo often is a sign of underlying scabies infestation.
  • Body lice are important disease vectors inrefugee populations.
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

Botanical Briefs: Fig Phytophotodermatitis (Ficus carica)

Article Type
Changed
Wed, 04/10/2024 - 10:12
Display Headline
Botanical Briefs: Fig Phytophotodermatitis (Ficus carica)

Plant Parts and Nomenclature

Ficus carica (common fig) is a deciduous shrub or small tree with smooth gray bark that can grow up to 10 m in height (Figure 1). It is characterized by many spreading branches, but the trunk rarely grows beyond a diameter of 7 in. Its hairy leaves are coarse on the upper side and soft underneath with 3 to 7 deep lobes that can extend up to 25 cm in length or width; the leaves grow individually, alternating along the sides of the branches. Fig trees often can be seen adorning yards, gardens, and parks, especially in tropical and subtropical climates. Ficus carica should not be confused with Ficus benjamina (weeping fig), a common ornamental tree that also is used to provide shade in hot climates, though both can cause phototoxic skin eruptions.

Ficus carica (common fig).
FIGURE 1. Ficus carica (common fig).

The common fig tree originated in the Mediterranean and western Asia1 and has been cultivated by humans since the second and third millennia bc for its fruit, which commonly is used to sweeten cookies, cakes, and jams.2 Figs are the most commonly mentioned food plant in the Bible, with at least 56 references in the Old and New Testaments.3 The “fruit” technically is a syconium—a hollow fleshy receptacle with a small opening at the apex partly closed by small scales. It can be obovoid, turbinate, or pear shaped; can be 1 to 4 inches long; and can vary in color from yellowish green to coppery, bronze, or dark purple (Figure 2).

Immature fruit of the common fig tree.
FIGURE 2. Immature fruit of the common fig tree.

Ficus carica is a member of the Moraceae family (derived from the Latin name for the mulberry tree), which includes 53 genera and approximately 1400 species, of which about 850 belong to the genus Ficus (the Latin name for a fig tree). The term carica likely comes from the Latin word carricare (to load) to describe a tree loaded with figs. Family members include trees, shrubs, lianas, and herbs that usually contain laticifers with a milky latex.

Traditional Uses

For centuries, components of the fig tree have been used in herbal teas and pastes to treat ailments ranging from sore throats to diarrhea, though there is no evidence to support their efficacy.4 Ancient Indians and Egyptians used plants such as the common fig tree containing furocoumarins to induce hyperpigmentation in vitiligo.5

Phototoxic Components

The leaves and sap of the common fig tree contain psoralens, which are members of the furocoumarin group of chemical compounds and are the source of its phototoxicity. The fruit does not contain psoralens.6-9 The tree also produces proteolytic enzymes such as protease, amylase, ficin, triterpenoids, and lipodiastase that enhance its phototoxic effects.8 Exposure to UV light between 320 and 400 nm following contact with these phototoxic components triggers a reaction in the skin over the course of 1 to 3 days.5 The psoralens bind in epidermal cells, cross-link the DNA, and cause cell-membrane destruction, leading to edema and necrosis.10 The delay in symptoms may be attributed to the time needed to synthesize acute-phase reaction proteins such as tumor necrosis factor α and IL-1.11 In spring and summer months, an increased concentration of psoralens in the leaves and sap contribute to an increased incidence of phytophotodermatitis.9 Humidity and sweat also increase the percutaneous absorption of psoralens.12,13

Allergens

Fig trees produce a latex protein that can cause cross-reactive hypersensitivity reactions in those allergic to F benjamina latex and rubber latex.6 The latex proteins in fig trees can act as airborne respiratory allergens. Ingestion of figs can produce anaphylactic reactions in those sensitized to rubber latex and F benjamina latex.7 Other plant families associated with phototoxic reactions include Rutaceae (lemon, lime, bitter orange), Apiaceae (formerly Umbelliferae)(carrot, parsnip, parsley, dill, celery, hogweed), and Fabaceae (prairie turnip).

 

 

Cutaneous Manifestations

Most cases of fig phytophotodermatitis begin with burning, pain, and/or itching within hours of sunlight exposure in areas of the skin that encountered components of the fig tree, often in a linear pattern. The affected areas become erythematous and edematous with formation of bullae and unilocular vesicles over the course of 1 to 3 days.12,14,15 Lesions may extend beyond the region of contact with the fig tree as they spread across the skin due to sweat or friction, and pain may linger even after the lesions resolve.12,13,16 Adults who handle fig trees (eg, pruning) are susceptible to phototoxic reactions, especially those using chain saws or other mechanisms that result in spray exposure, as the photosensitizing sap permeates the wood and bark of the entire tree.17 Similarly, children who handle fig leaves or sap during outdoor play can develop bullous eruptions. Severe cases have resulted in hospital admission after prolonged exposure.16 Additionally, irritant dermatitis may arise from contact with the trichomes or “hairs” on various parts of the plant.

Leaves and milky sap of the common fig tree.
Image provided with permission by Scott Norton, MD, MPH, MSc (Washington, DC).
FIGURE 3. Leaves and milky sap of the common fig tree.

Patients who use natural remedies containing components of the fig tree without the supervision of a medical provider put themselves at risk for unsafe or unwanted adverse effects, such as phytophotodermatitis.12,15,16,18 An entire family presented with burns after they applied fig leaf extract to the skin prior to tanning outside in the sun.19 A 42-year-old woman acquired a severe burn covering 81% of the body surface after topically applying fig leaf tea to the skin as a tanning agent.20 A subset of patients ingesting or applying fig tree components for conditions such as vitiligo, dermatitis, onychomycosis, and motor retardation developed similar cutaneous reactions.13,14,21,22 Lesions resembling finger marks can raise concerns for potential abuse or neglect in children.22

The differential diagnosis for fig phytophotodermatitis includes sunburn, chemical burns, drug-related photosensitivity, infectious lesions (eg, herpes simplex, bullous impetigo, Lyme disease, superficial lymphangitis), connective tissue disease (eg, systemic lupus erythematosus), contact dermatitis, and nonaccidental trauma.12,15,18 Compared to sunburn, phytophotodermatitis tends to increase in severity over days following exposure and heals with dramatic hyperpigmentation, which also prompts visits to dermatology.12

Treatment

Treatment of fig phytophotodermatitis chiefly is symptomatic, including analgesia, appropriate wound care, and infection prophylaxis. Topical and systemic corticosteroids may aid in the resolution of moderate to severe reactions.15,23,24 Even severe injuries over small areas or mild injuries to a high percentage of the total body surface area may require treatment in a burn unit. Patients should be encouraged to use mineral-based sunscreens on the affected areas to reduce the risk for hyperpigmentation. Individuals who regularly handle fig trees should use contact barriers including gloves and protective clothing (eg, long-sleeved shirts, long pants).

References
  1. Ikegami H, Nogata H, Hirashima K, et al. Analysis of genetic diversity among European and Asian fig varieties (Ficus carica L.) using ISSR, RAPD, and SSR markers. Genetic Resources and Crop Evolution. 2009;56:201-209.
  2. Zohary D, Spiegel-Roy P. Beginnings of fruit growing in the Old World. Science. 1975;187:319-327.
  3. Young R. Young’s Analytical Concordance. Thomas Nelson; 1982.
  4. Duke JA. Handbook of Medicinal Herbs. CRC Press; 2002.
  5. Pathak MA, Fitzpatrick TB. Bioassay of natural and synthetic furocoumarins (psoralens). J Invest Dermatol. 1959;32:509-518.
  6. Focke M, Hemmer W, Wöhrl S, et al. Cross-reactivity between Ficus benjamina latex and fig fruit in patients with clinical fig allergy. Clin Exp Allergy. 2003;33:971-977.
  7. Hemmer W, Focke M, Götz M, et al. Sensitization to Ficus benjamina: relationship to natural rubber latex allergy and identification of foods implicated in the Ficus-fruit syndrome. Clin Exp Allergy. 2004;34:1251-1258.
  8. Bonamonte D, Foti C, Lionetti N, et al. Photoallergic contact dermatitis to 8-methoxypsoralen in Ficus carica. Contact Dermatitis. 2010;62:343-348.
  9. Zaynoun ST, Aftimos BG, Abi Ali L, et al. Ficus carica; isolation and quantification of the photoactive components. Contact Dermatitis. 1984;11:21-25.
  10. Tessman JW, Isaacs ST, Hearst JE. Photochemistry of the furan-side 8-methoxypsoralen-thymidine monoadduct inside the DNA helix. conversion to diadduct and to pyrone-side monoadduct. Biochemistry. 1985;24:1669-1676.
  11. Geary P. Burns related to the use of psoralens as a tanning agent. Burns. 1996;22:636-637.
  12. Redgrave N, Solomon J. Severe phytophotodermatitis from fig sap: a little known phenomenon. BMJ Case Rep. 2021;14:E238745.
  13. Ozdamar E, Ozbek S, Akin S. An unusual cause of burn injury: fig leaf decoction used as a remedy for a dermatitis of unknown etiology. J Burn Care Rehabil. 2003;24:229-233; discussion 228.
  14. Berakha GJ, Lefkovits G. Psoralen phototherapy and phototoxicity. Ann Plast Surg. 1985;14:458-461.
  15. Papazoglou A, Mantadakis E. Fig tree leaves phytophotodermatitis. J Pediatr. 2021;239:244-245.
  16. Imen MS, Ahmadabadi A, Tavousi SH, et al. The curious cases of burn by fig tree leaves. Indian J Dermatol. 2019;64:71-73.
  17. Rouaiguia-Bouakkaz S, Amira-Guebailia H, Rivière C, et al. Identification and quantification of furanocoumarins in stem bark and wood of eight Algerian varieties of Ficus carica by RP-HPLC-DAD and RP-HPLC-DAD-MS. Nat Prod Commun. 2013;8:485-486.
  18. Oliveira AA, Morais J, Pires O, et al. Fig tree induced phytophotodermatitis. BMJ Case Rep. 2020;13:E233392.
  19. Bassioukas K, Stergiopoulou C, Hatzis J. Erythrodermic phytophotodermatitis after application of aqueous fig-leaf extract as an artificial suntan promoter and sunbathing. Contact Dermatitis. 2004;51:94-95.
  20. Sforza M, Andjelkov K, Zaccheddu R. Severe burn on 81% of body surface after sun tanning. Ulus Travma Acil Cerrahi Derg. 2013;19:383-384.
  21. Son JH, Jin H, You HS, et al. Five cases of phytophotodermatitis caused by fig leaves and relevant literature review. Ann Dermatol. 2017;29:86-90.
  22. Abali AE, Aka M, Aydogan C, et al. Burns or phytophotodermatitis, abuse or neglect: confusing aspects of skin lesions caused by the superstitious use of fig leaves. J Burn Care Res. 2012;33:E309-E312.
  23. Picard C, Morice C, Moreau A, et al. Phytophotodermatitis in children: a difficult diagnosis mimicking other dermatitis. 2017;5:1-3.
  24. Enjolras O, Soupre V, Picard A. Uncommon benign infantile vascular tumors. Adv Dermatol. 2008;24:105-124.
Article PDF
Author and Disclosure Information

Drs. Barker and Elston are from the Medical University of South Carolina, Charleston. Dr. Barker is from the Department of Internal Medicine, and Dr. Elston is from the Department of Dermatology and Dermatologic Surgery. Dr. McGovern is from Fort Wayne Dermatology Consultants, Indiana.

