LayerRx Mapping ID
748
Slot System
Featured Buckets
Featured Buckets Admin
Medscape Lead Concept
5000331

Isotretinoin-Induced Skin Fragility in an Aerialist

Article Type
Changed
Tue, 07/02/2024 - 17:02
Display Headline
Isotretinoin-Induced Skin Fragility in an Aerialist

Isotretinoin was introduced more than 3 decades ago and marked a major advancement in the treatment of severe refractory cystic acne. The most common adverse effects linked to isotretinoin usage are mucocutaneous in nature, manifesting as xerosis and cheilitis.1 Skin fragility and poor wound healing also have been reported.2-6 Current recommendations for avoiding these adverse effects include refraining from waxing, laser procedures, and other elective cutaneous procedures for at least 6 months.7 We present a case of isotretinoin-induced cutaneous fragility resulting in blistering and erosions on the palms of a competitive aerial trapeze artist.

Case Report

A 25-year-old woman presented for follow-up during week 12 of isotretinoin therapy (40 mg twice daily) prescribed for acne. She reported peeling of the skin on the palms following intense aerial acrobatic workouts. She had been a performing aerialist for many years and had never sustained a similar injury. The wounds were painful and led to decreased activity. She had no notable medical history. Physical examination of the palms revealed erosions in a distribution that corresponded to horizontal bar contact and friction (Figure). The patient was advised on proper wound care, application of emollients, and minimizing friction. She completed the course of isotretinoin and has continued aerialist activity without recurrence of skin fragility.

Comment

Skin fragility is a well-known adverse effect of isotretinoin therapy.8 Pavlis and Lieblich9 reported skin fragility in a young wrestler who experienced similar skin erosions due to isotretinoin therapy. The proposed mechanism of isotretinoin-induced skin fragility is multifactorial. It involves an apoptotic effect on sebocytes,5 which results in reduced stratum corneum hydration and an associated increase in transepidermal water loss.6,10,11 Retinoids also are known to cause thinning of the skin, likely due to the disadhesion of both the epidermis and the stratum corneum, which was demonstrated by the easy removal of cornified cells through tape stripping in hairless mice treated with isotretinoin.12 In further investigations, human patients and hairless mice treated with isotretinoin readily developed friction blisters through pencil eraser abrasion.13 Examination of the friction blisters using light and electron microscopy revealed fraying or loss of the stratum corneum and viable epidermis as well as loss of desmosomes and tonofilaments. Additionally, intracellular and intercellular deposits of an unidentified amorphous material were noted.13

mawufroslijiphastophalocigofreliwunachusafregufrofraswidugoslehuvihucrebrisodrinimobiswosidibestephoprestumaswustech
%3Cp%3EA%20and%20B%2C%20Erosions%20on%20the%20palms%20due%20to%20isotretinoin%20induced%20skin%20fragility.%3C%2Fp%3E

Overall, the origin of skin fragility induced by isotretinoin is supported by its effect on sebocytes, increased transepidermal water loss, and profound disruption of the integrity of the epidermis, resulting in an elevated risk for inadvertent skin damage. Patients were encouraged to avoid cosmetic procedures in prior case reports,14-16 and because our case demonstrates the risk for cutaneous injury in athletes due to isotretinoin-induced skin fragility, we propose an extension of these warnings to encompass athletes receiving isotretinoin treatment. Offering early guidance on wound prevention is of paramount importance in maintaining athletic performance and minimizing painful injuries.

References
  1. Rajput I, Anjankar VP. Side effects of treating acne vulgaris with isotretinoin: a systematic review. Cureus. 2024;16:E55946. doi:10.7759/cureus.55946
  2. Hatami P, Balighi K, Asl HN, et al. Isotretinoin and timing of procedural interventions: clinical implications and practical points. J Cosmet Dermatol. 2023;22:2146-2149. doi:10.1111/jocd.15874
  3. McDonald KA, Shelley AJ, Alavi A. A systematic review on oral isotretinoin therapy and clinically observable wound healing in acne patients. J Cutan Med Surg. 2017;21:325-333. doi:10.1177/1203475417701419
  4. Layton A. The use of isotretinoin in acne. Dermatoendocrinol. 2009;1:162-169. doi:10.4161/derm.1.3.9364
  5. Zouboulis CC. Isotretinoin revisited: pluripotent effects on human sebaceous gland cells. J Invest Dermatol. 2006;126:2154-2156. doi:10.1038/sj.jid.5700418
  6. Kmiec´ ML, Pajor A, Broniarczyk-Dyła G. Evaluation of biophysical skin parameters and assessment of hair growth in patients with acne treated with isotretinoin. Postepy Dermatol Alergol. 2013;30:343-349. doi:10.5114/pdia.2013.39432
  7. Waldman A, Bolotin D, Arndt KA, et al. ASDS Guidelines Task Force: Consensus recommendations regarding the safety of lasers, dermabrasion, chemical peels, energy devices, and skin surgery during and after isotretinoin use. Dermatolog Surg. 2017;43:1249-1262. doi:10.1097/DSS.0000000000001166
  8. Aksoy H, Aksoy B, Calikoglu E. Systemic retinoids and scar dehiscence. Indian J Dermatol. 2019;64:68. doi:10.4103/ijd.IJD_148_18
  9. Pavlis MB, Lieblich L. Isotretinoin-induced skin fragility in a teenaged athlete: a case report. Cutis. 2013;92:33-34.
  10. Herane MI, Fuenzalida H, Zegpi E, et al. Specific gel-cream as adjuvant to oral isotretinoin improved hydration and prevented TEWL increase—a double-blind, randomized, placebo-controlled study. J Cosmet Dermatol. 2009;8:181-185. doi:10.1111/j.1473-2165.2009.00455.x
  11. Park KY, Ko EJ, Kim IS, et al. The effect of evening primrose oil for the prevention of xerotic cheilitis in acne patients being treated with isotretinoin: a pilot study. Ann Dermatol. 2014;26:706-712. doi:10.5021/ad.2014.26.6.706
  12. Elias PM, Fritsch PO, Lampe M, et al. Retinoid effects on epidermal structure, differentiation, and permeability. Lab Invest. 1981;44:531-540.
  13. Williams ML, Elias PM. Nature of skin fragility in patients receiving retinoids for systemic effect. Arch Dermatol. 1981;117:611-619.
  14. Rubenstein R, Roenigk HH, Stegman SJ, et al. Atypical keloids after dermabrasion of patients taking isotretinoin. J Am Acad Dermatol. 1986;15:280-285. doi:10.1016/S0190-9622(86)70167-9
  15. Zachariae H. Delayed wound healing and keloid formation following argon laser treatment or dermabrasion during isotretinoin treatment. Br J Dermatol. 1988;118:703-706. doi:10.1111/j.1365-2133.1988.tb02574.x
  16. Katz BE, Mac Farlane DF. Atypical facial scarring after isotretinoin therapy in a patient with previous dermabrasion. J Am Acad Dermatol. 1994;30:852-853. doi:10.1016/S0190-9622(94)70096-6
Article PDF
Author and Disclosure Information

 

From the University of South Florida, Tampa. Helana Ghali is from the Morsani College of Medicine, and Dr. Albers is from the Department of Dermatology and Cutaneous Surgery.

The authors report no conflict of interest.

Correspondence: Helana Ghali, BS, 560 Channelside Dr, Tampa, FL 33602 (ghali2@usf.edu).

Cutis. 2024 July;114(1):32-33. doi:10.12788/cutis.1042

Publications
Topics
Page Number
32-33
Sections
Author and Disclosure Information

 

From the University of South Florida, Tampa. Helana Ghali is from the Morsani College of Medicine, and Dr. Albers is from the Department of Dermatology and Cutaneous Surgery.

The authors report no conflict of interest.

Correspondence: Helana Ghali, BS, 560 Channelside Dr, Tampa, FL 33602 (ghali2@usf.edu).

Cutis. 2024 July;114(1):32-33. doi:10.12788/cutis.1042

Author and Disclosure Information

 

From the University of South Florida, Tampa. Helana Ghali is from the Morsani College of Medicine, and Dr. Albers is from the Department of Dermatology and Cutaneous Surgery.

The authors report no conflict of interest.

Correspondence: Helana Ghali, BS, 560 Channelside Dr, Tampa, FL 33602 (ghali2@usf.edu).

Cutis. 2024 July;114(1):32-33. doi:10.12788/cutis.1042

Article PDF
Article PDF

Isotretinoin was introduced more than 3 decades ago and marked a major advancement in the treatment of severe refractory cystic acne. The most common adverse effects linked to isotretinoin usage are mucocutaneous in nature, manifesting as xerosis and cheilitis.1 Skin fragility and poor wound healing also have been reported.2-6 Current recommendations for avoiding these adverse effects include refraining from waxing, laser procedures, and other elective cutaneous procedures for at least 6 months.7 We present a case of isotretinoin-induced cutaneous fragility resulting in blistering and erosions on the palms of a competitive aerial trapeze artist.

Case Report

A 25-year-old woman presented for follow-up during week 12 of isotretinoin therapy (40 mg twice daily) prescribed for acne. She reported peeling of the skin on the palms following intense aerial acrobatic workouts. She had been a performing aerialist for many years and had never sustained a similar injury. The wounds were painful and led to decreased activity. She had no notable medical history. Physical examination of the palms revealed erosions in a distribution that corresponded to horizontal bar contact and friction (Figure). The patient was advised on proper wound care, application of emollients, and minimizing friction. She completed the course of isotretinoin and has continued aerialist activity without recurrence of skin fragility.

Comment

Skin fragility is a well-known adverse effect of isotretinoin therapy.8 Pavlis and Lieblich9 reported skin fragility in a young wrestler who experienced similar skin erosions due to isotretinoin therapy. The proposed mechanism of isotretinoin-induced skin fragility is multifactorial. It involves an apoptotic effect on sebocytes,5 which results in reduced stratum corneum hydration and an associated increase in transepidermal water loss.6,10,11 Retinoids also are known to cause thinning of the skin, likely due to the disadhesion of both the epidermis and the stratum corneum, which was demonstrated by the easy removal of cornified cells through tape stripping in hairless mice treated with isotretinoin.12 In further investigations, human patients and hairless mice treated with isotretinoin readily developed friction blisters through pencil eraser abrasion.13 Examination of the friction blisters using light and electron microscopy revealed fraying or loss of the stratum corneum and viable epidermis as well as loss of desmosomes and tonofilaments. Additionally, intracellular and intercellular deposits of an unidentified amorphous material were noted.13

mawufroslijiphastophalocigofreliwunachusafregufrofraswidugoslehuvihucrebrisodrinimobiswosidibestephoprestumaswustech
%3Cp%3EA%20and%20B%2C%20Erosions%20on%20the%20palms%20due%20to%20isotretinoin%20induced%20skin%20fragility.%3C%2Fp%3E

Overall, the origin of skin fragility induced by isotretinoin is supported by its effect on sebocytes, increased transepidermal water loss, and profound disruption of the integrity of the epidermis, resulting in an elevated risk for inadvertent skin damage. Patients were encouraged to avoid cosmetic procedures in prior case reports,14-16 and because our case demonstrates the risk for cutaneous injury in athletes due to isotretinoin-induced skin fragility, we propose an extension of these warnings to encompass athletes receiving isotretinoin treatment. Offering early guidance on wound prevention is of paramount importance in maintaining athletic performance and minimizing painful injuries.

Isotretinoin was introduced more than 3 decades ago and marked a major advancement in the treatment of severe refractory cystic acne. The most common adverse effects linked to isotretinoin usage are mucocutaneous in nature, manifesting as xerosis and cheilitis.1 Skin fragility and poor wound healing also have been reported.2-6 Current recommendations for avoiding these adverse effects include refraining from waxing, laser procedures, and other elective cutaneous procedures for at least 6 months.7 We present a case of isotretinoin-induced cutaneous fragility resulting in blistering and erosions on the palms of a competitive aerial trapeze artist.

Case Report

A 25-year-old woman presented for follow-up during week 12 of isotretinoin therapy (40 mg twice daily) prescribed for acne. She reported peeling of the skin on the palms following intense aerial acrobatic workouts. She had been a performing aerialist for many years and had never sustained a similar injury. The wounds were painful and led to decreased activity. She had no notable medical history. Physical examination of the palms revealed erosions in a distribution that corresponded to horizontal bar contact and friction (Figure). The patient was advised on proper wound care, application of emollients, and minimizing friction. She completed the course of isotretinoin and has continued aerialist activity without recurrence of skin fragility.

Comment

Skin fragility is a well-known adverse effect of isotretinoin therapy.8 Pavlis and Lieblich9 reported skin fragility in a young wrestler who experienced similar skin erosions due to isotretinoin therapy. The proposed mechanism of isotretinoin-induced skin fragility is multifactorial. It involves an apoptotic effect on sebocytes,5 which results in reduced stratum corneum hydration and an associated increase in transepidermal water loss.6,10,11 Retinoids also are known to cause thinning of the skin, likely due to the disadhesion of both the epidermis and the stratum corneum, which was demonstrated by the easy removal of cornified cells through tape stripping in hairless mice treated with isotretinoin.12 In further investigations, human patients and hairless mice treated with isotretinoin readily developed friction blisters through pencil eraser abrasion.13 Examination of the friction blisters using light and electron microscopy revealed fraying or loss of the stratum corneum and viable epidermis as well as loss of desmosomes and tonofilaments. Additionally, intracellular and intercellular deposits of an unidentified amorphous material were noted.13

mawufroslijiphastophalocigofreliwunachusafregufrofraswidugoslehuvihucrebrisodrinimobiswosidibestephoprestumaswustech
%3Cp%3EA%20and%20B%2C%20Erosions%20on%20the%20palms%20due%20to%20isotretinoin%20induced%20skin%20fragility.%3C%2Fp%3E

Overall, the origin of skin fragility induced by isotretinoin is supported by its effect on sebocytes, increased transepidermal water loss, and profound disruption of the integrity of the epidermis, resulting in an elevated risk for inadvertent skin damage. Patients were encouraged to avoid cosmetic procedures in prior case reports,14-16 and because our case demonstrates the risk for cutaneous injury in athletes due to isotretinoin-induced skin fragility, we propose an extension of these warnings to encompass athletes receiving isotretinoin treatment. Offering early guidance on wound prevention is of paramount importance in maintaining athletic performance and minimizing painful injuries.

References
  1. Rajput I, Anjankar VP. Side effects of treating acne vulgaris with isotretinoin: a systematic review. Cureus. 2024;16:E55946. doi:10.7759/cureus.55946
  2. Hatami P, Balighi K, Asl HN, et al. Isotretinoin and timing of procedural interventions: clinical implications and practical points. J Cosmet Dermatol. 2023;22:2146-2149. doi:10.1111/jocd.15874
  3. McDonald KA, Shelley AJ, Alavi A. A systematic review on oral isotretinoin therapy and clinically observable wound healing in acne patients. J Cutan Med Surg. 2017;21:325-333. doi:10.1177/1203475417701419
  4. Layton A. The use of isotretinoin in acne. Dermatoendocrinol. 2009;1:162-169. doi:10.4161/derm.1.3.9364
  5. Zouboulis CC. Isotretinoin revisited: pluripotent effects on human sebaceous gland cells. J Invest Dermatol. 2006;126:2154-2156. doi:10.1038/sj.jid.5700418
  6. Kmiec´ ML, Pajor A, Broniarczyk-Dyła G. Evaluation of biophysical skin parameters and assessment of hair growth in patients with acne treated with isotretinoin. Postepy Dermatol Alergol. 2013;30:343-349. doi:10.5114/pdia.2013.39432
  7. Waldman A, Bolotin D, Arndt KA, et al. ASDS Guidelines Task Force: Consensus recommendations regarding the safety of lasers, dermabrasion, chemical peels, energy devices, and skin surgery during and after isotretinoin use. Dermatolog Surg. 2017;43:1249-1262. doi:10.1097/DSS.0000000000001166
  8. Aksoy H, Aksoy B, Calikoglu E. Systemic retinoids and scar dehiscence. Indian J Dermatol. 2019;64:68. doi:10.4103/ijd.IJD_148_18
  9. Pavlis MB, Lieblich L. Isotretinoin-induced skin fragility in a teenaged athlete: a case report. Cutis. 2013;92:33-34.
  10. Herane MI, Fuenzalida H, Zegpi E, et al. Specific gel-cream as adjuvant to oral isotretinoin improved hydration and prevented TEWL increase—a double-blind, randomized, placebo-controlled study. J Cosmet Dermatol. 2009;8:181-185. doi:10.1111/j.1473-2165.2009.00455.x
  11. Park KY, Ko EJ, Kim IS, et al. The effect of evening primrose oil for the prevention of xerotic cheilitis in acne patients being treated with isotretinoin: a pilot study. Ann Dermatol. 2014;26:706-712. doi:10.5021/ad.2014.26.6.706
  12. Elias PM, Fritsch PO, Lampe M, et al. Retinoid effects on epidermal structure, differentiation, and permeability. Lab Invest. 1981;44:531-540.
  13. Williams ML, Elias PM. Nature of skin fragility in patients receiving retinoids for systemic effect. Arch Dermatol. 1981;117:611-619.
  14. Rubenstein R, Roenigk HH, Stegman SJ, et al. Atypical keloids after dermabrasion of patients taking isotretinoin. J Am Acad Dermatol. 1986;15:280-285. doi:10.1016/S0190-9622(86)70167-9
  15. Zachariae H. Delayed wound healing and keloid formation following argon laser treatment or dermabrasion during isotretinoin treatment. Br J Dermatol. 1988;118:703-706. doi:10.1111/j.1365-2133.1988.tb02574.x
  16. Katz BE, Mac Farlane DF. Atypical facial scarring after isotretinoin therapy in a patient with previous dermabrasion. J Am Acad Dermatol. 1994;30:852-853. doi:10.1016/S0190-9622(94)70096-6
References
  1. Rajput I, Anjankar VP. Side effects of treating acne vulgaris with isotretinoin: a systematic review. Cureus. 2024;16:E55946. doi:10.7759/cureus.55946
  2. Hatami P, Balighi K, Asl HN, et al. Isotretinoin and timing of procedural interventions: clinical implications and practical points. J Cosmet Dermatol. 2023;22:2146-2149. doi:10.1111/jocd.15874
  3. McDonald KA, Shelley AJ, Alavi A. A systematic review on oral isotretinoin therapy and clinically observable wound healing in acne patients. J Cutan Med Surg. 2017;21:325-333. doi:10.1177/1203475417701419
  4. Layton A. The use of isotretinoin in acne. Dermatoendocrinol. 2009;1:162-169. doi:10.4161/derm.1.3.9364
  5. Zouboulis CC. Isotretinoin revisited: pluripotent effects on human sebaceous gland cells. J Invest Dermatol. 2006;126:2154-2156. doi:10.1038/sj.jid.5700418
  6. Kmiec´ ML, Pajor A, Broniarczyk-Dyła G. Evaluation of biophysical skin parameters and assessment of hair growth in patients with acne treated with isotretinoin. Postepy Dermatol Alergol. 2013;30:343-349. doi:10.5114/pdia.2013.39432
  7. Waldman A, Bolotin D, Arndt KA, et al. ASDS Guidelines Task Force: Consensus recommendations regarding the safety of lasers, dermabrasion, chemical peels, energy devices, and skin surgery during and after isotretinoin use. Dermatolog Surg. 2017;43:1249-1262. doi:10.1097/DSS.0000000000001166
  8. Aksoy H, Aksoy B, Calikoglu E. Systemic retinoids and scar dehiscence. Indian J Dermatol. 2019;64:68. doi:10.4103/ijd.IJD_148_18
  9. Pavlis MB, Lieblich L. Isotretinoin-induced skin fragility in a teenaged athlete: a case report. Cutis. 2013;92:33-34.
  10. Herane MI, Fuenzalida H, Zegpi E, et al. Specific gel-cream as adjuvant to oral isotretinoin improved hydration and prevented TEWL increase—a double-blind, randomized, placebo-controlled study. J Cosmet Dermatol. 2009;8:181-185. doi:10.1111/j.1473-2165.2009.00455.x
  11. Park KY, Ko EJ, Kim IS, et al. The effect of evening primrose oil for the prevention of xerotic cheilitis in acne patients being treated with isotretinoin: a pilot study. Ann Dermatol. 2014;26:706-712. doi:10.5021/ad.2014.26.6.706
  12. Elias PM, Fritsch PO, Lampe M, et al. Retinoid effects on epidermal structure, differentiation, and permeability. Lab Invest. 1981;44:531-540.
  13. Williams ML, Elias PM. Nature of skin fragility in patients receiving retinoids for systemic effect. Arch Dermatol. 1981;117:611-619.
  14. Rubenstein R, Roenigk HH, Stegman SJ, et al. Atypical keloids after dermabrasion of patients taking isotretinoin. J Am Acad Dermatol. 1986;15:280-285. doi:10.1016/S0190-9622(86)70167-9
  15. Zachariae H. Delayed wound healing and keloid formation following argon laser treatment or dermabrasion during isotretinoin treatment. Br J Dermatol. 1988;118:703-706. doi:10.1111/j.1365-2133.1988.tb02574.x
  16. Katz BE, Mac Farlane DF. Atypical facial scarring after isotretinoin therapy in a patient with previous dermabrasion. J Am Acad Dermatol. 1994;30:852-853. doi:10.1016/S0190-9622(94)70096-6
Page Number
32-33
Page Number
32-33
Publications
Publications
Topics
Article Type
Display Headline
Isotretinoin-Induced Skin Fragility in an Aerialist
Display Headline
Isotretinoin-Induced Skin Fragility in an Aerialist
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>Ghali</fileName> <TBEID>0C02F909.SIG</TBEID> <TBUniqueIdentifier>NJ_0C02F909</TBUniqueIdentifier> <newsOrJournal>Journal</newsOrJournal> <publisherName>Frontline Medical Communications Inc.</publisherName> <storyname>Case Report</storyname> <articleType>1</articleType> <TBLocation>Copyfitting-CT</TBLocation> <QCDate/> <firstPublished>20240702T152306</firstPublished> <LastPublished>20240702T152306</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240702T152305</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>Helana Ghali, BS</byline> <bylineText>Helana Ghali, BS; Sharon E. Albers, MD </bylineText> <bylineFull>Helana Ghali, BS</bylineFull> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType/> <journalDocType/> <linkLabel/> <pageRange>32-33</pageRange> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:"> <name/> <rightsInfo> <copyrightHolder> <name/> </copyrightHolder> <copyrightNotice/> </rightsInfo> </provider> <abstract/> <metaDescription>Isotretinoin was introduced more than 3 decades ago and marked a major advancement in the treatment of severe refractory cystic acne. The most common adverse ef</metaDescription> <articlePDF>302108</articlePDF> <teaserImage/> <title>Isotretinoin-Induced Skin Fragility in an Aerialist</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear>2024</pubPubdateYear> <pubPubdateMonth>July</pubPubdateMonth> <pubPubdateDay/> <pubVolume>114</pubVolume> <pubNumber>1</pubNumber> <wireChannels/> <primaryCMSID/> <CMSIDs> <CMSID>2307</CMSID> <CMSID>2159</CMSID> </CMSIDs> <keywords/> <seeAlsos/> <publications_g> <publicationData> <publicationCode>CT</publicationCode> <pubIssueName>July 2024</pubIssueName> <pubArticleType>Departments | 2159</pubArticleType> <pubTopics/> <pubCategories/> <pubSections> <pubSection>Case Letter | 2307<pubSubsection/></pubSection> </pubSections> <journalTitle>Cutis</journalTitle> <journalFullTitle>Cutis</journalFullTitle> <copyrightStatement>Copyright 2015 Frontline Medical Communications Inc., Parsippany, NJ, USA. All rights reserved.</copyrightStatement> </publicationData> </publications_g> <publications> <term canonical="true">12</term> </publications> <sections> <term canonical="true">45</term> </sections> <topics> <term canonical="true">171</term> <term>313</term> </topics> <links> <link> <itemClass qcode="ninat:composite"/> <altRep contenttype="application/pdf">images/18002763.pdf</altRep> <description role="drol:caption"/> <description role="drol:credit"/> </link> </links> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Isotretinoin-Induced Skin Fragility in an Aerialist</title> <deck/> </itemMeta> <itemContent> <p class="abstract">Isotretinoin is widely used for treatment of severe cystic acne; however, its use is accompanied by mucocutaneous adverse effects. The established protocol for conducting cutaneous procedures on patients undergoing current or recent treatment with isotretinoin recommends a cessation period of at least 6 months to mitigate risks for delayed wound healing and hypertrophic scarring due to medication-induced skin fragility. We present a unique case of isotretinoin-induced skin fragility resulting in blistering and erosions on the palms of a 25-year-old competitive aerial trapeze artist. This case highlights the underrecognized risk for skin vulnerability in athletes undergoing isotretinoin treatment and the importance of guiding athletes on heightened skin vulnerability during isotretinoin treatment. </p> <p>Isotretinoin was introduced more than 3 decades ago and marked a major advancement in the treatment of severe refractory cystic acne. The most common adverse effects linked to isotretinoin usage are mucocutaneous in nature, manifesting as xerosis and cheilitis.<sup>1</sup> Skin fragility and poor wound healing also have been reported.<sup>2-6</sup> Current recommendations for avoiding these adverse effects include refraining from waxing, laser procedures, and other elective cutaneous procedures for at least 6 months.<sup>7</sup> We present a case of isotretinoin-induced cutaneous fragility resulting in blistering and erosions on the palms of a competitive aerial trapeze artist. </p> <h3>Case Report</h3> <p>A 25-year-old woman presented for follow-up during week 12 of isotretinoin therapy (40 mg twice daily) prescribed for acne. She reported peeling of the skin on the palms following intense aerial acrobatic workouts. She had been a performing aerialist for many years and had never sustained a similar injury. The wounds were painful and led to decreased activity. She had no notable medical history. Physical examination of the palms revealed erosions in a distribution that corresponded to horizontal bar contact and friction (Figure). The patient was advised on proper wound care, application of emollients, and minimizing friction. She completed the course of isotretinoin and has continued aerialist activity without recurrence of skin fragility. </p> <h3>Comment</h3> <p>Skin fragility is a well-known adverse effect of isotretinoin therapy.<sup>8</sup> Pavlis and Lieblich<sup>9</sup> reported skin fragility in a young wrestler who experienced similar skin erosions due to isotretinoin therapy. The proposed mechanism of isotretinoin-induced skin fragility is multifactorial. It involves an apoptotic effect on sebocytes,<sup>5</sup> which results in reduced stratum corneum hydration and an associated increase in transepidermal water loss.<sup>6,10,11</sup> Retinoids also are known to cause thinning of the skin, likely due to the disadhesion of both the epidermis and the stratum corneum, which was demonstrated by the easy removal of cornified cells through tape stripping in hairless mice treated with isotretinoin.<sup>12</sup> In further investigations, human patients and hairless mice treated with isotretinoin readily developed friction blisters through pencil eraser abrasion.<sup>13</sup> Examination of the friction blisters using light and electron microscopy revealed fraying or loss of the stratum corneum and viable epidermis as well as loss of desmosomes and tonofilaments. Additionally, intracellular and intercellular deposits of an unidentified amorphous material were noted.<sup>13</sup> </p> <p>Overall, the origin of skin fragility induced by isotretinoin is supported by its effect on sebocytes, increased transepidermal water loss, and profound disruption of the integrity of the epidermis, resulting in an elevated risk for inadvertent skin damage. Patients were encouraged to avoid cosmetic procedures in prior case reports,<sup>14-16</sup> and because our case demonstrates the risk for cutaneous injury in athletes due to isotretinoin-induced skin fragility, we propose an extension of these warnings to encompass athletes receiving isotretinoin treatment. Offering early guidance on wound prevention is of paramount importance in maintaining athletic performance and minimizing painful injuries. </p> <h2>References</h2> <p class="reference"> 1. Rajput I, Anjankar VP. Side effects of treating acne vulgaris with isotretinoin: a systematic review. <i>Cureus</i>. 2024;16:E55946. doi:10.7759/cureus.55946<br/><br/> 2. Hatami P, Balighi K, Asl HN, et al. Isotretinoin and timing of procedural interventions: clinical implications and practical points. <i>J Cosmet Dermatol</i>. 2023;22:2146-2149. doi:10.1111/jocd.15874<br/><br/> 3. McDonald KA, Shelley AJ, Alavi A. A systematic review on oral isotretinoin therapy and clinically observable wound healing in acne patients. <i>J Cutan Med Surg</i>. 2017;21:325-333. doi:10.1177/1203475417701419<br/><br/> 4. Layton A. The use of isotretinoin in acne. <i>Dermatoendocrinol</i>. 2009;1:162-169. doi:10.4161/derm.1.3.9364<br/><br/> 5. Zouboulis CC. Isotretinoin revisited: pluripotent effects on human sebaceous gland cells. <i>J Invest Dermatol</i>. 2006;126:2154-2156. doi:10.1038/sj.jid.5700418<br/><br/> 6. Kmiec´ ML, Pajor A, Broniarczyk-Dyła G. Evaluation of biophysical skin parameters and assessment of hair growth in patients with acne treated with isotretinoin. <i>Postepy Dermatol Alergol</i>. 2013;30:343-349. doi:10.5114/pdia.2013.39432<br/><br/> 7. Waldman A, Bolotin D, Arndt KA, et al. ASDS Guidelines Task Force: Consensus recommendations regarding the safety of lasers, dermabrasion, chemical peels, energy devices, and skin surgery during and after isotretinoin use. <em>Dermatolog Surg</em>. 2017;43:1249-1262. doi:10.1097/DSS.0000000000001166<br/><br/> 8. Aksoy H, Aksoy B, Calikoglu E. Systemic retinoids and scar dehiscence. <i>Indian J Dermatol</i>. 2019;64:68. doi:10.4103/ijd.IJD_148_18<br/><br/> 9. Pavlis MB, Lieblich L. Isotretinoin-induced skin fragility in a teenaged athlete: a case report. <i>Cutis</i>. 2013;92:33-34. <br/><br/>10. Herane MI, Fuenzalida H, Zegpi E, et al. Specific gel-cream as adjuvant to oral isotretinoin improved hydration and prevented TEWL increase—a double-blind, randomized, placebo-controlled study. <i>J Cosmet Dermatol</i>. 2009;8:181-185. doi:10.1111/j.1473-2165.2009.00455.x <br/><br/>11. Park KY, Ko EJ, Kim IS, et al. The effect of evening primrose oil for the prevention of xerotic cheilitis in acne patients being treated with isotretinoin: a pilot study. <i>Ann Dermatol</i>. 2014;26:706-712. doi:10.5021/ad.2014.26.6.706<br/><br/>12. Elias PM, Fritsch PO, Lampe M, et al. Retinoid effects on epidermal structure, differentiation, and permeability. <i>Lab Invest</i>. 1981;44:531-540.<br/><br/>13. Williams ML, Elias PM. Nature of skin fragility in patients receiving retinoids for systemic effect. <i>Arch Dermatol</i>. 1981;117:611-619.<br/><br/>14. Rubenstein R, Roenigk HH, Stegman SJ, et al. Atypical keloids after dermabrasion of patients taking isotretinoin. <i>J Am Acad Dermatol</i>. 1986;15:280-285. doi:10.1016/S0190-9622(86)70167-9<br/><br/>15. Zachariae H. Delayed wound healing and keloid formation following argon laser treatment or dermabrasion during isotretinoin treatment. <i>Br J Dermatol</i>. 1988;118:703-706. doi:10.1111/j.1365-2133.1988.tb02574.x<br/><br/>16. Katz BE, Mac Farlane DF. Atypical facial scarring after isotretinoin therapy in a patient with previous dermabrasion<i>. J Am Acad Dermatol</i>. 1994;30:852-853. doi:10.1016/S0190-9622(94)70096-6</p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>in</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="insidehead">Practice <strong>Points</strong></p> <ul class="insidebody"> <li>Isotretinoin is used to treat severe nodulocystic acne but can cause adverse effects such as skin fragility, xerosis, and poor wound healing. </li> <li>Dermatologists should inform athletes of heightened skin vulnerability while undergoing isotretinoin treatment. </li> <li>Isotretinoin-induced skin fragility involves the effects of isotretinoin on sebocytes, transepidermal water loss, and disruption of the integrity of the epidermis.</li> </ul> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>bio</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="disclosure">From the University of South Florida, Tampa. Helana Ghali is from the Morsani College of Medicine, and Dr. Albers is from the Department of Dermatology and Cutaneous Surgery.</p> <p class="disclosure">The authors report no conflict of interest. <br/><br/>Correspondence: Helana Ghali, BS, 560 Channelside Dr, Tampa, FL 33602 (ghali2@usf.edu).<br/><br/><em>Cutis. </em>2024 July;114(1):32-33. doi:10.12788/cutis.1042</p> </itemContent> </newsItem> </itemSet></root>
Inside the Article

 

Practice Points

  • Isotretinoin is used to treat severe nodulocystic acne but can cause adverse effects such as skin fragility, xerosis, and poor wound healing.
  • Dermatologists should inform athletes of heightened skin vulnerability while undergoing isotretinoin treatment.
  • Isotretinoin-induced skin fragility involves the effects of isotretinoin on sebocytes, transepidermal water loss, and disruption of the integrity of the epidermis.
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
Image
Disable zoom
Off

Olive Oil Shows Promise for Wound Healing of Ulcers

Article Type
Changed
Fri, 06/14/2024 - 12:39
Display Headline
Olive Oil Shows Promise for Wound Healing of Ulcers

Olive oil is obtained by mechanical extraction from the fruit of the Olea europaea tree, which is believed to have originated from ancient Iran and Turkestan, later spreading to Anatolia, Syria, Palestine, and Israel. Mechanical extraction of the oil from the olive fruit involves pressure processing, centrifugation, and adhesion filtering.1 Refining of olive oil is done via alkali refining or physical refining, with physical refining being useful in removing oxidation by-products and pro-oxidant metals. Olive oil is composed mainly of triacylglycerols, which are glycerol esters attached to various fatty acids, with the most common fatty acid being the monounsaturated oleic acid. Additional fatty acids include palmitic acid, linoleic acid, stearic acid, and palmitoleic acid.2 Olive oil contains phenolic compounds, the main ones being oleuropein, hydroxytyrosol, and tyrosol. These phenolic compounds are proposed to be strong antioxidants and radical scavengers.3

Mediterranean countries are responsible for approximately 97% of the world’s olive cultivation.4 Olive oil historically was used as lamp fuel, lubricant, body ointment, and later as a source of edible oil.1 Recently, its potential uses in medicine have called for further exploration into other uses for olive oil.

The skin is the largest organ of the body and serves as a protective barrier against pathogens and harmful substances. Skin damage results in 3 main phases to aid in wound healing: inflammation, proliferation, and maturation. In proper skin healing, inflammation will stop once the harmful microbes are removed. However, an excess and prolongation of inflammation can result in delayed healing. Thus, interventions that can limit the amount of inflammation can help promote wound healing. Olive oil contains several anti-inflammatory molecules (compounds or chemicals), including phenolic compounds and omega-3 fatty acids.5 Studies also have shown that olive oil can promote re-epithelialization in tissues.6 Thus, use of olive oil in wound therapy has been of great interest.

This article will review studies that have investigated the use of olive oil for wound healing of diabetic foot ulcers, pressure ulcers, perineal ulcers, and chronic ulcers. To conduct a comprehensive scoping review of the literature on the effects of olive oil in wound healing, we utilized the resources of the Galter Health Sciences Library & Learning Center (Chicago, Illinois). Our search strategy was structured to encompass a range of relevant databases accessible through the library, including PubMed, Embase, and Web of Science. We formulated our search terms to be broad yet specific to our topic, combining keywords such as olive oil, wound healing, skin repair, and dermal therapy. The inclusion criteria were set to filter studies conducted from January 2000 to December 2019, focusing on clinical trials, observational studies, and review articles. We limited our search to articles published in English, which yielded a preliminary set of articles that were then screened based on their titles and abstracts. Full-text versions of potentially relevant studies were retrieved and assessed for eligibility. We included studies that specifically evaluated the effects of olive oil in wound healing, excluding those that did not directly relate to our research question or had insufficient data. The data extraction from these studies was conducted using a standardized form, capturing study design, population, intervention details, outcomes, and key findings. The synthesis of these data provided a comprehensive overview of the current evidence on the topic, aiding in the identification of gaps in knowledge and directions for future research.

Diabetic Foot Ulcers

Foot ulcers are common in patients with diabetes mellitus and are associated with notable morbidity and mortality. Foot ulcers can clinically manifest in various forms but are classically described as lesions with a deep sinus in the feet. Patients with diabetic foot ulcers are at risk for infection, and severe forms of the ulcers require amputation.7,8 Routine care of foot ulcers involves irrigation of the ulcer and surrounding area with normal saline solution daily, followed by a dressing with sterile gauze. Studies investigating the effect of olive oil on foot ulcers suggest that olive oil use for care and healing of foot ulcers is an area of interest.