The authors report no conflict of interest.

Correspondence: Catherine Shirer Barker, MD, 96 Jonathan Lucas St, Ste 807B, MSC 623, Charleston, SC 29425 (catherinesbarker@gmail.com).

Issue
Cutis - 113(4)
Publications
Topics
Page Number
167-169
Sections
Author and Disclosure Information

Drs. Barker and Elston are from the Medical University of South Carolina, Charleston. Dr. Barker is from the Department of Internal Medicine, and Dr. Elston is from the Department of Dermatology and Dermatologic Surgery. Dr. McGovern is from Fort Wayne Dermatology Consultants, Indiana.

The authors report no conflict of interest.

Correspondence: Catherine Shirer Barker, MD, 96 Jonathan Lucas St, Ste 807B, MSC 623, Charleston, SC 29425 (catherinesbarker@gmail.com).

Author and Disclosure Information

Drs. Barker and Elston are from the Medical University of South Carolina, Charleston. Dr. Barker is from the Department of Internal Medicine, and Dr. Elston is from the Department of Dermatology and Dermatologic Surgery. Dr. McGovern is from Fort Wayne Dermatology Consultants, Indiana.

The authors report no conflict of interest.

Correspondence: Catherine Shirer Barker, MD, 96 Jonathan Lucas St, Ste 807B, MSC 623, Charleston, SC 29425 (catherinesbarker@gmail.com).

Article PDF
Article PDF

Plant Parts and Nomenclature

Ficus carica (common fig) is a deciduous shrub or small tree with smooth gray bark that can grow up to 10 m in height (Figure 1). It is characterized by many spreading branches, but the trunk rarely grows beyond a diameter of 7 in. Its hairy leaves are coarse on the upper side and soft underneath with 3 to 7 deep lobes that can extend up to 25 cm in length or width; the leaves grow individually, alternating along the sides of the branches. Fig trees often can be seen adorning yards, gardens, and parks, especially in tropical and subtropical climates. Ficus carica should not be confused with Ficus benjamina (weeping fig), a common ornamental tree that also is used to provide shade in hot climates, though both can cause phototoxic skin eruptions.

Ficus carica (common fig).
FIGURE 1. Ficus carica (common fig).

The common fig tree originated in the Mediterranean and western Asia1 and has been cultivated by humans since the second and third millennia bc for its fruit, which commonly is used to sweeten cookies, cakes, and jams.2 Figs are the most commonly mentioned food plant in the Bible, with at least 56 references in the Old and New Testaments.3 The “fruit” technically is a syconium—a hollow fleshy receptacle with a small opening at the apex partly closed by small scales. It can be obovoid, turbinate, or pear shaped; can be 1 to 4 inches long; and can vary in color from yellowish green to coppery, bronze, or dark purple (Figure 2).

Immature fruit of the common fig tree.
FIGURE 2. Immature fruit of the common fig tree.

Ficus carica is a member of the Moraceae family (derived from the Latin name for the mulberry tree), which includes 53 genera and approximately 1400 species, of which about 850 belong to the genus Ficus (the Latin name for a fig tree). The term carica likely comes from the Latin word carricare (to load) to describe a tree loaded with figs. Family members include trees, shrubs, lianas, and herbs that usually contain laticifers with a milky latex.

Traditional Uses

For centuries, components of the fig tree have been used in herbal teas and pastes to treat ailments ranging from sore throats to diarrhea, though there is no evidence to support their efficacy.4 Ancient Indians and Egyptians used plants such as the common fig tree containing furocoumarins to induce hyperpigmentation in vitiligo.5

Phototoxic Components

The leaves and sap of the common fig tree contain psoralens, which are members of the furocoumarin group of chemical compounds and are the source of its phototoxicity. The fruit does not contain psoralens.6-9 The tree also produces proteolytic enzymes such as protease, amylase, ficin, triterpenoids, and lipodiastase that enhance its phototoxic effects.8 Exposure to UV light between 320 and 400 nm following contact with these phototoxic components triggers a reaction in the skin over the course of 1 to 3 days.5 The psoralens bind in epidermal cells, cross-link the DNA, and cause cell-membrane destruction, leading to edema and necrosis.10 The delay in symptoms may be attributed to the time needed to synthesize acute-phase reaction proteins such as tumor necrosis factor α and IL-1.11 In spring and summer months, an increased concentration of psoralens in the leaves and sap contribute to an increased incidence of phytophotodermatitis.9 Humidity and sweat also increase the percutaneous absorption of psoralens.12,13

Allergens

Fig trees produce a latex protein that can cause cross-reactive hypersensitivity reactions in those allergic to F benjamina latex and rubber latex.6 The latex proteins in fig trees can act as airborne respiratory allergens. Ingestion of figs can produce anaphylactic reactions in those sensitized to rubber latex and F benjamina latex.7 Other plant families associated with phototoxic reactions include Rutaceae (lemon, lime, bitter orange), Apiaceae (formerly Umbelliferae)(carrot, parsnip, parsley, dill, celery, hogweed), and Fabaceae (prairie turnip).

 

 

Cutaneous Manifestations

Most cases of fig phytophotodermatitis begin with burning, pain, and/or itching within hours of sunlight exposure in areas of the skin that encountered components of the fig tree, often in a linear pattern. The affected areas become erythematous and edematous with formation of bullae and unilocular vesicles over the course of 1 to 3 days.12,14,15 Lesions may extend beyond the region of contact with the fig tree as they spread across the skin due to sweat or friction, and pain may linger even after the lesions resolve.12,13,16 Adults who handle fig trees (eg, pruning) are susceptible to phototoxic reactions, especially those using chain saws or other mechanisms that result in spray exposure, as the photosensitizing sap permeates the wood and bark of the entire tree.17 Similarly, children who handle fig leaves or sap during outdoor play can develop bullous eruptions. Severe cases have resulted in hospital admission after prolonged exposure.16 Additionally, irritant dermatitis may arise from contact with the trichomes or “hairs” on various parts of the plant.

Leaves and milky sap of the common fig tree.
Image provided with permission by Scott Norton, MD, MPH, MSc (Washington, DC).
FIGURE 3. Leaves and milky sap of the common fig tree.

Patients who use natural remedies containing components of the fig tree without the supervision of a medical provider put themselves at risk for unsafe or unwanted adverse effects, such as phytophotodermatitis.12,15,16,18 An entire family presented with burns after they applied fig leaf extract to the skin prior to tanning outside in the sun.19 A 42-year-old woman acquired a severe burn covering 81% of the body surface after topically applying fig leaf tea to the skin as a tanning agent.20 A subset of patients ingesting or applying fig tree components for conditions such as vitiligo, dermatitis, onychomycosis, and motor retardation developed similar cutaneous reactions.13,14,21,22 Lesions resembling finger marks can raise concerns for potential abuse or neglect in children.22

The differential diagnosis for fig phytophotodermatitis includes sunburn, chemical burns, drug-related photosensitivity, infectious lesions (eg, herpes simplex, bullous impetigo, Lyme disease, superficial lymphangitis), connective tissue disease (eg, systemic lupus erythematosus), contact dermatitis, and nonaccidental trauma.12,15,18 Compared to sunburn, phytophotodermatitis tends to increase in severity over days following exposure and heals with dramatic hyperpigmentation, which also prompts visits to dermatology.12

Treatment

Treatment of fig phytophotodermatitis chiefly is symptomatic, including analgesia, appropriate wound care, and infection prophylaxis. Topical and systemic corticosteroids may aid in the resolution of moderate to severe reactions.15,23,24 Even severe injuries over small areas or mild injuries to a high percentage of the total body surface area may require treatment in a burn unit. Patients should be encouraged to use mineral-based sunscreens on the affected areas to reduce the risk for hyperpigmentation. Individuals who regularly handle fig trees should use contact barriers including gloves and protective clothing (eg, long-sleeved shirts, long pants).

Plant Parts and Nomenclature

Ficus carica (common fig) is a deciduous shrub or small tree with smooth gray bark that can grow up to 10 m in height (Figure 1). It is characterized by many spreading branches, but the trunk rarely grows beyond a diameter of 7 in. Its hairy leaves are coarse on the upper side and soft underneath with 3 to 7 deep lobes that can extend up to 25 cm in length or width; the leaves grow individually, alternating along the sides of the branches. Fig trees often can be seen adorning yards, gardens, and parks, especially in tropical and subtropical climates. Ficus carica should not be confused with Ficus benjamina (weeping fig), a common ornamental tree that also is used to provide shade in hot climates, though both can cause phototoxic skin eruptions.

Ficus carica (common fig).
FIGURE 1. Ficus carica (common fig).

The common fig tree originated in the Mediterranean and western Asia1 and has been cultivated by humans since the second and third millennia bc for its fruit, which commonly is used to sweeten cookies, cakes, and jams.2 Figs are the most commonly mentioned food plant in the Bible, with at least 56 references in the Old and New Testaments.3 The “fruit” technically is a syconium—a hollow fleshy receptacle with a small opening at the apex partly closed by small scales. It can be obovoid, turbinate, or pear shaped; can be 1 to 4 inches long; and can vary in color from yellowish green to coppery, bronze, or dark purple (Figure 2).

Immature fruit of the common fig tree.
FIGURE 2. Immature fruit of the common fig tree.

Ficus carica is a member of the Moraceae family (derived from the Latin name for the mulberry tree), which includes 53 genera and approximately 1400 species, of which about 850 belong to the genus Ficus (the Latin name for a fig tree). The term carica likely comes from the Latin word carricare (to load) to describe a tree loaded with figs. Family members include trees, shrubs, lianas, and herbs that usually contain laticifers with a milky latex.

Traditional Uses

For centuries, components of the fig tree have been used in herbal teas and pastes to treat ailments ranging from sore throats to diarrhea, though there is no evidence to support their efficacy.4 Ancient Indians and Egyptians used plants such as the common fig tree containing furocoumarins to induce hyperpigmentation in vitiligo.5

Phototoxic Components

The leaves and sap of the common fig tree contain psoralens, which are members of the furocoumarin group of chemical compounds and are the source of its phototoxicity. The fruit does not contain psoralens.6-9 The tree also produces proteolytic enzymes such as protease, amylase, ficin, triterpenoids, and lipodiastase that enhance its phototoxic effects.8 Exposure to UV light between 320 and 400 nm following contact with these phototoxic components triggers a reaction in the skin over the course of 1 to 3 days.5 The psoralens bind in epidermal cells, cross-link the DNA, and cause cell-membrane destruction, leading to edema and necrosis.10 The delay in symptoms may be attributed to the time needed to synthesize acute-phase reaction proteins such as tumor necrosis factor α and IL-1.11 In spring and summer months, an increased concentration of psoralens in the leaves and sap contribute to an increased incidence of phytophotodermatitis.9 Humidity and sweat also increase the percutaneous absorption of psoralens.12,13

Allergens

Fig trees produce a latex protein that can cause cross-reactive hypersensitivity reactions in those allergic to F benjamina latex and rubber latex.6 The latex proteins in fig trees can act as airborne respiratory allergens. Ingestion of figs can produce anaphylactic reactions in those sensitized to rubber latex and F benjamina latex.7 Other plant families associated with phototoxic reactions include Rutaceae (lemon, lime, bitter orange), Apiaceae (formerly Umbelliferae)(carrot, parsnip, parsley, dill, celery, hogweed), and Fabaceae (prairie turnip).

 

 

Cutaneous Manifestations

Most cases of fig phytophotodermatitis begin with burning, pain, and/or itching within hours of sunlight exposure in areas of the skin that encountered components of the fig tree, often in a linear pattern. The affected areas become erythematous and edematous with formation of bullae and unilocular vesicles over the course of 1 to 3 days.12,14,15 Lesions may extend beyond the region of contact with the fig tree as they spread across the skin due to sweat or friction, and pain may linger even after the lesions resolve.12,13,16 Adults who handle fig trees (eg, pruning) are susceptible to phototoxic reactions, especially those using chain saws or other mechanisms that result in spray exposure, as the photosensitizing sap permeates the wood and bark of the entire tree.17 Similarly, children who handle fig leaves or sap during outdoor play can develop bullous eruptions. Severe cases have resulted in hospital admission after prolonged exposure.16 Additionally, irritant dermatitis may arise from contact with the trichomes or “hairs” on various parts of the plant.

Leaves and milky sap of the common fig tree.
Image provided with permission by Scott Norton, MD, MPH, MSc (Washington, DC).
FIGURE 3. Leaves and milky sap of the common fig tree.

Patients who use natural remedies containing components of the fig tree without the supervision of a medical provider put themselves at risk for unsafe or unwanted adverse effects, such as phytophotodermatitis.12,15,16,18 An entire family presented with burns after they applied fig leaf extract to the skin prior to tanning outside in the sun.19 A 42-year-old woman acquired a severe burn covering 81% of the body surface after topically applying fig leaf tea to the skin as a tanning agent.20 A subset of patients ingesting or applying fig tree components for conditions such as vitiligo, dermatitis, onychomycosis, and motor retardation developed similar cutaneous reactions.13,14,21,22 Lesions resembling finger marks can raise concerns for potential abuse or neglect in children.22

The differential diagnosis for fig phytophotodermatitis includes sunburn, chemical burns, drug-related photosensitivity, infectious lesions (eg, herpes simplex, bullous impetigo, Lyme disease, superficial lymphangitis), connective tissue disease (eg, systemic lupus erythematosus), contact dermatitis, and nonaccidental trauma.12,15,18 Compared to sunburn, phytophotodermatitis tends to increase in severity over days following exposure and heals with dramatic hyperpigmentation, which also prompts visits to dermatology.12

Treatment

Treatment of fig phytophotodermatitis chiefly is symptomatic, including analgesia, appropriate wound care, and infection prophylaxis. Topical and systemic corticosteroids may aid in the resolution of moderate to severe reactions.15,23,24 Even severe injuries over small areas or mild injuries to a high percentage of the total body surface area may require treatment in a burn unit. Patients should be encouraged to use mineral-based sunscreens on the affected areas to reduce the risk for hyperpigmentation. Individuals who regularly handle fig trees should use contact barriers including gloves and protective clothing (eg, long-sleeved shirts, long pants).

References
  1. Ikegami H, Nogata H, Hirashima K, et al. Analysis of genetic diversity among European and Asian fig varieties (Ficus carica L.) using ISSR, RAPD, and SSR markers. Genetic Resources and Crop Evolution. 2009;56:201-209.
  2. Zohary D, Spiegel-Roy P. Beginnings of fruit growing in the Old World. Science. 1975;187:319-327.
  3. Young R. Young’s Analytical Concordance. Thomas Nelson; 1982.
  4. Duke JA. Handbook of Medicinal Herbs. CRC Press; 2002.
  5. Pathak MA, Fitzpatrick TB. Bioassay of natural and synthetic furocoumarins (psoralens). J Invest Dermatol. 1959;32:509-518.
  6. Focke M, Hemmer W, Wöhrl S, et al. Cross-reactivity between Ficus benjamina latex and fig fruit in patients with clinical fig allergy. Clin Exp Allergy. 2003;33:971-977.
  7. Hemmer W, Focke M, Götz M, et al. Sensitization to Ficus benjamina: relationship to natural rubber latex allergy and identification of foods implicated in the Ficus-fruit syndrome. Clin Exp Allergy. 2004;34:1251-1258.
  8. Bonamonte D, Foti C, Lionetti N, et al. Photoallergic contact dermatitis to 8-methoxypsoralen in Ficus carica. Contact Dermatitis. 2010;62:343-348.
  9. Zaynoun ST, Aftimos BG, Abi Ali L, et al. Ficus carica; isolation and quantification of the photoactive components. Contact Dermatitis. 1984;11:21-25.
  10. Tessman JW, Isaacs ST, Hearst JE. Photochemistry of the furan-side 8-methoxypsoralen-thymidine monoadduct inside the DNA helix. conversion to diadduct and to pyrone-side monoadduct. Biochemistry. 1985;24:1669-1676.
  11. Geary P. Burns related to the use of psoralens as a tanning agent. Burns. 1996;22:636-637.
  12. Redgrave N, Solomon J. Severe phytophotodermatitis from fig sap: a little known phenomenon. BMJ Case Rep. 2021;14:E238745.
  13. Ozdamar E, Ozbek S, Akin S. An unusual cause of burn injury: fig leaf decoction used as a remedy for a dermatitis of unknown etiology. J Burn Care Rehabil. 2003;24:229-233; discussion 228.
  14. Berakha GJ, Lefkovits G. Psoralen phototherapy and phototoxicity. Ann Plast Surg. 1985;14:458-461.
  15. Papazoglou A, Mantadakis E. Fig tree leaves phytophotodermatitis. J Pediatr. 2021;239:244-245.
  16. Imen MS, Ahmadabadi A, Tavousi SH, et al. The curious cases of burn by fig tree leaves. Indian J Dermatol. 2019;64:71-73.
  17. Rouaiguia-Bouakkaz S, Amira-Guebailia H, Rivière C, et al. Identification and quantification of furanocoumarins in stem bark and wood of eight Algerian varieties of Ficus carica by RP-HPLC-DAD and RP-HPLC-DAD-MS. Nat Prod Commun. 2013;8:485-486.
  18. Oliveira AA, Morais J, Pires O, et al. Fig tree induced phytophotodermatitis. BMJ Case Rep. 2020;13:E233392.
  19. Bassioukas K, Stergiopoulou C, Hatzis J. Erythrodermic phytophotodermatitis after application of aqueous fig-leaf extract as an artificial suntan promoter and sunbathing. Contact Dermatitis. 2004;51:94-95.
  20. Sforza M, Andjelkov K, Zaccheddu R. Severe burn on 81% of body surface after sun tanning. Ulus Travma Acil Cerrahi Derg. 2013;19:383-384.
  21. Son JH, Jin H, You HS, et al. Five cases of phytophotodermatitis caused by fig leaves and relevant literature review. Ann Dermatol. 2017;29:86-90.
  22. Abali AE, Aka M, Aydogan C, et al. Burns or phytophotodermatitis, abuse or neglect: confusing aspects of skin lesions caused by the superstitious use of fig leaves. J Burn Care Res. 2012;33:E309-E312.
  23. Picard C, Morice C, Moreau A, et al. Phytophotodermatitis in children: a difficult diagnosis mimicking other dermatitis. 2017;5:1-3.
  24. Enjolras O, Soupre V, Picard A. Uncommon benign infantile vascular tumors. Adv Dermatol. 2008;24:105-124.
References
  1. Ikegami H, Nogata H, Hirashima K, et al. Analysis of genetic diversity among European and Asian fig varieties (Ficus carica L.) using ISSR, RAPD, and SSR markers. Genetic Resources and Crop Evolution. 2009;56:201-209.
  2. Zohary D, Spiegel-Roy P. Beginnings of fruit growing in the Old World. Science. 1975;187:319-327.
  3. Young R. Young’s Analytical Concordance. Thomas Nelson; 1982.
  4. Duke JA. Handbook of Medicinal Herbs. CRC Press; 2002.
  5. Pathak MA, Fitzpatrick TB. Bioassay of natural and synthetic furocoumarins (psoralens). J Invest Dermatol. 1959;32:509-518.
  6. Focke M, Hemmer W, Wöhrl S, et al. Cross-reactivity between Ficus benjamina latex and fig fruit in patients with clinical fig allergy. Clin Exp Allergy. 2003;33:971-977.
  7. Hemmer W, Focke M, Götz M, et al. Sensitization to Ficus benjamina: relationship to natural rubber latex allergy and identification of foods implicated in the Ficus-fruit syndrome. Clin Exp Allergy. 2004;34:1251-1258.
  8. Bonamonte D, Foti C, Lionetti N, et al. Photoallergic contact dermatitis to 8-methoxypsoralen in Ficus carica. Contact Dermatitis. 2010;62:343-348.
  9. Zaynoun ST, Aftimos BG, Abi Ali L, et al. Ficus carica; isolation and quantification of the photoactive components. Contact Dermatitis. 1984;11:21-25.
  10. Tessman JW, Isaacs ST, Hearst JE. Photochemistry of the furan-side 8-methoxypsoralen-thymidine monoadduct inside the DNA helix. conversion to diadduct and to pyrone-side monoadduct. Biochemistry. 1985;24:1669-1676.
  11. Geary P. Burns related to the use of psoralens as a tanning agent. Burns. 1996;22:636-637.
  12. Redgrave N, Solomon J. Severe phytophotodermatitis from fig sap: a little known phenomenon. BMJ Case Rep. 2021;14:E238745.
  13. Ozdamar E, Ozbek S, Akin S. An unusual cause of burn injury: fig leaf decoction used as a remedy for a dermatitis of unknown etiology. J Burn Care Rehabil. 2003;24:229-233; discussion 228.
  14. Berakha GJ, Lefkovits G. Psoralen phototherapy and phototoxicity. Ann Plast Surg. 1985;14:458-461.
  15. Papazoglou A, Mantadakis E. Fig tree leaves phytophotodermatitis. J Pediatr. 2021;239:244-245.
  16. Imen MS, Ahmadabadi A, Tavousi SH, et al. The curious cases of burn by fig tree leaves. Indian J Dermatol. 2019;64:71-73.
  17. Rouaiguia-Bouakkaz S, Amira-Guebailia H, Rivière C, et al. Identification and quantification of furanocoumarins in stem bark and wood of eight Algerian varieties of Ficus carica by RP-HPLC-DAD and RP-HPLC-DAD-MS. Nat Prod Commun. 2013;8:485-486.
  18. Oliveira AA, Morais J, Pires O, et al. Fig tree induced phytophotodermatitis. BMJ Case Rep. 2020;13:E233392.
  19. Bassioukas K, Stergiopoulou C, Hatzis J. Erythrodermic phytophotodermatitis after application of aqueous fig-leaf extract as an artificial suntan promoter and sunbathing. Contact Dermatitis. 2004;51:94-95.
  20. Sforza M, Andjelkov K, Zaccheddu R. Severe burn on 81% of body surface after sun tanning. Ulus Travma Acil Cerrahi Derg. 2013;19:383-384.
  21. Son JH, Jin H, You HS, et al. Five cases of phytophotodermatitis caused by fig leaves and relevant literature review. Ann Dermatol. 2017;29:86-90.
  22. Abali AE, Aka M, Aydogan C, et al. Burns or phytophotodermatitis, abuse or neglect: confusing aspects of skin lesions caused by the superstitious use of fig leaves. J Burn Care Res. 2012;33:E309-E312.
  23. Picard C, Morice C, Moreau A, et al. Phytophotodermatitis in children: a difficult diagnosis mimicking other dermatitis. 2017;5:1-3.
  24. Enjolras O, Soupre V, Picard A. Uncommon benign infantile vascular tumors. Adv Dermatol. 2008;24:105-124.
Issue
Cutis - 113(4)
Issue
Cutis - 113(4)
Page Number
167-169
Page Number
167-169
Publications
Publications
Topics
Article Type
Display Headline
Botanical Briefs: Fig Phytophotodermatitis (Ficus carica)
Display Headline
Botanical Briefs: Fig Phytophotodermatitis (Ficus carica)
Sections
Inside the Article

Practice Points

  • Exposure to the components of the common fig tree (Ficus carica) can induce phytophotodermatitis.
  • Notable postinflammatory hyperpigmentation typically occurs in the healing stage of fig phytophotodermatitis.
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

Asymptomatic Erythematous Plaque in an Outdoorsman

Article Type
Changed
Wed, 04/24/2024 - 10:16
Display Headline
Asymptomatic Erythematous Plaque in an Outdoorsman

The Diagnosis: Erythema Migrans

The patient was clinically diagnosed with erythema migrans. He did not recall a tick bite but spent a lot of time outdoors. He was treated with 10 days of doxycycline 100 mg twice daily with complete resolution of the rash.