A double-blind, randomized clinical trial investigated the effects of topical olive oil on diabetic foot ulcers.9 A total of 34 patients with foot ulcers of Wagner grades 1 (superficial ulcers that involved the skin but not underlying tissue) or 2 (deeper ulcers penetrating to the ligaments and muscles but not the bone) that had remained open and did not improve for more than 3 months were recruited. The patients were randomly assigned to receive topical olive oil and routine care (intervention group) or to receive routine care (control group). Patients who received olive oil had oil poured on their ulcers with gauze wrapped around the ulcer that was soaked with olive oil. The clinical characteristics of the diabetic ulcer (eg, site, grade, size, status of healing) were assessed. The study revealed that after 4 weeks, olive oil significantly decreased ulcer area (P=.01) and ulcer depth (P=.02) compared with the control. Furthermore, there was a significant difference (P=.003) in complete ulcer healing between the olive oil and control groups: 73.3% (11/15) of patients in the olive oil group had complete ulcer healing, whereas 13.3% (2/15) of patients in the control group had complete ulcer healing.9 The positive effect of olive oil on the healing of diabetic foot ulcers encourages further investigation as a possible therapy for foot ulcers.

Another randomized controlled trial of 45 patients with diabetic foot ulcers of Wagner grades 1 or 2 investigated the effect of olive oil.10 Patients were randomly assigned to 1 of 3 groups for 1 month: the olive oil group, the honey group, or the control group. Patients in the olive oil group had their wounds dressed using gauze with olive oil daily, the patients in the honey group had their wounds dressed using gauze with honey daily, and the control group had routine care consisting of irrigation with saline solution and dressing with a sterile gauze. This study calculated a wound healing score based on a predefined checklist for diabetic foot ulcers through 4 variables: wound grading, color, surrounding tissue status, and drainage. Each variable had a maximum score of 100, contributing to a total possible score of 400, which indicated complete healing. A score of 50 signified ­deterioration. Wound healing was categorized as follows: (1) complete healing is indicated by a total score of 400; (2) partial healing was indicated by an increase of at least 30 points from the initial score; (3) lack of healing occurred when there was no change or less than a 30-point increase from the initial score; and (4) aggravation was noted when the score decreased by at least 10 points from the initial assessment. The study revealed that olive oil and honey treatments resulted in an increase in mean score, which indicated better wound healing. Patients in the olive oil group had a mean score of 253.0 before the intervention and 330.5 after the intervention (P<.0001); patients in the honey group had a mean score of 267.5 before the intervention and 371.5 after the intervention (P<.0001).10

There also have been case reports on combined olive oil and honey in diabetic foot ulcer management. Haghighian et al11 presented a case of a diabetic foot wound that healed completely within 2 weeks after the combined use of olive oil and honey wax. Zahmatkesh and Rashidi12 observed the healing of a diabetic foot wound over a month with daily dressings of a mixture of heated honey and olive oil, resulting in granulation tissue formation within 5 days. Microvascular changes, such as capillary basement membrane thickening, pericyte degeneration, and impairment of vasodilation and constriction, may contribute to inflammation in blood vessels, which can delay the healing of diabetic foot ulcers.7 Because olive oil and honey contain compounds that have antioxidative, antimicrobial, and anti-inflammatory properties, both may play a role in notably reducing inflammation and promoting the healing of foot ulcers.13

Pressure Ulcers

A pressure ulcer is a superficial skin injury that is caused by a prolonged period of pressure on the skin, in which the skin becomes red but there is no rupture. Prolonged periods of immobility resulting in a reduction or pause of blood supply are common causes of pressure ulcers.14 Studies have suggested that topical olive oil may be effective in prevention of pressure ulcers and should be incorporated as part of standard-of-care measures.

In a randomized, single-blind trial, 72 patients with the first stage of bedsore—which is a pressure ulcer—in the sacral, shoulder, heel, or other areas were randomly assigned to either the intervention or control group.14 Patients in the intervention group had 15 mL of olive oil rubbed on the wound for 20 minutes daily and then washed with tepid water. The Pressure Ulcer Scale for Healing tool was utilized to assess the healing status of the pressure ulcer. This tool considers wound surface size, exudate rate, and tissue type to provide a score of 0 to 17 (0=healed ulcer; 17=progression of ulcer). The mean score (SD) was lower in the olive oil group at days 4 and 7 compared with the control group (day 4: 7.50 [2.823] vs 9.50 [1.732]; day 7: 5.44 [3.806] vs 8.83 [2.864])(P<.001). Furthermore, between days 1 and 7, there was significant improvement in the olive oil group (mean difference, 3.56; P<.001) but no significant change in the control group (mean difference, 0.75; P=.052).14 The results indicate that patients in the olive oil group had a better ulcer healing status compared with patients in the control group.

In a noninferiority, randomized, double-blind clinical trial, olive oil was compared to a recommended skin care measure of hyperoxygenated fatty acids (HOFAs) for the prevention of pressure ulcers.15 The study consisted of 571 residents from several nursing homes who were at risk for pressure ulcers. Either olive oil or HOFA was applied to areas at risk for pressure ulcers, with 2 sprays of 0.2 mL per spray to each area every 12 hours. The participants were followed up for 30 days or until a pressure ulcer developed. Researchers performed skin assessments; the Braden Scale was used to assess the risk for pressure ulcers. The incidence difference of pressure ulcers in the olive oil group and HOFA group did not exceed in the noninferiority margin of 7%. Furthermore, Kaplan-Meier survival curves for the time until pressure ulcer onset showed a nonsignificant difference between the 2 groups.15 These findings suggest that olive oil is as effective as HOFA for the prevention of pressure ulcers. Although the mechanism of olive oil on prevention of pressure ulcers has not yet been determined, it has been suggested that anti-inflammatory compounds in olive oil, such as polyphenol and oleocanthal compounds, play an anti-inflammatory role.

Perineal Ulcers

Episiotomy is a surgical incision that is made to open the vagina during birth to aid in delivery of the baby. In contrast to spontaneous vaginal tears, an episiotomy allows for easier repair and healing of the laceration.16 Studies were conducted to investigate the effect of olive oil on women with lacerations after an episiotomy.

A total of 90 primigravid women who had undergone episiotomy were recruited and randomly assigned to 1 of 2 interventions: cold compression with gel packs for 20 minutes within 12 hours after delivery for up to 10 days, if necessary, or topical olive oil twice daily within 12 hours after delivery for up to 10 days.17 Although there was no significant difference in the structural features of the wound, there was a significant difference in the redness severity. After 10 days, the mean REEDA (redness, edema, ecchymosis, discharge, and apposition) score (SD), which assesses tissue healing, was 0.47 (0.96) in patients who received cold compression with gel packs and 0.20 (0.50) in patients who received topical olive oil (P=.04).17 This study suggests that there is the potential for olive oil to be used for wound healing after episiotomy.

A double-blind trial consisted of 60 women who had mediolateral episiotomy or perineal tear grades 1 and 2 who were randomly assigned to 1 of 2 groups for 10 days: olive oil sitz bath or distilled water sitz bath (control group). The results showed a significant difference in pain severity after 5 and 10 days (P<.05), wound redness after 5 days (P<.0001), and redness (P<.000) and edema (P<.05) 10 days after delivery.18 This study encourages further investigation of the benefits of olive oil for care after an episiotomy.

Chronic Ulcers

Chronic ulcers are other persistent wounds that do not respond to standard treatments and pose a notable health burden. Their development is influenced by factors such as oxidative stress, microbial infections, and the body’s immune response. A case series was conducted to investigate the wound healing effects of olive oil on chronic ulcers.19 Fourteen patients who were diagnosed with 1 or more chronic skin ulcers that had not healed with conventional treatment, such as cleansing, debridement, or infection control, were recruited. The mean (SD) of the patients’ Bates-Jensen Wound Assessment Tool score was 39.05 (4.23), indicating that these ulcers had been challenging to treat. In addition, the wounds in this study were found to be infected with bacteria. An ointment consisting of Ceratothoa oestroides olive oil extract was applied to the wounds after they were cleansed. The results showed that Bates-Jensen Wound Assessment Tool scores decreased by 14.7% to 67.5% (mean, 36%; median, 38%) after 3 months of treatment. Furthermore, 5 patients had a completely healed wound, indicating that C oestroides olive oil extract can regenerate chronic ulcers that do not respond to antibacterial agents.19 These results encourage further investigation of the role of C oestroides olive oil extract on healing properties and microbial control.

Final Thoughts

This review illuminated several key aspects of research on the role of olive oil in wound healing. Although the studies included in this review offer valuable insights, it is essential to acknowledge the variability in the quality of data presented. Several studies demonstrated robust methodology with clear definitions of outcomes and controlled conditions, providing high-quality evidence. However, other studies exhibited limitations, including small sample sizes and potential biases, which may affect the generalizability of the findings. Despite these limitations, the collective evidence suggests potential for olive oil in wound healing, warranting further investigation. Future research should aim for more standardized methodologies and larger, more diverse patient cohorts to validate these findings and explore the mechanisms underlying the therapeutic effects of olive oil.

References
  1. Emmons EW, Fedeli E, Firestone D. Olive oil introduction and history. In: Hui YH, ed. Bailey’s Industrial Oil & Fat Products, Vol. 2. Edible Oil and Fat Products: Edible Oils. 5th ed. John Wiley & Sons, Ltd; 241-269.
  2. Gorzynik-Debicka M, Przychodzen P, Cappello F, et al. Potential health benefits of olive oil and plant polyphenols. Int J Mol Sci. 2018;19:686. doi:10.3390/IJMS19030686
  3. Tuck KL, Hayball PJ. Major phenolic compounds in olive oil: metabolism and health effects. J Nutr Biochem. 2002;13:636-644. doi:10.1016/S0955-2863(02)00229-2
  4. Rabiei Z, Enferadi ST. Traceability of origin and authenticity of olive oil. In: Boskou D, ed. Olive Oil: Constituents, Quality, Health Properties and Bioconversions. InTech; 2012.
  5. Wardhana, Surachmanto ES, Datau EA. The role of omega-3 fatty acids contained in olive oil on chronic inflammation. Acta Med Indones. 2011;43:138-143.
  6. Aboui MM, Eidi A, Mortazavi P. Study of effect of olive oil on re-epithelialization of epithelial tissue in excision wound healing model in rats. J Comp Pathobiol. 2016;13:1875-1884.
  7. Aldana PC, Cartron AM, Khachemoune A. Reappraising diabetic foot ulcers: a focus on mechanisms of ulceration and clinical evaluation.Int J Low Extrem Wounds. 2022;21:294-302. doi:10.1177/1534734620944514
  8. Aldana PC, Khachemoune A. Diabetic foot ulcers: appraising standard of care and reviewing new trends in management. Am J Clin Dermatol. 2020;21:255-264. doi:10.1007/s40257-019-00495-x
  9. Nasiri M, Fayazi S, Jahani S, et al. The effect of topical olive oil on the healing of foot ulcer in patients with type 2 diabetes: a double-blind randomized clinical trial study in Iran. J Diabetes Metab Disord. 2015;14:38. doi:10.1186/S40200-015-0167-9
  10. Karimi Z, Behnammoghadam M, Rafiei H, et al. Impact of olive oil and honey on healing of diabetic foot: a randomized controlled trial. Clin Cosmet Investig Dermatol. 2019;12:347-354. doi:10.2147/CCID.S198577
  11. Haghighian HK, Koushan Y, Asgharzadeh A. Treatment of diabetic foot ulcer with propolis and olive oil: a case report. Knowl Health. 2012;6:35-38.
  12. Zahmatkesh M, Rashidi M. Case report of diabetic foot ulcer with topical honey and olive oil. J Med Plants. 2008;8:36-41.
  13. Cicerale S, Lucas LJ, Keast RS. Antimicrobial, antioxidant and anti-inflammatory phenolic activities in extra virgin olive oil. Curr Opin Biotechnol. 2012;23:129-135. doi:10.1016/J.COPBIO.2011.09.006
  14. Miraj S, Pourafzali S, Ahmadabadi ZV, et al. Effect of olive oil in preventing the development of pressure ulcer grade one in intensive care unit patients. Int J Prev Med. 2020;11:23. doi:10.4103/IJPVM.IJPVM_545_18
  15. Díaz‐Valenzuela A, García‐Fernández FP, Carmona Fernández P, et al. Effectiveness and safety of olive oil preparation for topical use in pressure ulcer prevention: multicentre, controlled, randomised, and double‐blinded clinical trial. Int Wound J. 2019;16:1314-1322. doi:10.1111/IWJ.13191
  16. Carroli G, Mignini L. Episiotomy for vaginal birth. Cochrane Database Syst Rev. 2009;CD000081. doi:10.1002/14651858.CD000081.PUB2
  17. Amani R, Kariman N, Mojab F, et al. Comparison of the effects of cold compress with gel packs and topical olive oil on episiotomy wound healing. J Babol Univ Med Sci. 2015;17:7-12. doi:10.22088/JBUMS.17.11.7
  18. Behmanesh F, Aghamohammadi A, Zeinalzadeh M, et al. Effects of olive oil sitz bath on improvement of perineal injury after delivery. Koomesh. 2013;14:309-315.
  19. Vitsos A, Tsagarousianos C, Vergos O, et al. Efficacy of a Ceratothoa oestroides olive oil extract in patients with chronic ulcers: a pilot study. Int J Low Extrem Wounds. 2019;18:309-316. doi:10.1177/1534734619856143
Article PDF
Author and Disclosure Information

 

Dr. Malik and Muhammad Taaha Hassan are from the Northwestern University Feinberg School of Medicine, Chicago, Illinois. Dr. Khachemoune is from Veterans Affairs Medical Center, Brooklyn, New York, and SUNY Downstate Medical Center, Brooklyn, New York.

The authors report no conflict of interest.

Correspondence: Amor Khachemoune, MD, SUNY Downstate, Veterans Affairs Medical Center, 800 Poly Pl, Brooklyn, NY 11209(amorkh@gmail.com).

Cutis. 2024 June;113(6):260-263. doi:10.12788/cutis.1035

Issue
Cutis - 113(6)
Publications
Topics
Page Number
260-263
Sections
Author and Disclosure Information

 

Dr. Malik and Muhammad Taaha Hassan are from the Northwestern University Feinberg School of Medicine, Chicago, Illinois. Dr. Khachemoune is from Veterans Affairs Medical Center, Brooklyn, New York, and SUNY Downstate Medical Center, Brooklyn, New York.

The authors report no conflict of interest.

Correspondence: Amor Khachemoune, MD, SUNY Downstate, Veterans Affairs Medical Center, 800 Poly Pl, Brooklyn, NY 11209(amorkh@gmail.com).

Cutis. 2024 June;113(6):260-263. doi:10.12788/cutis.1035

Author and Disclosure Information

 

Dr. Malik and Muhammad Taaha Hassan are from the Northwestern University Feinberg School of Medicine, Chicago, Illinois. Dr. Khachemoune is from Veterans Affairs Medical Center, Brooklyn, New York, and SUNY Downstate Medical Center, Brooklyn, New York.

The authors report no conflict of interest.

Correspondence: Amor Khachemoune, MD, SUNY Downstate, Veterans Affairs Medical Center, 800 Poly Pl, Brooklyn, NY 11209(amorkh@gmail.com).

Cutis. 2024 June;113(6):260-263. doi:10.12788/cutis.1035

Article PDF
Article PDF

Olive oil is obtained by mechanical extraction from the fruit of the Olea europaea tree, which is believed to have originated from ancient Iran and Turkestan, later spreading to Anatolia, Syria, Palestine, and Israel. Mechanical extraction of the oil from the olive fruit involves pressure processing, centrifugation, and adhesion filtering.1 Refining of olive oil is done via alkali refining or physical refining, with physical refining being useful in removing oxidation by-products and pro-oxidant metals. Olive oil is composed mainly of triacylglycerols, which are glycerol esters attached to various fatty acids, with the most common fatty acid being the monounsaturated oleic acid. Additional fatty acids include palmitic acid, linoleic acid, stearic acid, and palmitoleic acid.2 Olive oil contains phenolic compounds, the main ones being oleuropein, hydroxytyrosol, and tyrosol. These phenolic compounds are proposed to be strong antioxidants and radical scavengers.3

Mediterranean countries are responsible for approximately 97% of the world’s olive cultivation.4 Olive oil historically was used as lamp fuel, lubricant, body ointment, and later as a source of edible oil.1 Recently, its potential uses in medicine have called for further exploration into other uses for olive oil.

The skin is the largest organ of the body and serves as a protective barrier against pathogens and harmful substances. Skin damage results in 3 main phases to aid in wound healing: inflammation, proliferation, and maturation. In proper skin healing, inflammation will stop once the harmful microbes are removed. However, an excess and prolongation of inflammation can result in delayed healing. Thus, interventions that can limit the amount of inflammation can help promote wound healing. Olive oil contains several anti-inflammatory molecules (compounds or chemicals), including phenolic compounds and omega-3 fatty acids.5 Studies also have shown that olive oil can promote re-epithelialization in tissues.6 Thus, use of olive oil in wound therapy has been of great interest.

This article will review studies that have investigated the use of olive oil for wound healing of diabetic foot ulcers, pressure ulcers, perineal ulcers, and chronic ulcers. To conduct a comprehensive scoping review of the literature on the effects of olive oil in wound healing, we utilized the resources of the Galter Health Sciences Library & Learning Center (Chicago, Illinois). Our search strategy was structured to encompass a range of relevant databases accessible through the library, including PubMed, Embase, and Web of Science. We formulated our search terms to be broad yet specific to our topic, combining keywords such as olive oil, wound healing, skin repair, and dermal therapy. The inclusion criteria were set to filter studies conducted from January 2000 to December 2019, focusing on clinical trials, observational studies, and review articles. We limited our search to articles published in English, which yielded a preliminary set of articles that were then screened based on their titles and abstracts. Full-text versions of potentially relevant studies were retrieved and assessed for eligibility. We included studies that specifically evaluated the effects of olive oil in wound healing, excluding those that did not directly relate to our research question or had insufficient data. The data extraction from these studies was conducted using a standardized form, capturing study design, population, intervention details, outcomes, and key findings. The synthesis of these data provided a comprehensive overview of the current evidence on the topic, aiding in the identification of gaps in knowledge and directions for future research.

Diabetic Foot Ulcers

Foot ulcers are common in patients with diabetes mellitus and are associated with notable morbidity and mortality. Foot ulcers can clinically manifest in various forms but are classically described as lesions with a deep sinus in the feet. Patients with diabetic foot ulcers are at risk for infection, and severe forms of the ulcers require amputation.7,8 Routine care of foot ulcers involves irrigation of the ulcer and surrounding area with normal saline solution daily, followed by a dressing with sterile gauze. Studies investigating the effect of olive oil on foot ulcers suggest that olive oil use for care and healing of foot ulcers is an area of interest.

A double-blind, randomized clinical trial investigated the effects of topical olive oil on diabetic foot ulcers.9 A total of 34 patients with foot ulcers of Wagner grades 1 (superficial ulcers that involved the skin but not underlying tissue) or 2 (deeper ulcers penetrating to the ligaments and muscles but not the bone) that had remained open and did not improve for more than 3 months were recruited. The patients were randomly assigned to receive topical olive oil and routine care (intervention group) or to receive routine care (control group). Patients who received olive oil had oil poured on their ulcers with gauze wrapped around the ulcer that was soaked with olive oil. The clinical characteristics of the diabetic ulcer (eg, site, grade, size, status of healing) were assessed. The study revealed that after 4 weeks, olive oil significantly decreased ulcer area (P=.01) and ulcer depth (P=.02) compared with the control. Furthermore, there was a significant difference (P=.003) in complete ulcer healing between the olive oil and control groups: 73.3% (11/15) of patients in the olive oil group had complete ulcer healing, whereas 13.3% (2/15) of patients in the control group had complete ulcer healing.9 The positive effect of olive oil on the healing of diabetic foot ulcers encourages further investigation as a possible therapy for foot ulcers.

Another randomized controlled trial of 45 patients with diabetic foot ulcers of Wagner grades 1 or 2 investigated the effect of olive oil.10 Patients were randomly assigned to 1 of 3 groups for 1 month: the olive oil group, the honey group, or the control group. Patients in the olive oil group had their wounds dressed using gauze with olive oil daily, the patients in the honey group had their wounds dressed using gauze with honey daily, and the control group had routine care consisting of irrigation with saline solution and dressing with a sterile gauze. This study calculated a wound healing score based on a predefined checklist for diabetic foot ulcers through 4 variables: wound grading, color, surrounding tissue status, and drainage. Each variable had a maximum score of 100, contributing to a total possible score of 400, which indicated complete healing. A score of 50 signified ­deterioration. Wound healing was categorized as follows: (1) complete healing is indicated by a total score of 400; (2) partial healing was indicated by an increase of at least 30 points from the initial score; (3) lack of healing occurred when there was no change or less than a 30-point increase from the initial score; and (4) aggravation was noted when the score decreased by at least 10 points from the initial assessment. The study revealed that olive oil and honey treatments resulted in an increase in mean score, which indicated better wound healing. Patients in the olive oil group had a mean score of 253.0 before the intervention and 330.5 after the intervention (P<.0001); patients in the honey group had a mean score of 267.5 before the intervention and 371.5 after the intervention (P<.0001).10

There also have been case reports on combined olive oil and honey in diabetic foot ulcer management. Haghighian et al11 presented a case of a diabetic foot wound that healed completely within 2 weeks after the combined use of olive oil and honey wax. Zahmatkesh and Rashidi12 observed the healing of a diabetic foot wound over a month with daily dressings of a mixture of heated honey and olive oil, resulting in granulation tissue formation within 5 days. Microvascular changes, such as capillary basement membrane thickening, pericyte degeneration, and impairment of vasodilation and constriction, may contribute to inflammation in blood vessels, which can delay the healing of diabetic foot ulcers.7 Because olive oil and honey contain compounds that have antioxidative, antimicrobial, and anti-inflammatory properties, both may play a role in notably reducing inflammation and promoting the healing of foot ulcers.13

Pressure Ulcers

A pressure ulcer is a superficial skin injury that is caused by a prolonged period of pressure on the skin, in which the skin becomes red but there is no rupture. Prolonged periods of immobility resulting in a reduction or pause of blood supply are common causes of pressure ulcers.14 Studies have suggested that topical olive oil may be effective in prevention of pressure ulcers and should be incorporated as part of standard-of-care measures.

In a randomized, single-blind trial, 72 patients with the first stage of bedsore—which is a pressure ulcer—in the sacral, shoulder, heel, or other areas were randomly assigned to either the intervention or control group.14 Patients in the intervention group had 15 mL of olive oil rubbed on the wound for 20 minutes daily and then washed with tepid water. The Pressure Ulcer Scale for Healing tool was utilized to assess the healing status of the pressure ulcer. This tool considers wound surface size, exudate rate, and tissue type to provide a score of 0 to 17 (0=healed ulcer; 17=progression of ulcer). The mean score (SD) was lower in the olive oil group at days 4 and 7 compared with the control group (day 4: 7.50 [2.823] vs 9.50 [1.732]; day 7: 5.44 [3.806] vs 8.83 [2.864])(P<.001). Furthermore, between days 1 and 7, there was significant improvement in the olive oil group (mean difference, 3.56; P<.001) but no significant change in the control group (mean difference, 0.75; P=.052).14 The results indicate that patients in the olive oil group had a better ulcer healing status compared with patients in the control group.

In a noninferiority, randomized, double-blind clinical trial, olive oil was compared to a recommended skin care measure of hyperoxygenated fatty acids (HOFAs) for the prevention of pressure ulcers.15 The study consisted of 571 residents from several nursing homes who were at risk for pressure ulcers. Either olive oil or HOFA was applied to areas at risk for pressure ulcers, with 2 sprays of 0.2 mL per spray to each area every 12 hours. The participants were followed up for 30 days or until a pressure ulcer developed. Researchers performed skin assessments; the Braden Scale was used to assess the risk for pressure ulcers. The incidence difference of pressure ulcers in the olive oil group and HOFA group did not exceed in the noninferiority margin of 7%. Furthermore, Kaplan-Meier survival curves for the time until pressure ulcer onset showed a nonsignificant difference between the 2 groups.15 These findings suggest that olive oil is as effective as HOFA for the prevention of pressure ulcers. Although the mechanism of olive oil on prevention of pressure ulcers has not yet been determined, it has been suggested that anti-inflammatory compounds in olive oil, such as polyphenol and oleocanthal compounds, play an anti-inflammatory role.

Perineal Ulcers

Episiotomy is a surgical incision that is made to open the vagina during birth to aid in delivery of the baby. In contrast to spontaneous vaginal tears, an episiotomy allows for easier repair and healing of the laceration.16 Studies were conducted to investigate the effect of olive oil on women with lacerations after an episiotomy.

A total of 90 primigravid women who had undergone episiotomy were recruited and randomly assigned to 1 of 2 interventions: cold compression with gel packs for 20 minutes within 12 hours after delivery for up to 10 days, if necessary, or topical olive oil twice daily within 12 hours after delivery for up to 10 days.17 Although there was no significant difference in the structural features of the wound, there was a significant difference in the redness severity. After 10 days, the mean REEDA (redness, edema, ecchymosis, discharge, and apposition) score (SD), which assesses tissue healing, was 0.47 (0.96) in patients who received cold compression with gel packs and 0.20 (0.50) in patients who received topical olive oil (P=.04).17 This study suggests that there is the potential for olive oil to be used for wound healing after episiotomy.

A double-blind trial consisted of 60 women who had mediolateral episiotomy or perineal tear grades 1 and 2 who were randomly assigned to 1 of 2 groups for 10 days: olive oil sitz bath or distilled water sitz bath (control group). The results showed a significant difference in pain severity after 5 and 10 days (P<.05), wound redness after 5 days (P<.0001), and redness (P<.000) and edema (P<.05) 10 days after delivery.18 This study encourages further investigation of the benefits of olive oil for care after an episiotomy.

Chronic Ulcers

Chronic ulcers are other persistent wounds that do not respond to standard treatments and pose a notable health burden. Their development is influenced by factors such as oxidative stress, microbial infections, and the body’s immune response. A case series was conducted to investigate the wound healing effects of olive oil on chronic ulcers.19 Fourteen patients who were diagnosed with 1 or more chronic skin ulcers that had not healed with conventional treatment, such as cleansing, debridement, or infection control, were recruited. The mean (SD) of the patients’ Bates-Jensen Wound Assessment Tool score was 39.05 (4.23), indicating that these ulcers had been challenging to treat. In addition, the wounds in this study were found to be infected with bacteria. An ointment consisting of Ceratothoa oestroides olive oil extract was applied to the wounds after they were cleansed. The results showed that Bates-Jensen Wound Assessment Tool scores decreased by 14.7% to 67.5% (mean, 36%; median, 38%) after 3 months of treatment. Furthermore, 5 patients had a completely healed wound, indicating that C oestroides olive oil extract can regenerate chronic ulcers that do not respond to antibacterial agents.19 These results encourage further investigation of the role of C oestroides olive oil extract on healing properties and microbial control.

Final Thoughts

This review illuminated several key aspects of research on the role of olive oil in wound healing. Although the studies included in this review offer valuable insights, it is essential to acknowledge the variability in the quality of data presented. Several studies demonstrated robust methodology with clear definitions of outcomes and controlled conditions, providing high-quality evidence. However, other studies exhibited limitations, including small sample sizes and potential biases, which may affect the generalizability of the findings. Despite these limitations, the collective evidence suggests potential for olive oil in wound healing, warranting further investigation. Future research should aim for more standardized methodologies and larger, more diverse patient cohorts to validate these findings and explore the mechanisms underlying the therapeutic effects of olive oil.

Olive oil is obtained by mechanical extraction from the fruit of the Olea europaea tree, which is believed to have originated from ancient Iran and Turkestan, later spreading to Anatolia, Syria, Palestine, and Israel. Mechanical extraction of the oil from the olive fruit involves pressure processing, centrifugation, and adhesion filtering.1 Refining of olive oil is done via alkali refining or physical refining, with physical refining being useful in removing oxidation by-products and pro-oxidant metals. Olive oil is composed mainly of triacylglycerols, which are glycerol esters attached to various fatty acids, with the most common fatty acid being the monounsaturated oleic acid. Additional fatty acids include palmitic acid, linoleic acid, stearic acid, and palmitoleic acid.2 Olive oil contains phenolic compounds, the main ones being oleuropein, hydroxytyrosol, and tyrosol. These phenolic compounds are proposed to be strong antioxidants and radical scavengers.3

Mediterranean countries are responsible for approximately 97% of the world’s olive cultivation.4 Olive oil historically was used as lamp fuel, lubricant, body ointment, and later as a source of edible oil.1 Recently, its potential uses in medicine have called for further exploration into other uses for olive oil.

The skin is the largest organ of the body and serves as a protective barrier against pathogens and harmful substances. Skin damage results in 3 main phases to aid in wound healing: inflammation, proliferation, and maturation. In proper skin healing, inflammation will stop once the harmful microbes are removed. However, an excess and prolongation of inflammation can result in delayed healing. Thus, interventions that can limit the amount of inflammation can help promote wound healing. Olive oil contains several anti-inflammatory molecules (compounds or chemicals), including phenolic compounds and omega-3 fatty acids.5 Studies also have shown that olive oil can promote re-epithelialization in tissues.6 Thus, use of olive oil in wound therapy has been of great interest.

This article will review studies that have investigated the use of olive oil for wound healing of diabetic foot ulcers, pressure ulcers, perineal ulcers, and chronic ulcers. To conduct a comprehensive scoping review of the literature on the effects of olive oil in wound healing, we utilized the resources of the Galter Health Sciences Library & Learning Center (Chicago, Illinois). Our search strategy was structured to encompass a range of relevant databases accessible through the library, including PubMed, Embase, and Web of Science. We formulated our search terms to be broad yet specific to our topic, combining keywords such as olive oil, wound healing, skin repair, and dermal therapy. The inclusion criteria were set to filter studies conducted from January 2000 to December 2019, focusing on clinical trials, observational studies, and review articles. We limited our search to articles published in English, which yielded a preliminary set of articles that were then screened based on their titles and abstracts. Full-text versions of potentially relevant studies were retrieved and assessed for eligibility. We included studies that specifically evaluated the effects of olive oil in wound healing, excluding those that did not directly relate to our research question or had insufficient data. The data extraction from these studies was conducted using a standardized form, capturing study design, population, intervention details, outcomes, and key findings. The synthesis of these data provided a comprehensive overview of the current evidence on the topic, aiding in the identification of gaps in knowledge and directions for future research.

Diabetic Foot Ulcers

Foot ulcers are common in patients with diabetes mellitus and are associated with notable morbidity and mortality. Foot ulcers can clinically manifest in various forms but are classically described as lesions with a deep sinus in the feet. Patients with diabetic foot ulcers are at risk for infection, and severe forms of the ulcers require amputation.7,8 Routine care of foot ulcers involves irrigation of the ulcer and surrounding area with normal saline solution daily, followed by a dressing with sterile gauze. Studies investigating the effect of olive oil on foot ulcers suggest that olive oil use for care and healing of foot ulcers is an area of interest.

A double-blind, randomized clinical trial investigated the effects of topical olive oil on diabetic foot ulcers.9 A total of 34 patients with foot ulcers of Wagner grades 1 (superficial ulcers that involved the skin but not underlying tissue) or 2 (deeper ulcers penetrating to the ligaments and muscles but not the bone) that had remained open and did not improve for more than 3 months were recruited. The patients were randomly assigned to receive topical olive oil and routine care (intervention group) or to receive routine care (control group). Patients who received olive oil had oil poured on their ulcers with gauze wrapped around the ulcer that was soaked with olive oil. The clinical characteristics of the diabetic ulcer (eg, site, grade, size, status of healing) were assessed. The study revealed that after 4 weeks, olive oil significantly decreased ulcer area (P=.01) and ulcer depth (P=.02) compared with the control. Furthermore, there was a significant difference (P=.003) in complete ulcer healing between the olive oil and control groups: 73.3% (11/15) of patients in the olive oil group had complete ulcer healing, whereas 13.3% (2/15) of patients in the control group had complete ulcer healing.9 The positive effect of olive oil on the healing of diabetic foot ulcers encourages further investigation as a possible therapy for foot ulcers.

Another randomized controlled trial of 45 patients with diabetic foot ulcers of Wagner grades 1 or 2 investigated the effect of olive oil.10 Patients were randomly assigned to 1 of 3 groups for 1 month: the olive oil group, the honey group, or the control group. Patients in the olive oil group had their wounds dressed using gauze with olive oil daily, the patients in the honey group had their wounds dressed using gauze with honey daily, and the control group had routine care consisting of irrigation with saline solution and dressing with a sterile gauze. This study calculated a wound healing score based on a predefined checklist for diabetic foot ulcers through 4 variables: wound grading, color, surrounding tissue status, and drainage. Each variable had a maximum score of 100, contributing to a total possible score of 400, which indicated complete healing. A score of 50 signified ­deterioration. Wound healing was categorized as follows: (1) complete healing is indicated by a total score of 400; (2) partial healing was indicated by an increase of at least 30 points from the initial score; (3) lack of healing occurred when there was no change or less than a 30-point increase from the initial score; and (4) aggravation was noted when the score decreased by at least 10 points from the initial assessment. The study revealed that olive oil and honey treatments resulted in an increase in mean score, which indicated better wound healing. Patients in the olive oil group had a mean score of 253.0 before the intervention and 330.5 after the intervention (P<.0001); patients in the honey group had a mean score of 267.5 before the intervention and 371.5 after the intervention (P<.0001).10

There also have been case reports on combined olive oil and honey in diabetic foot ulcer management. Haghighian et al11 presented a case of a diabetic foot wound that healed completely within 2 weeks after the combined use of olive oil and honey wax. Zahmatkesh and Rashidi12 observed the healing of a diabetic foot wound over a month with daily dressings of a mixture of heated honey and olive oil, resulting in granulation tissue formation within 5 days. Microvascular changes, such as capillary basement membrane thickening, pericyte degeneration, and impairment of vasodilation and constriction, may contribute to inflammation in blood vessels, which can delay the healing of diabetic foot ulcers.7 Because olive oil and honey contain compounds that have antioxidative, antimicrobial, and anti-inflammatory properties, both may play a role in notably reducing inflammation and promoting the healing of foot ulcers.13

Pressure Ulcers

A pressure ulcer is a superficial skin injury that is caused by a prolonged period of pressure on the skin, in which the skin becomes red but there is no rupture. Prolonged periods of immobility resulting in a reduction or pause of blood supply are common causes of pressure ulcers.14 Studies have suggested that topical olive oil may be effective in prevention of pressure ulcers and should be incorporated as part of standard-of-care measures.

In a randomized, single-blind trial, 72 patients with the first stage of bedsore—which is a pressure ulcer—in the sacral, shoulder, heel, or other areas were randomly assigned to either the intervention or control group.14 Patients in the intervention group had 15 mL of olive oil rubbed on the wound for 20 minutes daily and then washed with tepid water. The Pressure Ulcer Scale for Healing tool was utilized to assess the healing status of the pressure ulcer. This tool considers wound surface size, exudate rate, and tissue type to provide a score of 0 to 17 (0=healed ulcer; 17=progression of ulcer). The mean score (SD) was lower in the olive oil group at days 4 and 7 compared with the control group (day 4: 7.50 [2.823] vs 9.50 [1.732]; day 7: 5.44 [3.806] vs 8.83 [2.864])(P<.001). Furthermore, between days 1 and 7, there was significant improvement in the olive oil group (mean difference, 3.56; P<.001) but no significant change in the control group (mean difference, 0.75; P=.052).14 The results indicate that patients in the olive oil group had a better ulcer healing status compared with patients in the control group.

In a noninferiority, randomized, double-blind clinical trial, olive oil was compared to a recommended skin care measure of hyperoxygenated fatty acids (HOFAs) for the prevention of pressure ulcers.15 The study consisted of 571 residents from several nursing homes who were at risk for pressure ulcers. Either olive oil or HOFA was applied to areas at risk for pressure ulcers, with 2 sprays of 0.2 mL per spray to each area every 12 hours. The participants were followed up for 30 days or until a pressure ulcer developed. Researchers performed skin assessments; the Braden Scale was used to assess the risk for pressure ulcers. The incidence difference of pressure ulcers in the olive oil group and HOFA group did not exceed in the noninferiority margin of 7%. Furthermore, Kaplan-Meier survival curves for the time until pressure ulcer onset showed a nonsignificant difference between the 2 groups.15 These findings suggest that olive oil is as effective as HOFA for the prevention of pressure ulcers. Although the mechanism of olive oil on prevention of pressure ulcers has not yet been determined, it has been suggested that anti-inflammatory compounds in olive oil, such as polyphenol and oleocanthal compounds, play an anti-inflammatory role.

Perineal Ulcers

Episiotomy is a surgical incision that is made to open the vagina during birth to aid in delivery of the baby. In contrast to spontaneous vaginal tears, an episiotomy allows for easier repair and healing of the laceration.16 Studies were conducted to investigate the effect of olive oil on women with lacerations after an episiotomy.

A total of 90 primigravid women who had undergone episiotomy were recruited and randomly assigned to 1 of 2 interventions: cold compression with gel packs for 20 minutes within 12 hours after delivery for up to 10 days, if necessary, or topical olive oil twice daily within 12 hours after delivery for up to 10 days.17 Although there was no significant difference in the structural features of the wound, there was a significant difference in the redness severity. After 10 days, the mean REEDA (redness, edema, ecchymosis, discharge, and apposition) score (SD), which assesses tissue healing, was 0.47 (0.96) in patients who received cold compression with gel packs and 0.20 (0.50) in patients who received topical olive oil (P=.04).17 This study suggests that there is the potential for olive oil to be used for wound healing after episiotomy.