Lyme disease is a spirochete infection caused by the Borrelia burgdorferi sensu lato species complex and transmitted by the Ixodidae tick family. It is the most common tick-borne disease in the United States and mostly is reported in the northeastern and upper midwestern states during the warmer seasons, but it is prevalent worldwide. In geographic areas where Lyme disease is common, the incidence is approximately 40 cases per 100,000 individuals.1 Our patient resided in coastal South Carolina. Lyme disease is more commonly reported in White individuals. The skin lesions may be more difficult to discern and diagnose in patients with darker skin types, leading to delayed diagnosis and treatment.2,3

Patients may be diagnosed with early localized, early disseminated, or late Lyme disease. Erythema migrans is the early localized form of the disease and is classically described as an erythematous targetlike plaque with raised borders arising at the site of the tick bite 1 to 2 weeks later.4 However, many patients simply have a homogeneous erythematous plaque with raised advancing borders ranging in size from 5 to 68 cm.5 In a 2022 study of 69 patients with suspected Lyme disease, only 35 (50.7%) were determined to truly have acute Lyme disease.6 Of them, only 2 (5.7%) had the classic ringwithin- a-ring pattern. Most plaques were uniform, pink, oval-shaped lesions with well-demarcated borders.6

The rash may present with a burning sensation, or patients may experience no symptoms at all, which can lead to delayed diagnosis and progression to late disease. Patients may develop malaise, fever, headache, body aches, or joint pain. Early disseminated disease manifests similarly. Patients with disseminated disease also may develop more serious complications, including lymphadenopathy; cranial nerve palsies; ocular involvement; meningitis; or cardiac abnormalities such as myocarditis, pericarditis, or arrhythmia. Late disease most often causes arthritis of the large joints, though it also can have cardiac or neurologic manifestations. Some patients with chronic disease—the majority of whom were diagnosed in Europe—may develop acrodermatitis chronica atrophicans with edematous blue-red plaques that become atrophic and hyperpigmented fibrotic plaques over the course of years.

Allergic contact dermatitis to a plant more likely would cause itchy or painful, oozy, weepy, vesicular lesions arranged in a linear pattern. A dermatophyte infection likely would cause a scaly eruption. Although our patient presented with a sharply demarcated, raised, erythematous lesion, the distribution did not follow normal clothing lines and would be unusual for a photosensitive drug eruption. Cellulitis likely would be associated with tenderness or warmth to the touch. Finally, southern tick-associated rash illness, which is associated with Amblyomma americanum (lone star tick) bites, may appear with a similar rash but few systemic symptoms. It also can be treated with tetracycline antibiotics.7

Our case in South Carolina demonstrates the importance of keeping Lyme disease in the differential. Clinicians should remember to ask patients about their travel history. In endemic areas, patients with erythema migrans can be started on treatment without waiting for serology. Patients with early Lyme disease may or may not have positive serologies at the time of presentation.6 Guidelines for the treatment of Lyme disease have been revised in recent years to decrease patient antibiotic exposure by reducing the number of days of antibiotic therapy.8 A recent randomized controlled trial found no significant difference in recurrence for patients treated with 7 days of doxycycline compared with 14 days.9 We typically prescribe a 10-day course of doxycycline, which also is adequate for concurrent rickettsial disease. Patients who develop malarialike symptoms should be evaluated for babesiosis, which is treated with clindamycin.

References
  1. Skar GL, Simonsen KA. Lyme disease. StatPearls [Internet]. Updated February 4, 2024. Accessed March 20, 2024. https://www.ncbi.nlm.nih.gov/books/NBK431066/
  2. Dennison R, Novak C, Rebman A, et al. Lyme disease with erythema migrans and seventh nerve palsy in an African-American man. Cureus. 2019;11:E6509.
  3. Bax CE, Clark AK, Oboite M, et al. A case of disseminated Lyme disease in a child with skin of color. Pediatr Dermatol. 2021;38 (suppl 2):140-141.
  4. Shah AS, Varatharaj Palraj BR. Multiple erythema migrans rashes characteristic of early disseminated lyme disease, before and after therapy. Mayo Clin Proc. 2019;94:172-173.
  5. Feder HM Jr, Abeles M, Bernstein M, et al. Diagnosis, treatment, and prognosis of erythema migrans and Lyme arthritis. Clin Dermatol. 2006;24:509-520.
  6. Schotthoefer AM, Green CB, Dempsey G, et al. The spectrum of erythema migrans in early Lyme disease: can we improve its recognition? Cureus. 2022;14:E30673.
  7. Strle F, Wormser GP. Early Lyme disease (erythema migrans) and its mimics (southern tick-associated rash illness and tick-associated rash illness). Infect Dis Clin North Am. 2022;36:523-539.
  8. Torbahn G, Hofmann H, Rücker G, et al. Efficacy and safety of antibiotic therapy in early cutaneous Lyme borreliosis: a network meta-analysis. JAMA Dermatol. 2018;154:1292-1303.
  9. Stupica D, Collinet-Adler S, Blagus R, et al. Treatment of erythema migrans with doxycycline for 7 days versus 14 days in Slovenia: a randomised open-label non-inferiority trial. Lancet Infect Dis. 2023;23:371-379.
Article PDF
Author and Disclosure Information

From the Medical University of South Carolina, Charleston. Dr. Barker is from the Department of Internal Medicine, and Dr. Elston is from the Department of Dermatology and Dermatologic Surgery.

The authors report no conflict of interest.

Correspondence: Catherine Shirer Barker, MD, 96 Jonathan Lucas St, Ste 807B, MSC 623, Charleston, SC 29425 (catherinesbarker@gmail.com).

Issue
Cutis - 113(4)
Publications
Topics
Page Number
183-184
Sections
Author and Disclosure Information

From the Medical University of South Carolina, Charleston. Dr. Barker is from the Department of Internal Medicine, and Dr. Elston is from the Department of Dermatology and Dermatologic Surgery.

The authors report no conflict of interest.

Correspondence: Catherine Shirer Barker, MD, 96 Jonathan Lucas St, Ste 807B, MSC 623, Charleston, SC 29425 (catherinesbarker@gmail.com).

Author and Disclosure Information

From the Medical University of South Carolina, Charleston. Dr. Barker is from the Department of Internal Medicine, and Dr. Elston is from the Department of Dermatology and Dermatologic Surgery.

The authors report no conflict of interest.

Correspondence: Catherine Shirer Barker, MD, 96 Jonathan Lucas St, Ste 807B, MSC 623, Charleston, SC 29425 (catherinesbarker@gmail.com).

Article PDF
Article PDF
Related Articles

The Diagnosis: Erythema Migrans

The patient was clinically diagnosed with erythema migrans. He did not recall a tick bite but spent a lot of time outdoors. He was treated with 10 days of doxycycline 100 mg twice daily with complete resolution of the rash.

Lyme disease is a spirochete infection caused by the Borrelia burgdorferi sensu lato species complex and transmitted by the Ixodidae tick family. It is the most common tick-borne disease in the United States and mostly is reported in the northeastern and upper midwestern states during the warmer seasons, but it is prevalent worldwide. In geographic areas where Lyme disease is common, the incidence is approximately 40 cases per 100,000 individuals.1 Our patient resided in coastal South Carolina. Lyme disease is more commonly reported in White individuals. The skin lesions may be more difficult to discern and diagnose in patients with darker skin types, leading to delayed diagnosis and treatment.2,3

Patients may be diagnosed with early localized, early disseminated, or late Lyme disease. Erythema migrans is the early localized form of the disease and is classically described as an erythematous targetlike plaque with raised borders arising at the site of the tick bite 1 to 2 weeks later.4 However, many patients simply have a homogeneous erythematous plaque with raised advancing borders ranging in size from 5 to 68 cm.5 In a 2022 study of 69 patients with suspected Lyme disease, only 35 (50.7%) were determined to truly have acute Lyme disease.6 Of them, only 2 (5.7%) had the classic ringwithin- a-ring pattern. Most plaques were uniform, pink, oval-shaped lesions with well-demarcated borders.6

The rash may present with a burning sensation, or patients may experience no symptoms at all, which can lead to delayed diagnosis and progression to late disease. Patients may develop malaise, fever, headache, body aches, or joint pain. Early disseminated disease manifests similarly. Patients with disseminated disease also may develop more serious complications, including lymphadenopathy; cranial nerve palsies; ocular involvement; meningitis; or cardiac abnormalities such as myocarditis, pericarditis, or arrhythmia. Late disease most often causes arthritis of the large joints, though it also can have cardiac or neurologic manifestations. Some patients with chronic disease—the majority of whom were diagnosed in Europe—may develop acrodermatitis chronica atrophicans with edematous blue-red plaques that become atrophic and hyperpigmented fibrotic plaques over the course of years.

Allergic contact dermatitis to a plant more likely would cause itchy or painful, oozy, weepy, vesicular lesions arranged in a linear pattern. A dermatophyte infection likely would cause a scaly eruption. Although our patient presented with a sharply demarcated, raised, erythematous lesion, the distribution did not follow normal clothing lines and would be unusual for a photosensitive drug eruption. Cellulitis likely would be associated with tenderness or warmth to the touch. Finally, southern tick-associated rash illness, which is associated with Amblyomma americanum (lone star tick) bites, may appear with a similar rash but few systemic symptoms. It also can be treated with tetracycline antibiotics.7

Our case in South Carolina demonstrates the importance of keeping Lyme disease in the differential. Clinicians should remember to ask patients about their travel history. In endemic areas, patients with erythema migrans can be started on treatment without waiting for serology. Patients with early Lyme disease may or may not have positive serologies at the time of presentation.6 Guidelines for the treatment of Lyme disease have been revised in recent years to decrease patient antibiotic exposure by reducing the number of days of antibiotic therapy.8 A recent randomized controlled trial found no significant difference in recurrence for patients treated with 7 days of doxycycline compared with 14 days.9 We typically prescribe a 10-day course of doxycycline, which also is adequate for concurrent rickettsial disease. Patients who develop malarialike symptoms should be evaluated for babesiosis, which is treated with clindamycin.

The Diagnosis: Erythema Migrans

The patient was clinically diagnosed with erythema migrans. He did not recall a tick bite but spent a lot of time outdoors. He was treated with 10 days of doxycycline 100 mg twice daily with complete resolution of the rash.

Lyme disease is a spirochete infection caused by the Borrelia burgdorferi sensu lato species complex and transmitted by the Ixodidae tick family. It is the most common tick-borne disease in the United States and mostly is reported in the northeastern and upper midwestern states during the warmer seasons, but it is prevalent worldwide. In geographic areas where Lyme disease is common, the incidence is approximately 40 cases per 100,000 individuals.1 Our patient resided in coastal South Carolina. Lyme disease is more commonly reported in White individuals. The skin lesions may be more difficult to discern and diagnose in patients with darker skin types, leading to delayed diagnosis and treatment.2,3

Patients may be diagnosed with early localized, early disseminated, or late Lyme disease. Erythema migrans is the early localized form of the disease and is classically described as an erythematous targetlike plaque with raised borders arising at the site of the tick bite 1 to 2 weeks later.4 However, many patients simply have a homogeneous erythematous plaque with raised advancing borders ranging in size from 5 to 68 cm.5 In a 2022 study of 69 patients with suspected Lyme disease, only 35 (50.7%) were determined to truly have acute Lyme disease.6 Of them, only 2 (5.7%) had the classic ringwithin- a-ring pattern. Most plaques were uniform, pink, oval-shaped lesions with well-demarcated borders.6

The rash may present with a burning sensation, or patients may experience no symptoms at all, which can lead to delayed diagnosis and progression to late disease. Patients may develop malaise, fever, headache, body aches, or joint pain. Early disseminated disease manifests similarly. Patients with disseminated disease also may develop more serious complications, including lymphadenopathy; cranial nerve palsies; ocular involvement; meningitis; or cardiac abnormalities such as myocarditis, pericarditis, or arrhythmia. Late disease most often causes arthritis of the large joints, though it also can have cardiac or neurologic manifestations. Some patients with chronic disease—the majority of whom were diagnosed in Europe—may develop acrodermatitis chronica atrophicans with edematous blue-red plaques that become atrophic and hyperpigmented fibrotic plaques over the course of years.

Allergic contact dermatitis to a plant more likely would cause itchy or painful, oozy, weepy, vesicular lesions arranged in a linear pattern. A dermatophyte infection likely would cause a scaly eruption. Although our patient presented with a sharply demarcated, raised, erythematous lesion, the distribution did not follow normal clothing lines and would be unusual for a photosensitive drug eruption. Cellulitis likely would be associated with tenderness or warmth to the touch. Finally, southern tick-associated rash illness, which is associated with Amblyomma americanum (lone star tick) bites, may appear with a similar rash but few systemic symptoms. It also can be treated with tetracycline antibiotics.7

Our case in South Carolina demonstrates the importance of keeping Lyme disease in the differential. Clinicians should remember to ask patients about their travel history. In endemic areas, patients with erythema migrans can be started on treatment without waiting for serology. Patients with early Lyme disease may or may not have positive serologies at the time of presentation.6 Guidelines for the treatment of Lyme disease have been revised in recent years to decrease patient antibiotic exposure by reducing the number of days of antibiotic therapy.8 A recent randomized controlled trial found no significant difference in recurrence for patients treated with 7 days of doxycycline compared with 14 days.9 We typically prescribe a 10-day course of doxycycline, which also is adequate for concurrent rickettsial disease. Patients who develop malarialike symptoms should be evaluated for babesiosis, which is treated with clindamycin.

References
  1. Skar GL, Simonsen KA. Lyme disease. StatPearls [Internet]. Updated February 4, 2024. Accessed March 20, 2024. https://www.ncbi.nlm.nih.gov/books/NBK431066/
  2. Dennison R, Novak C, Rebman A, et al. Lyme disease with erythema migrans and seventh nerve palsy in an African-American man. Cureus. 2019;11:E6509.
  3. Bax CE, Clark AK, Oboite M, et al. A case of disseminated Lyme disease in a child with skin of color. Pediatr Dermatol. 2021;38 (suppl 2):140-141.
  4. Shah AS, Varatharaj Palraj BR. Multiple erythema migrans rashes characteristic of early disseminated lyme disease, before and after therapy. Mayo Clin Proc. 2019;94:172-173.
  5. Feder HM Jr, Abeles M, Bernstein M, et al. Diagnosis, treatment, and prognosis of erythema migrans and Lyme arthritis. Clin Dermatol. 2006;24:509-520.
  6. Schotthoefer AM, Green CB, Dempsey G, et al. The spectrum of erythema migrans in early Lyme disease: can we improve its recognition? Cureus. 2022;14:E30673.
  7. Strle F, Wormser GP. Early Lyme disease (erythema migrans) and its mimics (southern tick-associated rash illness and tick-associated rash illness). Infect Dis Clin North Am. 2022;36:523-539.
  8. Torbahn G, Hofmann H, Rücker G, et al. Efficacy and safety of antibiotic therapy in early cutaneous Lyme borreliosis: a network meta-analysis. JAMA Dermatol. 2018;154:1292-1303.
  9. Stupica D, Collinet-Adler S, Blagus R, et al. Treatment of erythema migrans with doxycycline for 7 days versus 14 days in Slovenia: a randomised open-label non-inferiority trial. Lancet Infect Dis. 2023;23:371-379.
References
  1. Skar GL, Simonsen KA. Lyme disease. StatPearls [Internet]. Updated February 4, 2024. Accessed March 20, 2024. https://www.ncbi.nlm.nih.gov/books/NBK431066/
  2. Dennison R, Novak C, Rebman A, et al. Lyme disease with erythema migrans and seventh nerve palsy in an African-American man. Cureus. 2019;11:E6509.
  3. Bax CE, Clark AK, Oboite M, et al. A case of disseminated Lyme disease in a child with skin of color. Pediatr Dermatol. 2021;38 (suppl 2):140-141.
  4. Shah AS, Varatharaj Palraj BR. Multiple erythema migrans rashes characteristic of early disseminated lyme disease, before and after therapy. Mayo Clin Proc. 2019;94:172-173.
  5. Feder HM Jr, Abeles M, Bernstein M, et al. Diagnosis, treatment, and prognosis of erythema migrans and Lyme arthritis. Clin Dermatol. 2006;24:509-520.
  6. Schotthoefer AM, Green CB, Dempsey G, et al. The spectrum of erythema migrans in early Lyme disease: can we improve its recognition? Cureus. 2022;14:E30673.
  7. Strle F, Wormser GP. Early Lyme disease (erythema migrans) and its mimics (southern tick-associated rash illness and tick-associated rash illness). Infect Dis Clin North Am. 2022;36:523-539.
  8. Torbahn G, Hofmann H, Rücker G, et al. Efficacy and safety of antibiotic therapy in early cutaneous Lyme borreliosis: a network meta-analysis. JAMA Dermatol. 2018;154:1292-1303.
  9. Stupica D, Collinet-Adler S, Blagus R, et al. Treatment of erythema migrans with doxycycline for 7 days versus 14 days in Slovenia: a randomised open-label non-inferiority trial. Lancet Infect Dis. 2023;23:371-379.
Issue
Cutis - 113(4)
Issue
Cutis - 113(4)
Page Number
183-184
Page Number
183-184
Publications
Publications
Topics
Article Type
Display Headline
Asymptomatic Erythematous Plaque in an Outdoorsman
Display Headline
Asymptomatic Erythematous Plaque in an Outdoorsman
Sections
Questionnaire Body

A middle-aged man presented with a well-demarcated, hyperpigmented, erythematous patch with an annular erythematous border that extended from the mid-back to the lower back. The patient was otherwise asymptomatic. He was an avid gardener who resided in South Carolina and had recently adopted 2 puppies.

Asymptomatic erythematous plaque in an outdoorsman

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Tue, 04/02/2024 - 10:45
Un-Gate On Date
Tue, 04/02/2024 - 10:45
Use ProPublica
CFC Schedule Remove Status
Tue, 04/02/2024 - 10:45
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

Aquatic Antagonists: Scorpionfish Envenomation

Article Type
Changed
Wed, 03/06/2024 - 11:47
Display Headline
Aquatic Antagonists: Scorpionfish Envenomation

With the growing popularity of water sports and a proliferation of invasive species, human injuries from marine animal envenomation continue to rise.1-3 Members of the scorpionfish family Scorpaenidae are second only to stingrays as the leading cause of the 40,000 to 50,000 injuries annually from marine life worldwide.4 Because scorpionfish represent a growing threat and competition with native species, it has been suggested that they could replace endangered species on restaurant menus.5-8 Scorpionfish have been introduced by humans from tropical to temperate seas and are now common off the coast of California and the eastern coast from New York to Florida, as well as in the Caribbean, the Bahamas, and off the southern coast of Brazil. Victims of scorpionfish stings experience considerable pain and may require days to weeks to fully recover, highlighting the socioeconomic costs and burden of scorpionfish envenomation.9,10 Fishers, divers, swimmers, and aquarium owners are most often affected.

Family

The common term scorpionfish refers to both the family Scorpaenidae and the genus Scorpaena. Members of this family possess similar dorsal, anal, and pelvic fins, though they vary between genera in their size and the potency of the venom they insulate. Other familiar members include the genus Pterois (lionfish) and Synanceja (stonefish). Synanceja are the most venomous within the group, but scorpionfish stings more commonly arise from Pterois and Scorpaena.8 Because of the rare shapes and vibrant colors of scorpionfish, some traders and aquarium owners will seek and pay high prices for these fish, providing further opportunity for envenomation.11,12

Characteristics

Scorpionfish have with a high variation in color, ranging from lighter grays to intense reds depending on their geographic location and habitat. Synanceja are bland in coloration, blending in with rocks and gravel, but the more dramatic-appearing Scorpaena exhibit a large cranium and wide range of multicolored patterns (Figure 1).13Pterois serve as the most conspicuous member of the group with brightly colored red and white stripes (Figure 2). Scorpionfish commonly grow up to 19 inches long and boast 12 dorsal, 2 pelvic, and 3 anal spines housing 5 to 10 mg of venom.14 An integumentary sheath encapsulates each spine housing the glandular tissue that produces the potent venom.

Red scorpionfish (Scorpaena scrofa).
Photograph courtesy of Sylvain Le Bris (https://www.inaturalist.org/). Republished under the Creative Commons Attribution (CC BY-NC 4.0).
FIGURE 1. Red scorpionfish (Scorpaena scrofa).