A double-blind trial consisted of 60 women who had mediolateral episiotomy or perineal tear grades 1 and 2 who were randomly assigned to 1 of 2 groups for 10 days: olive oil sitz bath or distilled water sitz bath (control group). The results showed a significant difference in pain severity after 5 and 10 days (P<.05), wound redness after 5 days (P<.0001), and redness (P<.000) and edema (P<.05) 10 days after delivery.18 This study encourages further investigation of the benefits of olive oil for care after an episiotomy.

Chronic Ulcers

Chronic ulcers are other persistent wounds that do not respond to standard treatments and pose a notable health burden. Their development is influenced by factors such as oxidative stress, microbial infections, and the body’s immune response. A case series was conducted to investigate the wound healing effects of olive oil on chronic ulcers.19 Fourteen patients who were diagnosed with 1 or more chronic skin ulcers that had not healed with conventional treatment, such as cleansing, debridement, or infection control, were recruited. The mean (SD) of the patients’ Bates-Jensen Wound Assessment Tool score was 39.05 (4.23), indicating that these ulcers had been challenging to treat. In addition, the wounds in this study were found to be infected with bacteria. An ointment consisting of Ceratothoa oestroides olive oil extract was applied to the wounds after they were cleansed. The results showed that Bates-Jensen Wound Assessment Tool scores decreased by 14.7% to 67.5% (mean, 36%; median, 38%) after 3 months of treatment. Furthermore, 5 patients had a completely healed wound, indicating that C oestroides olive oil extract can regenerate chronic ulcers that do not respond to antibacterial agents.19 These results encourage further investigation of the role of C oestroides olive oil extract on healing properties and microbial control.

Final Thoughts

This review illuminated several key aspects of research on the role of olive oil in wound healing. Although the studies included in this review offer valuable insights, it is essential to acknowledge the variability in the quality of data presented. Several studies demonstrated robust methodology with clear definitions of outcomes and controlled conditions, providing high-quality evidence. However, other studies exhibited limitations, including small sample sizes and potential biases, which may affect the generalizability of the findings. Despite these limitations, the collective evidence suggests potential for olive oil in wound healing, warranting further investigation. Future research should aim for more standardized methodologies and larger, more diverse patient cohorts to validate these findings and explore the mechanisms underlying the therapeutic effects of olive oil.

References
  1. Emmons EW, Fedeli E, Firestone D. Olive oil introduction and history. In: Hui YH, ed. Bailey’s Industrial Oil & Fat Products, Vol. 2. Edible Oil and Fat Products: Edible Oils. 5th ed. John Wiley & Sons, Ltd; 241-269.
  2. Gorzynik-Debicka M, Przychodzen P, Cappello F, et al. Potential health benefits of olive oil and plant polyphenols. Int J Mol Sci. 2018;19:686. doi:10.3390/IJMS19030686
  3. Tuck KL, Hayball PJ. Major phenolic compounds in olive oil: metabolism and health effects. J Nutr Biochem. 2002;13:636-644. doi:10.1016/S0955-2863(02)00229-2
  4. Rabiei Z, Enferadi ST. Traceability of origin and authenticity of olive oil. In: Boskou D, ed. Olive Oil: Constituents, Quality, Health Properties and Bioconversions. InTech; 2012.
  5. Wardhana, Surachmanto ES, Datau EA. The role of omega-3 fatty acids contained in olive oil on chronic inflammation. Acta Med Indones. 2011;43:138-143.
  6. Aboui MM, Eidi A, Mortazavi P. Study of effect of olive oil on re-epithelialization of epithelial tissue in excision wound healing model in rats. J Comp Pathobiol. 2016;13:1875-1884.
  7. Aldana PC, Cartron AM, Khachemoune A. Reappraising diabetic foot ulcers: a focus on mechanisms of ulceration and clinical evaluation.Int J Low Extrem Wounds. 2022;21:294-302. doi:10.1177/1534734620944514
  8. Aldana PC, Khachemoune A. Diabetic foot ulcers: appraising standard of care and reviewing new trends in management. Am J Clin Dermatol. 2020;21:255-264. doi:10.1007/s40257-019-00495-x
  9. Nasiri M, Fayazi S, Jahani S, et al. The effect of topical olive oil on the healing of foot ulcer in patients with type 2 diabetes: a double-blind randomized clinical trial study in Iran. J Diabetes Metab Disord. 2015;14:38. doi:10.1186/S40200-015-0167-9
  10. Karimi Z, Behnammoghadam M, Rafiei H, et al. Impact of olive oil and honey on healing of diabetic foot: a randomized controlled trial. Clin Cosmet Investig Dermatol. 2019;12:347-354. doi:10.2147/CCID.S198577
  11. Haghighian HK, Koushan Y, Asgharzadeh A. Treatment of diabetic foot ulcer with propolis and olive oil: a case report. Knowl Health. 2012;6:35-38.
  12. Zahmatkesh M, Rashidi M. Case report of diabetic foot ulcer with topical honey and olive oil. J Med Plants. 2008;8:36-41.
  13. Cicerale S, Lucas LJ, Keast RS. Antimicrobial, antioxidant and anti-inflammatory phenolic activities in extra virgin olive oil. Curr Opin Biotechnol. 2012;23:129-135. doi:10.1016/J.COPBIO.2011.09.006
  14. Miraj S, Pourafzali S, Ahmadabadi ZV, et al. Effect of olive oil in preventing the development of pressure ulcer grade one in intensive care unit patients. Int J Prev Med. 2020;11:23. doi:10.4103/IJPVM.IJPVM_545_18
  15. Díaz‐Valenzuela A, García‐Fernández FP, Carmona Fernández P, et al. Effectiveness and safety of olive oil preparation for topical use in pressure ulcer prevention: multicentre, controlled, randomised, and double‐blinded clinical trial. Int Wound J. 2019;16:1314-1322. doi:10.1111/IWJ.13191
  16. Carroli G, Mignini L. Episiotomy for vaginal birth. Cochrane Database Syst Rev. 2009;CD000081. doi:10.1002/14651858.CD000081.PUB2
  17. Amani R, Kariman N, Mojab F, et al. Comparison of the effects of cold compress with gel packs and topical olive oil on episiotomy wound healing. J Babol Univ Med Sci. 2015;17:7-12. doi:10.22088/JBUMS.17.11.7
  18. Behmanesh F, Aghamohammadi A, Zeinalzadeh M, et al. Effects of olive oil sitz bath on improvement of perineal injury after delivery. Koomesh. 2013;14:309-315.
  19. Vitsos A, Tsagarousianos C, Vergos O, et al. Efficacy of a Ceratothoa oestroides olive oil extract in patients with chronic ulcers: a pilot study. Int J Low Extrem Wounds. 2019;18:309-316. doi:10.1177/1534734619856143
References
  1. Emmons EW, Fedeli E, Firestone D. Olive oil introduction and history. In: Hui YH, ed. Bailey’s Industrial Oil & Fat Products, Vol. 2. Edible Oil and Fat Products: Edible Oils. 5th ed. John Wiley & Sons, Ltd; 241-269.
  2. Gorzynik-Debicka M, Przychodzen P, Cappello F, et al. Potential health benefits of olive oil and plant polyphenols. Int J Mol Sci. 2018;19:686. doi:10.3390/IJMS19030686
  3. Tuck KL, Hayball PJ. Major phenolic compounds in olive oil: metabolism and health effects. J Nutr Biochem. 2002;13:636-644. doi:10.1016/S0955-2863(02)00229-2
  4. Rabiei Z, Enferadi ST. Traceability of origin and authenticity of olive oil. In: Boskou D, ed. Olive Oil: Constituents, Quality, Health Properties and Bioconversions. InTech; 2012.
  5. Wardhana, Surachmanto ES, Datau EA. The role of omega-3 fatty acids contained in olive oil on chronic inflammation. Acta Med Indones. 2011;43:138-143.
  6. Aboui MM, Eidi A, Mortazavi P. Study of effect of olive oil on re-epithelialization of epithelial tissue in excision wound healing model in rats. J Comp Pathobiol. 2016;13:1875-1884.
  7. Aldana PC, Cartron AM, Khachemoune A. Reappraising diabetic foot ulcers: a focus on mechanisms of ulceration and clinical evaluation.Int J Low Extrem Wounds. 2022;21:294-302. doi:10.1177/1534734620944514
  8. Aldana PC, Khachemoune A. Diabetic foot ulcers: appraising standard of care and reviewing new trends in management. Am J Clin Dermatol. 2020;21:255-264. doi:10.1007/s40257-019-00495-x
  9. Nasiri M, Fayazi S, Jahani S, et al. The effect of topical olive oil on the healing of foot ulcer in patients with type 2 diabetes: a double-blind randomized clinical trial study in Iran. J Diabetes Metab Disord. 2015;14:38. doi:10.1186/S40200-015-0167-9
  10. Karimi Z, Behnammoghadam M, Rafiei H, et al. Impact of olive oil and honey on healing of diabetic foot: a randomized controlled trial. Clin Cosmet Investig Dermatol. 2019;12:347-354. doi:10.2147/CCID.S198577
  11. Haghighian HK, Koushan Y, Asgharzadeh A. Treatment of diabetic foot ulcer with propolis and olive oil: a case report. Knowl Health. 2012;6:35-38.
  12. Zahmatkesh M, Rashidi M. Case report of diabetic foot ulcer with topical honey and olive oil. J Med Plants. 2008;8:36-41.
  13. Cicerale S, Lucas LJ, Keast RS. Antimicrobial, antioxidant and anti-inflammatory phenolic activities in extra virgin olive oil. Curr Opin Biotechnol. 2012;23:129-135. doi:10.1016/J.COPBIO.2011.09.006
  14. Miraj S, Pourafzali S, Ahmadabadi ZV, et al. Effect of olive oil in preventing the development of pressure ulcer grade one in intensive care unit patients. Int J Prev Med. 2020;11:23. doi:10.4103/IJPVM.IJPVM_545_18
  15. Díaz‐Valenzuela A, García‐Fernández FP, Carmona Fernández P, et al. Effectiveness and safety of olive oil preparation for topical use in pressure ulcer prevention: multicentre, controlled, randomised, and double‐blinded clinical trial. Int Wound J. 2019;16:1314-1322. doi:10.1111/IWJ.13191
  16. Carroli G, Mignini L. Episiotomy for vaginal birth. Cochrane Database Syst Rev. 2009;CD000081. doi:10.1002/14651858.CD000081.PUB2
  17. Amani R, Kariman N, Mojab F, et al. Comparison of the effects of cold compress with gel packs and topical olive oil on episiotomy wound healing. J Babol Univ Med Sci. 2015;17:7-12. doi:10.22088/JBUMS.17.11.7
  18. Behmanesh F, Aghamohammadi A, Zeinalzadeh M, et al. Effects of olive oil sitz bath on improvement of perineal injury after delivery. Koomesh. 2013;14:309-315.
  19. Vitsos A, Tsagarousianos C, Vergos O, et al. Efficacy of a Ceratothoa oestroides olive oil extract in patients with chronic ulcers: a pilot study. Int J Low Extrem Wounds. 2019;18:309-316. doi:10.1177/1534734619856143
Issue
Cutis - 113(6)
Issue
Cutis - 113(6)
Page Number
260-263
Page Number
260-263
Publications
Publications
Topics
Article Type
Display Headline
Olive Oil Shows Promise for Wound Healing of Ulcers
Display Headline
Olive Oil Shows Promise for Wound Healing of Ulcers
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>Malik olive</fileName> <TBEID>0C02F805.SIG</TBEID> <TBUniqueIdentifier>NJ_0C02F805</TBUniqueIdentifier> <newsOrJournal>Journal</newsOrJournal> <publisherName>Frontline Medical Communications Inc.</publisherName> <storyname>Malik_olive</storyname> <articleType>1</articleType> <TBLocation>Copyfitting-CT</TBLocation> <QCDate/> <firstPublished>20240607T122237</firstPublished> <LastPublished>20240607T122237</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240607T122236</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>Sara Malik, MD</byline> <bylineText>Sara Malik, MD; Muhammad Taaha Hassan, BHSc; Amor Khachemoune, MD</bylineText> <bylineFull>Sara Malik, MD</bylineFull> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType/> <journalDocType/> <linkLabel/> <pageRange>260-263</pageRange> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:"> <name/> <rightsInfo> <copyrightHolder> <name/> </copyrightHolder> <copyrightNotice/> </rightsInfo> </provider> <abstract/> <metaDescription>Olive oil is obtained by mechanical extraction from the fruit of the Olea europaea tree, which is believed to have originated from ancient Iran and Turkestan, l</metaDescription> <articlePDF>301781</articlePDF> <teaserImage/> <title>Olive Oil Shows Promise for Wound Healing of Ulcers</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear>2024</pubPubdateYear> <pubPubdateMonth>June</pubPubdateMonth> <pubPubdateDay/> <pubVolume>113</pubVolume> <pubNumber>6</pubNumber> <wireChannels/> <primaryCMSID/> <CMSIDs> <CMSID>2161</CMSID> </CMSIDs> <keywords/> <seeAlsos/> <publications_g> <publicationData> <publicationCode>CT</publicationCode> <pubIssueName>June 2024</pubIssueName> <pubArticleType>Original Articles | 2161</pubArticleType> <pubTopics/> <pubCategories/> <pubSections/> <journalTitle>Cutis</journalTitle> <journalFullTitle>Cutis</journalFullTitle> <copyrightStatement>Copyright 2015 Frontline Medical Communications Inc., Parsippany, NJ, USA. All rights reserved.</copyrightStatement> </publicationData> </publications_g> <publications> <term canonical="true">12</term> </publications> <sections> <term canonical="true">49</term> </sections> <topics> <term canonical="true">313</term> </topics> <links> <link> <itemClass qcode="ninat:composite"/> <altRep contenttype="application/pdf">images/1800274c.pdf</altRep> <description role="drol:caption"/> <description role="drol:credit"/> </link> </links> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Olive Oil Shows Promise for Wound Healing of Ulcers</title> <deck/> </itemMeta> <itemContent> <p class="abstract">In proper skin healing, inflammation will stop once the harmful microbes are removed. However, an excess and prolongation of inflammation can result in delayed healing. Thus, interventions that can limit the amount of inflammation can help promote wound healing. The use of olive oil in wound therapy has been of great interest. Herein, we will review studies that investigated the use of olive oil on diabetic foot ulcers, pressure ulcers, perineal ulcers, and chronic ulcers.</p> <p>Olive oil is obtained by mechanical extraction from the fruit of the <i>Olea europaea</i> tree, which is believed to have originated from ancient Iran and Turkestan, later spreading to Anatolia, Syria, Palestine, and Israel. Mechanical extraction of the oil from the olive fruit involves pressure processing, centrifugation, and adhesion filtering.<sup>1</sup> Refining of olive oil is done via alkali refining or physical refining, with physical refining being useful in removing oxidation by-products and pro-oxidant metals. Olive oil is composed mainly of triacylglycerols, which are glycerol esters attached to various fatty acids, with the most common fatty acid being the monounsaturated oleic acid. Additional fatty acids include palmitic acid, linoleic acid, stearic acid, and palmitoleic acid.<sup>2</sup> Olive oil contains phenolic compounds, the main ones being oleuropein, hydroxytyrosol, and tyrosol. These phenolic compounds are proposed to be strong antioxidants and radical scavengers.<sup>3</sup></p> <p>Mediterranean countries are responsible for approximately 97% of the world’s olive cultivation.<sup>4</sup> Olive oil historically was used as lamp fuel, lubricant, body ointment, and later as a source of edible oil.<sup>1</sup> Recently, its potential uses in medicine have called for further exploration into other uses for olive oil.<br/><br/>The skin is the largest organ of the body and serves as a protective barrier against pathogens and harmful substances. Skin damage results in 3 main phases to aid in wound healing: inflammation, proliferation, and maturation. In proper skin healing, inflammation will stop once the harmful microbes are removed. However, an excess and prolongation of inflammation can result in delayed healing. Thus, interventions that can limit the amount of inflammation can help promote wound healing. Olive oil contains several anti-inflammatory molecules (compounds or chemicals), including phenolic compounds and omega-3 fatty acids.<sup>5</sup> Studies also have shown that olive oil can promote re-epithelialization in tissues.<sup>6</sup> Thus, use of olive oil in wound therapy has been of great interest. <br/><br/>This article will review studies that have investigated the use of olive oil for wound healing of diabetic foot ulcers, pressure ulcers, perineal ulcers, and chronic ulcers. To conduct a comprehensive scoping review of the literature on the effects of olive oil in wound healing, we utilized the resources of the Galter Health Sciences Library &amp; Learning Center (Chicago, Illinois). Our search strategy was structured to encompass a range of relevant databases accessible through the library, including PubMed, Embase, and Web of Science. We formulated our search terms to be broad yet specific to our topic, combining keywords such as <i>olive oil</i>, <i>wound healing</i>, <i>skin repair</i>, and <i>dermal therapy</i>. The inclusion criteria were set to filter studies conducted from January 2000 to December 2019, focusing on clinical trials, observational studies, and review articles. We limited our search to articles published in English, which yielded a preliminary set of articles that were then screened based on their titles and abstracts. Full-text versions of potentially relevant studies were retrieved and assessed for eligibility. We included studies that specifically evaluated the effects of olive oil in wound healing, excluding those that did not directly relate to our research question or had insufficient data. The data extraction from these studies was conducted using a standardized form, capturing study design, population, intervention details, outcomes, and key findings. The synthesis of these data provided a comprehensive overview of the current evidence on the topic, aiding in the identification of gaps in knowledge and directions for future research.</p> <h3>Diabetic Foot Ulcers </h3> <p>Foot ulcers are common in patients with diabetes mellitus and are associated with notable morbidity and mortality. Foot ulcers can clinically manifest in various forms but are classically described as lesions with a deep sinus in the feet. Patients with diabetic foot ulcers are at risk for infection, and severe forms of the ulcers require amputation.<sup>7,8</sup> Routine care of foot ulcers involves irrigation of the ulcer and surrounding area with normal saline solution daily, followed by a dressing with sterile gauze. Studies investigating the effect of olive oil on foot ulcers suggest that olive oil use for care and healing of foot ulcers is an area of interest. </p> <p>A double-blind, randomized clinical trial investigated the effects of topical olive oil on diabetic foot ulcers.<sup>9</sup> A total of 34 patients with foot ulcers of Wagner grades 1 (superficial ulcers that involved the skin but not underlying tissue) or 2 (deeper ulcers penetrating to the ligaments and muscles but not the bone) that had remained open and did not improve for more than 3 months were recruited. The patients were randomly assigned to receive topical olive oil and routine care (intervention group) or to receive routine care (control group). Patients who received olive oil had oil poured on their ulcers with gauze wrapped around the ulcer that was soaked with olive oil. The clinical characteristics of the diabetic ulcer (eg, site, grade, size, status of healing) were assessed. The study revealed that after 4 weeks, olive oil significantly decreased ulcer area (<i>P</i><span class="body">=</span>.01) and ulcer depth (<i>P</i><span class="body">=</span>.02) compared with the control. Furthermore, there was a significant difference (<i>P</i><span class="body">=</span>.003) in complete ulcer healing between the olive oil and control groups: 73.3% (11/15) of patients in the olive oil group had complete ulcer healing, whereas 13.3% (2/15) of patients in the control group had complete ulcer healing.<sup>9</sup> The positive effect of olive oil on the healing of diabetic foot ulcers encourages further investigation as a possible therapy for foot ulcers. <br/><br/>Another randomized controlled trial of 45 patients with diabetic foot ulcers of Wagner grades 1 or 2 investigated the effect of olive oil.<sup>10</sup> Patients were randomly assigned to 1 of 3 groups for 1 month: the olive oil group, the honey group, or the control group. Patients in the olive oil group had their wounds dressed using gauze with olive oil daily, the patients in the honey group had their wounds dressed using gauze with honey daily, and the control group had routine care consisting of irrigation with saline solution and dressing with a sterile gauze. This study calculated a wound healing score based on a predefined checklist for diabetic foot ulcers through 4 variables: wound grading, color, surrounding tissue status, and drainage. Each variable had a maximum score of 100, contributing to a total possible score of 400, which indicated complete healing. A score of 50 signified ­deterioration. Wound healing was categorized as follows: (1) complete healing is indicated by a total score of 400; (2) partial healing was indicated by an increase of at least 30 points from the initial score; (3) lack of healing occurred when there was no change or less than a 30-point increase from the initial score; and (4) aggravation was noted when the score decreased by at least 10 points from the initial assessment. The study revealed that olive oil and honey treatments resulted in an increase in mean score, which indicated better wound healing. Patients in the olive oil group had a mean score of 253.0 before the intervention and 330.5 after the intervention (<i>P</i><span class="body">&lt;</span>.0001); patients in the honey group had a mean score of 267.5 before the intervention and 371.5 after the intervention (<i>P</i><span class="body">&lt;</span>.0001).<sup>10</sup> <br/><br/>There also have been case reports on combined olive oil and honey in diabetic foot ulcer management. Haghighian et al<sup>11</sup> presented a case of a diabetic foot wound that healed completely within 2 weeks after the combined use of olive oil and honey wax. Zahmatkesh and Rashidi<sup>12</sup> observed the healing of a diabetic foot wound over a month with daily dressings of a mixture of heated honey and olive oil, resulting in granulation tissue formation within 5 days. Microvascular changes, such as capillary basement membrane thickening, pericyte degeneration, and impairment of vasodilation and constriction, may contribute to inflammation in blood vessels, which can delay the healing of diabetic foot ulcers.<sup>7</sup> Because olive oil and honey contain compounds that have antioxidative, antimicrobial, and anti-inflammatory properties, both may play a role in notably reducing inflammation and promoting the healing of foot ulcers.<sup>13</sup> </p> <h3>Pressure Ulcers </h3> <p>A pressure ulcer is a superficial skin injury that is caused by a prolonged period of pressure on the skin, in which the skin becomes red but there is no rupture. Prolonged periods of immobility resulting in a reduction or pause of blood supply are common causes of pressure ulcers.<sup>14</sup> Studies have suggested that topical olive oil may be effective in prevention of pressure ulcers and should be incorporated as part of standard-of-care measures.</p> <p>In a randomized, single-blind trial, 72 patients with the first stage of bedsore—which is a pressure ulcer—in the sacral, shoulder, heel, or other areas were randomly assigned to either the intervention or control group.<sup>14</sup> Patients in the intervention group had 15 mL of olive oil rubbed on the wound for 20 minutes daily and then washed with tepid water. The Pressure Ulcer Scale for Healing tool was utilized to assess the healing status of the pressure ulcer. This tool considers wound surface size, exudate rate, and tissue type to provide a score of 0 to 17 (0<span class="body">=</span>healed ulcer; 17<span class="body">=</span>progression of ulcer). The mean score (SD) was lower in the olive oil group at days 4 and 7 compared with the control group (day 4: 7.50 [2.823] vs 9.50 [1.732]; day 7: 5.44 [3.806] vs 8.83 [2.864])(<i>P</i><span class="body">&lt;</span>.001). Furthermore, between days 1 and 7, there was significant improvement in the olive oil group (mean difference, 3.56; <i>P</i><span class="body">&lt;</span>.001) but no significant change in the control group (mean difference, 0.75; <i>P</i><span class="body">=</span>.052).<sup>14</sup> The results indicate that patients in the olive oil group had a better ulcer healing status compared with patients in the control group. <br/><br/>In a noninferiority, randomized, double-blind clinical trial, olive oil was compared to a recommended skin care measure of hyperoxygenated fatty acids (HOFAs) for the prevention of pressure ulcers.<sup>15</sup> The study consisted of 571 residents from several nursing homes who were at risk for pressure ulcers. Either olive oil or HOFA was applied to areas at risk for pressure ulcers, with 2 sprays of 0.2 mL per spray to each area every 12 hours. The participants were followed up for 30 days or until a pressure ulcer developed. Researchers performed skin assessments; the Braden Scale was used to assess the risk for pressure ulcers. The incidence difference of pressure ulcers in the olive oil group and HOFA group did not exceed in the noninferiority margin of 7%. Furthermore, Kaplan-Meier survival curves for the time until pressure ulcer onset showed a nonsignificant difference between the 2 groups.<sup>15</sup> These findings suggest that olive oil is as effective as HOFA for the prevention of pressure ulcers. Although the mechanism of olive oil on prevention of pressure ulcers has not yet been determined, it has been suggested that anti-inflammatory compounds in olive oil, such as polyphenol and oleocanthal compounds, play an anti-inflammatory role. </p> <h3>Perineal Ulcers</h3> <p>Episiotomy is a surgical incision that is made to open the vagina during birth to aid in delivery of the baby. In contrast to spontaneous vaginal tears, an episiotomy allows for easier repair and healing of the laceration.<sup>16</sup> Studies were conducted to investigate the effect of olive oil on women with lacerations after an episiotomy. </p> <p>A total of 90 primigravid women who had undergone episiotomy were recruited and randomly assigned to 1 of 2 interventions: cold compression with gel packs for 20 minutes within 12 hours after delivery for up to 10 days, if necessary, or topical olive oil twice daily within 12 hours after delivery for up to 10 days.<sup>17</sup> Although there was no significant difference in the structural features of the wound, there was a significant difference in the redness severity. After 10 days, the mean REEDA (redness, edema, ecchymosis, discharge, and apposition) score (SD), which assesses tissue healing, was 0.47 (0.96) in patients who received cold compression with gel packs and 0.20 (0.50) in patients who received topical olive oil (<i>P</i><span class="body">=</span>.04).<sup>17</sup> This study suggests that there is the potential for olive oil to be used for wound healing after episiotomy. <br/><br/>A double-blind trial consisted of 60 women who had mediolateral episiotomy or perineal tear grades 1 and 2 who were randomly assigned to 1 of 2 groups for 10 days: olive oil sitz bath or distilled water sitz bath (control group). The results showed a significant difference in pain severity after 5 and 10 days (<i>P</i><span class="body">&lt;</span>.05), wound redness after 5 days (<i>P</i><span class="body">&lt;</span>.0001), and redness (<i>P</i><span class="body">&lt;</span>.000) and edema (<i>P</i><span class="body">&lt;</span>.05) 10 days after delivery.<sup>18</sup> This study encourages further investigation of the benefits of olive oil for care after an episiotomy. </p> <h3>Chronic Ulcers</h3> <p>Chronic ulcers are other persistent wounds that do not respond to standard treatments and pose a notable health burden. Their development is influenced by factors such as oxidative stress, microbial infections, and the body’s immune response. A case series was conducted to investigate the wound healing effects of olive oil on chronic ulcers.<sup>19</sup> Fourteen patients who were diagnosed with 1 or more chronic skin ulcers that had not healed with conventional treatment, such as cleansing, debridement, or infection control, were recruited. The mean (SD) of the patients’ <hl name="17866"/>Bates-Jensen Wound Assessment Tool score was 39.05 (4.23), indicating that these ulcers had been challenging to treat. In addition, the wounds in this study were found to be infected with bacteria. An ointment consisting of <i>Ceratothoa oestroides</i> olive oil extract was applied to the wounds after they were cleansed. The results showed that Bates-Jensen Wound Assessment Tool scores decreased by 14.7% to 67.5% (mean, 36%; median, 38%) after 3 months of treatment. Furthermore, 5 patients had a completely healed wound, indicating that <i>C oestroides</i> olive oil extract can regenerate chronic ulcers that do not respond to antibacterial agents.<sup>19</sup> These results encourage further investigation of the role of <i>C oestroides</i> olive oil extract on healing properties and microbial control. </p> <h3>Final Thoughts</h3> <p>This review illuminated several key aspects of research on the role of olive oil in wound healing. Although the studies included in this review offer valuable insights, it is essential to acknowledge the variability in the quality of data presented. Several studies demonstrated robust methodology with clear definitions of outcomes and controlled conditions, providing high-quality evidence. However, other studies exhibited limitations, including small sample sizes and potential biases, which may affect the generalizability of the findings. Despite these limitations, the collective evidence suggests potential for olive oil in wound healing, warranting further investigation. Future research should aim for more standardized methodologies and larger, more diverse patient cohorts to validate these findings and explore the mechanisms underlying the therapeutic effects of olive oil.</p> <h2>References </h2> <p class="reference"> 1. Emmons EW, Fedeli E, Firestone D. Olive oil introduction and history. In: Hui YH, ed. <i>Bailey’s Industrial Oil &amp; Fat Products, Vol. 2. Edible Oil and Fat Products: Edible Oils</i>. 5th ed. John Wiley &amp; Sons, Ltd; 241-269.<br/><br/> 2. Gorzynik-Debicka M, Przychodzen P, Cappello F, et al. Potential health benefits of olive oil and plant polyphenols. <i>Int J Mol Sci</i>. 2018;19:686. doi:10.3390/IJMS19030686<br/><br/> 3. Tuck KL, Hayball PJ. Major phenolic compounds in olive oil: metabolism and health effects. <i>J Nutr Biochem</i>. 2002;13:636-644. doi:10.1016/S0955-2863(02)00229-2<br/><br/> 4. Rabiei Z, Enferadi ST. Traceability of origin and authenticity of olive oil. In: Boskou D, ed. <i>Olive Oil: Constituents, Quality, Health Properties and Bioconversions</i>. InTech; 2012. <br/><br/> 5. Wardhana, Surachmanto ES, Datau EA. The role of omega-3 fatty acids contained in olive oil on chronic inflammation. <i>Acta Med Indones</i>. 2011;43:138-143.<br/><br/> 6. Aboui MM, Eidi A, Mortazavi P. Study of effect of olive oil on re-epithelialization of epithelial tissue in excision wound healing model in rats. <i>J Comp Pathobiol</i>. 2016;13:1875-1884.</p> <p class="reference"> 7. Aldana PC, Cartron AM, Khachemoune A. Reappraising diabetic foot ulcers: a focus on mechanisms of ulceration and clinical evaluation.<i>Int J Low Extrem Wounds</i>. 2022;21:294-302. doi:10.1177/1534734620944514 <br/><br/> 8. Aldana PC, Khachemoune A. Diabetic foot ulcers: appraising standard of care and reviewing new trends in management. <i>Am J Clin Dermatol</i>. 2020;21:255-264. doi:10.1007/s40257-019-00495-x<br/><br/> 9. Nasiri M, Fayazi S, Jahani S, et al. The effect of topical olive oil on the healing of foot ulcer in patients with type 2 diabetes: a double-blind randomized clinical trial study in Iran. <i>J Diabetes Metab Disord</i>. 2015;14:38. doi:10.1186/S40200-015-0167-9<br/><br/>10. Karimi Z, Behnammoghadam M, Rafiei H, et al. Impact of olive oil and honey on healing of diabetic foot: a randomized controlled trial. <i>Clin Cosmet Investig Dermatol</i>. 2019;12:347-354. doi:10.2147/CCID.S198577<br/><br/>11. Haghighian HK, Koushan Y, Asgharzadeh A. Treatment of diabetic foot ulcer with propolis and olive oil: a case report. <i>Knowl Health</i>. 2012;6:35-38.<br/><br/>12. Zahmatkesh M, Rashidi M. Case report of diabetic foot ulcer with topical honey and olive oil. <i>J Med Plants</i>. 2008;8:36-41.<br/><br/>13. Cicerale S, Lucas LJ, Keast RS. Antimicrobial, antioxidant and anti-inflammatory phenolic activities in extra virgin olive oil. <i>Curr Opin Biotechnol</i>. 2012;23:129-135. doi:10.1016/J.COPBIO.2011.09.006<br/><br/>14. Miraj S, Pourafzali S, Ahmadabadi ZV, et al. Effect of olive oil in preventing the development of pressure ulcer grade one in intensive care unit patients. <i>Int J Prev Med</i>. 2020;11:23. doi:10.4103/IJPVM.IJPVM_545_18<br/><br/>15. Díaz‐Valenzuela A, García‐Fernández FP, Carmona Fernández P, et al. Effectiveness and safety of olive oil preparation for topical use in pressure ulcer prevention: multicentre, controlled, randomised, and double‐blinded clinical trial. <i>Int Wound J</i>. 2019;16:1314-1322. doi:10.1111/IWJ.13191<br/><br/>16. Carroli G, Mignini L. Episiotomy for vaginal birth. <i>Cochrane Database Syst Rev</i>. 2009;CD000081. doi:10.1002/14651858.CD000081.PUB2<br/><br/>17. Amani R, Kariman N, Mojab F, et al. Comparison of the effects of cold compress with gel packs and topical olive oil on episiotomy wound healing. <i>J Babol Univ Med Sci</i>. 2015;17:7-12. doi:10.22088/JBUMS.17.11.7<br/><br/>18. Behmanesh F, Aghamohammadi A, Zeinalzadeh M, et al. Effects of olive oil sitz bath on improvement of perineal injury after delivery. <i>Koomesh</i>. 2013;14:309-315. <br/><br/>19. Vitsos A, Tsagarousianos C, Vergos O, et al. Efficacy of a <i>Ceratothoa </i><i>oestroides </i>olive oil extract in patients with chronic ulcers: a pilot study. <i>Int </i><i>J Low Extrem Wounds</i>. 2019;18:309-316. doi:10.1177/1534734619856143</p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>bio</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="disclosure">Dr. Malik and Muhammad Taaha Hassan are from the Northwestern University Feinberg School of Medicine, Chicago, Illinois. Dr. Khachemoune is from Veterans Affairs Medical Center, Brooklyn, New York, and SUNY Downstate Medical Center, Brooklyn, New York.</p> <p class="disclosure">The authors report no conflict of interest.<br/><br/>Correspondence: Amor Khachemoune, MD, SUNY Downstate, Veterans Affairs Medical Center, 800 Poly Pl, Brooklyn, NY 11209(amorkh@gmail.com).<br/><br/><em>Cutis.</em> 2024 June;113(6):260-263. doi:10.12788/cutis.1035</p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>in</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="insidehead">Practice <strong>Points</strong></p> <ul class="insidebody"> <li>Interventions that effectively reduce excessive and prolonged inflammation can help promote timely wound healing. Consider integrating anti-inflammatory treatments into wound care protocols to enhance healing outcomes.</li> <li>Utilization of olive oil in wound therapy, particularly for conditions such as diabetic foot ulcers, pressure ulcers, perineal ulcers, and chronic ulcers, has shown promise for promoting healing.</li> </ul> </itemContent> </newsItem> </itemSet></root>
Inside the Article

Practice Points

  • Interventions that effectively reduce excessive and prolonged inflammation can help promote timely wound healing. Consider integrating anti-inflammatory treatments into wound care protocols to enhance healing outcomes.
  • Utilization of olive oil in wound therapy, particularly for conditions such as diabetic foot ulcers, pressure ulcers, perineal ulcers, and chronic ulcers, has shown promise for promoting healing.
  • Regularly review and incorporate findings from recent studies on the use of olive oil and other novel interventions in wound therapy to ensure the application of the most current and effective treatment strategies.
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
Image
Disable zoom
Off

Analysis Finds Minority of Chronic Wounds Treated by Dermatologists

Article Type
Changed
Mon, 06/03/2024 - 15:14

Between 2011 and 2019, chronic cutaneous wounds accounted for about one third of all health care visits for cutaneous wounds, and the most common diagnoses were open wounds of the thumb without nail damage. However, fewer than 8% of chronic wounds were managed by dermatologists during this time.

Those are among key findings from an analysis of National Ambulatory Medical Care Survey (NAMCS) data between 2011 and 2019 presented as a late-breaking abstract at the annual meeting of the Society for Investigative Dermatology. “Cutaneous wounds were estimated to account for 28.1 to 96.1 billion dollars in US health care costs in 2014,” one of the study authors, Rithi Chandy, MD, MS, a research fellow at the Center for Dermatology Research at Wake Forest University School of Medicine, Winston-Salem, North Carolina, said in an interview following the meeting. “By examining national trends in patient visits and treatment, we may be able to better inform health care utilization for cutaneous wounds.”

bo
Dr. Rithi Chandy


Dr. Chandy and colleagues analyzed de-identified patient data from the 2011 to 2019 NAMCS for acute and chronic wound diagnoses, medications prescribed, and physician specialty categories. During the time studied, 5.76 billion patient visits were made, including 45.1 million visits for cutaneous wounds. Of these, the most common diagnoses were open wounds of the thumb without nail damage (7.96%), the lower leg (5.75%), nonpressure chronic ulcers of other parts of the foot (5.08%), and open wounds of the ear (5%).

Among all visits for cutaneous wounds, about one third were chronic cutaneous wounds, with the following descriptions: “Nonpressure chronic ulcer of other part of foot” (17.8%); “nonpressure chronic ulcer of skin, not elsewhere classified” (9.38%); and “ulcer of lower limbs, excluding decubitus, unspecified” (8.72%). “The frequency of patient visits per year during the study period remained stable for both acute and chronic wounds,” Dr. Chandy said. The number of visits for which antimicrobials were used was stable over time for both acute and chronic cutaneous wounds, with the exception of increased use of antivirals for chronic cutaneous wounds, he added.

Specifically, prescriptions were issued in 156 million visits over the time studied, most commonly cephalexin (4.22%), topical silver sulfadiazine (1.59%), topical mupirocin (1.12%), and miscellaneous antibiotics (1.18%).

“Our data shows that topical mupirocin is the most commonly used topical antimicrobial for cutaneous wounds,” Dr. Chandy said. “However, there are reports of emerging bacterial resistance to mupirocin. Our data can inform ongoing efforts to promote antimicrobial stewardship and drug development to provide alternative options that are less likely to induce antimicrobial resistance.”

In findings limited to specialty-specific NAMCS data available from 2011 and from 2013 to 2016, dermatologists managed 3.85% of overall cutaneous wounds, 2.35% of acute wounds, and 7.39% of chronic wounds. By contrast, Dr. Chandy said, 21.1% of chronic wounds were managed by general/family practice physicians, 20.7% by internists, 6.84% by general surgeons, and 5.65% by orthopedic surgeons.

“As dermatologists are experts in the structure and function of the skin and are trained to manage cutaneous disorders including wound healing, we [believe that] dermatologists are equipped with the skill set” for managing wounds, especially for chronic ulcers, he said. The decline in dermatologists who specialize in wound care, he added, “underscores the need for structured dermatology fellowship programs to prepare next-generation dermatologists to address this shortage and ensure dermatology leadership in cutaneous wound healing.”

Dr. Chandy acknowledged certain limitations of the study, including the potential for misclassification of diagnoses or medications prescribed and the fact that the NAMCS database is unable to provide insight into individual patient experiences such as continual cutaneous wound management for the same patient over time.

In the opinion of Shari R. Lipner, MD, PhD, associate professor of clinical dermatology and director of the Nail Division at Weill Cornell Medicine, New York, who was asked to comment on the study, the most interesting finding was that dermatologists cared for a small minority of patients with cutaneous wounds. “It would be interesting to know whether this is due to dermatologist shortages or knowledge gaps on the part of primary care physicians or patients that dermatologists are trained to care for wounds,” Dr. Lipner told this news organization. Other unanswered questions, she noted, “are patient demographics, geographic locations, and comorbidities.”

One of the study authors, Steven R. Feldman, MD, PhD, professor of dermatology at Wake Forest University, disclosed that he has received research, speaking and/or consulting support from numerous pharmaceutical companies. No other authors reported having relevant disclosures. Dr. Lipner reported having no disclosures.

A version of this article appeared on Medscape.com .

Publications
Topics
Sections

Between 2011 and 2019, chronic cutaneous wounds accounted for about one third of all health care visits for cutaneous wounds, and the most common diagnoses were open wounds of the thumb without nail damage. However, fewer than 8% of chronic wounds were managed by dermatologists during this time.

Those are among key findings from an analysis of National Ambulatory Medical Care Survey (NAMCS) data between 2011 and 2019 presented as a late-breaking abstract at the annual meeting of the Society for Investigative Dermatology. “Cutaneous wounds were estimated to account for 28.1 to 96.1 billion dollars in US health care costs in 2014,” one of the study authors, Rithi Chandy, MD, MS, a research fellow at the Center for Dermatology Research at Wake Forest University School of Medicine, Winston-Salem, North Carolina, said in an interview following the meeting. “By examining national trends in patient visits and treatment, we may be able to better inform health care utilization for cutaneous wounds.”

bo
Dr. Rithi Chandy


Dr. Chandy and colleagues analyzed de-identified patient data from the 2011 to 2019 NAMCS for acute and chronic wound diagnoses, medications prescribed, and physician specialty categories. During the time studied, 5.76 billion patient visits were made, including 45.1 million visits for cutaneous wounds. Of these, the most common diagnoses were open wounds of the thumb without nail damage (7.96%), the lower leg (5.75%), nonpressure chronic ulcers of other parts of the foot (5.08%), and open wounds of the ear (5%).

Among all visits for cutaneous wounds, about one third were chronic cutaneous wounds, with the following descriptions: “Nonpressure chronic ulcer of other part of foot” (17.8%); “nonpressure chronic ulcer of skin, not elsewhere classified” (9.38%); and “ulcer of lower limbs, excluding decubitus, unspecified” (8.72%). “The frequency of patient visits per year during the study period remained stable for both acute and chronic wounds,” Dr. Chandy said. The number of visits for which antimicrobials were used was stable over time for both acute and chronic cutaneous wounds, with the exception of increased use of antivirals for chronic cutaneous wounds, he added.

Specifically, prescriptions were issued in 156 million visits over the time studied, most commonly cephalexin (4.22%), topical silver sulfadiazine (1.59%), topical mupirocin (1.12%), and miscellaneous antibiotics (1.18%).

“Our data shows that topical mupirocin is the most commonly used topical antimicrobial for cutaneous wounds,” Dr. Chandy said. “However, there are reports of emerging bacterial resistance to mupirocin. Our data can inform ongoing efforts to promote antimicrobial stewardship and drug development to provide alternative options that are less likely to induce antimicrobial resistance.”

In findings limited to specialty-specific NAMCS data available from 2011 and from 2013 to 2016, dermatologists managed 3.85% of overall cutaneous wounds, 2.35% of acute wounds, and 7.39% of chronic wounds. By contrast, Dr. Chandy said, 21.1% of chronic wounds were managed by general/family practice physicians, 20.7% by internists, 6.84% by general surgeons, and 5.65% by orthopedic surgeons.

“As dermatologists are experts in the structure and function of the skin and are trained to manage cutaneous disorders including wound healing, we [believe that] dermatologists are equipped with the skill set” for managing wounds, especially for chronic ulcers, he said. The decline in dermatologists who specialize in wound care, he added, “underscores the need for structured dermatology fellowship programs to prepare next-generation dermatologists to address this shortage and ensure dermatology leadership in cutaneous wound healing.”

Dr. Chandy acknowledged certain limitations of the study, including the potential for misclassification of diagnoses or medications prescribed and the fact that the NAMCS database is unable to provide insight into individual patient experiences such as continual cutaneous wound management for the same patient over time.

In the opinion of Shari R. Lipner, MD, PhD, associate professor of clinical dermatology and director of the Nail Division at Weill Cornell Medicine, New York, who was asked to comment on the study, the most interesting finding was that dermatologists cared for a small minority of patients with cutaneous wounds. “It would be interesting to know whether this is due to dermatologist shortages or knowledge gaps on the part of primary care physicians or patients that dermatologists are trained to care for wounds,” Dr. Lipner told this news organization. Other unanswered questions, she noted, “are patient demographics, geographic locations, and comorbidities.”

One of the study authors, Steven R. Feldman, MD, PhD, professor of dermatology at Wake Forest University, disclosed that he has received research, speaking and/or consulting support from numerous pharmaceutical companies. No other authors reported having relevant disclosures. Dr. Lipner reported having no disclosures.

A version of this article appeared on Medscape.com .

Between 2011 and 2019, chronic cutaneous wounds accounted for about one third of all health care visits for cutaneous wounds, and the most common diagnoses were open wounds of the thumb without nail damage. However, fewer than 8% of chronic wounds were managed by dermatologists during this time.

Those are among key findings from an analysis of National Ambulatory Medical Care Survey (NAMCS) data between 2011 and 2019 presented as a late-breaking abstract at the annual meeting of the Society for Investigative Dermatology. “Cutaneous wounds were estimated to account for 28.1 to 96.1 billion dollars in US health care costs in 2014,” one of the study authors, Rithi Chandy, MD, MS, a research fellow at the Center for Dermatology Research at Wake Forest University School of Medicine, Winston-Salem, North Carolina, said in an interview following the meeting. “By examining national trends in patient visits and treatment, we may be able to better inform health care utilization for cutaneous wounds.”

bo
Dr. Rithi Chandy


Dr. Chandy and colleagues analyzed de-identified patient data from the 2011 to 2019 NAMCS for acute and chronic wound diagnoses, medications prescribed, and physician specialty categories. During the time studied, 5.76 billion patient visits were made, including 45.1 million visits for cutaneous wounds. Of these, the most common diagnoses were open wounds of the thumb without nail damage (7.96%), the lower leg (5.75%), nonpressure chronic ulcers of other parts of the foot (5.08%), and open wounds of the ear (5%).

Among all visits for cutaneous wounds, about one third were chronic cutaneous wounds, with the following descriptions: “Nonpressure chronic ulcer of other part of foot” (17.8%); “nonpressure chronic ulcer of skin, not elsewhere classified” (9.38%); and “ulcer of lower limbs, excluding decubitus, unspecified” (8.72%). “The frequency of patient visits per year during the study period remained stable for both acute and chronic wounds,” Dr. Chandy said. The number of visits for which antimicrobials were used was stable over time for both acute and chronic cutaneous wounds, with the exception of increased use of antivirals for chronic cutaneous wounds, he added.

Specifically, prescriptions were issued in 156 million visits over the time studied, most commonly cephalexin (4.22%), topical silver sulfadiazine (1.59%), topical mupirocin (1.12%), and miscellaneous antibiotics (1.18%).

“Our data shows that topical mupirocin is the most commonly used topical antimicrobial for cutaneous wounds,” Dr. Chandy said. “However, there are reports of emerging bacterial resistance to mupirocin. Our data can inform ongoing efforts to promote antimicrobial stewardship and drug development to provide alternative options that are less likely to induce antimicrobial resistance.”

In findings limited to specialty-specific NAMCS data available from 2011 and from 2013 to 2016, dermatologists managed 3.85% of overall cutaneous wounds, 2.35% of acute wounds, and 7.39% of chronic wounds. By contrast, Dr. Chandy said, 21.1% of chronic wounds were managed by general/family practice physicians, 20.7% by internists, 6.84% by general surgeons, and 5.65% by orthopedic surgeons.

“As dermatologists are experts in the structure and function of the skin and are trained to manage cutaneous disorders including wound healing, we [believe that] dermatologists are equipped with the skill set” for managing wounds, especially for chronic ulcers, he said. The decline in dermatologists who specialize in wound care, he added, “underscores the need for structured dermatology fellowship programs to prepare next-generation dermatologists to address this shortage and ensure dermatology leadership in cutaneous wound healing.”

Dr. Chandy acknowledged certain limitations of the study, including the potential for misclassification of diagnoses or medications prescribed and the fact that the NAMCS database is unable to provide insight into individual patient experiences such as continual cutaneous wound management for the same patient over time.

In the opinion of Shari R. Lipner, MD, PhD, associate professor of clinical dermatology and director of the Nail Division at Weill Cornell Medicine, New York, who was asked to comment on the study, the most interesting finding was that dermatologists cared for a small minority of patients with cutaneous wounds. “It would be interesting to know whether this is due to dermatologist shortages or knowledge gaps on the part of primary care physicians or patients that dermatologists are trained to care for wounds,” Dr. Lipner told this news organization. Other unanswered questions, she noted, “are patient demographics, geographic locations, and comorbidities.”

One of the study authors, Steven R. Feldman, MD, PhD, professor of dermatology at Wake Forest University, disclosed that he has received research, speaking and/or consulting support from numerous pharmaceutical companies. No other authors reported having relevant disclosures. Dr. Lipner reported having no disclosures.

A version of this article appeared on Medscape.com .

Publications
Publications
Topics
Article Type
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>168261</fileName> <TBEID>0C0505D9.SIG</TBEID> <TBUniqueIdentifier>MD_0C0505D9</TBUniqueIdentifier> <newsOrJournal>News</newsOrJournal> <publisherName>Frontline Medical Communications</publisherName> <storyname/> <articleType>2</articleType> <TBLocation>QC Done-All Pubs</TBLocation> <QCDate>20240603T150242</QCDate> <firstPublished>20240603T151018</firstPublished> <LastPublished>20240603T151018</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240603T151018</CMSDate> <articleSource>FROM SID 2024</articleSource> <facebookInfo/> <meetingNumber>3047-24</meetingNumber> <byline>Doug Brunk</byline> <bylineText>DOUG BRUNK</bylineText> <bylineFull>DOUG BRUNK</bylineFull> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType/> <journalDocType/> <linkLabel/> <pageRange/> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:imng"> <name>IMNG Medical Media</name> <rightsInfo> <copyrightHolder> <name>Frontline Medical News</name> </copyrightHolder> <copyrightNotice>Copyright (c) 2015 Frontline Medical News, a Frontline Medical Communications Inc. company. All rights reserved. This material may not be published, broadcast, copied, or otherwise reproduced or distributed without the prior written permission of Frontline Medical Communications Inc.</copyrightNotice> </rightsInfo> </provider> <abstract/> <metaDescription>Between 2011 and 2019, chronic cutaneous wounds accounted for about one third of all health care visits for cutaneous wounds, and the most common diagnoses were</metaDescription> <articlePDF/> <teaserImage>301720</teaserImage> <teaser>The study also looked at costs, and what antimicrobials were used to treat wounds between 2011 and 2019.</teaser> <title>Analysis Finds Minority of Chronic Wounds Treated by Dermatologists</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear/> <pubPubdateMonth/> <pubPubdateDay/> <pubVolume/> <pubNumber/> <wireChannels/> <primaryCMSID/> <CMSIDs/> <keywords/> <seeAlsos/> <publications_g> <publicationData> <publicationCode>skin</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> <publicationData> <publicationCode>fp</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> <publicationData> <publicationCode>im</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> </publications_g> <publications> <term>13</term> <term canonical="true">15</term> <term>21</term> </publications> <sections> <term>53</term> <term canonical="true">39313</term> </sections> <topics> <term canonical="true">313</term> <term>203</term> <term>234</term> </topics> <links> <link> <itemClass qcode="ninat:picture"/> <altRep contenttype="image/jpeg">images/240129cc.jpg</altRep> <description role="drol:caption">Dr. Rithi Chandy</description> <description role="drol:credit"/> </link> </links> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Analysis Finds Minority of Chronic Wounds Treated by Dermatologists</title> <deck/> </itemMeta> <itemContent> <p><span class="tag metaDescription">Between 2011 and 2019, chronic cutaneous wounds accounted for about one third of all health care visits for cutaneous wounds, and the most common diagnoses were open wounds of the thumb without nail damage</span>. However, fewer than 8% of chronic wounds were managed by dermatologists during this time.</p> <p>Those are among key findings from an analysis of National Ambulatory Medical Care Survey (<span class="Hyperlink"><a href="https://www.cdc.gov/nchs/ahcd/index.htm">NAMCS</a></span>) data between 2011 and 2019 presented as a late-breaking abstract at the <span class="Hyperlink"><a href="https://www.sidannualmeeting.org/">annual meeting of the Society for Investigative Dermatology</a></span>. “Cutaneous wounds were estimated to account for 28.1 to 96.1 billion dollars in US health care costs in 2014,” one of the study authors, Rithi Chandy, MD, MS, a research fellow at the <span class="Hyperlink"><a href="https://school.wakehealth.edu/departments/dermatology/center-for-dermatology-research">Center for Dermatology Research</a></span> at Wake Forest University School of Medicine, Winston-Salem, North Carolina, said in an interview following the meeting. “By examining national trends in patient visits and treatment, we may be able to better inform health care utilization for cutaneous wounds.”[[{"fid":"301720","view_mode":"medstat_image_flush_left","fields":{"format":"medstat_image_flush_left","field_file_image_alt_text[und][0][value]":"Rithi Chandy, MD, research fellow at the Center for Dermatology Research at Wake Forest University School of Medicine, Winston-Salem, North Carolina","field_file_image_credit[und][0][value]":"","field_file_image_caption[und][0][value]":"Dr. Rithi Chandy"},"type":"media","attributes":{"class":"media-element file-medstat_image_flush_left"}}]]<br/><br/>Dr. Chandy and colleagues analyzed de-identified patient data from the 2011 to 2019 NAMCS for acute and chronic wound diagnoses, medications prescribed, and physician specialty categories. During the time studied, 5.76 billion patient visits were made, including 45.1 million visits for cutaneous wounds. Of these, the most common diagnoses were open wounds of the thumb without nail damage (7.96%), the lower leg (5.75%), nonpressure chronic ulcers of other parts of the foot (5.08%), and open wounds of the ear (5%).<br/><br/>Among all visits for cutaneous wounds, about one third were chronic cutaneous wounds, with the following descriptions: “Nonpressure chronic ulcer of other part of foot” (17.8%); “nonpressure chronic ulcer of skin, not elsewhere classified” (9.38%); and “ulcer of lower limbs, excluding decubitus, unspecified” (8.72%). “The frequency of patient visits per year during the study period remained stable for both acute and chronic wounds,” Dr. Chandy said. The number of visits for which antimicrobials were used was stable over time for both acute and chronic cutaneous wounds, with the exception of increased use of antivirals for chronic cutaneous wounds, he added.<br/><br/>Specifically, prescriptions were issued in 156 million visits over the time studied, most commonly cephalexin (4.22%), topical silver sulfadiazine (1.59%), <span class="Hyperlink">topical mupirocin</span> (1.12%), and miscellaneous antibiotics (1.18%).<br/><br/>“Our data shows that topical mupirocin is the most commonly used topical antimicrobial for cutaneous wounds,” Dr. Chandy said. “However, there are reports of emerging bacterial resistance to mupirocin. Our data can inform ongoing efforts to promote antimicrobial stewardship and drug development to provide alternative options that are less likely to induce antimicrobial resistance.”<br/><br/>In findings limited to specialty-specific NAMCS data available from 2011 and from 2013 to 2016, dermatologists managed 3.85% of overall cutaneous wounds, 2.35% of acute wounds, and 7.39% of chronic wounds. By contrast, Dr. Chandy said, 21.1% of chronic wounds were managed by general/family practice physicians, 20.7% by internists, 6.84% by general surgeons, and 5.65% by orthopedic surgeons.<br/><br/>“As dermatologists are experts in the structure and function of the skin and are trained to manage cutaneous disorders including wound healing, we [believe that] dermatologists are equipped with the skill set” for managing wounds, especially for chronic ulcers, he said. The decline in dermatologists who specialize in wound care, he added, “underscores the need for structured dermatology fellowship programs to prepare next-generation dermatologists to address this shortage and ensure dermatology leadership in cutaneous wound healing.”<br/><br/>Dr. Chandy acknowledged certain limitations of the study, including the potential for misclassification of diagnoses or medications prescribed and the fact that the NAMCS database is unable to provide insight into individual patient experiences such as continual cutaneous wound management for the same patient over time.<br/><br/>In the opinion of <span class="Hyperlink">Shari R. Lipner, MD, PhD</span>, associate professor of clinical dermatology and director of the Nail Division at Weill Cornell Medicine, New York, who was asked to comment on the study, the most interesting finding was that dermatologists cared for a small minority of patients with cutaneous wounds. “It would be interesting to know whether this is due to dermatologist shortages or knowledge gaps on the part of primary care physicians or patients that dermatologists are trained to care for wounds,” Dr. Lipner told this news organization. Other unanswered questions, she noted, “are patient demographics, geographic locations, and comorbidities.”<br/><br/>One of the study authors, Steven R. Feldman, MD, PhD, professor of dermatology at Wake Forest University, disclosed that he has received research, speaking and/or consulting support from numerous pharmaceutical companies. No other authors reported having relevant disclosures. Dr. Lipner reported having no disclosures.<span class="end"/></p> <p> <em> <span class="Emphasis">A version of this article appeared on </span> <span class="Hyperlink"> <a href="https://www.medscape.com/viewarticle/dermatologists-manage-minority-patients-chronic-wounds-study-2024a1000a5m">Medscape.com</a> </span> <span class="Emphasis">.</span> </em> </p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>teaser</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> </itemContent> </newsItem> </itemSet></root>
Article Source

FROM SID 2024

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

Gene Therapy for Dystrophic EB: Extension Study Results Reported

Article Type
Changed
Fri, 05/03/2024 - 14:23

In an extension study of patients with dystrophic epidermolysis bullosa (DEB) treated with the topical gene therapy beremagene geperpavec, wound closure rates and adverse events were similar to those seen in the phase 3 study and no new safety signals were identified.

The results were presented by Amy S. Paller, MD, during a late-breaking session at the annual meeting of the American Academy of Dermatology.

In May 2023, beremagene geperpavec, marketed as Vyjuvek (formerly known as B-VEC) was approved by the US Food and Drug Administration (FDA) for the treatment of wounds in patients 6 months of age and older with DEB, a rare genetic blistering disorder caused by COL7A1 gene variants. The therapy uses a nonreplicating herpes simplex virus type 1 (HSV-1) vector to deliver the COL7A1 gene directly to skin cells, restoring the COL7 protein fibrils that stabilize skin structure. It is designed to be used repetitively, to heal a single wound, or on more than one wound.

In the pivotal study of patients with DEB, the gene therapy, delivered in a topical gel, was administered once a week for 6 months to one wound and placebo was applied to another wound for each participant. The proportion of wounds treated with beremagene geperpavec that healed was significantly higher than among placebo-treated wounds at 3 and 6 months (68% vs. 23% at 3 months, P = .003) and 65% vs. 26% at 6 months (P = .012), with no serious adverse events related to treatment.

The prospective, open label, uncontrolled extension study included 24 patients from the phase 3 study and 23 treatment-naive patients from five US sites. Their mean age was 16 years (range, 6 months to 46 years).

Of the 47 patients, 29 (62%) were on treatment for more than 1 year (the longest was about 2 years), and the mean duration of treatment was 475 days; 5 patients withdrew from the study for reasons not related to treatment.

Their types of adverse events (AEs) were similar to those seen in the phase 3 study and were consistent with what would be expected in patients with DEB, said Dr. Paller, professor and chair of dermatology, Northwestern University, Chicago. One patient experienced two wound hemorrhages that were possibly related to treatment, but there were no treatment-related AEs, no deaths or treatment discontinuations because of an AE, and no serious AEs thought to be related to treatment.

Wounds that were evaluated in the phase 3 study showed “a high durability of closure with continued treatment,” according to Dr. Paller. There were enough data on 19 of the 24 patients who had been in the phase 3 trial to evaluate wound closure, defined as “complete wound closure based on comparison to the exact wound area selected at baseline” at the beginning of the phase 3 study.

In the extension study, wound closure rates were almost 90% at baseline, 84.2% at 3 months, 61.1% at 6 months, 82.4% at 9 months, and 62.5% at 12 months, which was comparable to the rates observed in the third (86.4%) and sixth (73.7%) months of the phase 3 study, Dr. Paller said.

Patient-reported outcomes indicated that quality of life and satisfaction with treatment were preserved with continued treatment.The extension study was terminated in July 2023, after FDA approval, when patients could be transitioned to the commercially available treatment.Dr. Paller disclosed being an investigator (funds to institution) for multiple pharmaceutical companies, including the manufacturer of beremagene geperpavec, Krystal Biotech, which funded the study.

Meeting/Event
Publications
Topics
Sections
Meeting/Event
Meeting/Event

In an extension study of patients with dystrophic epidermolysis bullosa (DEB) treated with the topical gene therapy beremagene geperpavec, wound closure rates and adverse events were similar to those seen in the phase 3 study and no new safety signals were identified.

The results were presented by Amy S. Paller, MD, during a late-breaking session at the annual meeting of the American Academy of Dermatology.

In May 2023, beremagene geperpavec, marketed as Vyjuvek (formerly known as B-VEC) was approved by the US Food and Drug Administration (FDA) for the treatment of wounds in patients 6 months of age and older with DEB, a rare genetic blistering disorder caused by COL7A1 gene variants. The therapy uses a nonreplicating herpes simplex virus type 1 (HSV-1) vector to deliver the COL7A1 gene directly to skin cells, restoring the COL7 protein fibrils that stabilize skin structure. It is designed to be used repetitively, to heal a single wound, or on more than one wound.

In the pivotal study of patients with DEB, the gene therapy, delivered in a topical gel, was administered once a week for 6 months to one wound and placebo was applied to another wound for each participant. The proportion of wounds treated with beremagene geperpavec that healed was significantly higher than among placebo-treated wounds at 3 and 6 months (68% vs. 23% at 3 months, P = .003) and 65% vs. 26% at 6 months (P = .012), with no serious adverse events related to treatment.

The prospective, open label, uncontrolled extension study included 24 patients from the phase 3 study and 23 treatment-naive patients from five US sites. Their mean age was 16 years (range, 6 months to 46 years).

Of the 47 patients, 29 (62%) were on treatment for more than 1 year (the longest was about 2 years), and the mean duration of treatment was 475 days; 5 patients withdrew from the study for reasons not related to treatment.

Their types of adverse events (AEs) were similar to those seen in the phase 3 study and were consistent with what would be expected in patients with DEB, said Dr. Paller, professor and chair of dermatology, Northwestern University, Chicago. One patient experienced two wound hemorrhages that were possibly related to treatment, but there were no treatment-related AEs, no deaths or treatment discontinuations because of an AE, and no serious AEs thought to be related to treatment.

Wounds that were evaluated in the phase 3 study showed “a high durability of closure with continued treatment,” according to Dr. Paller. There were enough data on 19 of the 24 patients who had been in the phase 3 trial to evaluate wound closure, defined as “complete wound closure based on comparison to the exact wound area selected at baseline” at the beginning of the phase 3 study.

In the extension study, wound closure rates were almost 90% at baseline, 84.2% at 3 months, 61.1% at 6 months, 82.4% at 9 months, and 62.5% at 12 months, which was comparable to the rates observed in the third (86.4%) and sixth (73.7%) months of the phase 3 study, Dr. Paller said.

Patient-reported outcomes indicated that quality of life and satisfaction with treatment were preserved with continued treatment.The extension study was terminated in July 2023, after FDA approval, when patients could be transitioned to the commercially available treatment.Dr. Paller disclosed being an investigator (funds to institution) for multiple pharmaceutical companies, including the manufacturer of beremagene geperpavec, Krystal Biotech, which funded the study.

In an extension study of patients with dystrophic epidermolysis bullosa (DEB) treated with the topical gene therapy beremagene geperpavec, wound closure rates and adverse events were similar to those seen in the phase 3 study and no new safety signals were identified.

The results were presented by Amy S. Paller, MD, during a late-breaking session at the annual meeting of the American Academy of Dermatology.

In May 2023, beremagene geperpavec, marketed as Vyjuvek (formerly known as B-VEC) was approved by the US Food and Drug Administration (FDA) for the treatment of wounds in patients 6 months of age and older with DEB, a rare genetic blistering disorder caused by COL7A1 gene variants. The therapy uses a nonreplicating herpes simplex virus type 1 (HSV-1) vector to deliver the COL7A1 gene directly to skin cells, restoring the COL7 protein fibrils that stabilize skin structure. It is designed to be used repetitively, to heal a single wound, or on more than one wound.

In the pivotal study of patients with DEB, the gene therapy, delivered in a topical gel, was administered once a week for 6 months to one wound and placebo was applied to another wound for each participant. The proportion of wounds treated with beremagene geperpavec that healed was significantly higher than among placebo-treated wounds at 3 and 6 months (68% vs. 23% at 3 months, P = .003) and 65% vs. 26% at 6 months (P = .012), with no serious adverse events related to treatment.

The prospective, open label, uncontrolled extension study included 24 patients from the phase 3 study and 23 treatment-naive patients from five US sites. Their mean age was 16 years (range, 6 months to 46 years).

Of the 47 patients, 29 (62%) were on treatment for more than 1 year (the longest was about 2 years), and the mean duration of treatment was 475 days; 5 patients withdrew from the study for reasons not related to treatment.

Their types of adverse events (AEs) were similar to those seen in the phase 3 study and were consistent with what would be expected in patients with DEB, said Dr. Paller, professor and chair of dermatology, Northwestern University, Chicago. One patient experienced two wound hemorrhages that were possibly related to treatment, but there were no treatment-related AEs, no deaths or treatment discontinuations because of an AE, and no serious AEs thought to be related to treatment.

Wounds that were evaluated in the phase 3 study showed “a high durability of closure with continued treatment,” according to Dr. Paller. There were enough data on 19 of the 24 patients who had been in the phase 3 trial to evaluate wound closure, defined as “complete wound closure based on comparison to the exact wound area selected at baseline” at the beginning of the phase 3 study.

In the extension study, wound closure rates were almost 90% at baseline, 84.2% at 3 months, 61.1% at 6 months, 82.4% at 9 months, and 62.5% at 12 months, which was comparable to the rates observed in the third (86.4%) and sixth (73.7%) months of the phase 3 study, Dr. Paller said.

Patient-reported outcomes indicated that quality of life and satisfaction with treatment were preserved with continued treatment.The extension study was terminated in July 2023, after FDA approval, when patients could be transitioned to the commercially available treatment.Dr. Paller disclosed being an investigator (funds to institution) for multiple pharmaceutical companies, including the manufacturer of beremagene geperpavec, Krystal Biotech, which funded the study.

Publications
Publications
Topics
Article Type
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>167880</fileName> <TBEID>0C04FDB9.SIG</TBEID> <TBUniqueIdentifier>MD_0C04FDB9</TBUniqueIdentifier> <newsOrJournal>News</newsOrJournal> <publisherName>Frontline Medical Communications</publisherName> <storyname/> <articleType>2</articleType> <TBLocation>QC Done-All Pubs</TBLocation> <QCDate>20240501T094221</QCDate> <firstPublished>20240503T142013</firstPublished> <LastPublished>20240503T142013</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240503T142013</CMSDate> <articleSource>FROM AAD 2024</articleSource> <facebookInfo/> <meetingNumber>2884-24</meetingNumber> <byline>Elizabeth Mechcatie</byline> <bylineText>ELIZABETH MECHCATIE</bylineText> <bylineFull>ELIZABETH MECHCATIE</bylineFull> <bylineTitleText>MDedge News</bylineTitleText> <USOrGlobal/> <wireDocType/> <newsDocType>News</newsDocType> <journalDocType/> <linkLabel/> <pageRange/> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:imng"> <name>IMNG Medical Media</name> <rightsInfo> <copyrightHolder> <name>Frontline Medical News</name> </copyrightHolder> <copyrightNotice>Copyright (c) 2015 Frontline Medical News, a Frontline Medical Communications Inc. company. All rights reserved. This material may not be published, broadcast, copied, or otherwise reproduced or distributed without the prior written permission of Frontline Medical Communications Inc.</copyrightNotice> </rightsInfo> </provider> <abstract/> <metaDescription>In an extension study of patients with dystrophic epidermolysis bullosa (DEB) treated with the topical gene therapy beremagene geperpavec, wound closure rates a</metaDescription> <articlePDF/> <teaserImage/> <teaser>Patient-reported outcomes indicated that quality of life and satisfaction with treatment were preserved with continued treatment.</teaser> <title>Gene Therapy for Dystrophic EB: Extension Study Results Reported</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear/> <pubPubdateMonth/> <pubPubdateDay/> <pubVolume/> <pubNumber/> <wireChannels/> <primaryCMSID/> <CMSIDs/> <keywords/> <seeAlsos/> <publications_g> <publicationData> <publicationCode>skin</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> <publicationData> <publicationCode>pn</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> </publications_g> <publications> <term canonical="true">13</term> <term>25</term> </publications> <sections> <term>53</term> <term canonical="true">39313</term> </sections> <topics> <term>285</term> <term canonical="true">313</term> <term>203</term> </topics> <links/> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Gene Therapy for Dystrophic EB: Extension Study Results Reported</title> <deck/> </itemMeta> <itemContent> <p><span class="tag metaDescription">In an extension study of patients with dystrophic epidermolysis bullosa (DEB) treated with the topical gene therapy <span class="Hyperlink"><a href="https://reference.medscape.com/drug/vyjuvek-beremagene-geperpavec-4000316">beremagene geperpavec</a></span>, wound closure rates and adverse events were similar to those seen in the phase 3 study</span> and no new safety signals were identified. <br/><br/>The results were presented by Amy S. Paller, MD, during a late-breaking session at the annual meeting of the American Academy of Dermatology.</p> <p>In May 2023, b<span class="Hyperlink">eremagene geperpavec, marketed as Vyjuvek (formerly known as </span>B-VEC) was <span class="Hyperlink"><a href="https://www.fda.gov/news-events/press-announcements/fda-approves-first-topical-gene-therapy-treatment-wounds-patients-dystrophic-epidermolysis-bullosa">approved</a></span> by the US Food and Drug Administration (FDA) for the treatment of wounds in patients 6 months of age and older with DEB, a rare genetic blistering disorder caused by <em>COL7A1</em> gene variants. The therapy uses a nonreplicating herpes simplex virus type 1 (HSV-1) vector to deliver the <em>COL7A1</em> gene directly to skin cells, restoring the COL7 protein fibrils that stabilize skin structure. It is designed to be used repetitively, to heal a single wound, or on more than one wound. <br/><br/>In the pivotal study of patients with DEB, the gene therapy, delivered in a topical gel, was administered once a week for 6 months to one wound and placebo was applied to another wound for each participant. The proportion of wounds treated with b<span class="Hyperlink">eremagene geperpavec that healed was </span>significantly higher than among placebo-treated wounds at 3 and 6 months (68% vs. 23% at 3 months, <em>P</em> = .003) and 65% vs. 26% at 6 months<em> (P</em> = .012), with no serious adverse events related to treatment.<br/><br/>The prospective, open label, uncontrolled extension <span class="Hyperlink"><a href="https://clinicaltrials.gov/study/NCT04917874?term=NCT04917874&amp;rank=1">study</a></span> included 24 patients from the phase 3 study and 23 treatment-naive patients from five US sites. Their mean age was 16 years (range, 6 months to 46 years).<br/><br/>Of the 47 patients, 29 (62%) were on treatment for more than 1 year (the longest was about 2 years), and the mean duration of treatment was 475 days; 5 patients withdrew from the study for reasons not related to treatment.<br/><br/>Their types of adverse events (AEs) were similar to those seen in the phase 3 study and were consistent with what would be expected in patients with DEB, said <span class="Hyperlink"><a href="https://www.feinberg.northwestern.edu/faculty-profiles/az/profile.html?xid=16616">Dr. Paller</a></span>, professor and chair of dermatology, Northwestern University, Chicago. One patient experienced two wound hemorrhages that were possibly related to treatment, but there were no treatment-related AEs, no deaths or treatment discontinuations because of an AE, and no serious AEs thought to be related to treatment. <br/><br/>Wounds that were evaluated in the phase 3 study showed “a high durability of closure with continued treatment,” according to Dr. Paller. There were enough data on 19 of the 24 patients who had been in the phase 3 trial to evaluate wound closure, defined as “complete wound closure based on comparison to the exact wound area selected at baseline” at the beginning of the phase 3 study.<br/><br/>In the extension study, wound closure rates were almost 90% at baseline, 84.2% at 3 months, 61.1% at 6 months, 82.4% at 9 months, and 62.5% at 12 months, which was comparable to the rates observed in the third (86.4%) and sixth (73.7%) months of the phase 3 study, Dr. Paller said.<br/><br/>Patient-reported outcomes indicated that quality of life and satisfaction with treatment were preserved with continued treatment.The extension study was terminated in July 2023, after FDA approval, when patients could be transitioned to the commercially available treatment.Dr. Paller disclosed being an investigator (funds to institution) for multiple pharmaceutical companies, including the manufacturer of b<span class="Hyperlink">eremagene geperpavec, </span>Krystal Biotech, which funded the study.</p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>teaser</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> </itemContent> </newsItem> </itemSet></root>
Article Source

FROM AAD 2024

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

FDA Requests More Information for RDEB Rx Under Review

Article Type
Changed
Fri, 05/03/2024 - 13:22

 

The Food and Drug Administration (FDA) has issued a complete response letter regarding the Biologics License Application (BLA) for prademagene zamikeracel (pz-cel), which is under review for the treatment of patients with recessive dystrophic epidermolysis bullosa (RDEB), requesting more information from the manufacturer.

Pz-cel, which comprises autologous, COL7A1 gene–corrected epidermal sheets, is being evaluated for its ability to enable normal type VII collagen expression in a patient’s skin cells and to facilitate wound healing and pain reduction in wounds in patients with RDEB after a one-time application procedure. The cause of RDEB is a defect in the COL7A1 gene that “results in the inability to produce type VII collagen,” a press release from the manufacturer noted.

[embed:render:related:node:264410]

On April 22, 2024, the manufacturer Abeona Therapeutics announced that following a meeting with the FDA in March and in a subsequent request for information, the agency requires additional information to satisfy certain Chemistry Manufacturing and Controls requirements before the BLA for pz-cel can be approved. According to a press release from the company, the information pertains to validation requirements for certain manufacturing and release testing methods, including some that were observed during the FDA’s pre-licensing inspection.

The complete response letter did not identify any issues related to the clinical efficacy or safety data in the BLA, and the FDA did not request any new clinical trials or clinical data to support approval, according to the company.

The company anticipates completing the BLA resubmission in the third quarter of 2024. The application is supported by clinical efficacy and safety data from the pivotal phase 3 VIITAL study and a phase 1/2a study in patients with RDEB.

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

Publications
Topics
Sections

 

The Food and Drug Administration (FDA) has issued a complete response letter regarding the Biologics License Application (BLA) for prademagene zamikeracel (pz-cel), which is under review for the treatment of patients with recessive dystrophic epidermolysis bullosa (RDEB), requesting more information from the manufacturer.

Pz-cel, which comprises autologous, COL7A1 gene–corrected epidermal sheets, is being evaluated for its ability to enable normal type VII collagen expression in a patient’s skin cells and to facilitate wound healing and pain reduction in wounds in patients with RDEB after a one-time application procedure. The cause of RDEB is a defect in the COL7A1 gene that “results in the inability to produce type VII collagen,” a press release from the manufacturer noted.