Toxin Properties

Unlike Pterois and Synanceja, Scorpaena do not have venom ducts around their glands, complicating the work of marine biologists aiming to extract and study the venomous toxins. Several studies have managed to isolate scorpionfish venom and overcome its unstable heat-labile nature to investigate its biologic properties.15-20 Several high-molecular-weight proteins (50–800 kDa) comprise the venom, including hyaluronidase, integrin-inhibiting factors, capillary permeability factor, proteases, and some less-understood cytolytic toxins. These factors provoke the inflammatory, proteolytic, hemorrhagic, cardiovascular, and hemolytic biologic activities at both the local and systemic levels, directing damage to wounded tissues and inducing vascular and tissue permeability to reach cellular processes far and wide. Mediators of inflammation include tumor necrosis factor, IL-6, and monocyte chemoattractant protein 1, followed by neutrophils and other mononuclear cells, initiating the immune response at the wound site. Toxin potency remains for up to 2 days after fish death.1

Lionfish (Pterois volitans).
Photograph courtesy of 808_Diver (https://www.inaturalist.org/). Republished under the Creative Commons Attribution (CC BY-NC 4.0).
FIGURE 2. Lionfish (Pterois volitans).

Clinical Manifestation

Physicians may be guided by clinical symptoms in identifying scorpionfish stings, as the patient may not know the identity of their marine assailant. Initially, individuals punctured by scorpionfish spikes will experience an acute pain and burning sensation at the puncture site that may be accompanied by systemic symptoms such as nausea, vomiting, diarrhea, tachycardia, hypotension, loss of consciousness, difficulty breathing, and delirium.9,21-23 The pain will intensify and radiate distal to the site of envenomation, and the wound may exhibit vesiculation, erythema, bruising, pallor, and notable edema.4,24 Pain intensity peaks at 30 to 90 minutes after envenomation, and other systemic symptoms generally last for 24 to 48 hours.25 If patients do not seek prompt treatment, secondary infection may ensue, and the lingering venom in the blister may cause dermal necrosis, paresthesia, and anesthesia. Chronic sequelae may include joint contractures, compartment syndrome, necrotic ulcers, and chronic neuropathy.1

Management

Treatment of scorpionfish stings primarily is palliative and aimed at symptom reduction. Patients should immediately treat wounds with hot but not scalding water immersion.26,27 Given the thermolabile components of scorpionfish venom, the most effective treatment is to soak the affected limb in water 42 °C to 45 °C for 30 to 90 minutes. Any higher temperature may pose risk for scalding burns. Children should be monitored throughout treatment.28 If hot water immersion does not provide relief, oral analgesics may be considered. Stonefish antivenom is available and may be used for any scorpionfish sting given the shared biologic properties between genera. Providers evaluating stings could use sterile irrigation to clean wounds and search for foreign bodies including spine fragments; probing should be accomplished by instruments rather than a gloved finger. Providers should consider culturing wounds and prescribing antibiotics for suspected secondary infections. A tetanus toxoid history also should be elicited, and patients may have a booster administered, as indicated.29

References
  1. Rensch G, Murphy-Lavoie HM. Lionfish, scorpionfish, and stonefish toxicity. StatPearls. StatPearls Publishing; May 10, 2022.
  2. Cearnal L. Red lionfish and ciguatoxin: menace spreading through western hemisphere. Ann Emerg Med. 2012;60:21A-22A. doi:10.1016/j.annemergmed.2012.05.022
  3. Côté IM, Green SJ. Potential effects of climate change on a marine invasion: the importance of current context. Curr Zool. 2012;58:1-8. doi:10.1093/czoolo/58.1.1
  4. Venomology of scorpionfishes. In: Santhanam R. Biology and Ecology of Venomous Marine Scorpionfishes. Academic Press; 2019:263-278.
  5. Ferri J, Staglicˇic´ N, Matić-Skoko S. The black scorpionfish, Scorpaena porcus (Scorpaenidae): could it serve as reliable indicator of Mediterranean coastal communities’ health? Ecol Indicators. 2012;18:25-30. doi:10.1016/j.ecolind.2011.11.004
  6. Santhanam R. Biology and Ecology of Venomous Marine Scorpionfishes. Academic Press; 2019.
  7. Morris JA, Akins JL. Feeding ecology of invasive lionfish (Pterois volitans) in the Bahamian Archipelago. Environ Biol Fishes. 2009;86:389-398. doi:10.1007/s10641-009-9538-8 
  8. Albins MA, Hixon MA. Worst case scenario: potential long-term effects of invasive predatory lionfish (Pterois volitans) on Atlantic and Caribbean coral-reef communities. Environ Biol Fishes. 2013;96:1151–1157. doi:10.1007/s10641-011-9795-1
  9. Haddad V Jr, Martins IA, Makyama HM. Injuries caused by scorpionfishes (Scorpaena plumieri Bloch, 1789 and Scorpaena brasiliensis Cuvier, 1829) in the Southwestern Atlantic Ocean (Brazilian coast): epidemiologic, clinic and therapeutic aspects of 23 stings in humans. Toxicon. 2003;42:79-83. doi:10.1016/s0041-0101(03)00103-x
  10. Campos FV, Menezes TN, Malacarne PF, et al. A review on the Scorpaena plumieri fish venom and its bioactive compounds. J Venom Anim Toxins Incl Trop Dis. 2016;22:35. doi:10.1186/s40409-016-0090-7
  11. Needleman RK, Neylan IP, Erickson TB. Environmental and ecological effects of climate change on venomous marine and amphibious species in the wilderness. Wilderness Environ Med. 2018;29:343-356. doi:10.1016/j.wem.2018.04.003
  12. Aldred B, Erickson T, Lipscomb J. Lionfish envenomations in an urban wilderness. Wilderness Environ Med. 1996;7:291-296. doi:10.1580/1080-6032(1996)007[0291:leiauw]2.3.co;2
  13. Stewart J, Hughes JM. Life-history traits of the southern hemisphere eastern red scorpionfish, Scorpaena cardinalis (Scorpaenidae: Scorpaeninae). Mar Freshw Res. 2010;61:1290-1297. doi:10.1071/MF10040
  14. Auerbach PS. Marine envenomations. N Engl J Med. 1991;325:486-493. doi:10.1056/NEJM199108153250707
  15. Andrich F, Carnielli JB, Cassoli JS, et al. A potent vasoactive cytolysin isolated from Scorpaena plumieri scorpionfish venom. Toxicon. 2010;56:487-496. doi:10.1016/j.toxicon.2010.05.003
  16. Gomes HL, Andrich F, Mauad H, et al. Cardiovascular effects of scorpionfish (Scorpaena plumieri) venom. Toxicon. 2010;55(2-3):580-589. doi:10.1016/j.toxicon.2009.10.012
  17. Menezes TN, Carnielli JB, Gomes HL, et al. Local inflammatory response induced by scorpionfish Scorpaena plumieri venom in mice. Toxicon. 2012;60:4-11. doi:10.1016/j.toxicon.2012.03.008
  18. Schaeffer RC Jr, Carlson RW, Russell FE. Some chemical properties of the venom of the scorpionfish Scorpaena guttata. Toxicon. 1971;9:69-78. doi:10.1016/0041-0101(71)90045-6
  19. Khalil AM, Wahsha MA, Abu Khadra KM, et al. Biochemical and histopathological effects of the stonefish (Synanceia verrucosa) venom in rats. Toxicon. 2018;142:45-51. doi:10.1016/j.toxicon.2017.12.052
  20. Mouchbahani-Constance S, Lesperance LS, Petitjean H, et al. Lionfish venom elicits pain predominantly through the activation of nonpeptidergic nociceptors. Pain. 2018;159:2255-2266. doi:10.1097/j.pain.0000000000001326
  21. Ottuso P. Aquatic dermatology: encounters with the denizens of the deep (and not so deep)—a review. part II: the vertebrates, single-celled organisms, and aquatic biotoxins. Int J Dermatol. 2013;52:268-278. doi:10.1111/j.1365-4632.2011.05426.x
  22. Bayley HH. Injuries caused by scorpion fish. Trans R Soc Trop Med Hyg. 1940;34:227-230. doi:10.1016/s0035-9203(40)90072-4
  23. González D. Epidemiological and clinical aspects of certain venomous animals of Spain. Toxicon. 1982;20:925-928. doi:10.1016/0041-0101(82)90080-0
  24. Halstead BW. Injurious effects from the sting of the scorpionfish, Scorpaena guttata. with report of a case. Calif Med. 1951;74:395-396.
  25. Vasievich MP, Villarreal JD, Tomecki KJ. Got the travel bug? a review of common infections, infestations, bites, and stings among returning travelers. Am J Clin Dermatol. 2016;17:451-462. doi:10.1007/s40257-016-0203-7
  26. Barnett S, Saggiomo S, Smout M, et al. Heat deactivation of the stonefish Synanceia horrida venom—implications for first-aid management. Diving Hyperb Med. 2017;47:155-158. doi:10.28920/dhm47.3.155-158
  27. Russell FE. Weever fish sting: the last word. Br Med J (Clin Res Ed). 1983;287:981-982. doi:10.1136/bmj.287.6397.981-c
  28. Tomlinson H, Elston DM. Aquatic antagonists: lionfish (Pterois volitans). Cutis. 2018;102:232-234.
  29. Hornbeak KB, Auerbach PS. Marine envenomation. Emerg Med Clin North Am. 2017;35:321-337. doi:10.1016/j.emc.2016.12.004
Article PDF
Author and Disclosure Information

Shawn Afvari is from the New York Medical College School of Medicine, Valhalla. Dr. Elston is from the Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston.

The authors report no conflict of interest.

Correspondence: Shawn Afvari, BS (safvari@student.nymc.edu).

Issue
Cutis - 113(3)
Publications
Topics
Page Number
133-134,136
Sections
Author and Disclosure Information

Shawn Afvari is from the New York Medical College School of Medicine, Valhalla. Dr. Elston is from the Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston.

The authors report no conflict of interest.

Correspondence: Shawn Afvari, BS (safvari@student.nymc.edu).

Author and Disclosure Information

Shawn Afvari is from the New York Medical College School of Medicine, Valhalla. Dr. Elston is from the Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston.

The authors report no conflict of interest.

Correspondence: Shawn Afvari, BS (safvari@student.nymc.edu).

Article PDF
Article PDF

With the growing popularity of water sports and a proliferation of invasive species, human injuries from marine animal envenomation continue to rise.1-3 Members of the scorpionfish family Scorpaenidae are second only to stingrays as the leading cause of the 40,000 to 50,000 injuries annually from marine life worldwide.4 Because scorpionfish represent a growing threat and competition with native species, it has been suggested that they could replace endangered species on restaurant menus.5-8 Scorpionfish have been introduced by humans from tropical to temperate seas and are now common off the coast of California and the eastern coast from New York to Florida, as well as in the Caribbean, the Bahamas, and off the southern coast of Brazil. Victims of scorpionfish stings experience considerable pain and may require days to weeks to fully recover, highlighting the socioeconomic costs and burden of scorpionfish envenomation.9,10 Fishers, divers, swimmers, and aquarium owners are most often affected.

Family

The common term scorpionfish refers to both the family Scorpaenidae and the genus Scorpaena. Members of this family possess similar dorsal, anal, and pelvic fins, though they vary between genera in their size and the potency of the venom they insulate. Other familiar members include the genus Pterois (lionfish) and Synanceja (stonefish). Synanceja are the most venomous within the group, but scorpionfish stings more commonly arise from Pterois and Scorpaena.8 Because of the rare shapes and vibrant colors of scorpionfish, some traders and aquarium owners will seek and pay high prices for these fish, providing further opportunity for envenomation.11,12

Characteristics

Scorpionfish have with a high variation in color, ranging from lighter grays to intense reds depending on their geographic location and habitat. Synanceja are bland in coloration, blending in with rocks and gravel, but the more dramatic-appearing Scorpaena exhibit a large cranium and wide range of multicolored patterns (Figure 1).13Pterois serve as the most conspicuous member of the group with brightly colored red and white stripes (Figure 2). Scorpionfish commonly grow up to 19 inches long and boast 12 dorsal, 2 pelvic, and 3 anal spines housing 5 to 10 mg of venom.14 An integumentary sheath encapsulates each spine housing the glandular tissue that produces the potent venom.