[embed:render:related:node:264410]

On April 22, 2024, the manufacturer Abeona Therapeutics announced that following a meeting with the FDA in March and in a subsequent request for information, the agency requires additional information to satisfy certain Chemistry Manufacturing and Controls requirements before the BLA for pz-cel can be approved. According to a press release from the company, the information pertains to validation requirements for certain manufacturing and release testing methods, including some that were observed during the FDA’s pre-licensing inspection.

The complete response letter did not identify any issues related to the clinical efficacy or safety data in the BLA, and the FDA did not request any new clinical trials or clinical data to support approval, according to the company.

The company anticipates completing the BLA resubmission in the third quarter of 2024. The application is supported by clinical efficacy and safety data from the pivotal phase 3 VIITAL study and a phase 1/2a study in patients with RDEB.

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

 

The Food and Drug Administration (FDA) has issued a complete response letter regarding the Biologics License Application (BLA) for prademagene zamikeracel (pz-cel), which is under review for the treatment of patients with recessive dystrophic epidermolysis bullosa (RDEB), requesting more information from the manufacturer.

Pz-cel, which comprises autologous, COL7A1 gene–corrected epidermal sheets, is being evaluated for its ability to enable normal type VII collagen expression in a patient’s skin cells and to facilitate wound healing and pain reduction in wounds in patients with RDEB after a one-time application procedure. The cause of RDEB is a defect in the COL7A1 gene that “results in the inability to produce type VII collagen,” a press release from the manufacturer noted.

[embed:render:related:node:264410]

On April 22, 2024, the manufacturer Abeona Therapeutics announced that following a meeting with the FDA in March and in a subsequent request for information, the agency requires additional information to satisfy certain Chemistry Manufacturing and Controls requirements before the BLA for pz-cel can be approved. According to a press release from the company, the information pertains to validation requirements for certain manufacturing and release testing methods, including some that were observed during the FDA’s pre-licensing inspection.

The complete response letter did not identify any issues related to the clinical efficacy or safety data in the BLA, and the FDA did not request any new clinical trials or clinical data to support approval, according to the company.

The company anticipates completing the BLA resubmission in the third quarter of 2024. The application is supported by clinical efficacy and safety data from the pivotal phase 3 VIITAL study and a phase 1/2a study in patients with RDEB.

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

Publications
Publications
Topics
Article Type
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>167875</fileName> <TBEID>0C04FD98.SIG</TBEID> <TBUniqueIdentifier>MD_0C04FD98</TBUniqueIdentifier> <newsOrJournal>News</newsOrJournal> <publisherName>Frontline Medical Communications</publisherName> <storyname/> <articleType>2</articleType> <TBLocation>QC Done-All Pubs</TBLocation> <QCDate>20240430T142208</QCDate> <firstPublished>20240503T131759</firstPublished> <LastPublished>20240503T131759</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240503T131759</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>Doug Brunk</byline> <bylineText> DOUG BRUNK </bylineText> <bylineFull> DOUG BRUNK </bylineFull> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType>News</newsDocType> <journalDocType/> <linkLabel/> <pageRange/> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:imng"> <name>IMNG Medical Media</name> <rightsInfo> <copyrightHolder> <name>Frontline Medical News</name> </copyrightHolder> <copyrightNotice>Copyright (c) 2015 Frontline Medical News, a Frontline Medical Communications Inc. company. All rights reserved. This material may not be published, broadcast, copied, or otherwise reproduced or distributed without the prior written permission of Frontline Medical Communications Inc.</copyrightNotice> </rightsInfo> </provider> <abstract/> <metaDescription>(FDA) has issued a complete response letter regarding the Biologics License Application (BLA) for prademagene zamikeracel (pz-cel), which is under review for th</metaDescription> <articlePDF/> <teaserImage/> <teaser>Pz-cel, which comprises autologous, <em>COL7A1</em> gene–corrected epidermal sheets, is being evaluated for its ability to enable normal type VII collagen expression in a patient’s skin cells and to facilitate wound healing and pain reduction.</teaser> <title>FDA Requests More Information for RDEB Rx Under Review</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear/> <pubPubdateMonth/> <pubPubdateDay/> <pubVolume/> <pubNumber/> <wireChannels/> <primaryCMSID/> <CMSIDs/> <keywords/> <seeAlsos/> <publications_g> <publicationData> <publicationCode>skin</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> <publicationData> <publicationCode>fp</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> <publicationData> <publicationCode>im</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> <publicationData> <publicationCode>pn</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> </publications_g> <publications> <term canonical="true">13</term> <term>15</term> <term>21</term> <term>25</term> </publications> <sections> <term>37225</term> <term canonical="true">39313</term> </sections> <topics> <term>313</term> <term canonical="true">285</term> <term>271</term> <term>203</term> </topics> <links/> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>FDA Requests More Information for RDEB Rx Under Review</title> <deck/> </itemMeta> <itemContent> <p>The Food and Drug Administration<span class="tag metaDescription"> (FDA) has issued a complete response letter regarding the Biologics License Application (BLA) for prademagene zamikeracel (pz-cel), which is under review for the treatment of patients with recessive dystrophic epidermolysis bullosa</span> (RDEB), requesting more information from the manufacturer.</p> <p>Pz-cel, which comprises autologous, <em><a href="https://medlineplus.gov/genetics/gene/col7a1/">COL7A1</a></em> gene–corrected epidermal sheets, is being evaluated for its ability to enable normal type VII collagen expression in a patient’s skin cells and to facilitate wound healing and pain reduction in wounds in patients with RDEB after a one-time application procedure. The cause of RDEB is a defect in the <em>COL7A1</em> gene that “results in the inability to produce type VII collagen,” a press release from the manufacturer noted.<br/><br/>On April 22, 2024, the manufacturer Abeona Therapeutics announced that following a meeting with the FDA in March and in a subsequent request for information, the agency requires additional information to satisfy certain Chemistry Manufacturing and Controls requirements before the BLA for pz-cel can be approved. According to <a href="https://investors.abeonatherapeutics.com/press-releases/detail/276/abeona-therapeutics-provides-regulatory-update-on-pz-cel">a press release</a> from the company, the information pertains to validation requirements for certain manufacturing and release testing methods, including some that were observed during the FDA’s pre-licensing inspection.<br/><br/>The complete response letter did not identify any issues related to the clinical efficacy or safety data in the BLA, and the FDA did not request any new clinical trials or clinical data to support approval, according to the company.<br/><br/>The company anticipates completing the BLA resubmission in the third quarter of 2024. The application is supported by clinical efficacy and safety data from the pivotal <a href="https://classic.clinicaltrials.gov/ct2/show/NCT04227106">phase 3 VIITAL study</a> and a <a href="https://clinicaltrials.gov/study/NCT01263379">phase 1/2a study</a> in patients with RDEB.<span class="end"/></p> <p> <em>A version of this article first appeared on <span class="Hyperlink"><a href="https://www.medscape.com/viewarticle/fda-seeks-more-information-about-rdeb-rx-under-review-2024a1000836?src=">Medscape.com</a></span>.</em> </p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>teaser</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> </itemContent> </newsItem> </itemSet></root>
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

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.

Afvari_scorpionfish_1.jpg
%3Cp%3E%3Cstrong%3EFIGURE%201.%3C%2Fstrong%3E%20Red%20scorpionfish%20(%3Cem%3EScorpaena%20scrofa%3C%2Fem%3E).%3C%2Fp%3E

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

Afvari_scorpionfish_2.jpg
%3Cp%3E%3Cstrong%3EFIGURE%202.%3C%2Fstrong%3E%20Lionfish%20(%3Cem%3EPterois%20volitans%3C%2Fem%3E).%3C%2Fp%3E

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.

Afvari_scorpionfish_1.jpg
%3Cp%3E%3Cstrong%3EFIGURE%201.%3C%2Fstrong%3E%20Red%20scorpionfish%20(%3Cem%3EScorpaena%20scrofa%3C%2Fem%3E).%3C%2Fp%3E

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

Afvari_scorpionfish_2.jpg
%3Cp%3E%3Cstrong%3EFIGURE%202.%3C%2Fstrong%3E%20Lionfish%20(%3Cem%3EPterois%20volitans%3C%2Fem%3E).%3C%2Fp%3E

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.

Afvari_scorpionfish_1.jpg
%3Cp%3E%3Cstrong%3EFIGURE%201.%3C%2Fstrong%3E%20Red%20scorpionfish%20(%3Cem%3EScorpaena%20scrofa%3C%2Fem%3E).%3C%2Fp%3E

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

Afvari_scorpionfish_2.jpg
%3Cp%3E%3Cstrong%3EFIGURE%202.%3C%2Fstrong%3E%20Lionfish%20(%3Cem%3EPterois%20volitans%3C%2Fem%3E).%3C%2Fp%3E

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
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>Afvari scorpionfish</fileName> <TBEID>0C02F309.SIG</TBEID> <TBUniqueIdentifier>NJ_0C02F309</TBUniqueIdentifier> <newsOrJournal>Journal</newsOrJournal> <publisherName>Frontline Medical Communications Inc.</publisherName> <storyname>Afvari scorpionfish</storyname> <articleType>1</articleType> <TBLocation>Copyfitting-CT</TBLocation> <QCDate/> <firstPublished>20240305T083640</firstPublished> <LastPublished>20240305T083640</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240305T083639</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>Shawn Afvari, BS; Dirk M. Elston, MD</byline> <bylineText>Shawn Afvari, BS; Dirk M. Elston, MD</bylineText> <bylineFull>Shawn Afvari, BS; Dirk M. Elston, MD</bylineFull> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType/> <journalDocType/> <linkLabel/> <pageRange>133-134,136</pageRange> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:"> <name/> <rightsInfo> <copyrightHolder> <name/> </copyrightHolder> <copyrightNotice/> </rightsInfo> </provider> <abstract/> <metaDescription>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</metaDescription> <articlePDF>300457</articlePDF> <teaserImage/> <title>Aquatic Antagonists: Scorpionfish Envenomation</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear>2024</pubPubdateYear> <pubPubdateMonth>March</pubPubdateMonth> <pubPubdateDay/> <pubVolume>113</pubVolume> <pubNumber>3</pubNumber> <wireChannels/> <primaryCMSID/> <CMSIDs> <CMSID>2159</CMSID> </CMSIDs> <keywords> <keyword>wounds</keyword> <keyword> scorpionfish envenomation</keyword> </keywords> <seeAlsos/> <publications_g> <publicationData> <publicationCode>CT</publicationCode> <pubIssueName>March 2024</pubIssueName> <pubArticleType>Departments | 2159</pubArticleType> <pubTopics/> <pubCategories/> <pubSections/> <journalTitle>Cutis</journalTitle> <journalFullTitle>Cutis</journalFullTitle> <copyrightStatement>Copyright 2015 Frontline Medical Communications Inc., Parsippany, NJ, USA. All rights reserved.</copyrightStatement> </publicationData> </publications_g> <publications> <term canonical="true">12</term> </publications> <sections> <term canonical="true">60</term> </sections> <topics> <term canonical="true">313</term> </topics> <links> <link> <itemClass qcode="ninat:composite"/> <altRep contenttype="application/pdf">images/180026e1.pdf</altRep> <description role="drol:caption"/> <description role="drol:credit"/> </link> </links> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Aquatic Antagonists: Scorpionfish Envenomation</title> <deck/> </itemMeta> <itemContent> <p class="abstract">Scorpionfish are among the most venomous creatures found in American and Caribbean seas. Their envenomation is responsible for considerable morbidity and socioeconomic burden associated with marine animal injuries. Avoiding physical contact with scorpionfish through proper identification prevails as the chief prevention method for stings. This article discusses common features of scorpionfish as well as the clinical presentation and treatment options following exposure to its toxins.</p> <p> <em><em>Cutis.</em> 2024;113:133-134, 136.</em> </p> <p>With the growing popularity of water sports and a proliferation of invasive species, human injuries from marine animal envenomation continue to rise.<sup>1-3</sup> 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.<sup>4</sup> Because scorpionfish represent a growing threat and competition with native species, it has been suggested that they could replace endangered species on restaurant menus.<sup>5-8</sup> 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.<sup>9,10</sup> Fishers, divers, swimmers, and aquarium owners are most often affected. </p> <h3>Family</h3> <p>The common term <i>scorpionfish</i> refers to both the family Scorpaenidae and the genus <i>Scorpaena</i>. 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 <i>Pterois</i> (lionfish) and <i>Synanceja</i> (stonefish). <i>Synanceja</i> are the most venomous within the group, but scorpionfish stings more commonly arise from <i>Pterois </i>and <i>Scorpaena.</i><sup>8</sup><i> </i>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.<sup>11,12</sup> </p> <h3>Characteristics</h3> <p>Scorpionfish have with a high variation in color, ranging from lighter grays to intense reds depending on their geographic location and habitat. <i>Synanceja</i> are bland in coloration, blending in with rocks and gravel, but the more dramatic-appearing <i>Scorpaena </i>exhibit a large cranium and wide range of multicolored patterns (Figure 1).<sup>13</sup> <i>Pterois</i> 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.<sup>14</sup> An integumentary sheath encapsulates each spine housing the glandular tissue that produces the potent venom. </p> <h3>Toxin Properties </h3> <p>Unlike <i>Pterois </i>and<i> Synanceja, Scorpaena </i>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.<sup>15-20</sup> 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.<sup>1</sup> </p> <h3>Clinical Manifestation</h3> <p>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.<sup>9,21-23</sup> The pain will intensify and radiate distal to the site of envenomation, and the wound may exhibit vesiculation, erythema, bruising, pallor, and notable edema.<sup>4,24</sup> Pain intensity peaks at 30 to 90 minutes after envenomation, and other systemic symptoms generally last for 24 to 48 hours.<sup>25</sup> 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.<sup>1</sup></p> <h3>Management </h3> <p>Treatment of scorpionfish stings primarily is palliative and aimed at symptom reduction. Patients should immediately treat wounds with hot but not scalding water immersion.<sup>26,27</sup> Given the thermolabile components of scorpionfish venom, the most effective treatment is to soak the affected limb in water 42 <span class="body">°</span>C to 45 <span class="body">°</span>C for 30 to 90 minutes. Any higher temperature may pose risk for scalding burns. Children should be monitored throughout treatment.<sup>28</sup> 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.<sup>29</sup></p> <h2>References</h2> <p class="reference"> 1. Rensch G, Murphy-Lavoie HM. Lionfish, scorpionfish, and stonefish toxicity. <i>StatPearls</i>. StatPearls Publishing; May 10, 2022.<br/><br/> 2. Cearnal L. Red lionfish and ciguatoxin: menace spreading through western hemisphere. <i>Ann Emerg Med</i>. 2012;60:21A-22A. doi:10.1016/j.annemergmed.2012.05.022<br/><br/> 3. Côté IM, Green SJ. Potential effects of climate change on a marine invasion: the importance of current context. <i>Curr Zool.</i> 2012;58:1-8. doi:10.1093/czoolo/58.1.1<br/><br/> 4. Venomology of scorpionfishes. In: Santhanam R. <i>Biology and Ecology of Venomous Marine Scorpionfishes</i>. Academic Press; 2019:263-278.<br/><br/> 5. Ferri J, Staglicˇic´ N, Matić-Skoko S. The black scorpionfish, <i>Scorpaena porcus</i> (Scorpaenidae): could it serve as reliable indicator of Mediterranean coastal communities’ health? <i>Ecol Indicators</i>. 2012;18:25-30. doi:10.1016/j.ecolind.2011.11.004 <br/><br/> 6. Santhanam R.<i> Biology and Ecology of Venomous Marine Scorpionfishes</i>. Academic Press; 2019.<br/><br/> 7. Morris JA, Akins JL. Feeding ecology of invasive lionfish (<i>Pterois volitans</i>) in the Bahamian Archipelago. <i>Environ Biol Fishes</i>. 2009;86:389-398. doi:10.1007/s10641-009-9538-8 <br/><br/> <br/><br/> 8. Albins MA, Hixon MA. Worst case scenario: potential long-term effects of invasive predatory lionfish (<i>Pterois volitans</i>) on Atlantic and Caribbean coral-reef communities. <i>Environ Biol Fishes. </i>2013;96:1151–1157. doi:10.1007/s10641-011-9795-1</p> <p class="reference"> 9. Haddad V Jr, Martins IA, Makyama HM. Injuries caused by scorpionfishes (<i>Scorpaena plumieri</i> Bloch, 1789 and <i>Scorpaena brasiliensis</i> Cuvier, 1829) in the Southwestern Atlantic Ocean (Brazilian coast): epidemiologic, clinic and therapeutic aspects of 23 stings in humans. <i>Toxicon</i>. 2003;42:79-83. doi:10.1016/s0041-0101(03)00103-x<br/><br/>10. Campos FV, Menezes TN, Malacarne PF, et al. A review on the <i>Scorpaena plumieri</i> fish venom and its bioactive compounds. <i>J Venom Anim Toxins Incl Trop Dis</i>. 2016;22:35. doi:10.1186/s40409-016-0090-7<br/><br/>11. Needleman RK, Neylan IP, Erickson TB. Environmental and ecological effects of climate change on venomous marine and amphibious species in the wilderness. <i>Wilderness Environ Med</i>. 2018;29:343-356. doi:10.1016/j.wem.2018.04.003<br/><br/>12. Aldred B, Erickson T, Lipscomb J. Lionfish envenomations in an urban wilderness. <i>Wilderness Environ Med</i>. 1996;7:291-296. doi:10.1580/1080-6032(1996)007[0291:leiauw]2.3.co;2<br/><br/>13. Stewart J, Hughes JM. Life-history traits of the southern hemisphere eastern red scorpionfish, <i>Scorpaena cardinalis</i> (Scorpaenidae: Scorpaeninae). <i>Mar Freshw Res. </i>2010;61:1290-1297. doi:10.1071/MF10040<br/><br/>14. Auerbach PS. Marine envenomations. <i>N Engl J Med</i>. 1991;325:486-493. doi:10.1056/NEJM199108153250707<br/><br/>15. Andrich F, Carnielli JB, Cassoli JS, et al. A potent vasoactive cytolysin isolated from <i>Scorpaena plumieri</i> scorpionfish venom. <i>Toxicon</i>. 2010;56:487-496. doi:10.1016/j.toxicon.2010.05.003<br/><br/>16. Gomes HL, Andrich F, Mauad H, et al. Cardiovascular effects of scorpionfish (<i>Scorpaena plumieri</i>) venom. <i>Toxicon</i>. 2010;55(2-3):580-589. doi:10.1016/j.toxicon.2009.10.012<br/><br/>17. Menezes TN, Carnielli JB, Gomes HL, et al. Local inflammatory response induced by scorpionfish <i>Scorpaena plumieri</i> venom in mice. <i>Toxicon</i>. 2012;60:4-11. doi:10.1016/j.toxicon.2012.03.008<br/><br/>18. Schaeffer RC Jr, Carlson RW, Russell FE. Some chemical properties of the venom of the scorpionfish <i>Scorpaena guttata</i>. <i>Toxicon</i>. 1971;9:69-78. doi:10.1016/0041-0101(71)90045-6<br/><br/>19. Khalil AM, Wahsha MA, Abu Khadra KM, et al. Biochemical and histopathological effects of the stonefish (<i>Synanceia verrucosa</i>) venom in rats. <i>Toxicon</i>. 2018;142:45-51. doi:10.1016/j.toxicon.2017.12.052<br/><br/>20. Mouchbahani-Constance S, Lesperance LS, Petitjean H, et al. Lionfish venom elicits pain predominantly through the activation of nonpeptidergic nociceptors. <i>Pain</i>. 2018;159:2255-2266. doi:10.1097/j.pain.0000000000001326<br/><br/>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. <i>Int J Dermatol</i>. 2013;52:268-278. doi:10.1111/j.1365-4632.2011.05426.x<br/><br/>22. Bayley HH. Injuries caused by scorpion fish. <i>Trans R Soc Trop Med Hyg.</i> 1940;34:227-230. doi:10.1016/s0035-9203(40)90072-4 <br/><br/>23. González D. Epidemiological and clinical aspects of certain venomous animals of Spain. <i>Toxicon</i>. 1982;20:925-928. doi:10.1016/0041-0101(82)90080-0<br/><br/>24. Halstead BW. Injurious effects from the sting of the scorpionfish, <i>Scorpaena guttata</i>. with report of a case. <i>Calif Med</i>. 1951;74:395-396.<br/><br/>25. Vasievich MP, Villarreal JD, Tomecki KJ. Got the travel bug? a review of common infections, infestations, bites, and stings among returning travelers. <i>Am J Clin Dermatol</i>. 2016;17:451-462. doi:10.1007/s40257-016-0203-7<br/><br/>26. Barnett S, Saggiomo S, Smout M, et al. Heat deactivation of the stonefish <i>Synanceia horrida</i> venom—implications for first-aid management. <i>Diving Hyperb Med</i>. 2017;47:155-158. doi:10.28920/dhm47.3.155-158<br/><br/>27. Russell FE. Weever fish sting: the last word. <i>Br Med J (Clin Res Ed)</i>. 1983;287:981-982. doi:10.1136/bmj.287.6397.981-c<br/><br/>28. Tomlinson H, Elston DM. Aquatic antagonists: lionfish (<i>Pterois volitans</i>). <i>Cutis</i>. 2018;102:232-234.<br/><br/>29. Hornbeak KB, Auerbach PS. Marine envenomation. <i>Emerg Med Clin North Am</i>. 2017;35:321-337. doi:10.1016/j.emc.2016.12.004 </p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>bio</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="disclosure">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.</p> <p class="disclosure">The authors report no conflict of interest.<br/><br/>Correspondence: Shawn Afvari, BS (safvari@student.nymc.edu).doi:10.12788/cutis.0973</p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>in</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="insidehead">Practice <strong>Points</strong></p> <ul class="insidebody"> <li>As some species of scorpionfish proliferate, providers may see an increase in envenomation cases. </li> <li>Physicians should suspect scorpionfish stings based on clinical symptoms and physical examination. </li> </ul> </itemContent> </newsItem> </itemSet></root>
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
Image
Teambase ID
180026E1.SIG
Disable zoom
Off

Wound Healing: Cellular Review With Specific Attention to Postamputation Care

Article Type
Changed
Wed, 03/06/2024 - 11:45
Display Headline
Wound Healing: Cellular Review With Specific Attention to Postamputation Care
IN PARTNERSHIP WITH THE ASSOCIATION OF MILITARY DERMATOLOGISTS

Restoring skin integrity and balance after injury is vital for survival, serving as a crucial defense mechanism against potential infections by preventing the entry of harmful pathogens. Moreover, proper healing is essential for restoring normal tissue function, allowing damaged tissues to repair and, in an ideal scenario, regenerate. Timely healing helps reduce the risk for complications, such as chronic wounds, which could lead to more severe issues if left untreated. Additionally, pain relief often is associated with effective wound healing as inflammatory responses diminish during the repair process.

The immune system plays a pivotal role in wound healing, influencing various repair mechanisms and ultimately determining the extent of scarring. Although inflammation is present throughout the repair response, recent studies have challenged the conventional belief of an inverse correlation between the intensity of inflammation and regenerative capacity. Inflammatory signals were found to be crucial for timely repair and fundamental processes in regeneration, possibly presenting a paradigm shift in the understanding of immunology.1-4 The complexities of wound healing are exemplified when evaluating and treating postamputation wounds. To address such a task, one needs a firm understanding of the science behind healing wounds and what can go wrong along the way.

Phases of Wound Healing

Wound healing is a complex process that involves a series of sequential yet overlapping phases, including hemostasis/inflammation, proliferation, and remodeling.

Hemostasis/Inflammation—The initial stage of wound healing involves hemostasis, in which the primary objective is to prevent blood loss and initiate inflammation. Platelets arrive at the wound site, forming a provisional clot that is crucial for subsequent healing phases.4-6 Platelets halt bleeding as well as act as a medium for cell migration and adhesion; they also are a source of growth factors and proinflammatory cytokines that herald the inflammatory response.4-7

Inflammation is characterized by the infiltration of immune cells, particularly neutrophils and macrophages. Neutrophils act as the first line of defense, clearing debris and preventing infection. Macrophages follow, phagocytizing apoptotic cells and releasing growth factors such as tumor necrosis factor α, vascular endothelial growth factor, and matrix metalloprotease 9, which stimulate the next phase.4-6,8 Typically, the hemostasis and inflammatory phase starts approximately 6 to 8 hours after wound origin and lasts 3 to 4 days.4,6,7

Proliferation—Following hemostasis and inflammation, the wound transitions into the proliferation phase, which is marked by the development of granulation tissue—a dynamic amalgamation of fibroblasts, endothelial cells, and inflammatory cells.1,4-8 Fibroblasts play a central role in synthesizing collagen, the primary structural protein in connective tissue. They also orchestrate synthesis of vitronectin, fibronectin, fibrin, and tenascin.4-6,8 Simultaneously, angiogenesis takes place, involving the creation of new blood vessels to supply essential nutrients and oxygen to the healing tissue.4,7,9 Growth factors such as transforming growth factor β and vascular endothelial growth factor coordinate cellular activities and foster tissue repair.4-6,8 The proliferation phase extends over days to weeks, laying the groundwork for subsequent tissue restructuring.

Remodeling—The final stage of wound healing is remodeling, an extended process that may persist for several months or, in some cases, years. Throughout this phase, the initially deposited collagen, predominantly type III collagen, undergoes transformation into mature type I collagen.4-6,8 This transformation is critical for reinstating the tissue’s strength and functionality. The balance between collagen synthesis and degradation is delicate, regulated by matrix metalloproteinases and inhibitors of metalloproteinases.4-8 Fibroblasts, myofibroblasts, and other cells coordinate this intricate process of tissue reorganization.4-7

 

 

The eventual outcome of the remodeling phase determines the appearance and functionality of the healed tissue. Any disruption in this phase can lead to complications, such as chronic wounds and hypertrophic scars/keloids.4-6 These abnormal healing processes are characterized by localized inflammation, heightened fibroblast function, and excessive accumulation of the extracellular matrix.4-8

Molecular Mechanisms

Comprehensive investigations—both in vivo and in vitro—have explored the intricate molecular mechanisms involved in heightened wound healing. Transforming growth factor β takes center stage as a crucial factor, prompting the transformation of fibroblasts into myofibroblasts and contributing to the deposition of extracellular matrix.2,4-8,10 Transforming growth factor β activates non-Smad signaling pathways, such as MAPK (mitogen-activated protein kinase) and PI3K (phosphoinositide 3-kinase), influencing processes associated with fibrosis.5,11 Furthermore, microRNAs play a pivotal role in posttranscriptional regulation, influencing both transforming growth factor β signaling and fibroblast behavior.12-16

The involvement of prostaglandins is crucial in wound healing. Prostaglandin E2 plays a notable role and is positively correlated with the rate of wound healing.5 The cyclooxygenase pathway, pivotal for prostaglandin synthesis, becomes a target for inflammation control.4,5,10 Although aspirin and nonsteroidal anti-inflammatory drugs commonly are employed, their impact on wound healing remains controversial, as inhibition of cyclooxygenase may disrupt normal repair processes.5,17,18

Wound healing exhibits variations depending on age. Fetal skin regeneration is marked by the restoration of normal dermal architecture, including adnexal structures, nerves, vessels, and muscle.4-6 The distinctive characteristics of fetal wound healing include a unique profile of growth factors, a diminished inflammatory response, reduced biomechanical stress, and a distinct extracellular matrix composition.19 These factors contribute to a lower propensity for scar formation compared to the healing processes observed in adults. Fetal and adult wound healing differ fundamentally in their extracellular matrix composition, inflammatory cells, and cytokine levels.4-6,19 Adult wounds feature myofibroblasts, which are absent in fetal wounds, contributing to heightened mechanical tension.5 Delving deeper into the biochemical basis of fetal wound healing holds promise for mitigating scar formation in adults.

Takeaways From Other Species

Much of the biochemical knowledge of wound healing, especially regenerative wound healing, is known from other species. Geckos provide a unique model for studying regenerative repair in tails and nonregenerative healing in limbs after amputation. Scar-free wound healing is characterized by rapid wound closure, delayed blood vessel development, and collagen deposition, which contrasts with the hypervascular granulation tissue seen in scarring wounds.20 Scar-free wound healing and regeneration are intrinsic properties of the lizard tail and are unaffected by the location or method of detachment.21

Compared to amphibians with extraordinary regenerative capacity, data suggest the lack of regenerative capacity in mammals may come from a desynchronization of the fine-tuned interplay of progenitor cells such as blastema and differentiated cells.22,23 In mice, the response to amputation is specific to the level: cutting through the distal third of the terminal phalanx elicits a regeneration response, yielding a new digit tip resembling the lost one, while an amputation through the distal third of the intermediate phalanx triggers a wound healing and scarring response.24

Wound Healing Following Limb Amputation

Limb amputation represents a profound change in an individual’s life, impacting daily activities and overall well-being. There are many causes of amputation, but the most common include cardiovascular diseases, diabetes mellitus, cancer, and trauma.25-27 Trauma represents a relatively common cause within the US Military due to the overall young population as well as inherent risks of uniformed service.25,27 Advances in protective gear and combat casualty care have led to an increased number of individuals surviving with extremity injuries requiring amputation, particularly among younger service members, with a subgroup experiencing multiple amputations.27-29

 

 

Numerous factors play a crucial role in the healing and function of postamputation wounds. The level of amputation is a key determinant influencing both functional outcomes and the healing process. Achieving a balance between preserving function and removing damaged tissue is essential. A study investigating cardiac function and oxygen consumption in 25 patients with peripheral vascular disease found higher-level amputations resulted in decreased walking speed and cadence, along with increased oxygen consumption per meter walked.30

Selecting the appropriate amputation level is vital to optimize functional outcomes without compromising wound healing. Successful prosthetic limb fitting depends largely on the length of the residual stump to support the body load and suspend the prosthesis. For long bone amputations, maintaining at least 12-cm clearance above the knee joint in transfemoral amputees and 10-cm below the knee joint in transtibial amputees is critical for maximizing functional outcomes.31

Surgical technique also is paramount. The goal is to minimize the risk for pressure ulcers by avoiding bony spurs and muscle imbalances. Shaping the muscle and residual limb is essential for proper prosthesis fitting. Attention to neurovascular structures, such as burying nerve ends to prevent neuropathic pain during prosthesis wear, is crucial.32 In extremity amputations, surgeons often resort to free flap transfer techniques for stump reconstruction. In a study of 31 patients with severe lower extremity injuries undergoing various amputations, the use of latissimus dorsi myocutaneous flaps, alone or in combination with serratus anterior muscle flaps, resulted in fewer instances of deep ulceration and allowed for earlier prosthesis wear.33

Addressing Barriers to Wound Healing

Multiple barriers to successful wound healing are encountered in the amputee population. Amputations from trauma have a less-controlled initiation, which carries with it a higher risk for infection, poor wound healing, and other complications.

Infection—Infection often is one of the first hurdles encountered in postamputation wound healing. Critical first steps in infection prevention include thorough cleaning of soiled traumatic wounds and appropriate tissue debridement coupled with scrupulous sterile technique and postoperative monitoring for signs and symptoms of infection.

In a retrospective study of 223 combat-related major lower extremity amputations (initial and revision) between 2009 and 2015, the use of intrawound antibiotic powder at the time of closure demonstrated a 13% absolute risk reduction in deep infection rates, which was particularly notable in revision amputations, with a number needed to treat of 8 for initial amputations and 4 for revision amputations on previously infected limbs.34 Intra-operative antibiotic powder may represent a cheap and easy consideration for this special population of amputees. Postamputation antibiotic prophylaxis for infection prevention is an area of controversy. For nontraumatic infections, data suggest antibiotic prophylaxis may not decrease infection rates in these patients.35,36

Interestingly, a study by Azarbal et al37 aimed to investigate the correlation between nasal methicillin-resistant Staphylococcus aureus (MRSA) colonization and other patient factors with wound occurrence following major lower extremity amputation. The study found MRSA colonization was associated with higher rates of overall wound occurrence as well as wound occurrence due to wound infection. These data suggest nasal MRSA eradication may improve postoperative wound outcomes after major lower extremity amputation.37

 

 

Dressing Choice—The dressing chosen for a residual limb also is of paramount importance following amputation. The personalized and dynamic management of postamputation wounds and skin involves achieving optimal healing through a dressing that sustains appropriate moisture levels, addresses edema, helps prevent contractures, and safeguards the limb.38 From the start, using negative pressure wound dressings after surgical amputation can decrease wound-related complications.39

Topical oxygen therapy following amputation also shows promise. In a retrospective case series by Kalliainen et al,40 topical oxygen therapy applied to 58 wounds in 32 patients over 9 months demonstrated positive outcomes in promoting wound healing, with 38 wounds (66%) healing completely with the use of topical oxygen. Minimal complications and no detrimental effects were observed.40

Current recommendations suggest that non–weight-bearing removable rigid dressings are the superior postoperative management for transtibial amputations compared to soft dressings, offering benefits such as faster healing, reduced limb edema, earlier ambulation, preparatory shaping for prosthetic use, and prevention of knee flexion contractures.41-46 Similarly, adding a silicone liner following amputation significantly reduced the duration of prosthetic rehabilitation compared with a conventional soft dressing program in one study (P<.05).47

Specifically targeting wound edema, a case series by Hoskins et al48 investigated the impact of prostheses with vacuum-assisted suspension on the size of residual limb wounds in individuals with transtibial amputation. Well-fitting sockets with vacuum-assisted suspension did not impede wound healing, and the results suggest the potential for continued prosthesis use during the healing process.48 However, a study by Johannesson et al49 compared the outcomes of transtibial amputation patients using a vacuum-formed rigid dressing and a conventional rigid plaster dressing, finding no significant differences in wound healing, time to prosthetic fitting, or functional outcomes with the prosthesis between the 2 groups. When comparing elastic bandaging, pneumatic prosthesis, and temporary prosthesis on postoperative stump management, temporary prosthesis led to a decrease in stump volume, quicker transition to a permanent prosthesis, and improved quality of life compared with elastic bandaging and pneumatic prosthetics.50

The type of material in dressings may contribute to utility in amputation wounds. Keratin-based wound dressings show promise for wound healing, especially in recalcitrant vascular wounds.51 There also are numerous proprietary wound dressings available for patients, at least one of which has particularly thorough data. In a retrospective study of more than 2 million lower extremity wounds across 644 institutions, a proprietary bioactive human skin allograft (TheraSkin [LifeNet Health]) demonstrated higher healing rates, greater percentage area reductions, lower amputations, reduced recidivism, higher treatment completion, and fewer medical transfers compared with standard of care alone.52

Postamputation Dermatologic Concerns

After the postamputation wound heals, a notable concern is the prevalence of skin diseases affecting residual limbs. The stump site in amputees, marked by a delicate cutaneous landscape vulnerable to skin diseases, faces challenges arising from amputation-induced damage to various structures.53

When integrated into a prosthesis socket, the altered skin must acclimate to a humid environment and endure forces for which it is not well suited, especially during movement.53 Amputation remarkably alters normal tissue perfusion, which can lead to aberrant blood and lymphatic circulation in residual limbs.27,53 This compromised skin, often associated with a history of vascular disease, diabetes mellitus, or malignancy, becomes immunocompromised, heightening the risk for dermatologic issues such as inflammation, infection, and malignancies.53 Unlike the resilient volar skin on palms and soles, stump skin lacks adaptation to withstand the compressive forces generated during ambulation, sometimes leading to skin disease and pain that result in abandonment of the prosthesis.53,54 Mechanical forces on the skin, especially in active patients eager to resume pre-injury lifestyles, contribute to skin breakdown. The dynamic nature of the residual limb, including muscle atrophy, gait changes, and weight fluctuations, complicates the prosthetic fitting process. Prosthesis abandonment remains a challenge, despite modern technologic advancements.

 

 

The occurrence of heterotopic ossification (extraskeletal bone formation) is another notable issue in military amputees.27,55-57 Poor prosthetic fit can lead to skin degradation, necessitating further surgery to address mispositioned bone formations. Orthopedic monitoring supplemented by appropriate imaging studies can benefit postamputation patients by detecting and preventing heterotopic ossification in its early stages.

Dermatologic issues, especially among lower limb amputees, are noteworthy, with a substantial percentage experiencing complications related to socket prosthetics, such as heat, sweating, sores, and skin irritation. Up to 41% of patients are seen regularly for a secondary skin disorder following amputation.58 As one might expect, persistent wounds, blisters, ulcers, and abscesses are some of the most typical cutaneous abnormalities affecting residual limbs with prostheses.27,58 More rare skin conditions also are documented in residual limbs, including cutaneous granuloma, verrucous carcinoma, bullous pemphigoid, and angiodermatitis.27,59-61

Treatments offered in the dermatology clinic often are similar to patients who have not had an amputation. For instance, hyperhidrosis can be treated with prescription antiperspirant, topical aluminum chloride, topical glycopyrronium, botulinum toxin, and iontophoresis, which can greatly decrease skin irritation and malodor. Subcutaneous neurotoxins such as botulinum toxin are especially useful for hyperhidrosis following amputation because a single treatment can last 3 to 6 months, whereas topicals must be applied multiple times per day and can be inherently irritating to the skin.27,62 Furthermore, ablative fractional resurfacing lasers also can help stimulate new collagen growth, increase skin mobility on residual limbs, smooth jagged scars, and aid prosthetic fitting.27,63 Perforated prosthetic liners also may be useful to address issues such as excessive sweating, demonstrating improvements in skin health, reduced sweating problems, and potential avoidance of surgical interventions.64

When comorbid skin conditions are at bay, preventive measures for excessive wound healing necessitate early recognition and timely intervention for residual limbs. Preventive techniques encompass the use of silicone gel sheeting, hypoallergenic microporous tape, and intralesional steroid injections.

Psychological Concerns—An overarching issue following amputation is the psychological toll the process imposes on the patient. Psychological concerns, including anxiety and depression, present additional challenges impacting residual limb hygiene and prosthetic maintenance. Chronic wounds are devastating to patients. These patients consistently express feeling ostracized from their community and anxious about unemployment, leaking fluid, or odor from the wound, as well as other social stigmata.62 Depression and anxiety can hinder a patient’s ability to care for their wound and make them more susceptible to the myriad issues that can ensue.

Recent Developments in Wound Healing

Wound healing is ripe for innovation that could assuage ailments that impact patients following amputation. A 2022 study by Abu El Hawa et al65 illustrated advanced progression in wound healing for patients taking statins, even though the statin group had increased age and number of comorbidities compared with patients not taking statins.

Nasseri and Sharifi66 showed the potential of antimicrobial peptides—small proteins with cationic charges and amphipathic structures exhibiting electrostatic interaction with microbial cell membranes—in promoting wound healing, particularly defensins and cathelicidin LL-37.They also discussed innovative delivery systems, such as nanoparticles and electrospun fibrous scaffolds, highlighting their potential as possibly more effective therapeutics than antibiotics, especially in the context of diabetic wound closure.66 Aimed at increased angiogenesis in the proliferative phase, there is evidence that N-acetylcysteine can increase amputation stump perfusion with the goal of better long-term wound healing and more efficient scar formation.67

Stem cell therapy, particularly employing cells from the human amniotic membrane, represents an auspicious avenue for antifibrotic treatment. Amniotic epithelial cells and amniotic mesenchymal cells, with their self-renewal and multilineage differentiation capabilities, exhibit anti-inflammatory and antifibrotic properties.4,5 A study by Dong et al68 aimed to assess the efficacy of cell therapy, particularly differentiated progenitor cell–based graft transplantation or autologous stem cell injection, in treating refractory skin injuries such as nonrevascularizable critical limb ischemic ulcers, venous leg ulcers, and diabetic lower limb ulcers. The findings demonstrated cell therapy effectively reduced the size of ulcers, improved wound closure rates, and decreased major amputation rates compared with standard therapy. Of note, cell therapy had limited impact on alleviating pain in patients with critical limb ischemia-related cutaneous ulcers.68

Final Thoughts

Wound care following amputation is a multidisciplinary endeavor, necessitating collaboration between many health care professionals. Dermatologists play a crucial role in providing routine care as well as addressing wound healing and related skin issues among amputation patients. As the field progresses, dermatologists are well positioned to make notable contributions and ensure enhanced outcomes, resulting in a better quality of life for patients facing the challenges of limb amputation and prosthetic use.

References
  1. Brockes JP, Kumar A. Comparative aspects of animal regeneration. Annu Rev Cell Dev Biol. 2008;24:525-549. doi:10.1146/annurev.cellbio.24.110707.175336
  2. Eming SA, Hammerschmidt M, Krieg T, et al. Interrelation of immunity and tissue repair or regeneration. Semin Cell Dev Biol. 2009;20:517-527. doi:10.1016/j.semcdb.2009.04.009
  3. Eming SA. Evolution of immune pathways in regeneration and repair: recent concepts and translational perspectives. Semin Immunol. 2014;26:275-276. doi:10.1016/j.smim.2014.09.001
  4. Bolognia JL, Jorizzo JJ, Schaffer JV, et al. Dermatology. 4th edition. Elsevier; 2018.
  5. Wang PH, Huang BS, Horng HC, et al. Wound healing. J Chin Med Assoc JCMA. 2018;81:94-101. doi:10.1016/j.jcma.2017.11.002
  6. Velnar T, Bailey T, Smrkolj V. The wound healing process: an overview of the cellular and molecular mechanisms. J Int Med Res. 2009;37:1528-1542. doi:10.1177/147323000903700531
  7. Gurtner GC, Werner S, Barrandon Y, et al. Wound repair and regeneration. Nature. 2008;453:314-321. doi:10.1038/nature07039
  8. Eming SA, Martin P, Tomic-Canic M. Wound repair and regeneration: mechanisms, signaling, and translation. Sci Transl Med. 2014;6:265sr6. doi:10.1126/scitranslmed.3009337
  9. Eming SA, Brachvogel B, Odorisio T, et al. Regulation of angiogenesis: wound healing as a model. Prog Histochem Cytochem. 2007;42:115-170. doi:10.1016/j.proghi.2007.06.001
  10. Janis JE, Harrison B. Wound healing: part I. basic science. Plast Reconstr Surg. 2016;138(3 suppl):9S-17S. doi:10.1097/PRS.0000000000002773
  11. Profyris C, Tziotzios C, Do Vale I. Cutaneous scarring: pathophysiology, molecular mechanisms, and scar reduction therapeutics. part I: the molecular basis of scar formation. J Am Acad Dermatol. 2012;66:1-10; quiz 11-12. doi:10.1016/j.jaad.2011.05.055
  12. Kwan P, Ding J, Tredget EE. MicroRNA 181b regulates decorin production by dermal fibroblasts and may be a potential therapy for hypertrophic scar. PLoS One. 2015;10:e0123054. doi:10.1371/journal.pone.0123054
  13. Ben W, Yang Y, Yuan J, et al. Human papillomavirus 16 E6 modulates the expression of host microRNAs in cervical cancer. Taiwan J Obstet Gynecol. 2015;54:364-370. doi:10.1016/j.tjog.2014.06.007
  14. Yu EH, Tu HF, Wu CH, et al. MicroRNA-21 promotes perineural invasion and impacts survival in patients with oral carcinoma. J Chin Med Assoc JCMA. 2017;80:383-388. doi:10.1016/j.jcma.2017.01.003
  15. Wen KC, Sung PL, Yen MS, et al. MicroRNAs regulate several functions of normal tissues and malignancies. Taiwan J Obstet Gynecol. 2013;52:465-469. doi:10.1016/j.tjog.2013.10.002
  16. Babalola O, Mamalis A, Lev-Tov H, et al. The role of microRNAs in skin fibrosis. Arch Dermatol Res. 2013;305:763-776. doi:10.1007/s00403-013-1410-1
  17. Hofer M, Hoferová Z, Falk M. Pharmacological modulation of radiation damage. does it exist a chance for other substances than hematopoietic growth factors and cytokines? Int J Mol Sci. 2017;18:1385. doi:10.3390/ijms18071385
  18. Darby IA, Weller CD. Aspirin treatment for chronic wounds: potential beneficial and inhibitory effects. Wound Repair Regen. 2017;25:7-12. doi:10.1111/wrr.12502
  19. Khalid KA, Nawi AFM, Zulkifli N, et al. Aging and wound healing of the skin: a review of clinical and pathophysiological hallmarks. Life. 2022;12:2142. doi:10.3390/life12122142
  20. Peacock HM, Gilbert EAB, Vickaryous MK. Scar‐free cutaneous wound healing in the leopard gecko, Eublepharis macularius. J Anat. 2015;227:596-610. doi:10.1111/joa.12368
  21. Delorme SL, Lungu IM, Vickaryous MK. Scar‐free wound healing and regeneration following tail loss in the leopard gecko, Eublepharis macularius. Anat Rec. 2012;295:1575-1595. doi:10.1002/ar.22490
  22. Brunauer R, Xia IG, Asrar SN, et al. Aging delays epimorphic regeneration in mice. J Gerontol Ser A Biol Sci Med Sci. 2021;76:1726-1733. doi:10.1093/gerona/glab131
  23. Dolan CP, Yang TJ, Zimmel K, et al. Epimorphic regeneration of the mouse digit tip is finite. Stem Cell Res Ther. 2022;13:62. doi:10.1186/s13287-022-02741-2
  24. Simkin J, Han M, Yu L, et al. The mouse digit tip: from wound healing to regeneration. Methods Mol Biol Clifton NJ. 2013;1037:419-435. doi:10.1007/978-1-62703-505-7_24
  25. Ziegler-Graham K, MacKenzie EJ, Ephraim PL, et al. Estimating the prevalence of limb loss in the United States: 2005 to 2050. Arch Phys Med Rehabil. 2008;89:422-429. doi:10.1016/j.apmr.2007.11.005
  26. Dudek NL, Marks MB, Marshall SC, et al. Dermatologic conditions associated with use of a lower-extremity prosthesis. Arch Phys Med Rehabil. 2005;86:659-663. doi:10.1016/j.apmr.2004.09.003
  27. Lannan FM, Meyerle JH. The dermatologist’s role in amputee skin care. Cutis. 2019;103:86-90.
  28. Dougherty AL, Mohrle CR, Galarneau MR, et al. Battlefield extremity injuries in Operation Iraqi Freedom. Injury. 2009;40:772-777. doi:10.1016/j.injury.2009.02.014
  29. Epstein RA, Heinemann AW, McFarland LV. Quality of life for veterans and servicemembers with major traumatic limb loss from Vietnam and OIF/OEF conflicts. J Rehabil Res Dev. 2010;47:373-385. doi:10.1682/jrrd.2009.03.0023
  30. Pinzur MS, Gold J, Schwartz D, et al. Energy demands for walking in dysvascular amputees as related to the level of amputation. Orthopedics. 1992;15:1033-1036; discussion 1036-1037. doi:10.3928/0147-7447-19920901-07
  31. Robinson V, Sansam K, Hirst L, et al. Major lower limb amputation–what, why and how to achieve the best results. Orthop Trauma. 2010;24:276-285. doi:10.1016/j.mporth.2010.03.017
  32. Lu S, Wang C, Zhong W, et al. Amputation stump revision using a free sural neurocutaneous perforator flap. Ann Plast Surg. 2016;76:83-87. doi:10.1097/SAP.0000000000000211
  33. Kim SW, Jeon SB, Hwang KT, et al. Coverage of amputation stumps using a latissimus dorsi flap with a serratus anterior muscle flap: a comparative study. Ann Plast Surg. 2016;76:88-93. doi:10.1097/SAP.0000000000000220
  34. Pavey GJ, Formby PM, Hoyt BW, et al. Intrawound antibiotic powder decreases frequency of deep infection and severity of heterotopic ossification in combat lower extremity amputations. Clin Orthop. 2019;477:802-810. doi:10.1007/s11999.0000000000000090
  35. Dunkel N, Belaieff W, Assal M, et al. Wound dehiscence and stump infection after lower limb amputation: risk factors and association with antibiotic use. J Orthop Sci Off J Jpn Orthop Assoc. 2012;17:588-594. doi:10.1007/s00776-012-0245-5
  36. Rubin G, Orbach H, Rinott M, et al. The use of prophylactic antibiotics in treatment of fingertip amputation: a randomized prospective trial. Am J Emerg Med. 2015;33:645-647. doi:10.1016/j.ajem.2015.02.002
  37. Azarbal AF, Harris S, Mitchell EL, et al. Nasal methicillin-resistant Staphylococcus aureus colonization is associated with increased wound occurrence after major lower extremity amputation. J Vasc Surg. 2015;62:401-405. doi:10.1016/j.jvs.2015.02.052
  38. Kwasniewski M, Mitchel D. Post amputation skin and wound care. Phys Med Rehabil Clin N Am. 2022;33:857-870. doi:10.1016/j.pmr.2022.06.010
  39. Chang H, Maldonado TS, Rockman CB, et al. Closed incision negative pressure wound therapy may decrease wound complications in major lower extremity amputations. J Vasc Surg. 2021;73:1041-1047. doi:10.1016/j.jvs.2020.07.061
  40. Kalliainen LK, Gordillo GM, Schlanger R, et al. Topical oxygen as an adjunct to wound healing: a clinical case series. Pathophysiol Off J Int Soc Pathophysiol. 2003;9:81-87. doi:10.1016/s0928-4680(02)00079-2
  41. Reichmann JP, Stevens PM, Rheinstein J, et al. Removable rigid dressings for postoperative management of transtibial amputations: a review of published evidence. PM R. 2018;10:516-523. doi:10.1016/j.pmrj.2017.10.002
  42. MacLean N, Fick GH. The effect of semirigid dressings on below-knee amputations. Phys Ther. 1994;74:668-673. doi:10.1093/ptj/74.7.668
  43. Koonalinthip N, Sukthongsa A, Janchai S. Comparison of removable rigid dressing and elastic bandage for residual limb maturation in transtibial amputees: a randomized controlled trial. Arch Phys Med Rehabil. 2020;101:1683-1688. doi:10.1016/j.apmr.2020.05.009
  44. Taylor L, Cavenett S, Stepien JM, et al. Removable rigid dressings: a retrospective case-note audit to determine the validity of post-amputation application. Prosthet Orthot Int. 2008;32:223-230. doi:10.1080/03093640802016795
  45. Sumpio B, Shine SR, Mahler D, et al. A comparison of immediate postoperative rigid and soft dressings for below-knee amputations. Ann Vasc Surg. 2013;27:774-780. doi:10.1016/j.avsg.2013.03.007
  46. van Velzen AD, Nederhand MJ, Emmelot CH, et al. Early treatment of trans-tibial amputees: retrospective analysis of early fitting and elastic bandaging. Prosthet Orthot Int. 2005;29:3-12. doi:10.1080/17461550500069588
  47. Chin T, Toda M. Results of prosthetic rehabilitation on managing transtibial vascular amputation with silicone liner after wound closure. J Int Med Res. 2016;44:957-967. doi:10.1177/0300060516647554
  48. Hoskins RD, Sutton EE, Kinor D, et al. Using vacuum-assisted suspension to manage residual limb wounds in persons with transtibial amputation: a case series. Prosthet Orthot Int. 2014;38:68-74. doi:10.1177/0309364613487547
  49. Johannesson A, Larsson GU, Oberg T, et al. Comparison of vacuum-formed removable rigid dressing with conventional rigid dressing after transtibial amputation: similar outcome in a randomized controlled trial involving 27 patients. Acta Orthop. 2008;79:361-369. doi:10.1080/17453670710015265
  50. Alsancak S, Köse SK, Altınkaynak H. Effect of elastic bandaging and prosthesis on the decrease in stump volume. Acta Orthop Traumatol Turc. 2011;45:14-22. doi:10.3944/AOTT.2011.2365
  51. Than MP, Smith RA, Hammond C, et al. Keratin-based wound care products for treatment of resistant vascular wounds. J Clin Aesthetic Dermatol. 2012;5:31-35.
  52. Gurtner GC, Garcia AD, Bakewell K, et al. A retrospective matched‐cohort study of 3994 lower extremity wounds of multiple etiologies across 644 institutions comparing a bioactive human skin allograft, TheraSkin, plus standard of care, to standard of care alone. Int Wound J. 2020;17:55-64. doi:10.1111/iwj.13231
  53. Buikema KES, Meyerle JH. Amputation stump: privileged harbor for infections, tumors, and immune disorders. Clin Dermatol. 2014;32:670-677. doi:10.1016/j.clindermatol.2014.04.015
  54. Yang NB, Garza LA, Foote CE, et al. High prevalence of stump dermatoses 38 years or more after amputation. Arch Dermatol. 2012;148:1283-1286. doi:10.1001/archdermatol.2012.3004
  55. Potter BK, Burns TC, Lacap AP, et al. Heterotopic ossification following traumatic and combat-related amputations. Prevalence, risk factors, and preliminary results of excision. J Bone Joint Surg Am. 2007;89:476-486. doi:10.2106/JBJS.F.00412
  56. Edwards DS, Kuhn KM, Potter BK, et al. Heterotopic ossification: a review of current understanding, treatment, and future. J Orthop Trauma. 2016;30(suppl 3):S27-S30. doi:10.1097/BOT.0000000000000666
  57. Tintle SM, Shawen SB, Forsberg JA, et al. Reoperation after combat-related major lower extremity amputations. J Orthop Trauma. 2014;28:232-237. doi:10.1097/BOT.0b013e3182a53130
  58. Bui KM, Raugi GJ, Nguyen VQ, et al. Skin problems in individuals with lower-limb loss: literature review and proposed classification system. J Rehabil Res Dev. 2009;46:1085-1090. doi:10.1682/jrrd.2009.04.0052
  59. Turan H, Bas¸kan EB, Adim SB, et al. Acroangiodermatitis in a below-knee amputation stump. Clin Exp Dermatol. 2011;36:560-561. doi:10.1111/j.1365-2230.2011.04037.x
  60. Lin CH, Ma H, Chung MT, et al. Granulomatous cutaneous lesions associated with risperidone-induced hyperprolactinemia in an amputated upper limb. Int J Dermatol. 2012;51:75-78. doi:10.1111/j.1365-4632.2011.04906.x
  61. Schwartz RA, Bagley MP, Janniger CK, et al. Verrucous carcinoma of a leg amputation stump. Dermatologica. 1991;182:193-195. doi:10.1159/000247782
  62. Campanati A, Diotallevi F, Radi G, et al. Efficacy and safety of botulinum toxin B in focal hyperhidrosis: a narrative review. Toxins. 2023;15:147. doi:10.3390/toxins15020147
  63. Anderson RR, Donelan MB, Hivnor C, et al. Laser treatment of traumatic scars with an emphasis on ablative fractional laser resurfacing: consensus report. JAMA Dermatol. 2014;150:187-193. doi:10.1001/jamadermatol.2013.7761
  64. McGrath M, McCarthy J, Gallego A, et al. The influence of perforated prosthetic liners on residual limb wound healing: a case report. Can Prosthet Orthot J. 2019;2:32723. doi:10.33137/cpoj.v2i1.32723
  65. Abu El Hawa AA, Klein D, Bekeny JC, et al. The impact of statins on wound healing: an ally in treating the highly comorbid patient. J Wound Care. 2022;31(suppl 2):S36-S41. doi:10.12968/jowc.2022.31.Sup2.S36
  66. Nasseri S, Sharifi M. Therapeutic potential of antimicrobial peptides for wound healing. Int J Pept Res Ther. 2022;28:38. doi:10.1007/s10989-021-10350-5
  67. Lee JV, Engel C, Tay S, et al. N-Acetyl-Cysteine treatment after lower extremity amputation improves areas of perfusion defect and wound healing outcomes. J Vasc Surg. 2021;73:39-40. doi:10.1016/j.jvs.2020.12.025
  68. Dong Y, Yang Q, Sun X. Comprehensive analysis of cell therapy on chronic skin wound healing: a meta-analysis. Hum Gene Ther. 2021;32:787-795. doi:10.1089/hum.2020.275
Article PDF
Author and Disclosure Information

From the Naval Medical Center San Diego, California.

The authors report no conflict of interest.

All authors are military service members. This work was prepared as part of their official duties. Title 17 U.S.C. 105 provides that “Copyright protection under this title is not available for any work of the United States Government.” Title 17 U.S.C. 101 defines a United States Government work as a work prepared by a military service member or employee of the United States Government as part of that person’s official duties.

The views expressed in this article are those of the authors and do not necessarily reflect the official policy or position of the Department of the Navy, the Department of Defense, or the US Government.

Correspondence: David S. Kirwin, MD, Naval Medical Center San Diego Dermatology Department, 1261 34th St, Unit 31, San Diego, CA 92102 (dsk247@cornell.edu).

Issue
Cutis - 113(3)
Publications
Topics
Page Number
125-131
Sections
Author and Disclosure Information

From the Naval Medical Center San Diego, California.

The authors report no conflict of interest.

All authors are military service members. This work was prepared as part of their official duties. Title 17 U.S.C. 105 provides that “Copyright protection under this title is not available for any work of the United States Government.” Title 17 U.S.C. 101 defines a United States Government work as a work prepared by a military service member or employee of the United States Government as part of that person’s official duties.

The views expressed in this article are those of the authors and do not necessarily reflect the official policy or position of the Department of the Navy, the Department of Defense, or the US Government.

Correspondence: David S. Kirwin, MD, Naval Medical Center San Diego Dermatology Department, 1261 34th St, Unit 31, San Diego, CA 92102 (dsk247@cornell.edu).

Author and Disclosure Information

From the Naval Medical Center San Diego, California.

The authors report no conflict of interest.

All authors are military service members. This work was prepared as part of their official duties. Title 17 U.S.C. 105 provides that “Copyright protection under this title is not available for any work of the United States Government.” Title 17 U.S.C. 101 defines a United States Government work as a work prepared by a military service member or employee of the United States Government as part of that person’s official duties.

The views expressed in this article are those of the authors and do not necessarily reflect the official policy or position of the Department of the Navy, the Department of Defense, or the US Government.

Correspondence: David S. Kirwin, MD, Naval Medical Center San Diego Dermatology Department, 1261 34th St, Unit 31, San Diego, CA 92102 (dsk247@cornell.edu).

Article PDF
Article PDF
IN PARTNERSHIP WITH THE ASSOCIATION OF MILITARY DERMATOLOGISTS
IN PARTNERSHIP WITH THE ASSOCIATION OF MILITARY DERMATOLOGISTS

Restoring skin integrity and balance after injury is vital for survival, serving as a crucial defense mechanism against potential infections by preventing the entry of harmful pathogens. Moreover, proper healing is essential for restoring normal tissue function, allowing damaged tissues to repair and, in an ideal scenario, regenerate. Timely healing helps reduce the risk for complications, such as chronic wounds, which could lead to more severe issues if left untreated. Additionally, pain relief often is associated with effective wound healing as inflammatory responses diminish during the repair process.

The immune system plays a pivotal role in wound healing, influencing various repair mechanisms and ultimately determining the extent of scarring. Although inflammation is present throughout the repair response, recent studies have challenged the conventional belief of an inverse correlation between the intensity of inflammation and regenerative capacity. Inflammatory signals were found to be crucial for timely repair and fundamental processes in regeneration, possibly presenting a paradigm shift in the understanding of immunology.1-4 The complexities of wound healing are exemplified when evaluating and treating postamputation wounds. To address such a task, one needs a firm understanding of the science behind healing wounds and what can go wrong along the way.

Phases of Wound Healing

Wound healing is a complex process that involves a series of sequential yet overlapping phases, including hemostasis/inflammation, proliferation, and remodeling.

Hemostasis/Inflammation—The initial stage of wound healing involves hemostasis, in which the primary objective is to prevent blood loss and initiate inflammation. Platelets arrive at the wound site, forming a provisional clot that is crucial for subsequent healing phases.4-6 Platelets halt bleeding as well as act as a medium for cell migration and adhesion; they also are a source of growth factors and proinflammatory cytokines that herald the inflammatory response.4-7

Inflammation is characterized by the infiltration of immune cells, particularly neutrophils and macrophages. Neutrophils act as the first line of defense, clearing debris and preventing infection. Macrophages follow, phagocytizing apoptotic cells and releasing growth factors such as tumor necrosis factor α, vascular endothelial growth factor, and matrix metalloprotease 9, which stimulate the next phase.4-6,8 Typically, the hemostasis and inflammatory phase starts approximately 6 to 8 hours after wound origin and lasts 3 to 4 days.4,6,7

Proliferation—Following hemostasis and inflammation, the wound transitions into the proliferation phase, which is marked by the development of granulation tissue—a dynamic amalgamation of fibroblasts, endothelial cells, and inflammatory cells.1,4-8 Fibroblasts play a central role in synthesizing collagen, the primary structural protein in connective tissue. They also orchestrate synthesis of vitronectin, fibronectin, fibrin, and tenascin.4-6,8 Simultaneously, angiogenesis takes place, involving the creation of new blood vessels to supply essential nutrients and oxygen to the healing tissue.4,7,9 Growth factors such as transforming growth factor β and vascular endothelial growth factor coordinate cellular activities and foster tissue repair.4-6,8 The proliferation phase extends over days to weeks, laying the groundwork for subsequent tissue restructuring.

Remodeling—The final stage of wound healing is remodeling, an extended process that may persist for several months or, in some cases, years. Throughout this phase, the initially deposited collagen, predominantly type III collagen, undergoes transformation into mature type I collagen.4-6,8 This transformation is critical for reinstating the tissue’s strength and functionality. The balance between collagen synthesis and degradation is delicate, regulated by matrix metalloproteinases and inhibitors of metalloproteinases.4-8 Fibroblasts, myofibroblasts, and other cells coordinate this intricate process of tissue reorganization.4-7

 

 

The eventual outcome of the remodeling phase determines the appearance and functionality of the healed tissue. Any disruption in this phase can lead to complications, such as chronic wounds and hypertrophic scars/keloids.4-6 These abnormal healing processes are characterized by localized inflammation, heightened fibroblast function, and excessive accumulation of the extracellular matrix.4-8

Molecular Mechanisms

Comprehensive investigations—both in vivo and in vitro—have explored the intricate molecular mechanisms involved in heightened wound healing. Transforming growth factor β takes center stage as a crucial factor, prompting the transformation of fibroblasts into myofibroblasts and contributing to the deposition of extracellular matrix.2,4-8,10 Transforming growth factor β activates non-Smad signaling pathways, such as MAPK (mitogen-activated protein kinase) and PI3K (phosphoinositide 3-kinase), influencing processes associated with fibrosis.5,11 Furthermore, microRNAs play a pivotal role in posttranscriptional regulation, influencing both transforming growth factor β signaling and fibroblast behavior.12-16

The involvement of prostaglandins is crucial in wound healing. Prostaglandin E2 plays a notable role and is positively correlated with the rate of wound healing.5 The cyclooxygenase pathway, pivotal for prostaglandin synthesis, becomes a target for inflammation control.4,5,10 Although aspirin and nonsteroidal anti-inflammatory drugs commonly are employed, their impact on wound healing remains controversial, as inhibition of cyclooxygenase may disrupt normal repair processes.5,17,18

Wound healing exhibits variations depending on age. Fetal skin regeneration is marked by the restoration of normal dermal architecture, including adnexal structures, nerves, vessels, and muscle.4-6 The distinctive characteristics of fetal wound healing include a unique profile of growth factors, a diminished inflammatory response, reduced biomechanical stress, and a distinct extracellular matrix composition.19 These factors contribute to a lower propensity for scar formation compared to the healing processes observed in adults. Fetal and adult wound healing differ fundamentally in their extracellular matrix composition, inflammatory cells, and cytokine levels.4-6,19 Adult wounds feature myofibroblasts, which are absent in fetal wounds, contributing to heightened mechanical tension.5 Delving deeper into the biochemical basis of fetal wound healing holds promise for mitigating scar formation in adults.

Takeaways From Other Species

Much of the biochemical knowledge of wound healing, especially regenerative wound healing, is known from other species. Geckos provide a unique model for studying regenerative repair in tails and nonregenerative healing in limbs after amputation. Scar-free wound healing is characterized by rapid wound closure, delayed blood vessel development, and collagen deposition, which contrasts with the hypervascular granulation tissue seen in scarring wounds.20 Scar-free wound healing and regeneration are intrinsic properties of the lizard tail and are unaffected by the location or method of detachment.21

Compared to amphibians with extraordinary regenerative capacity, data suggest the lack of regenerative capacity in mammals may come from a desynchronization of the fine-tuned interplay of progenitor cells such as blastema and differentiated cells.22,23 In mice, the response to amputation is specific to the level: cutting through the distal third of the terminal phalanx elicits a regeneration response, yielding a new digit tip resembling the lost one, while an amputation through the distal third of the intermediate phalanx triggers a wound healing and scarring response.24

Wound Healing Following Limb Amputation

Limb amputation represents a profound change in an individual’s life, impacting daily activities and overall well-being. There are many causes of amputation, but the most common include cardiovascular diseases, diabetes mellitus, cancer, and trauma.25-27 Trauma represents a relatively common cause within the US Military due to the overall young population as well as inherent risks of uniformed service.25,27 Advances in protective gear and combat casualty care have led to an increased number of individuals surviving with extremity injuries requiring amputation, particularly among younger service members, with a subgroup experiencing multiple amputations.27-29

 

 

Numerous factors play a crucial role in the healing and function of postamputation wounds. The level of amputation is a key determinant influencing both functional outcomes and the healing process. Achieving a balance between preserving function and removing damaged tissue is essential. A study investigating cardiac function and oxygen consumption in 25 patients with peripheral vascular disease found higher-level amputations resulted in decreased walking speed and cadence, along with increased oxygen consumption per meter walked.30

Selecting the appropriate amputation level is vital to optimize functional outcomes without compromising wound healing. Successful prosthetic limb fitting depends largely on the length of the residual stump to support the body load and suspend the prosthesis. For long bone amputations, maintaining at least 12-cm clearance above the knee joint in transfemoral amputees and 10-cm below the knee joint in transtibial amputees is critical for maximizing functional outcomes.31

Surgical technique also is paramount. The goal is to minimize the risk for pressure ulcers by avoiding bony spurs and muscle imbalances. Shaping the muscle and residual limb is essential for proper prosthesis fitting. Attention to neurovascular structures, such as burying nerve ends to prevent neuropathic pain during prosthesis wear, is crucial.32 In extremity amputations, surgeons often resort to free flap transfer techniques for stump reconstruction. In a study of 31 patients with severe lower extremity injuries undergoing various amputations, the use of latissimus dorsi myocutaneous flaps, alone or in combination with serratus anterior muscle flaps, resulted in fewer instances of deep ulceration and allowed for earlier prosthesis wear.33

Addressing Barriers to Wound Healing

Multiple barriers to successful wound healing are encountered in the amputee population. Amputations from trauma have a less-controlled initiation, which carries with it a higher risk for infection, poor wound healing, and other complications.

Infection—Infection often is one of the first hurdles encountered in postamputation wound healing. Critical first steps in infection prevention include thorough cleaning of soiled traumatic wounds and appropriate tissue debridement coupled with scrupulous sterile technique and postoperative monitoring for signs and symptoms of infection.

In a retrospective study of 223 combat-related major lower extremity amputations (initial and revision) between 2009 and 2015, the use of intrawound antibiotic powder at the time of closure demonstrated a 13% absolute risk reduction in deep infection rates, which was particularly notable in revision amputations, with a number needed to treat of 8 for initial amputations and 4 for revision amputations on previously infected limbs.34 Intra-operative antibiotic powder may represent a cheap and easy consideration for this special population of amputees. Postamputation antibiotic prophylaxis for infection prevention is an area of controversy. For nontraumatic infections, data suggest antibiotic prophylaxis may not decrease infection rates in these patients.35,36

Interestingly, a study by Azarbal et al37 aimed to investigate the correlation between nasal methicillin-resistant Staphylococcus aureus (MRSA) colonization and other patient factors with wound occurrence following major lower extremity amputation. The study found MRSA colonization was associated with higher rates of overall wound occurrence as well as wound occurrence due to wound infection. These data suggest nasal MRSA eradication may improve postoperative wound outcomes after major lower extremity amputation.37

 

 

Dressing Choice—The dressing chosen for a residual limb also is of paramount importance following amputation. The personalized and dynamic management of postamputation wounds and skin involves achieving optimal healing through a dressing that sustains appropriate moisture levels, addresses edema, helps prevent contractures, and safeguards the limb.38 From the start, using negative pressure wound dressings after surgical amputation can decrease wound-related complications.39

Topical oxygen therapy following amputation also shows promise. In a retrospective case series by Kalliainen et al,40 topical oxygen therapy applied to 58 wounds in 32 patients over 9 months demonstrated positive outcomes in promoting wound healing, with 38 wounds (66%) healing completely with the use of topical oxygen. Minimal complications and no detrimental effects were observed.40

Current recommendations suggest that non–weight-bearing removable rigid dressings are the superior postoperative management for transtibial amputations compared to soft dressings, offering benefits such as faster healing, reduced limb edema, earlier ambulation, preparatory shaping for prosthetic use, and prevention of knee flexion contractures.41-46 Similarly, adding a silicone liner following amputation significantly reduced the duration of prosthetic rehabilitation compared with a conventional soft dressing program in one study (P<.05).47

Specifically targeting wound edema, a case series by Hoskins et al48 investigated the impact of prostheses with vacuum-assisted suspension on the size of residual limb wounds in individuals with transtibial amputation. Well-fitting sockets with vacuum-assisted suspension did not impede wound healing, and the results suggest the potential for continued prosthesis use during the healing process.48 However, a study by Johannesson et al49 compared the outcomes of transtibial amputation patients using a vacuum-formed rigid dressing and a conventional rigid plaster dressing, finding no significant differences in wound healing, time to prosthetic fitting, or functional outcomes with the prosthesis between the 2 groups. When comparing elastic bandaging, pneumatic prosthesis, and temporary prosthesis on postoperative stump management, temporary prosthesis led to a decrease in stump volume, quicker transition to a permanent prosthesis, and improved quality of life compared with elastic bandaging and pneumatic prosthetics.50

The type of material in dressings may contribute to utility in amputation wounds. Keratin-based wound dressings show promise for wound healing, especially in recalcitrant vascular wounds.51 There also are numerous proprietary wound dressings available for patients, at least one of which has particularly thorough data. In a retrospective study of more than 2 million lower extremity wounds across 644 institutions, a proprietary bioactive human skin allograft (TheraSkin [LifeNet Health]) demonstrated higher healing rates, greater percentage area reductions, lower amputations, reduced recidivism, higher treatment completion, and fewer medical transfers compared with standard of care alone.52

Postamputation Dermatologic Concerns

After the postamputation wound heals, a notable concern is the prevalence of skin diseases affecting residual limbs. The stump site in amputees, marked by a delicate cutaneous landscape vulnerable to skin diseases, faces challenges arising from amputation-induced damage to various structures.53

When integrated into a prosthesis socket, the altered skin must acclimate to a humid environment and endure forces for which it is not well suited, especially during movement.53 Amputation remarkably alters normal tissue perfusion, which can lead to aberrant blood and lymphatic circulation in residual limbs.27,53 This compromised skin, often associated with a history of vascular disease, diabetes mellitus, or malignancy, becomes immunocompromised, heightening the risk for dermatologic issues such as inflammation, infection, and malignancies.53 Unlike the resilient volar skin on palms and soles, stump skin lacks adaptation to withstand the compressive forces generated during ambulation, sometimes leading to skin disease and pain that result in abandonment of the prosthesis.53,54 Mechanical forces on the skin, especially in active patients eager to resume pre-injury lifestyles, contribute to skin breakdown. The dynamic nature of the residual limb, including muscle atrophy, gait changes, and weight fluctuations, complicates the prosthetic fitting process. Prosthesis abandonment remains a challenge, despite modern technologic advancements.

 

 

The occurrence of heterotopic ossification (extraskeletal bone formation) is another notable issue in military amputees.27,55-57 Poor prosthetic fit can lead to skin degradation, necessitating further surgery to address mispositioned bone formations. Orthopedic monitoring supplemented by appropriate imaging studies can benefit postamputation patients by detecting and preventing heterotopic ossification in its early stages.

Dermatologic issues, especially among lower limb amputees, are noteworthy, with a substantial percentage experiencing complications related to socket prosthetics, such as heat, sweating, sores, and skin irritation. Up to 41% of patients are seen regularly for a secondary skin disorder following amputation.58 As one might expect, persistent wounds, blisters, ulcers, and abscesses are some of the most typical cutaneous abnormalities affecting residual limbs with prostheses.27,58 More rare skin conditions also are documented in residual limbs, including cutaneous granuloma, verrucous carcinoma, bullous pemphigoid, and angiodermatitis.27,59-61

Treatments offered in the dermatology clinic often are similar to patients who have not had an amputation. For instance, hyperhidrosis can be treated with prescription antiperspirant, topical aluminum chloride, topical glycopyrronium, botulinum toxin, and iontophoresis, which can greatly decrease skin irritation and malodor. Subcutaneous neurotoxins such as botulinum toxin are especially useful for hyperhidrosis following amputation because a single treatment can last 3 to 6 months, whereas topicals must be applied multiple times per day and can be inherently irritating to the skin.27,62 Furthermore, ablative fractional resurfacing lasers also can help stimulate new collagen growth, increase skin mobility on residual limbs, smooth jagged scars, and aid prosthetic fitting.27,63 Perforated prosthetic liners also may be useful to address issues such as excessive sweating, demonstrating improvements in skin health, reduced sweating problems, and potential avoidance of surgical interventions.64

When comorbid skin conditions are at bay, preventive measures for excessive wound healing necessitate early recognition and timely intervention for residual limbs. Preventive techniques encompass the use of silicone gel sheeting, hypoallergenic microporous tape, and intralesional steroid injections.

Psychological Concerns—An overarching issue following amputation is the psychological toll the process imposes on the patient. Psychological concerns, including anxiety and depression, present additional challenges impacting residual limb hygiene and prosthetic maintenance. Chronic wounds are devastating to patients. These patients consistently express feeling ostracized from their community and anxious about unemployment, leaking fluid, or odor from the wound, as well as other social stigmata.62 Depression and anxiety can hinder a patient’s ability to care for their wound and make them more susceptible to the myriad issues that can ensue.

Recent Developments in Wound Healing

Wound healing is ripe for innovation that could assuage ailments that impact patients following amputation. A 2022 study by Abu El Hawa et al65 illustrated advanced progression in wound healing for patients taking statins, even though the statin group had increased age and number of comorbidities compared with patients not taking statins.