Red scorpionfish (Scorpaena scrofa).
Photograph courtesy of Sylvain Le Bris (https://www.inaturalist.org/). Republished under the Creative Commons Attribution (CC BY-NC 4.0).
FIGURE 1. Red scorpionfish (Scorpaena scrofa).

Toxin Properties

Unlike Pterois and Synanceja, Scorpaena do not have venom ducts around their glands, complicating the work of marine biologists aiming to extract and study the venomous toxins. Several studies have managed to isolate scorpionfish venom and overcome its unstable heat-labile nature to investigate its biologic properties.15-20 Several high-molecular-weight proteins (50–800 kDa) comprise the venom, including hyaluronidase, integrin-inhibiting factors, capillary permeability factor, proteases, and some less-understood cytolytic toxins. These factors provoke the inflammatory, proteolytic, hemorrhagic, cardiovascular, and hemolytic biologic activities at both the local and systemic levels, directing damage to wounded tissues and inducing vascular and tissue permeability to reach cellular processes far and wide. Mediators of inflammation include tumor necrosis factor, IL-6, and monocyte chemoattractant protein 1, followed by neutrophils and other mononuclear cells, initiating the immune response at the wound site. Toxin potency remains for up to 2 days after fish death.1

Lionfish (Pterois volitans).
Photograph courtesy of 808_Diver (https://www.inaturalist.org/). Republished under the Creative Commons Attribution (CC BY-NC 4.0).
FIGURE 2. Lionfish (Pterois volitans).

Clinical Manifestation

Physicians may be guided by clinical symptoms in identifying scorpionfish stings, as the patient may not know the identity of their marine assailant. Initially, individuals punctured by scorpionfish spikes will experience an acute pain and burning sensation at the puncture site that may be accompanied by systemic symptoms such as nausea, vomiting, diarrhea, tachycardia, hypotension, loss of consciousness, difficulty breathing, and delirium.9,21-23 The pain will intensify and radiate distal to the site of envenomation, and the wound may exhibit vesiculation, erythema, bruising, pallor, and notable edema.4,24 Pain intensity peaks at 30 to 90 minutes after envenomation, and other systemic symptoms generally last for 24 to 48 hours.25 If patients do not seek prompt treatment, secondary infection may ensue, and the lingering venom in the blister may cause dermal necrosis, paresthesia, and anesthesia. Chronic sequelae may include joint contractures, compartment syndrome, necrotic ulcers, and chronic neuropathy.1

Management

Treatment of scorpionfish stings primarily is palliative and aimed at symptom reduction. Patients should immediately treat wounds with hot but not scalding water immersion.26,27 Given the thermolabile components of scorpionfish venom, the most effective treatment is to soak the affected limb in water 42 °C to 45 °C for 30 to 90 minutes. Any higher temperature may pose risk for scalding burns. Children should be monitored throughout treatment.28 If hot water immersion does not provide relief, oral analgesics may be considered. Stonefish antivenom is available and may be used for any scorpionfish sting given the shared biologic properties between genera. Providers evaluating stings could use sterile irrigation to clean wounds and search for foreign bodies including spine fragments; probing should be accomplished by instruments rather than a gloved finger. Providers should consider culturing wounds and prescribing antibiotics for suspected secondary infections. A tetanus toxoid history also should be elicited, and patients may have a booster administered, as indicated.29

With the growing popularity of water sports and a proliferation of invasive species, human injuries from marine animal envenomation continue to rise.1-3 Members of the scorpionfish family Scorpaenidae are second only to stingrays as the leading cause of the 40,000 to 50,000 injuries annually from marine life worldwide.4 Because scorpionfish represent a growing threat and competition with native species, it has been suggested that they could replace endangered species on restaurant menus.5-8 Scorpionfish have been introduced by humans from tropical to temperate seas and are now common off the coast of California and the eastern coast from New York to Florida, as well as in the Caribbean, the Bahamas, and off the southern coast of Brazil. Victims of scorpionfish stings experience considerable pain and may require days to weeks to fully recover, highlighting the socioeconomic costs and burden of scorpionfish envenomation.9,10 Fishers, divers, swimmers, and aquarium owners are most often affected.

Family

The common term scorpionfish refers to both the family Scorpaenidae and the genus Scorpaena. Members of this family possess similar dorsal, anal, and pelvic fins, though they vary between genera in their size and the potency of the venom they insulate. Other familiar members include the genus Pterois (lionfish) and Synanceja (stonefish). Synanceja are the most venomous within the group, but scorpionfish stings more commonly arise from Pterois and Scorpaena.8 Because of the rare shapes and vibrant colors of scorpionfish, some traders and aquarium owners will seek and pay high prices for these fish, providing further opportunity for envenomation.11,12

Characteristics

Scorpionfish have with a high variation in color, ranging from lighter grays to intense reds depending on their geographic location and habitat. Synanceja are bland in coloration, blending in with rocks and gravel, but the more dramatic-appearing Scorpaena exhibit a large cranium and wide range of multicolored patterns (Figure 1).13Pterois serve as the most conspicuous member of the group with brightly colored red and white stripes (Figure 2). Scorpionfish commonly grow up to 19 inches long and boast 12 dorsal, 2 pelvic, and 3 anal spines housing 5 to 10 mg of venom.14 An integumentary sheath encapsulates each spine housing the glandular tissue that produces the potent venom.

Red scorpionfish (Scorpaena scrofa).
Photograph courtesy of Sylvain Le Bris (https://www.inaturalist.org/). Republished under the Creative Commons Attribution (CC BY-NC 4.0).
FIGURE 1. Red scorpionfish (Scorpaena scrofa).

Toxin Properties

Unlike Pterois and Synanceja, Scorpaena do not have venom ducts around their glands, complicating the work of marine biologists aiming to extract and study the venomous toxins. Several studies have managed to isolate scorpionfish venom and overcome its unstable heat-labile nature to investigate its biologic properties.15-20 Several high-molecular-weight proteins (50–800 kDa) comprise the venom, including hyaluronidase, integrin-inhibiting factors, capillary permeability factor, proteases, and some less-understood cytolytic toxins. These factors provoke the inflammatory, proteolytic, hemorrhagic, cardiovascular, and hemolytic biologic activities at both the local and systemic levels, directing damage to wounded tissues and inducing vascular and tissue permeability to reach cellular processes far and wide. Mediators of inflammation include tumor necrosis factor, IL-6, and monocyte chemoattractant protein 1, followed by neutrophils and other mononuclear cells, initiating the immune response at the wound site. Toxin potency remains for up to 2 days after fish death.1

Lionfish (Pterois volitans).
Photograph courtesy of 808_Diver (https://www.inaturalist.org/). Republished under the Creative Commons Attribution (CC BY-NC 4.0).
FIGURE 2. Lionfish (Pterois volitans).

Clinical Manifestation

Physicians may be guided by clinical symptoms in identifying scorpionfish stings, as the patient may not know the identity of their marine assailant. Initially, individuals punctured by scorpionfish spikes will experience an acute pain and burning sensation at the puncture site that may be accompanied by systemic symptoms such as nausea, vomiting, diarrhea, tachycardia, hypotension, loss of consciousness, difficulty breathing, and delirium.9,21-23 The pain will intensify and radiate distal to the site of envenomation, and the wound may exhibit vesiculation, erythema, bruising, pallor, and notable edema.4,24 Pain intensity peaks at 30 to 90 minutes after envenomation, and other systemic symptoms generally last for 24 to 48 hours.25 If patients do not seek prompt treatment, secondary infection may ensue, and the lingering venom in the blister may cause dermal necrosis, paresthesia, and anesthesia. Chronic sequelae may include joint contractures, compartment syndrome, necrotic ulcers, and chronic neuropathy.1

Management

Treatment of scorpionfish stings primarily is palliative and aimed at symptom reduction. Patients should immediately treat wounds with hot but not scalding water immersion.26,27 Given the thermolabile components of scorpionfish venom, the most effective treatment is to soak the affected limb in water 42 °C to 45 °C for 30 to 90 minutes. Any higher temperature may pose risk for scalding burns. Children should be monitored throughout treatment.28 If hot water immersion does not provide relief, oral analgesics may be considered. Stonefish antivenom is available and may be used for any scorpionfish sting given the shared biologic properties between genera. Providers evaluating stings could use sterile irrigation to clean wounds and search for foreign bodies including spine fragments; probing should be accomplished by instruments rather than a gloved finger. Providers should consider culturing wounds and prescribing antibiotics for suspected secondary infections. A tetanus toxoid history also should be elicited, and patients may have a booster administered, as indicated.29