Nasseri and Sharifi66 showed the potential of antimicrobial peptides—small proteins with cationic charges and amphipathic structures exhibiting electrostatic interaction with microbial cell membranes—in promoting wound healing, particularly defensins and cathelicidin LL-37.They also discussed innovative delivery systems, such as nanoparticles and electrospun fibrous scaffolds, highlighting their potential as possibly more effective therapeutics than antibiotics, especially in the context of diabetic wound closure.66 Aimed at increased angiogenesis in the proliferative phase, there is evidence that N-acetylcysteine can increase amputation stump perfusion with the goal of better long-term wound healing and more efficient scar formation.67

Stem cell therapy, particularly employing cells from the human amniotic membrane, represents an auspicious avenue for antifibrotic treatment. Amniotic epithelial cells and amniotic mesenchymal cells, with their self-renewal and multilineage differentiation capabilities, exhibit anti-inflammatory and antifibrotic properties.4,5 A study by Dong et al68 aimed to assess the efficacy of cell therapy, particularly differentiated progenitor cell–based graft transplantation or autologous stem cell injection, in treating refractory skin injuries such as nonrevascularizable critical limb ischemic ulcers, venous leg ulcers, and diabetic lower limb ulcers. The findings demonstrated cell therapy effectively reduced the size of ulcers, improved wound closure rates, and decreased major amputation rates compared with standard therapy. Of note, cell therapy had limited impact on alleviating pain in patients with critical limb ischemia-related cutaneous ulcers.68

Final Thoughts

Wound care following amputation is a multidisciplinary endeavor, necessitating collaboration between many health care professionals. Dermatologists play a crucial role in providing routine care as well as addressing wound healing and related skin issues among amputation patients. As the field progresses, dermatologists are well positioned to make notable contributions and ensure enhanced outcomes, resulting in a better quality of life for patients facing the challenges of limb amputation and prosthetic use.

Restoring skin integrity and balance after injury is vital for survival, serving as a crucial defense mechanism against potential infections by preventing the entry of harmful pathogens. Moreover, proper healing is essential for restoring normal tissue function, allowing damaged tissues to repair and, in an ideal scenario, regenerate. Timely healing helps reduce the risk for complications, such as chronic wounds, which could lead to more severe issues if left untreated. Additionally, pain relief often is associated with effective wound healing as inflammatory responses diminish during the repair process.

The immune system plays a pivotal role in wound healing, influencing various repair mechanisms and ultimately determining the extent of scarring. Although inflammation is present throughout the repair response, recent studies have challenged the conventional belief of an inverse correlation between the intensity of inflammation and regenerative capacity. Inflammatory signals were found to be crucial for timely repair and fundamental processes in regeneration, possibly presenting a paradigm shift in the understanding of immunology.1-4 The complexities of wound healing are exemplified when evaluating and treating postamputation wounds. To address such a task, one needs a firm understanding of the science behind healing wounds and what can go wrong along the way.

Phases of Wound Healing

Wound healing is a complex process that involves a series of sequential yet overlapping phases, including hemostasis/inflammation, proliferation, and remodeling.

Hemostasis/Inflammation—The initial stage of wound healing involves hemostasis, in which the primary objective is to prevent blood loss and initiate inflammation. Platelets arrive at the wound site, forming a provisional clot that is crucial for subsequent healing phases.4-6 Platelets halt bleeding as well as act as a medium for cell migration and adhesion; they also are a source of growth factors and proinflammatory cytokines that herald the inflammatory response.4-7

Inflammation is characterized by the infiltration of immune cells, particularly neutrophils and macrophages. Neutrophils act as the first line of defense, clearing debris and preventing infection. Macrophages follow, phagocytizing apoptotic cells and releasing growth factors such as tumor necrosis factor α, vascular endothelial growth factor, and matrix metalloprotease 9, which stimulate the next phase.4-6,8 Typically, the hemostasis and inflammatory phase starts approximately 6 to 8 hours after wound origin and lasts 3 to 4 days.4,6,7

Proliferation—Following hemostasis and inflammation, the wound transitions into the proliferation phase, which is marked by the development of granulation tissue—a dynamic amalgamation of fibroblasts, endothelial cells, and inflammatory cells.1,4-8 Fibroblasts play a central role in synthesizing collagen, the primary structural protein in connective tissue. They also orchestrate synthesis of vitronectin, fibronectin, fibrin, and tenascin.4-6,8 Simultaneously, angiogenesis takes place, involving the creation of new blood vessels to supply essential nutrients and oxygen to the healing tissue.4,7,9 Growth factors such as transforming growth factor β and vascular endothelial growth factor coordinate cellular activities and foster tissue repair.4-6,8 The proliferation phase extends over days to weeks, laying the groundwork for subsequent tissue restructuring.

Remodeling—The final stage of wound healing is remodeling, an extended process that may persist for several months or, in some cases, years. Throughout this phase, the initially deposited collagen, predominantly type III collagen, undergoes transformation into mature type I collagen.4-6,8 This transformation is critical for reinstating the tissue’s strength and functionality. The balance between collagen synthesis and degradation is delicate, regulated by matrix metalloproteinases and inhibitors of metalloproteinases.4-8 Fibroblasts, myofibroblasts, and other cells coordinate this intricate process of tissue reorganization.4-7

 

 

The eventual outcome of the remodeling phase determines the appearance and functionality of the healed tissue. Any disruption in this phase can lead to complications, such as chronic wounds and hypertrophic scars/keloids.4-6 These abnormal healing processes are characterized by localized inflammation, heightened fibroblast function, and excessive accumulation of the extracellular matrix.4-8

Molecular Mechanisms

Comprehensive investigations—both in vivo and in vitro—have explored the intricate molecular mechanisms involved in heightened wound healing. Transforming growth factor β takes center stage as a crucial factor, prompting the transformation of fibroblasts into myofibroblasts and contributing to the deposition of extracellular matrix.2,4-8,10 Transforming growth factor β activates non-Smad signaling pathways, such as MAPK (mitogen-activated protein kinase) and PI3K (phosphoinositide 3-kinase), influencing processes associated with fibrosis.5,11 Furthermore, microRNAs play a pivotal role in posttranscriptional regulation, influencing both transforming growth factor β signaling and fibroblast behavior.12-16

The involvement of prostaglandins is crucial in wound healing. Prostaglandin E2 plays a notable role and is positively correlated with the rate of wound healing.5 The cyclooxygenase pathway, pivotal for prostaglandin synthesis, becomes a target for inflammation control.4,5,10 Although aspirin and nonsteroidal anti-inflammatory drugs commonly are employed, their impact on wound healing remains controversial, as inhibition of cyclooxygenase may disrupt normal repair processes.5,17,18

Wound healing exhibits variations depending on age. Fetal skin regeneration is marked by the restoration of normal dermal architecture, including adnexal structures, nerves, vessels, and muscle.4-6 The distinctive characteristics of fetal wound healing include a unique profile of growth factors, a diminished inflammatory response, reduced biomechanical stress, and a distinct extracellular matrix composition.19 These factors contribute to a lower propensity for scar formation compared to the healing processes observed in adults. Fetal and adult wound healing differ fundamentally in their extracellular matrix composition, inflammatory cells, and cytokine levels.4-6,19 Adult wounds feature myofibroblasts, which are absent in fetal wounds, contributing to heightened mechanical tension.5 Delving deeper into the biochemical basis of fetal wound healing holds promise for mitigating scar formation in adults.

Takeaways From Other Species

Much of the biochemical knowledge of wound healing, especially regenerative wound healing, is known from other species. Geckos provide a unique model for studying regenerative repair in tails and nonregenerative healing in limbs after amputation. Scar-free wound healing is characterized by rapid wound closure, delayed blood vessel development, and collagen deposition, which contrasts with the hypervascular granulation tissue seen in scarring wounds.20 Scar-free wound healing and regeneration are intrinsic properties of the lizard tail and are unaffected by the location or method of detachment.21

Compared to amphibians with extraordinary regenerative capacity, data suggest the lack of regenerative capacity in mammals may come from a desynchronization of the fine-tuned interplay of progenitor cells such as blastema and differentiated cells.22,23 In mice, the response to amputation is specific to the level: cutting through the distal third of the terminal phalanx elicits a regeneration response, yielding a new digit tip resembling the lost one, while an amputation through the distal third of the intermediate phalanx triggers a wound healing and scarring response.24

Wound Healing Following Limb Amputation

Limb amputation represents a profound change in an individual’s life, impacting daily activities and overall well-being. There are many causes of amputation, but the most common include cardiovascular diseases, diabetes mellitus, cancer, and trauma.25-27 Trauma represents a relatively common cause within the US Military due to the overall young population as well as inherent risks of uniformed service.25,27 Advances in protective gear and combat casualty care have led to an increased number of individuals surviving with extremity injuries requiring amputation, particularly among younger service members, with a subgroup experiencing multiple amputations.27-29

 

 

Numerous factors play a crucial role in the healing and function of postamputation wounds. The level of amputation is a key determinant influencing both functional outcomes and the healing process. Achieving a balance between preserving function and removing damaged tissue is essential. A study investigating cardiac function and oxygen consumption in 25 patients with peripheral vascular disease found higher-level amputations resulted in decreased walking speed and cadence, along with increased oxygen consumption per meter walked.30

Selecting the appropriate amputation level is vital to optimize functional outcomes without compromising wound healing. Successful prosthetic limb fitting depends largely on the length of the residual stump to support the body load and suspend the prosthesis. For long bone amputations, maintaining at least 12-cm clearance above the knee joint in transfemoral amputees and 10-cm below the knee joint in transtibial amputees is critical for maximizing functional outcomes.31

Surgical technique also is paramount. The goal is to minimize the risk for pressure ulcers by avoiding bony spurs and muscle imbalances. Shaping the muscle and residual limb is essential for proper prosthesis fitting. Attention to neurovascular structures, such as burying nerve ends to prevent neuropathic pain during prosthesis wear, is crucial.32 In extremity amputations, surgeons often resort to free flap transfer techniques for stump reconstruction. In a study of 31 patients with severe lower extremity injuries undergoing various amputations, the use of latissimus dorsi myocutaneous flaps, alone or in combination with serratus anterior muscle flaps, resulted in fewer instances of deep ulceration and allowed for earlier prosthesis wear.33

Addressing Barriers to Wound Healing

Multiple barriers to successful wound healing are encountered in the amputee population. Amputations from trauma have a less-controlled initiation, which carries with it a higher risk for infection, poor wound healing, and other complications.

Infection—Infection often is one of the first hurdles encountered in postamputation wound healing. Critical first steps in infection prevention include thorough cleaning of soiled traumatic wounds and appropriate tissue debridement coupled with scrupulous sterile technique and postoperative monitoring for signs and symptoms of infection.

In a retrospective study of 223 combat-related major lower extremity amputations (initial and revision) between 2009 and 2015, the use of intrawound antibiotic powder at the time of closure demonstrated a 13% absolute risk reduction in deep infection rates, which was particularly notable in revision amputations, with a number needed to treat of 8 for initial amputations and 4 for revision amputations on previously infected limbs.34 Intra-operative antibiotic powder may represent a cheap and easy consideration for this special population of amputees. Postamputation antibiotic prophylaxis for infection prevention is an area of controversy. For nontraumatic infections, data suggest antibiotic prophylaxis may not decrease infection rates in these patients.35,36

Interestingly, a study by Azarbal et al37 aimed to investigate the correlation between nasal methicillin-resistant Staphylococcus aureus (MRSA) colonization and other patient factors with wound occurrence following major lower extremity amputation. The study found MRSA colonization was associated with higher rates of overall wound occurrence as well as wound occurrence due to wound infection. These data suggest nasal MRSA eradication may improve postoperative wound outcomes after major lower extremity amputation.37

 

 

Dressing Choice—The dressing chosen for a residual limb also is of paramount importance following amputation. The personalized and dynamic management of postamputation wounds and skin involves achieving optimal healing through a dressing that sustains appropriate moisture levels, addresses edema, helps prevent contractures, and safeguards the limb.38 From the start, using negative pressure wound dressings after surgical amputation can decrease wound-related complications.39

Topical oxygen therapy following amputation also shows promise. In a retrospective case series by Kalliainen et al,40 topical oxygen therapy applied to 58 wounds in 32 patients over 9 months demonstrated positive outcomes in promoting wound healing, with 38 wounds (66%) healing completely with the use of topical oxygen. Minimal complications and no detrimental effects were observed.40

Current recommendations suggest that non–weight-bearing removable rigid dressings are the superior postoperative management for transtibial amputations compared to soft dressings, offering benefits such as faster healing, reduced limb edema, earlier ambulation, preparatory shaping for prosthetic use, and prevention of knee flexion contractures.41-46 Similarly, adding a silicone liner following amputation significantly reduced the duration of prosthetic rehabilitation compared with a conventional soft dressing program in one study (P<.05).47

Specifically targeting wound edema, a case series by Hoskins et al48 investigated the impact of prostheses with vacuum-assisted suspension on the size of residual limb wounds in individuals with transtibial amputation. Well-fitting sockets with vacuum-assisted suspension did not impede wound healing, and the results suggest the potential for continued prosthesis use during the healing process.48 However, a study by Johannesson et al49 compared the outcomes of transtibial amputation patients using a vacuum-formed rigid dressing and a conventional rigid plaster dressing, finding no significant differences in wound healing, time to prosthetic fitting, or functional outcomes with the prosthesis between the 2 groups. When comparing elastic bandaging, pneumatic prosthesis, and temporary prosthesis on postoperative stump management, temporary prosthesis led to a decrease in stump volume, quicker transition to a permanent prosthesis, and improved quality of life compared with elastic bandaging and pneumatic prosthetics.50

The type of material in dressings may contribute to utility in amputation wounds. Keratin-based wound dressings show promise for wound healing, especially in recalcitrant vascular wounds.51 There also are numerous proprietary wound dressings available for patients, at least one of which has particularly thorough data. In a retrospective study of more than 2 million lower extremity wounds across 644 institutions, a proprietary bioactive human skin allograft (TheraSkin [LifeNet Health]) demonstrated higher healing rates, greater percentage area reductions, lower amputations, reduced recidivism, higher treatment completion, and fewer medical transfers compared with standard of care alone.52

Postamputation Dermatologic Concerns

After the postamputation wound heals, a notable concern is the prevalence of skin diseases affecting residual limbs. The stump site in amputees, marked by a delicate cutaneous landscape vulnerable to skin diseases, faces challenges arising from amputation-induced damage to various structures.53

When integrated into a prosthesis socket, the altered skin must acclimate to a humid environment and endure forces for which it is not well suited, especially during movement.53 Amputation remarkably alters normal tissue perfusion, which can lead to aberrant blood and lymphatic circulation in residual limbs.27,53 This compromised skin, often associated with a history of vascular disease, diabetes mellitus, or malignancy, becomes immunocompromised, heightening the risk for dermatologic issues such as inflammation, infection, and malignancies.53 Unlike the resilient volar skin on palms and soles, stump skin lacks adaptation to withstand the compressive forces generated during ambulation, sometimes leading to skin disease and pain that result in abandonment of the prosthesis.53,54 Mechanical forces on the skin, especially in active patients eager to resume pre-injury lifestyles, contribute to skin breakdown. The dynamic nature of the residual limb, including muscle atrophy, gait changes, and weight fluctuations, complicates the prosthetic fitting process. Prosthesis abandonment remains a challenge, despite modern technologic advancements.

 

 

The occurrence of heterotopic ossification (extraskeletal bone formation) is another notable issue in military amputees.27,55-57 Poor prosthetic fit can lead to skin degradation, necessitating further surgery to address mispositioned bone formations. Orthopedic monitoring supplemented by appropriate imaging studies can benefit postamputation patients by detecting and preventing heterotopic ossification in its early stages.

Dermatologic issues, especially among lower limb amputees, are noteworthy, with a substantial percentage experiencing complications related to socket prosthetics, such as heat, sweating, sores, and skin irritation. Up to 41% of patients are seen regularly for a secondary skin disorder following amputation.58 As one might expect, persistent wounds, blisters, ulcers, and abscesses are some of the most typical cutaneous abnormalities affecting residual limbs with prostheses.27,58 More rare skin conditions also are documented in residual limbs, including cutaneous granuloma, verrucous carcinoma, bullous pemphigoid, and angiodermatitis.27,59-61

Treatments offered in the dermatology clinic often are similar to patients who have not had an amputation. For instance, hyperhidrosis can be treated with prescription antiperspirant, topical aluminum chloride, topical glycopyrronium, botulinum toxin, and iontophoresis, which can greatly decrease skin irritation and malodor. Subcutaneous neurotoxins such as botulinum toxin are especially useful for hyperhidrosis following amputation because a single treatment can last 3 to 6 months, whereas topicals must be applied multiple times per day and can be inherently irritating to the skin.27,62 Furthermore, ablative fractional resurfacing lasers also can help stimulate new collagen growth, increase skin mobility on residual limbs, smooth jagged scars, and aid prosthetic fitting.27,63 Perforated prosthetic liners also may be useful to address issues such as excessive sweating, demonstrating improvements in skin health, reduced sweating problems, and potential avoidance of surgical interventions.64

When comorbid skin conditions are at bay, preventive measures for excessive wound healing necessitate early recognition and timely intervention for residual limbs. Preventive techniques encompass the use of silicone gel sheeting, hypoallergenic microporous tape, and intralesional steroid injections.

Psychological Concerns—An overarching issue following amputation is the psychological toll the process imposes on the patient. Psychological concerns, including anxiety and depression, present additional challenges impacting residual limb hygiene and prosthetic maintenance. Chronic wounds are devastating to patients. These patients consistently express feeling ostracized from their community and anxious about unemployment, leaking fluid, or odor from the wound, as well as other social stigmata.62 Depression and anxiety can hinder a patient’s ability to care for their wound and make them more susceptible to the myriad issues that can ensue.

Recent Developments in Wound Healing

Wound healing is ripe for innovation that could assuage ailments that impact patients following amputation. A 2022 study by Abu El Hawa et al65 illustrated advanced progression in wound healing for patients taking statins, even though the statin group had increased age and number of comorbidities compared with patients not taking statins.

Nasseri and Sharifi66 showed the potential of antimicrobial peptides—small proteins with cationic charges and amphipathic structures exhibiting electrostatic interaction with microbial cell membranes—in promoting wound healing, particularly defensins and cathelicidin LL-37.They also discussed innovative delivery systems, such as nanoparticles and electrospun fibrous scaffolds, highlighting their potential as possibly more effective therapeutics than antibiotics, especially in the context of diabetic wound closure.66 Aimed at increased angiogenesis in the proliferative phase, there is evidence that N-acetylcysteine can increase amputation stump perfusion with the goal of better long-term wound healing and more efficient scar formation.67

Stem cell therapy, particularly employing cells from the human amniotic membrane, represents an auspicious avenue for antifibrotic treatment. Amniotic epithelial cells and amniotic mesenchymal cells, with their self-renewal and multilineage differentiation capabilities, exhibit anti-inflammatory and antifibrotic properties.4,5 A study by Dong et al68 aimed to assess the efficacy of cell therapy, particularly differentiated progenitor cell–based graft transplantation or autologous stem cell injection, in treating refractory skin injuries such as nonrevascularizable critical limb ischemic ulcers, venous leg ulcers, and diabetic lower limb ulcers. The findings demonstrated cell therapy effectively reduced the size of ulcers, improved wound closure rates, and decreased major amputation rates compared with standard therapy. Of note, cell therapy had limited impact on alleviating pain in patients with critical limb ischemia-related cutaneous ulcers.68

Final Thoughts

Wound care following amputation is a multidisciplinary endeavor, necessitating collaboration between many health care professionals. Dermatologists play a crucial role in providing routine care as well as addressing wound healing and related skin issues among amputation patients. As the field progresses, dermatologists are well positioned to make notable contributions and ensure enhanced outcomes, resulting in a better quality of life for patients facing the challenges of limb amputation and prosthetic use.