References
  1. Rensch G, Murphy-Lavoie HM. Lionfish, scorpionfish, and stonefish toxicity. StatPearls. StatPearls Publishing; May 10, 2022.
  2. Cearnal L. Red lionfish and ciguatoxin: menace spreading through western hemisphere. Ann Emerg Med. 2012;60:21A-22A. doi:10.1016/j.annemergmed.2012.05.022
  3. Côté IM, Green SJ. Potential effects of climate change on a marine invasion: the importance of current context. Curr Zool. 2012;58:1-8. doi:10.1093/czoolo/58.1.1
  4. Venomology of scorpionfishes. In: Santhanam R. Biology and Ecology of Venomous Marine Scorpionfishes. Academic Press; 2019:263-278.
  5. Ferri J, Staglicˇic´ N, Matić-Skoko S. The black scorpionfish, Scorpaena porcus (Scorpaenidae): could it serve as reliable indicator of Mediterranean coastal communities’ health? Ecol Indicators. 2012;18:25-30. doi:10.1016/j.ecolind.2011.11.004
  6. Santhanam R. Biology and Ecology of Venomous Marine Scorpionfishes. Academic Press; 2019.
  7. Morris JA, Akins JL. Feeding ecology of invasive lionfish (Pterois volitans) in the Bahamian Archipelago. Environ Biol Fishes. 2009;86:389-398. doi:10.1007/s10641-009-9538-8 
  8. Albins MA, Hixon MA. Worst case scenario: potential long-term effects of invasive predatory lionfish (Pterois volitans) on Atlantic and Caribbean coral-reef communities. Environ Biol Fishes. 2013;96:1151–1157. doi:10.1007/s10641-011-9795-1
  9. Haddad V Jr, Martins IA, Makyama HM. Injuries caused by scorpionfishes (Scorpaena plumieri Bloch, 1789 and Scorpaena brasiliensis Cuvier, 1829) in the Southwestern Atlantic Ocean (Brazilian coast): epidemiologic, clinic and therapeutic aspects of 23 stings in humans. Toxicon. 2003;42:79-83. doi:10.1016/s0041-0101(03)00103-x
  10. Campos FV, Menezes TN, Malacarne PF, et al. A review on the Scorpaena plumieri fish venom and its bioactive compounds. J Venom Anim Toxins Incl Trop Dis. 2016;22:35. doi:10.1186/s40409-016-0090-7
  11. Needleman RK, Neylan IP, Erickson TB. Environmental and ecological effects of climate change on venomous marine and amphibious species in the wilderness. Wilderness Environ Med. 2018;29:343-356. doi:10.1016/j.wem.2018.04.003
  12. Aldred B, Erickson T, Lipscomb J. Lionfish envenomations in an urban wilderness. Wilderness Environ Med. 1996;7:291-296. doi:10.1580/1080-6032(1996)007[0291:leiauw]2.3.co;2
  13. Stewart J, Hughes JM. Life-history traits of the southern hemisphere eastern red scorpionfish, Scorpaena cardinalis (Scorpaenidae: Scorpaeninae). Mar Freshw Res. 2010;61:1290-1297. doi:10.1071/MF10040
  14. Auerbach PS. Marine envenomations. N Engl J Med. 1991;325:486-493. doi:10.1056/NEJM199108153250707
  15. Andrich F, Carnielli JB, Cassoli JS, et al. A potent vasoactive cytolysin isolated from Scorpaena plumieri scorpionfish venom. Toxicon. 2010;56:487-496. doi:10.1016/j.toxicon.2010.05.003
  16. Gomes HL, Andrich F, Mauad H, et al. Cardiovascular effects of scorpionfish (Scorpaena plumieri) venom. Toxicon. 2010;55(2-3):580-589. doi:10.1016/j.toxicon.2009.10.012
  17. Menezes TN, Carnielli JB, Gomes HL, et al. Local inflammatory response induced by scorpionfish Scorpaena plumieri venom in mice. Toxicon. 2012;60:4-11. doi:10.1016/j.toxicon.2012.03.008
  18. Schaeffer RC Jr, Carlson RW, Russell FE. Some chemical properties of the venom of the scorpionfish Scorpaena guttata. Toxicon. 1971;9:69-78. doi:10.1016/0041-0101(71)90045-6
  19. Khalil AM, Wahsha MA, Abu Khadra KM, et al. Biochemical and histopathological effects of the stonefish (Synanceia verrucosa) venom in rats. Toxicon. 2018;142:45-51. doi:10.1016/j.toxicon.2017.12.052
  20. Mouchbahani-Constance S, Lesperance LS, Petitjean H, et al. Lionfish venom elicits pain predominantly through the activation of nonpeptidergic nociceptors. Pain. 2018;159:2255-2266. doi:10.1097/j.pain.0000000000001326
  21. Ottuso P. Aquatic dermatology: encounters with the denizens of the deep (and not so deep)—a review. part II: the vertebrates, single-celled organisms, and aquatic biotoxins. Int J Dermatol. 2013;52:268-278. doi:10.1111/j.1365-4632.2011.05426.x
  22. Bayley HH. Injuries caused by scorpion fish. Trans R Soc Trop Med Hyg. 1940;34:227-230. doi:10.1016/s0035-9203(40)90072-4
  23. González D. Epidemiological and clinical aspects of certain venomous animals of Spain. Toxicon. 1982;20:925-928. doi:10.1016/0041-0101(82)90080-0
  24. Halstead BW. Injurious effects from the sting of the scorpionfish, Scorpaena guttata. with report of a case. Calif Med. 1951;74:395-396.
  25. Vasievich MP, Villarreal JD, Tomecki KJ. Got the travel bug? a review of common infections, infestations, bites, and stings among returning travelers. Am J Clin Dermatol. 2016;17:451-462. doi:10.1007/s40257-016-0203-7
  26. Barnett S, Saggiomo S, Smout M, et al. Heat deactivation of the stonefish Synanceia horrida venom—implications for first-aid management. Diving Hyperb Med. 2017;47:155-158. doi:10.28920/dhm47.3.155-158
  27. Russell FE. Weever fish sting: the last word. Br Med J (Clin Res Ed). 1983;287:981-982. doi:10.1136/bmj.287.6397.981-c
  28. Tomlinson H, Elston DM. Aquatic antagonists: lionfish (Pterois volitans). Cutis. 2018;102:232-234.
  29. Hornbeak KB, Auerbach PS. Marine envenomation. Emerg Med Clin North Am. 2017;35:321-337. doi:10.1016/j.emc.2016.12.004
References
  1. Rensch G, Murphy-Lavoie HM. Lionfish, scorpionfish, and stonefish toxicity. StatPearls. StatPearls Publishing; May 10, 2022.
  2. Cearnal L. Red lionfish and ciguatoxin: menace spreading through western hemisphere. Ann Emerg Med. 2012;60:21A-22A. doi:10.1016/j.annemergmed.2012.05.022
  3. Côté IM, Green SJ. Potential effects of climate change on a marine invasion: the importance of current context. Curr Zool. 2012;58:1-8. doi:10.1093/czoolo/58.1.1
  4. Venomology of scorpionfishes. In: Santhanam R. Biology and Ecology of Venomous Marine Scorpionfishes. Academic Press; 2019:263-278.
  5. Ferri J, Staglicˇic´ N, Matić-Skoko S. The black scorpionfish, Scorpaena porcus (Scorpaenidae): could it serve as reliable indicator of Mediterranean coastal communities’ health? Ecol Indicators. 2012;18:25-30. doi:10.1016/j.ecolind.2011.11.004
  6. Santhanam R. Biology and Ecology of Venomous Marine Scorpionfishes. Academic Press; 2019.
  7. Morris JA, Akins JL. Feeding ecology of invasive lionfish (Pterois volitans) in the Bahamian Archipelago. Environ Biol Fishes. 2009;86:389-398. doi:10.1007/s10641-009-9538-8 
  8. Albins MA, Hixon MA. Worst case scenario: potential long-term effects of invasive predatory lionfish (Pterois volitans) on Atlantic and Caribbean coral-reef communities. Environ Biol Fishes. 2013;96:1151–1157. doi:10.1007/s10641-011-9795-1
  9. Haddad V Jr, Martins IA, Makyama HM. Injuries caused by scorpionfishes (Scorpaena plumieri Bloch, 1789 and Scorpaena brasiliensis Cuvier, 1829) in the Southwestern Atlantic Ocean (Brazilian coast): epidemiologic, clinic and therapeutic aspects of 23 stings in humans. Toxicon. 2003;42:79-83. doi:10.1016/s0041-0101(03)00103-x
  10. Campos FV, Menezes TN, Malacarne PF, et al. A review on the Scorpaena plumieri fish venom and its bioactive compounds. J Venom Anim Toxins Incl Trop Dis. 2016;22:35. doi:10.1186/s40409-016-0090-7
  11. Needleman RK, Neylan IP, Erickson TB. Environmental and ecological effects of climate change on venomous marine and amphibious species in the wilderness. Wilderness Environ Med. 2018;29:343-356. doi:10.1016/j.wem.2018.04.003
  12. Aldred B, Erickson T, Lipscomb J. Lionfish envenomations in an urban wilderness. Wilderness Environ Med. 1996;7:291-296. doi:10.1580/1080-6032(1996)007[0291:leiauw]2.3.co;2
  13. Stewart J, Hughes JM. Life-history traits of the southern hemisphere eastern red scorpionfish, Scorpaena cardinalis (Scorpaenidae: Scorpaeninae). Mar Freshw Res. 2010;61:1290-1297. doi:10.1071/MF10040
  14. Auerbach PS. Marine envenomations. N Engl J Med. 1991;325:486-493. doi:10.1056/NEJM199108153250707
  15. Andrich F, Carnielli JB, Cassoli JS, et al. A potent vasoactive cytolysin isolated from Scorpaena plumieri scorpionfish venom. Toxicon. 2010;56:487-496. doi:10.1016/j.toxicon.2010.05.003
  16. Gomes HL, Andrich F, Mauad H, et al. Cardiovascular effects of scorpionfish (Scorpaena plumieri) venom. Toxicon. 2010;55(2-3):580-589. doi:10.1016/j.toxicon.2009.10.012
  17. Menezes TN, Carnielli JB, Gomes HL, et al. Local inflammatory response induced by scorpionfish Scorpaena plumieri venom in mice. Toxicon. 2012;60:4-11. doi:10.1016/j.toxicon.2012.03.008
  18. Schaeffer RC Jr, Carlson RW, Russell FE. Some chemical properties of the venom of the scorpionfish Scorpaena guttata. Toxicon. 1971;9:69-78. doi:10.1016/0041-0101(71)90045-6
  19. Khalil AM, Wahsha MA, Abu Khadra KM, et al. Biochemical and histopathological effects of the stonefish (Synanceia verrucosa) venom in rats. Toxicon. 2018;142:45-51. doi:10.1016/j.toxicon.2017.12.052
  20. Mouchbahani-Constance S, Lesperance LS, Petitjean H, et al. Lionfish venom elicits pain predominantly through the activation of nonpeptidergic nociceptors. Pain. 2018;159:2255-2266. doi:10.1097/j.pain.0000000000001326
  21. Ottuso P. Aquatic dermatology: encounters with the denizens of the deep (and not so deep)—a review. part II: the vertebrates, single-celled organisms, and aquatic biotoxins. Int J Dermatol. 2013;52:268-278. doi:10.1111/j.1365-4632.2011.05426.x
  22. Bayley HH. Injuries caused by scorpion fish. Trans R Soc Trop Med Hyg. 1940;34:227-230. doi:10.1016/s0035-9203(40)90072-4
  23. González D. Epidemiological and clinical aspects of certain venomous animals of Spain. Toxicon. 1982;20:925-928. doi:10.1016/0041-0101(82)90080-0
  24. Halstead BW. Injurious effects from the sting of the scorpionfish, Scorpaena guttata. with report of a case. Calif Med. 1951;74:395-396.
  25. Vasievich MP, Villarreal JD, Tomecki KJ. Got the travel bug? a review of common infections, infestations, bites, and stings among returning travelers. Am J Clin Dermatol. 2016;17:451-462. doi:10.1007/s40257-016-0203-7
  26. Barnett S, Saggiomo S, Smout M, et al. Heat deactivation of the stonefish Synanceia horrida venom—implications for first-aid management. Diving Hyperb Med. 2017;47:155-158. doi:10.28920/dhm47.3.155-158
  27. Russell FE. Weever fish sting: the last word. Br Med J (Clin Res Ed). 1983;287:981-982. doi:10.1136/bmj.287.6397.981-c
  28. Tomlinson H, Elston DM. Aquatic antagonists: lionfish (Pterois volitans). Cutis. 2018;102:232-234.
  29. Hornbeak KB, Auerbach PS. Marine envenomation. Emerg Med Clin North Am. 2017;35:321-337. doi:10.1016/j.emc.2016.12.004
Issue
Cutis - 113(3)
Issue
Cutis - 113(3)
Page Number
133-134,136
Page Number
133-134,136
Publications
Publications
Topics
Article Type
Display Headline
Aquatic Antagonists: Scorpionfish Envenomation
Display Headline
Aquatic Antagonists: Scorpionfish Envenomation
Sections
Inside the Article

Practice Points

  • As some species of scorpionfish proliferate, providers may see an increase in envenomation cases.
  • Physicians should suspect scorpionfish stings based on clinical symptoms and physical examination.
  • Scorpionfish toxins are thermolabile, and patients can find symptom relief by immediately immersing the affected area in hot water (42 °C–45 °C) for 30 to 90 minutes.
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