References
  1. Brockes JP, Kumar A. Comparative aspects of animal regeneration. Annu Rev Cell Dev Biol. 2008;24:525-549. doi:10.1146/annurev.cellbio.24.110707.175336
  2. Eming SA, Hammerschmidt M, Krieg T, et al. Interrelation of immunity and tissue repair or regeneration. Semin Cell Dev Biol. 2009;20:517-527. doi:10.1016/j.semcdb.2009.04.009
  3. Eming SA. Evolution of immune pathways in regeneration and repair: recent concepts and translational perspectives. Semin Immunol. 2014;26:275-276. doi:10.1016/j.smim.2014.09.001
  4. Bolognia JL, Jorizzo JJ, Schaffer JV, et al. Dermatology. 4th edition. Elsevier; 2018.
  5. Wang PH, Huang BS, Horng HC, et al. Wound healing. J Chin Med Assoc JCMA. 2018;81:94-101. doi:10.1016/j.jcma.2017.11.002
  6. Velnar T, Bailey T, Smrkolj V. The wound healing process: an overview of the cellular and molecular mechanisms. J Int Med Res. 2009;37:1528-1542. doi:10.1177/147323000903700531
  7. Gurtner GC, Werner S, Barrandon Y, et al. Wound repair and regeneration. Nature. 2008;453:314-321. doi:10.1038/nature07039
  8. Eming SA, Martin P, Tomic-Canic M. Wound repair and regeneration: mechanisms, signaling, and translation. Sci Transl Med. 2014;6:265sr6. doi:10.1126/scitranslmed.3009337
  9. Eming SA, Brachvogel B, Odorisio T, et al. Regulation of angiogenesis: wound healing as a model. Prog Histochem Cytochem. 2007;42:115-170. doi:10.1016/j.proghi.2007.06.001
  10. Janis JE, Harrison B. Wound healing: part I. basic science. Plast Reconstr Surg. 2016;138(3 suppl):9S-17S. doi:10.1097/PRS.0000000000002773
  11. Profyris C, Tziotzios C, Do Vale I. Cutaneous scarring: pathophysiology, molecular mechanisms, and scar reduction therapeutics. part I: the molecular basis of scar formation. J Am Acad Dermatol. 2012;66:1-10; quiz 11-12. doi:10.1016/j.jaad.2011.05.055
  12. Kwan P, Ding J, Tredget EE. MicroRNA 181b regulates decorin production by dermal fibroblasts and may be a potential therapy for hypertrophic scar. PLoS One. 2015;10:e0123054. doi:10.1371/journal.pone.0123054
  13. Ben W, Yang Y, Yuan J, et al. Human papillomavirus 16 E6 modulates the expression of host microRNAs in cervical cancer. Taiwan J Obstet Gynecol. 2015;54:364-370. doi:10.1016/j.tjog.2014.06.007
  14. Yu EH, Tu HF, Wu CH, et al. MicroRNA-21 promotes perineural invasion and impacts survival in patients with oral carcinoma. J Chin Med Assoc JCMA. 2017;80:383-388. doi:10.1016/j.jcma.2017.01.003
  15. Wen KC, Sung PL, Yen MS, et al. MicroRNAs regulate several functions of normal tissues and malignancies. Taiwan J Obstet Gynecol. 2013;52:465-469. doi:10.1016/j.tjog.2013.10.002
  16. Babalola O, Mamalis A, Lev-Tov H, et al. The role of microRNAs in skin fibrosis. Arch Dermatol Res. 2013;305:763-776. doi:10.1007/s00403-013-1410-1
  17. Hofer M, Hoferová Z, Falk M. Pharmacological modulation of radiation damage. does it exist a chance for other substances than hematopoietic growth factors and cytokines? Int J Mol Sci. 2017;18:1385. doi:10.3390/ijms18071385
  18. Darby IA, Weller CD. Aspirin treatment for chronic wounds: potential beneficial and inhibitory effects. Wound Repair Regen. 2017;25:7-12. doi:10.1111/wrr.12502
  19. Khalid KA, Nawi AFM, Zulkifli N, et al. Aging and wound healing of the skin: a review of clinical and pathophysiological hallmarks. Life. 2022;12:2142. doi:10.3390/life12122142
  20. Peacock HM, Gilbert EAB, Vickaryous MK. Scar‐free cutaneous wound healing in the leopard gecko, Eublepharis macularius. J Anat. 2015;227:596-610. doi:10.1111/joa.12368
  21. Delorme SL, Lungu IM, Vickaryous MK. Scar‐free wound healing and regeneration following tail loss in the leopard gecko, Eublepharis macularius. Anat Rec. 2012;295:1575-1595. doi:10.1002/ar.22490
  22. Brunauer R, Xia IG, Asrar SN, et al. Aging delays epimorphic regeneration in mice. J Gerontol Ser A Biol Sci Med Sci. 2021;76:1726-1733. doi:10.1093/gerona/glab131
  23. Dolan CP, Yang TJ, Zimmel K, et al. Epimorphic regeneration of the mouse digit tip is finite. Stem Cell Res Ther. 2022;13:62. doi:10.1186/s13287-022-02741-2
  24. Simkin J, Han M, Yu L, et al. The mouse digit tip: from wound healing to regeneration. Methods Mol Biol Clifton NJ. 2013;1037:419-435. doi:10.1007/978-1-62703-505-7_24
  25. Ziegler-Graham K, MacKenzie EJ, Ephraim PL, et al. Estimating the prevalence of limb loss in the United States: 2005 to 2050. Arch Phys Med Rehabil. 2008;89:422-429. doi:10.1016/j.apmr.2007.11.005
  26. Dudek NL, Marks MB, Marshall SC, et al. Dermatologic conditions associated with use of a lower-extremity prosthesis. Arch Phys Med Rehabil. 2005;86:659-663. doi:10.1016/j.apmr.2004.09.003
  27. Lannan FM, Meyerle JH. The dermatologist’s role in amputee skin care. Cutis. 2019;103:86-90.
  28. Dougherty AL, Mohrle CR, Galarneau MR, et al. Battlefield extremity injuries in Operation Iraqi Freedom. Injury. 2009;40:772-777. doi:10.1016/j.injury.2009.02.014
  29. Epstein RA, Heinemann AW, McFarland LV. Quality of life for veterans and servicemembers with major traumatic limb loss from Vietnam and OIF/OEF conflicts. J Rehabil Res Dev. 2010;47:373-385. doi:10.1682/jrrd.2009.03.0023
  30. Pinzur MS, Gold J, Schwartz D, et al. Energy demands for walking in dysvascular amputees as related to the level of amputation. Orthopedics. 1992;15:1033-1036; discussion 1036-1037. doi:10.3928/0147-7447-19920901-07
  31. Robinson V, Sansam K, Hirst L, et al. Major lower limb amputation–what, why and how to achieve the best results. Orthop Trauma. 2010;24:276-285. doi:10.1016/j.mporth.2010.03.017
  32. Lu S, Wang C, Zhong W, et al. Amputation stump revision using a free sural neurocutaneous perforator flap. Ann Plast Surg. 2016;76:83-87. doi:10.1097/SAP.0000000000000211
  33. Kim SW, Jeon SB, Hwang KT, et al. Coverage of amputation stumps using a latissimus dorsi flap with a serratus anterior muscle flap: a comparative study. Ann Plast Surg. 2016;76:88-93. doi:10.1097/SAP.0000000000000220
  34. Pavey GJ, Formby PM, Hoyt BW, et al. Intrawound antibiotic powder decreases frequency of deep infection and severity of heterotopic ossification in combat lower extremity amputations. Clin Orthop. 2019;477:802-810. doi:10.1007/s11999.0000000000000090
  35. Dunkel N, Belaieff W, Assal M, et al. Wound dehiscence and stump infection after lower limb amputation: risk factors and association with antibiotic use. J Orthop Sci Off J Jpn Orthop Assoc. 2012;17:588-594. doi:10.1007/s00776-012-0245-5
  36. Rubin G, Orbach H, Rinott M, et al. The use of prophylactic antibiotics in treatment of fingertip amputation: a randomized prospective trial. Am J Emerg Med. 2015;33:645-647. doi:10.1016/j.ajem.2015.02.002
  37. Azarbal AF, Harris S, Mitchell EL, et al. Nasal methicillin-resistant Staphylococcus aureus colonization is associated with increased wound occurrence after major lower extremity amputation. J Vasc Surg. 2015;62:401-405. doi:10.1016/j.jvs.2015.02.052
  38. Kwasniewski M, Mitchel D. Post amputation skin and wound care. Phys Med Rehabil Clin N Am. 2022;33:857-870. doi:10.1016/j.pmr.2022.06.010
  39. Chang H, Maldonado TS, Rockman CB, et al. Closed incision negative pressure wound therapy may decrease wound complications in major lower extremity amputations. J Vasc Surg. 2021;73:1041-1047. doi:10.1016/j.jvs.2020.07.061
  40. Kalliainen LK, Gordillo GM, Schlanger R, et al. Topical oxygen as an adjunct to wound healing: a clinical case series. Pathophysiol Off J Int Soc Pathophysiol. 2003;9:81-87. doi:10.1016/s0928-4680(02)00079-2
  41. Reichmann JP, Stevens PM, Rheinstein J, et al. Removable rigid dressings for postoperative management of transtibial amputations: a review of published evidence. PM R. 2018;10:516-523. doi:10.1016/j.pmrj.2017.10.002
  42. MacLean N, Fick GH. The effect of semirigid dressings on below-knee amputations. Phys Ther. 1994;74:668-673. doi:10.1093/ptj/74.7.668
  43. Koonalinthip N, Sukthongsa A, Janchai S. Comparison of removable rigid dressing and elastic bandage for residual limb maturation in transtibial amputees: a randomized controlled trial. Arch Phys Med Rehabil. 2020;101:1683-1688. doi:10.1016/j.apmr.2020.05.009
  44. Taylor L, Cavenett S, Stepien JM, et al. Removable rigid dressings: a retrospective case-note audit to determine the validity of post-amputation application. Prosthet Orthot Int. 2008;32:223-230. doi:10.1080/03093640802016795
  45. Sumpio B, Shine SR, Mahler D, et al. A comparison of immediate postoperative rigid and soft dressings for below-knee amputations. Ann Vasc Surg. 2013;27:774-780. doi:10.1016/j.avsg.2013.03.007
  46. van Velzen AD, Nederhand MJ, Emmelot CH, et al. Early treatment of trans-tibial amputees: retrospective analysis of early fitting and elastic bandaging. Prosthet Orthot Int. 2005;29:3-12. doi:10.1080/17461550500069588
  47. Chin T, Toda M. Results of prosthetic rehabilitation on managing transtibial vascular amputation with silicone liner after wound closure. J Int Med Res. 2016;44:957-967. doi:10.1177/0300060516647554
  48. Hoskins RD, Sutton EE, Kinor D, et al. Using vacuum-assisted suspension to manage residual limb wounds in persons with transtibial amputation: a case series. Prosthet Orthot Int. 2014;38:68-74. doi:10.1177/0309364613487547
  49. Johannesson A, Larsson GU, Oberg T, et al. Comparison of vacuum-formed removable rigid dressing with conventional rigid dressing after transtibial amputation: similar outcome in a randomized controlled trial involving 27 patients. Acta Orthop. 2008;79:361-369. doi:10.1080/17453670710015265
  50. Alsancak S, Köse SK, Altınkaynak H. Effect of elastic bandaging and prosthesis on the decrease in stump volume. Acta Orthop Traumatol Turc. 2011;45:14-22. doi:10.3944/AOTT.2011.2365
  51. Than MP, Smith RA, Hammond C, et al. Keratin-based wound care products for treatment of resistant vascular wounds. J Clin Aesthetic Dermatol. 2012;5:31-35.
  52. Gurtner GC, Garcia AD, Bakewell K, et al. A retrospective matched‐cohort study of 3994 lower extremity wounds of multiple etiologies across 644 institutions comparing a bioactive human skin allograft, TheraSkin, plus standard of care, to standard of care alone. Int Wound J. 2020;17:55-64. doi:10.1111/iwj.13231
  53. Buikema KES, Meyerle JH. Amputation stump: privileged harbor for infections, tumors, and immune disorders. Clin Dermatol. 2014;32:670-677. doi:10.1016/j.clindermatol.2014.04.015
  54. Yang NB, Garza LA, Foote CE, et al. High prevalence of stump dermatoses 38 years or more after amputation. Arch Dermatol. 2012;148:1283-1286. doi:10.1001/archdermatol.2012.3004
  55. Potter BK, Burns TC, Lacap AP, et al. Heterotopic ossification following traumatic and combat-related amputations. Prevalence, risk factors, and preliminary results of excision. J Bone Joint Surg Am. 2007;89:476-486. doi:10.2106/JBJS.F.00412
  56. Edwards DS, Kuhn KM, Potter BK, et al. Heterotopic ossification: a review of current understanding, treatment, and future. J Orthop Trauma. 2016;30(suppl 3):S27-S30. doi:10.1097/BOT.0000000000000666
  57. Tintle SM, Shawen SB, Forsberg JA, et al. Reoperation after combat-related major lower extremity amputations. J Orthop Trauma. 2014;28:232-237. doi:10.1097/BOT.0b013e3182a53130
  58. Bui KM, Raugi GJ, Nguyen VQ, et al. Skin problems in individuals with lower-limb loss: literature review and proposed classification system. J Rehabil Res Dev. 2009;46:1085-1090. doi:10.1682/jrrd.2009.04.0052
  59. Turan H, Bas¸kan EB, Adim SB, et al. Acroangiodermatitis in a below-knee amputation stump. Clin Exp Dermatol. 2011;36:560-561. doi:10.1111/j.1365-2230.2011.04037.x
  60. Lin CH, Ma H, Chung MT, et al. Granulomatous cutaneous lesions associated with risperidone-induced hyperprolactinemia in an amputated upper limb. Int J Dermatol. 2012;51:75-78. doi:10.1111/j.1365-4632.2011.04906.x
  61. Schwartz RA, Bagley MP, Janniger CK, et al. Verrucous carcinoma of a leg amputation stump. Dermatologica. 1991;182:193-195. doi:10.1159/000247782
  62. Campanati A, Diotallevi F, Radi G, et al. Efficacy and safety of botulinum toxin B in focal hyperhidrosis: a narrative review. Toxins. 2023;15:147. doi:10.3390/toxins15020147
  63. Anderson RR, Donelan MB, Hivnor C, et al. Laser treatment of traumatic scars with an emphasis on ablative fractional laser resurfacing: consensus report. JAMA Dermatol. 2014;150:187-193. doi:10.1001/jamadermatol.2013.7761
  64. McGrath M, McCarthy J, Gallego A, et al. The influence of perforated prosthetic liners on residual limb wound healing: a case report. Can Prosthet Orthot J. 2019;2:32723. doi:10.33137/cpoj.v2i1.32723
  65. Abu El Hawa AA, Klein D, Bekeny JC, et al. The impact of statins on wound healing: an ally in treating the highly comorbid patient. J Wound Care. 2022;31(suppl 2):S36-S41. doi:10.12968/jowc.2022.31.Sup2.S36
  66. Nasseri S, Sharifi M. Therapeutic potential of antimicrobial peptides for wound healing. Int J Pept Res Ther. 2022;28:38. doi:10.1007/s10989-021-10350-5
  67. Lee JV, Engel C, Tay S, et al. N-Acetyl-Cysteine treatment after lower extremity amputation improves areas of perfusion defect and wound healing outcomes. J Vasc Surg. 2021;73:39-40. doi:10.1016/j.jvs.2020.12.025
  68. Dong Y, Yang Q, Sun X. Comprehensive analysis of cell therapy on chronic skin wound healing: a meta-analysis. Hum Gene Ther. 2021;32:787-795. doi:10.1089/hum.2020.275
References
  1. Brockes JP, Kumar A. Comparative aspects of animal regeneration. Annu Rev Cell Dev Biol. 2008;24:525-549. doi:10.1146/annurev.cellbio.24.110707.175336
  2. Eming SA, Hammerschmidt M, Krieg T, et al. Interrelation of immunity and tissue repair or regeneration. Semin Cell Dev Biol. 2009;20:517-527. doi:10.1016/j.semcdb.2009.04.009
  3. Eming SA. Evolution of immune pathways in regeneration and repair: recent concepts and translational perspectives. Semin Immunol. 2014;26:275-276. doi:10.1016/j.smim.2014.09.001
  4. Bolognia JL, Jorizzo JJ, Schaffer JV, et al. Dermatology. 4th edition. Elsevier; 2018.
  5. Wang PH, Huang BS, Horng HC, et al. Wound healing. J Chin Med Assoc JCMA. 2018;81:94-101. doi:10.1016/j.jcma.2017.11.002
  6. Velnar T, Bailey T, Smrkolj V. The wound healing process: an overview of the cellular and molecular mechanisms. J Int Med Res. 2009;37:1528-1542. doi:10.1177/147323000903700531
  7. Gurtner GC, Werner S, Barrandon Y, et al. Wound repair and regeneration. Nature. 2008;453:314-321. doi:10.1038/nature07039
  8. Eming SA, Martin P, Tomic-Canic M. Wound repair and regeneration: mechanisms, signaling, and translation. Sci Transl Med. 2014;6:265sr6. doi:10.1126/scitranslmed.3009337
  9. Eming SA, Brachvogel B, Odorisio T, et al. Regulation of angiogenesis: wound healing as a model. Prog Histochem Cytochem. 2007;42:115-170. doi:10.1016/j.proghi.2007.06.001
  10. Janis JE, Harrison B. Wound healing: part I. basic science. Plast Reconstr Surg. 2016;138(3 suppl):9S-17S. doi:10.1097/PRS.0000000000002773
  11. Profyris C, Tziotzios C, Do Vale I. Cutaneous scarring: pathophysiology, molecular mechanisms, and scar reduction therapeutics. part I: the molecular basis of scar formation. J Am Acad Dermatol. 2012;66:1-10; quiz 11-12. doi:10.1016/j.jaad.2011.05.055
  12. Kwan P, Ding J, Tredget EE. MicroRNA 181b regulates decorin production by dermal fibroblasts and may be a potential therapy for hypertrophic scar. PLoS One. 2015;10:e0123054. doi:10.1371/journal.pone.0123054
  13. Ben W, Yang Y, Yuan J, et al. Human papillomavirus 16 E6 modulates the expression of host microRNAs in cervical cancer. Taiwan J Obstet Gynecol. 2015;54:364-370. doi:10.1016/j.tjog.2014.06.007
  14. Yu EH, Tu HF, Wu CH, et al. MicroRNA-21 promotes perineural invasion and impacts survival in patients with oral carcinoma. J Chin Med Assoc JCMA. 2017;80:383-388. doi:10.1016/j.jcma.2017.01.003
  15. Wen KC, Sung PL, Yen MS, et al. MicroRNAs regulate several functions of normal tissues and malignancies. Taiwan J Obstet Gynecol. 2013;52:465-469. doi:10.1016/j.tjog.2013.10.002
  16. Babalola O, Mamalis A, Lev-Tov H, et al. The role of microRNAs in skin fibrosis. Arch Dermatol Res. 2013;305:763-776. doi:10.1007/s00403-013-1410-1
  17. Hofer M, Hoferová Z, Falk M. Pharmacological modulation of radiation damage. does it exist a chance for other substances than hematopoietic growth factors and cytokines? Int J Mol Sci. 2017;18:1385. doi:10.3390/ijms18071385
  18. Darby IA, Weller CD. Aspirin treatment for chronic wounds: potential beneficial and inhibitory effects. Wound Repair Regen. 2017;25:7-12. doi:10.1111/wrr.12502
  19. Khalid KA, Nawi AFM, Zulkifli N, et al. Aging and wound healing of the skin: a review of clinical and pathophysiological hallmarks. Life. 2022;12:2142. doi:10.3390/life12122142
  20. Peacock HM, Gilbert EAB, Vickaryous MK. Scar‐free cutaneous wound healing in the leopard gecko, Eublepharis macularius. J Anat. 2015;227:596-610. doi:10.1111/joa.12368
  21. Delorme SL, Lungu IM, Vickaryous MK. Scar‐free wound healing and regeneration following tail loss in the leopard gecko, Eublepharis macularius. Anat Rec. 2012;295:1575-1595. doi:10.1002/ar.22490
  22. Brunauer R, Xia IG, Asrar SN, et al. Aging delays epimorphic regeneration in mice. J Gerontol Ser A Biol Sci Med Sci. 2021;76:1726-1733. doi:10.1093/gerona/glab131
  23. Dolan CP, Yang TJ, Zimmel K, et al. Epimorphic regeneration of the mouse digit tip is finite. Stem Cell Res Ther. 2022;13:62. doi:10.1186/s13287-022-02741-2
  24. Simkin J, Han M, Yu L, et al. The mouse digit tip: from wound healing to regeneration. Methods Mol Biol Clifton NJ. 2013;1037:419-435. doi:10.1007/978-1-62703-505-7_24
  25. Ziegler-Graham K, MacKenzie EJ, Ephraim PL, et al. Estimating the prevalence of limb loss in the United States: 2005 to 2050. Arch Phys Med Rehabil. 2008;89:422-429. doi:10.1016/j.apmr.2007.11.005
  26. Dudek NL, Marks MB, Marshall SC, et al. Dermatologic conditions associated with use of a lower-extremity prosthesis. Arch Phys Med Rehabil. 2005;86:659-663. doi:10.1016/j.apmr.2004.09.003
  27. Lannan FM, Meyerle JH. The dermatologist’s role in amputee skin care. Cutis. 2019;103:86-90.
  28. Dougherty AL, Mohrle CR, Galarneau MR, et al. Battlefield extremity injuries in Operation Iraqi Freedom. Injury. 2009;40:772-777. doi:10.1016/j.injury.2009.02.014
  29. Epstein RA, Heinemann AW, McFarland LV. Quality of life for veterans and servicemembers with major traumatic limb loss from Vietnam and OIF/OEF conflicts. J Rehabil Res Dev. 2010;47:373-385. doi:10.1682/jrrd.2009.03.0023
  30. Pinzur MS, Gold J, Schwartz D, et al. Energy demands for walking in dysvascular amputees as related to the level of amputation. Orthopedics. 1992;15:1033-1036; discussion 1036-1037. doi:10.3928/0147-7447-19920901-07
  31. Robinson V, Sansam K, Hirst L, et al. Major lower limb amputation–what, why and how to achieve the best results. Orthop Trauma. 2010;24:276-285. doi:10.1016/j.mporth.2010.03.017
  32. Lu S, Wang C, Zhong W, et al. Amputation stump revision using a free sural neurocutaneous perforator flap. Ann Plast Surg. 2016;76:83-87. doi:10.1097/SAP.0000000000000211
  33. Kim SW, Jeon SB, Hwang KT, et al. Coverage of amputation stumps using a latissimus dorsi flap with a serratus anterior muscle flap: a comparative study. Ann Plast Surg. 2016;76:88-93. doi:10.1097/SAP.0000000000000220
  34. Pavey GJ, Formby PM, Hoyt BW, et al. Intrawound antibiotic powder decreases frequency of deep infection and severity of heterotopic ossification in combat lower extremity amputations. Clin Orthop. 2019;477:802-810. doi:10.1007/s11999.0000000000000090
  35. Dunkel N, Belaieff W, Assal M, et al. Wound dehiscence and stump infection after lower limb amputation: risk factors and association with antibiotic use. J Orthop Sci Off J Jpn Orthop Assoc. 2012;17:588-594. doi:10.1007/s00776-012-0245-5
  36. Rubin G, Orbach H, Rinott M, et al. The use of prophylactic antibiotics in treatment of fingertip amputation: a randomized prospective trial. Am J Emerg Med. 2015;33:645-647. doi:10.1016/j.ajem.2015.02.002
  37. Azarbal AF, Harris S, Mitchell EL, et al. Nasal methicillin-resistant Staphylococcus aureus colonization is associated with increased wound occurrence after major lower extremity amputation. J Vasc Surg. 2015;62:401-405. doi:10.1016/j.jvs.2015.02.052
  38. Kwasniewski M, Mitchel D. Post amputation skin and wound care. Phys Med Rehabil Clin N Am. 2022;33:857-870. doi:10.1016/j.pmr.2022.06.010
  39. Chang H, Maldonado TS, Rockman CB, et al. Closed incision negative pressure wound therapy may decrease wound complications in major lower extremity amputations. J Vasc Surg. 2021;73:1041-1047. doi:10.1016/j.jvs.2020.07.061
  40. Kalliainen LK, Gordillo GM, Schlanger R, et al. Topical oxygen as an adjunct to wound healing: a clinical case series. Pathophysiol Off J Int Soc Pathophysiol. 2003;9:81-87. doi:10.1016/s0928-4680(02)00079-2
  41. Reichmann JP, Stevens PM, Rheinstein J, et al. Removable rigid dressings for postoperative management of transtibial amputations: a review of published evidence. PM R. 2018;10:516-523. doi:10.1016/j.pmrj.2017.10.002
  42. MacLean N, Fick GH. The effect of semirigid dressings on below-knee amputations. Phys Ther. 1994;74:668-673. doi:10.1093/ptj/74.7.668
  43. Koonalinthip N, Sukthongsa A, Janchai S. Comparison of removable rigid dressing and elastic bandage for residual limb maturation in transtibial amputees: a randomized controlled trial. Arch Phys Med Rehabil. 2020;101:1683-1688. doi:10.1016/j.apmr.2020.05.009
  44. Taylor L, Cavenett S, Stepien JM, et al. Removable rigid dressings: a retrospective case-note audit to determine the validity of post-amputation application. Prosthet Orthot Int. 2008;32:223-230. doi:10.1080/03093640802016795
  45. Sumpio B, Shine SR, Mahler D, et al. A comparison of immediate postoperative rigid and soft dressings for below-knee amputations. Ann Vasc Surg. 2013;27:774-780. doi:10.1016/j.avsg.2013.03.007
  46. van Velzen AD, Nederhand MJ, Emmelot CH, et al. Early treatment of trans-tibial amputees: retrospective analysis of early fitting and elastic bandaging. Prosthet Orthot Int. 2005;29:3-12. doi:10.1080/17461550500069588
  47. Chin T, Toda M. Results of prosthetic rehabilitation on managing transtibial vascular amputation with silicone liner after wound closure. J Int Med Res. 2016;44:957-967. doi:10.1177/0300060516647554
  48. Hoskins RD, Sutton EE, Kinor D, et al. Using vacuum-assisted suspension to manage residual limb wounds in persons with transtibial amputation: a case series. Prosthet Orthot Int. 2014;38:68-74. doi:10.1177/0309364613487547
  49. Johannesson A, Larsson GU, Oberg T, et al. Comparison of vacuum-formed removable rigid dressing with conventional rigid dressing after transtibial amputation: similar outcome in a randomized controlled trial involving 27 patients. Acta Orthop. 2008;79:361-369. doi:10.1080/17453670710015265
  50. Alsancak S, Köse SK, Altınkaynak H. Effect of elastic bandaging and prosthesis on the decrease in stump volume. Acta Orthop Traumatol Turc. 2011;45:14-22. doi:10.3944/AOTT.2011.2365
  51. Than MP, Smith RA, Hammond C, et al. Keratin-based wound care products for treatment of resistant vascular wounds. J Clin Aesthetic Dermatol. 2012;5:31-35.
  52. Gurtner GC, Garcia AD, Bakewell K, et al. A retrospective matched‐cohort study of 3994 lower extremity wounds of multiple etiologies across 644 institutions comparing a bioactive human skin allograft, TheraSkin, plus standard of care, to standard of care alone. Int Wound J. 2020;17:55-64. doi:10.1111/iwj.13231
  53. Buikema KES, Meyerle JH. Amputation stump: privileged harbor for infections, tumors, and immune disorders. Clin Dermatol. 2014;32:670-677. doi:10.1016/j.clindermatol.2014.04.015
  54. Yang NB, Garza LA, Foote CE, et al. High prevalence of stump dermatoses 38 years or more after amputation. Arch Dermatol. 2012;148:1283-1286. doi:10.1001/archdermatol.2012.3004
  55. Potter BK, Burns TC, Lacap AP, et al. Heterotopic ossification following traumatic and combat-related amputations. Prevalence, risk factors, and preliminary results of excision. J Bone Joint Surg Am. 2007;89:476-486. doi:10.2106/JBJS.F.00412
  56. Edwards DS, Kuhn KM, Potter BK, et al. Heterotopic ossification: a review of current understanding, treatment, and future. J Orthop Trauma. 2016;30(suppl 3):S27-S30. doi:10.1097/BOT.0000000000000666
  57. Tintle SM, Shawen SB, Forsberg JA, et al. Reoperation after combat-related major lower extremity amputations. J Orthop Trauma. 2014;28:232-237. doi:10.1097/BOT.0b013e3182a53130
  58. Bui KM, Raugi GJ, Nguyen VQ, et al. Skin problems in individuals with lower-limb loss: literature review and proposed classification system. J Rehabil Res Dev. 2009;46:1085-1090. doi:10.1682/jrrd.2009.04.0052
  59. Turan H, Bas¸kan EB, Adim SB, et al. Acroangiodermatitis in a below-knee amputation stump. Clin Exp Dermatol. 2011;36:560-561. doi:10.1111/j.1365-2230.2011.04037.x
  60. Lin CH, Ma H, Chung MT, et al. Granulomatous cutaneous lesions associated with risperidone-induced hyperprolactinemia in an amputated upper limb. Int J Dermatol. 2012;51:75-78. doi:10.1111/j.1365-4632.2011.04906.x
  61. Schwartz RA, Bagley MP, Janniger CK, et al. Verrucous carcinoma of a leg amputation stump. Dermatologica. 1991;182:193-195. doi:10.1159/000247782
  62. Campanati A, Diotallevi F, Radi G, et al. Efficacy and safety of botulinum toxin B in focal hyperhidrosis: a narrative review. Toxins. 2023;15:147. doi:10.3390/toxins15020147
  63. Anderson RR, Donelan MB, Hivnor C, et al. Laser treatment of traumatic scars with an emphasis on ablative fractional laser resurfacing: consensus report. JAMA Dermatol. 2014;150:187-193. doi:10.1001/jamadermatol.2013.7761
  64. McGrath M, McCarthy J, Gallego A, et al. The influence of perforated prosthetic liners on residual limb wound healing: a case report. Can Prosthet Orthot J. 2019;2:32723. doi:10.33137/cpoj.v2i1.32723
  65. Abu El Hawa AA, Klein D, Bekeny JC, et al. The impact of statins on wound healing: an ally in treating the highly comorbid patient. J Wound Care. 2022;31(suppl 2):S36-S41. doi:10.12968/jowc.2022.31.Sup2.S36
  66. Nasseri S, Sharifi M. Therapeutic potential of antimicrobial peptides for wound healing. Int J Pept Res Ther. 2022;28:38. doi:10.1007/s10989-021-10350-5
  67. Lee JV, Engel C, Tay S, et al. N-Acetyl-Cysteine treatment after lower extremity amputation improves areas of perfusion defect and wound healing outcomes. J Vasc Surg. 2021;73:39-40. doi:10.1016/j.jvs.2020.12.025
  68. Dong Y, Yang Q, Sun X. Comprehensive analysis of cell therapy on chronic skin wound healing: a meta-analysis. Hum Gene Ther. 2021;32:787-795. doi:10.1089/hum.2020.275
Issue
Cutis - 113(3)
Issue
Cutis - 113(3)
Page Number
125-131
Page Number
125-131
Publications
Publications
Topics
Article Type
Display Headline
Wound Healing: Cellular Review With Specific Attention to Postamputation Care
Display Headline
Wound Healing: Cellular Review With Specific Attention to Postamputation Care
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>Kirwin</fileName> <TBEID>0C02F2F4.SIG</TBEID> <TBUniqueIdentifier>NJ_0C02F2F4</TBUniqueIdentifier> <newsOrJournal>Journal</newsOrJournal> <publisherName>Frontline Medical Communications Inc.</publisherName> <storyname>Military Derm</storyname> <articleType>1</articleType> <TBLocation>Copyfitting-CT</TBLocation> <QCDate/> <firstPublished>20240305T083438</firstPublished> <LastPublished>20240305T083438</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240305T083437</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>Kirwin</byline> <bylineText>David S. Kirwin, MD; Harrison E. Diaz, MD; Travis C. Frantz, MD; Cory F. Janney, MD; Curtis L. Hardy, DO; W. Hugh Lyford, MD</bylineText> <bylineFull>Kirwin</bylineFull> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType/> <journalDocType/> <linkLabel/> <pageRange>125-131</pageRange> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:"> <name/> <rightsInfo> <copyrightHolder> <name/> </copyrightHolder> <copyrightNotice/> </rightsInfo> </provider> <abstract/> <metaDescription>Restoring skin integrity and balance after injury is vital for survival, serving as a crucial defense mechanism against potential infections by preventing the e</metaDescription> <articlePDF>300455</articlePDF> <teaserImage/> <title>Wound Healing: Cellular Review With Specific Attention to Postamputation Care</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear>2024</pubPubdateYear> <pubPubdateMonth>March</pubPubdateMonth> <pubPubdateDay/> <pubVolume>113</pubVolume> <pubNumber>3</pubNumber> <wireChannels/> <primaryCMSID/> <CMSIDs> <CMSID>21173</CMSID> <CMSID>2159</CMSID> </CMSIDs> <keywords> <keyword>wounds</keyword> <keyword> postamputation care</keyword> </keywords> <seeAlsos/> <publications_g> <publicationData> <publicationCode>CT</publicationCode> <pubIssueName>March 2024</pubIssueName> <pubArticleType>Departments | 2159</pubArticleType> <pubTopics/> <pubCategories/> <pubSections> <pubSection>Military Dermatology | 21173<pubSubsection/></pubSection> </pubSections> <journalTitle>Cutis</journalTitle> <journalFullTitle>Cutis</journalFullTitle> <copyrightStatement>Copyright 2015 Frontline Medical Communications Inc., Parsippany, NJ, USA. All rights reserved.</copyrightStatement> </publicationData> </publications_g> <publications> <term canonical="true">12</term> </publications> <sections> <term canonical="true">38668</term> </sections> <topics> <term canonical="true">313</term> </topics> <links> <link> <itemClass qcode="ninat:composite"/> <altRep contenttype="application/pdf">images/180026df.pdf</altRep> <description role="drol:caption"/> <description role="drol:credit"/> </link> </links> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Wound Healing: Cellular Review With Specific Attention to Postamputation Care</title> <deck/> </itemMeta> <itemContent> <p class="abstract">Wound healing is crucial for survival, prevention of infection, and restoration of tissue function. The immune system drives this process with 3 main phases: inflammation, proliferation, and remodeling. Keloids and hypertrophic scars reveal disruptions in these phases, underscoring the balance needed for healing. Limb amputation, a life-changing event, demands careful consideration for healing and function. Factors such as amputation level, surgical technique, and prosthetic fitting shape outcomes, while complications such as heterotopic ossification challenge recovery. Treatment advances including statins and stem cell therapy hold promise, with dermatologists poised to contribute substantially to postamputation care.</p> <p> <em><i>Cutis</i>. 2024;113:125-131.</em> </p> <p>Restoring skin integrity and balance after injury is vital for survival, serving as a crucial defense mechanism against potential infections by preventing the entry of harmful pathogens. Moreover, proper healing is essential for restoring normal tissue function, allowing damaged tissues to repair and, in an ideal scenario, regenerate. Timely healing helps reduce the risk for complications, such as chronic wounds, which could lead to more severe issues if left untreated. Additionally, pain relief often is associated with effective wound healing as inflammatory responses diminish during the repair process. </p> <p>The immune system plays a pivotal role in wound healing, influencing various repair mechanisms and ultimately determining the extent of scarring. Although inflammation is present throughout the repair response, recent studies have challenged the conventional belief of an inverse correlation between the intensity of inflammation and regenerative capacity. Inflammatory signals were found to be crucial for timely repair and fundamental processes in regeneration, possibly presenting a paradigm shift in the understanding of immunology.<sup>1-4</sup> The complexities of wound healing are exemplified when evaluating and treating postamputation wounds. To address such a task, one needs a firm understanding of the science behind healing wounds and what can go wrong along the way. </p> <h3>Phases of Wound Healing</h3> <p>Wound healing is a complex process that involves a series of sequential yet overlapping phases, including hemostasis/inflammation, proliferation, and remodeling. </p> <p><i>Hemostasis/Inflammation—</i>The initial stage of wound healing involves hemostasis, in which the primary objective is to prevent blood loss and initiate inflammation. Platelets arrive at the wound site, forming a provisional clot that is crucial for subsequent healing phases.<sup>4-6</sup> Platelets halt bleeding as well as act as a medium for cell migration and adhesion; they also are a source of growth factors and proinflammatory cytokines that herald the inflammatory response.<sup>4-7</sup> <br/><br/>Inflammation is characterized by the infiltration of immune cells, particularly neutrophils and macrophages. Neutrophils act as the first line of defense, clearing debris and preventing infection. Macrophages follow, phagocytizing apoptotic cells and releasing growth factors such as tumor necrosis factor α, vascular endothelial growth factor, and matrix metalloprotease 9, which stimulate the next phase.<sup>4-6,8</sup> Typically, the hemostasis and inflammatory phase starts approximately 6 to 8 hours after wound origin and lasts 3 to 4 days.<sup>4,6,7<br/><br/></sup><i>Proliferation—</i>Following hemostasis and inflammation, the wound transitions into the proliferation phase, which is marked by the development of granulation tissue—a dynamic amalgamation of fibroblasts, endothelial cells, and inflammatory cells.<sup>1,4-8</sup> Fibroblasts play a central role in synthesizing collagen, the primary structural protein in connective tissue. They also orchestrate synthesis of vitronectin, fibronectin, fibrin, and tenascin.<sup>4-6,8</sup> Simultaneously, angiogenesis takes place, involving the creation of new blood vessels to supply essential nutrients and oxygen to the healing tissue.<sup>4,7,9</sup> Growth factors such as transforming growth factor <span class="body">β</span> and vascular endothelial growth factor coordinate cellular activities and foster tissue repair.<sup>4-6,8</sup> The proliferation phase extends over days to weeks, laying the groundwork for subsequent tissue restructuring.<br/><br/><i>Remodeling—</i>The final stage of wound healing is remodeling, an extended process that may persist for several months or, in some cases, years. Throughout this phase, the initially deposited collagen, predominantly type III collagen, undergoes transformation into mature type I collagen.<sup>4-6,8</sup> This transformation is critical for reinstating the tissue’s strength and functionality. The balance between collagen synthesis and degradation is delicate, regulated by matrix metalloproteinases and inhibitors of metalloproteinases.<sup>4-8</sup> Fibroblasts, myofibroblasts, and other cells coordinate this intricate process of tissue reorganization.<sup>4-7</sup> <br/><br/>The eventual outcome of the remodeling phase determines the appearance and functionality of the healed tissue. Any disruption in this phase can lead to complications, such as chronic wounds and hypertrophic scars/keloids.<sup>4-6</sup> These abnormal healing processes are characterized by localized inflammation, heightened fibroblast function, and excessive accumulation of the extracellular matrix.<sup>4-8</sup></p> <h3>Molecular Mechanisms</h3> <p>Comprehensive investigations—both in vivo and in vitro—have explored the intricate molecular mechanisms involved in heightened wound healing. Transforming growth factor <span class="body">β</span> takes center stage as a crucial factor, prompting the transformation of fibroblasts into myofibroblasts and contributing to the deposition of extracellular matrix.<sup>2,4-8,10</sup> Transforming growth factor <span class="body">β </span>activates non-Smad signaling pathways, such as MAPK (mitogen-activated protein kinase) and PI3K (phosphoinositide 3-kinase), influencing processes associated with fibrosis.<sup>5,11</sup> Furthermore, microRNAs play a pivotal role in posttranscriptional regulation, influencing both transforming growth factor <span class="body">β</span> signaling and fibroblast behavior.<sup>12-16</sup></p> <p>The involvement of prostaglandins is crucial in wound healing. Prostaglandin E2 plays a notable role and is positively correlated with the rate of wound healing.<sup>5</sup> The cyclooxygenase pathway, pivotal for prostaglandin synthesis, becomes a target for inflammation control.<sup>4,5,10</sup> Although aspirin and nonsteroidal anti-inflammatory drugs commonly are employed, their impact on wound healing remains controversial, as inhibition of cyclooxygenase may disrupt normal repair processes.<sup>5,17,18<br/><br/></sup>Wound healing exhibits variations depending on age. Fetal skin regeneration is marked by the restoration of normal dermal architecture, including adnexal structures, nerves, vessels, and muscle.<sup>4-6</sup> The distinctive characteristics of fetal wound healing include a unique profile of growth factors, a diminished inflammatory response, reduced biomechanical stress, and a distinct extracellular matrix composition.<sup>19</sup> These factors contribute to a lower propensity for scar formation compared to the healing processes observed in adults. Fetal and adult wound healing differ fundamentally in their extracellular matrix composition, inflammatory cells, and cytokine levels.<sup>4-6,19</sup> Adult wounds feature myofibroblasts, which are absent in fetal wounds, contributing to heightened mechanical tension.<sup>5</sup> Delving deeper into the biochemical basis of fetal wound healing holds promise for mitigating scar formation in adults. </p> <h3>Takeaways From Other Species</h3> <p>Much of the biochemical knowledge of wound healing, especially regenerative wound healing, is known from other species. Geckos provide a unique model for studying regenerative repair in tails and nonregenerative healing in limbs after amputation. Scar-free wound healing is characterized by rapid wound closure, delayed blood vessel development, and collagen deposition, which contrasts with the hypervascular granulation tissue seen in scarring wounds.<sup>20</sup> Scar-free wound healing and regeneration are intrinsic properties of the lizard tail and are unaffected by the location or method of detachment.<sup>21</sup></p> <p>Compared to amphibians with extraordinary regenerative capacity, data suggest the lack of regenerative capacity in mammals may come from a desynchronization of the fine-tuned interplay of progenitor cells such as blastema and differentiated cells.<sup>22,23</sup> In mice, the response to amputation is specific to the level: cutting through the distal third of the terminal phalanx elicits a regeneration response, yielding a new digit tip resembling the lost one, while an amputation through the distal third of the intermediate phalanx triggers a wound healing and scarring response.<sup>24</sup> </p> <h3>Wound Healing Following Limb Amputation</h3> <p>Limb amputation represents a profound change in an individual’s life, impacting daily activities and overall well-being. There are many causes of amputation, but the most common include cardiovascular diseases, diabetes mellitus, cancer, and trauma.<sup>25-27</sup> Trauma represents a relatively common cause within the US Military due to the overall young population as well as inherent risks of uniformed service.<sup>25,27</sup> Advances in protective gear and combat casualty care have led to an increased number of individuals surviving with extremity injuries requiring amputation, particularly among younger service members, with a subgroup experiencing multiple amputations.<sup>27-29</sup> </p> <p>Numerous factors play a crucial role in the healing and function of postamputation wounds. The level of amputation is a key determinant influencing both functional outcomes and the healing process. Achieving a balance between preserving function and removing damaged tissue is essential. A study investigating cardiac function and oxygen consumption in 25 patients with peripheral vascular disease found higher-level amputations resulted in decreased walking speed and cadence, along with increased oxygen consumption per meter walked.<sup>30</sup> <br/><br/>Selecting the appropriate amputation level is vital to optimize functional outcomes without compromising wound healing. Successful prosthetic limb fitting depends largely on the length of the residual stump to support the body load and suspend the prosthesis. For long bone amputations, maintaining at least 12-cm clearance above the knee joint in transfemoral amputees and 10-cm below the knee joint in transtibial amputees is critical for maximizing functional outcomes.<sup>31</sup> <br/><br/>Surgical technique also is paramount. The goal is to minimize the risk for pressure ulcers by avoiding bony spurs and muscle imbalances. Shaping the muscle and residual limb is essential for proper prosthesis fitting. Attention to neurovascular structures, such as burying nerve ends to prevent neuropathic pain during prosthesis wear, is crucial.<sup>32</sup> In extremity amputations, surgeons often resort to free flap transfer techniques for stump reconstruction. In a study of 31 patients with severe lower extremity injuries undergoing various amputations, the use of latissimus dorsi myocutaneous flaps, alone or in combination with serratus anterior muscle flaps, resulted in fewer instances of deep ulceration and allowed for earlier prosthesis wear.<sup>33</sup></p> <h3>Addressing Barriers to Wound Healing</h3> <p>Multiple barriers to successful wound healing are encountered in the amputee population. Amputations from trauma have a less-controlled initiation, which carries with it a higher risk for infection, poor wound healing, and other complications. </p> <p><i>Infection—</i>Infection often is one of the first hurdles encountered in postamputation wound healing. Critical first steps in infection prevention include thorough cleaning of soiled traumatic wounds and appropriate tissue debridement coupled with scrupulous sterile technique and postoperative monitoring for signs and symptoms of infection. <br/><br/>In a retrospective study of 223 combat-related major lower extremity amputations (initial and revision) between 2009 and 2015, the use of intrawound antibiotic powder at the time of closure demonstrated a 13% absolute risk reduction in deep infection rates, which was particularly notable in revision amputations, with a number needed to treat of 8 for initial amputations and 4 for revision amputations on previously infected limbs.<sup>34</sup> Intra-operative antibiotic powder may represent a cheap and easy consideration for this special population of amputees. Postamputation antibiotic prophylaxis for infection prevention is an area of controversy. For nontraumatic infections, data suggest antibiotic prophylaxis may not decrease infection rates in these patients.<sup>35,36<br/><br/></sup>Interestingly, a study by Azarbal et al<sup>37</sup> aimed to investigate the correlation between nasal methicillin-resistant <i>Staphylococcus aureus</i> (MRSA) colonization and other patient factors with wound occurrence following major lower extremity amputation. The study found MRSA colonization was associated with higher rates of overall wound occurrence as well as wound occurrence due to wound infection. These data suggest nasal MRSA eradication may improve postoperative wound outcomes after major lower extremity amputation.<sup>37</sup> <br/><br/><i>Dressing Choice—</i>The dressing chosen for a residual limb also is of paramount importance following amputation. The personalized and dynamic management of postamputation wounds and skin involves achieving optimal healing through a dressing that sustains appropriate moisture levels, addresses edema, helps prevent contractures, and safeguards the limb.<sup>38</sup> From the start, using negative pressure wound dressings after surgical amputation can decrease wound-related complications.<sup>39</sup> <br/><br/>Topical oxygen therapy following amputation also shows promise. In a retrospective case series by Kalliainen et al,<sup>40</sup> topical oxygen therapy applied to 58 wounds in 32 patients over 9 months demonstrated positive outcomes in promoting wound healing, with 38 wounds (66%) healing completely with the use of topical oxygen. Minimal complications and no detrimental effects were observed.<sup>40<br/><br/></sup>Current recommendations suggest that non–weight-bearing removable rigid dressings are the superior postoperative management for transtibial amputations compared to soft dressings, offering benefits such as faster healing, reduced limb edema, earlier ambulation, preparatory shaping for prosthetic use, and prevention of knee flexion contractures.<sup>41-46</sup> Similarly, adding a silicone liner following amputation significantly reduced the duration of prosthetic rehabilitation compared with a conventional soft dressing program in one study (<i>P</i><span class="body">&lt;</span>.05).<sup>47</sup> <br/><br/>Specifically targeting wound edema, a case series by Hoskins et al<sup>48</sup> investigated the impact of prostheses with vacuum-assisted suspension on the size of residual limb wounds in individuals with transtibial amputation. Well-fitting sockets with vacuum-assisted suspension did not impede wound healing, and the results suggest the potential for continued prosthesis use during the healing process.<sup>48</sup> However, a study by Johannesson et al<sup>49</sup> compared the outcomes of transtibial amputation patients using a vacuum-formed rigid dressing and a conventional rigid plaster dressing, finding no significant differences in wound healing, time to prosthetic fitting, or functional outcomes with the prosthesis between the 2 groups. When comparing elastic bandaging, pneumatic prosthesis, and temporary prosthesis on postoperative stump management, temporary prosthesis led to a decrease in stump volume, quicker transition to a permanent prosthesis, and improved quality of life compared with elastic bandaging and pneumatic prosthetics.<sup>50</sup> <br/><br/>The type of material in dressings may contribute to utility in amputation wounds. Keratin-based wound dressings show promise for wound healing, especially in recalcitrant vascular wounds.<sup>51</sup> There also are numerous proprietary wound dressings available for patients, at least one of which has particularly thorough data. In a retrospective study of more than 2 million lower extremity wounds across 644 institutions, a proprietary bioactive human skin allograft (TheraSkin [LifeNet Health]) demonstrated higher healing rates, greater percentage area reductions, lower amputations, reduced recidivism, higher treatment completion, and fewer medical transfers compared with standard of care alone.<sup>52</sup> </p> <h3>Postamputation Dermatologic Concerns</h3> <p>After the postamputation wound heals, a notable concern is the prevalence of skin diseases affecting residual limbs. The stump site in amputees, marked by a delicate cutaneous landscape vulnerable to skin diseases, faces challenges arising from amputation-induced damage to various structures.<sup>53</sup> </p> <p>When integrated into a prosthesis socket, the altered skin must acclimate to a humid environment and endure forces for which it is not well suited, especially during movement.<sup>53</sup> Amputation remarkably alters normal tissue perfusion, which can lead to aberrant blood and lymphatic circulation in residual limbs.<sup>27,53</sup> This compromised skin, often associated with a history of vascular disease, diabetes mellitus, or malignancy, becomes immunocompromised, heightening the risk for dermatologic issues such as inflammation, infection, and malignancies.<sup>53</sup> Unlike the resilient volar skin on palms and soles, stump skin lacks adaptation to withstand the compressive forces generated during ambulation, sometimes leading to skin disease and pain that result in abandonment of the prosthesis.<sup>53,54</sup> Mechanical forces on the skin, especially in active patients eager to resume pre-injury lifestyles, contribute to skin breakdown. The dynamic nature of the residual limb, including muscle atrophy, gait changes, and weight fluctuations, complicates the prosthetic fitting process. Prosthesis abandonment remains a challenge, despite modern technologic advancements. <br/><br/>The occurrence of heterotopic ossification (extraskeletal bone formation) is another notable issue in military amputees.<sup>27,55-57</sup> Poor prosthetic fit can lead to skin degradation, necessitating further surgery to address mispositioned bone formations. Orthopedic monitoring supplemented by appropriate imaging studies can benefit postamputation patients by detecting and preventing heterotopic ossification in its early stages. <br/><br/>Dermatologic issues, especially among lower limb amputees, are noteworthy, with a substantial percentage experiencing complications related to socket prosthetics, such as heat, sweating, sores, and skin irritation. Up to 41% of patients are seen regularly for a secondary skin disorder following amputation.<sup>58</sup> As one might expect, persistent wounds, blisters, ulcers, and abscesses are some of the most typical cutaneous abnormalities affecting residual limbs with prostheses.<sup>27,58</sup> More rare skin conditions also are documented in residual limbs, including cutaneous granuloma, verrucous carcinoma, bullous pemphigoid, and angiodermatitis.<sup>27,59-61</sup> <br/><br/>Treatments offered in the dermatology clinic often are similar to patients who have not had an amputation. For instance, hyperhidrosis can be treated with prescription antiperspirant, topical aluminum chloride, topical glycopyrronium, botulinum toxin, and iontophoresis, which can greatly decrease skin irritation and malodor. Subcutaneous neurotoxins such as botulinum toxin are especially useful for hyperhidrosis following amputation because a single treatment can last 3 to 6 months, whereas topicals must be applied multiple times per day and can be inherently irritating to the skin.<sup>27,62</sup> Furthermore, ablative fractional resurfacing lasers also can help stimulate new collagen growth, increase skin mobility on residual limbs, smooth jagged scars, and aid prosthetic fitting.<sup>27,63</sup> Perforated prosthetic liners also may be useful to address issues such as excessive sweating, demonstrating improvements in skin health, reduced sweating problems, and potential avoidance of surgical interventions.<sup>64</sup> <br/><br/>When comorbid skin conditions are at bay, preventive measures for excessive wound healing necessitate early recognition and timely intervention for residual limbs. Preventive techniques encompass the use of silicone gel sheeting, hypoallergenic microporous tape, and intralesional steroid injections. <br/><br/><i>Psychological Concerns—</i>An overarching issue following amputation is the psychological toll the process imposes on the patient. Psychological concerns, including anxiety and depression, present additional challenges impacting residual limb hygiene and prosthetic maintenance. Chronic wounds are devastating to patients. These patients consistently express feeling ostracized from their community and anxious about unemployment, leaking fluid, or odor from the wound, as well as other social stigmata.<sup>62</sup> Depression and anxiety can hinder a patient’s ability to care for their wound and make them more susceptible to the myriad issues that can ensue. </p> <h3>Recent Developments in Wound Healing </h3> <p>Wound healing is ripe for innovation that could assuage ailments that impact patients following amputation. A 2022 study by Abu El Hawa et al<sup>65</sup> illustrated advanced progression in wound healing for patients taking statins, even though the statin group had increased age and number of comorbidities compared with patients not taking statins.</p> <p>Nasseri and Sharifi<sup>66</sup> showed the potential of antimicrobial peptides—small proteins with cationic charges and amphipathic structures exhibiting electrostatic interaction with microbial cell membranes—in promoting wound healing, particularly defensins and cathelicidin LL-37.They also discussed innovative delivery systems, such as nanoparticles and electrospun fibrous scaffolds, highlighting their potential as possibly more effective therapeutics than antibiotics, especially in the context of diabetic wound closure.<sup>66</sup> Aimed at increased angiogenesis in the proliferative phase, there is evidence that N-acetylcysteine can increase amputation stump perfusion with the goal of better long-term wound healing and more efficient scar formation.<sup>67</sup> <br/><br/>Stem cell therapy, particularly employing cells from the human amniotic membrane, represents an auspicious avenue for antifibrotic treatment. Amniotic epithelial cells and amniotic mesenchymal cells, with their self-renewal and multilineage differentiation capabilities, exhibit anti-inflammatory and antifibrotic properties.<sup>4,5</sup> A study by Dong et al<sup>68</sup> aimed to assess the efficacy of cell therapy, particularly differentiated progenitor cell–based graft transplantation or autologous stem cell injection, in treating refractory skin injuries such as nonrevascularizable critical limb ischemic ulcers, venous leg ulcers, and diabetic lower limb ulcers. The findings demonstrated cell therapy effectively reduced the size of ulcers, improved wound closure rates, and decreased major amputation rates compared with standard therapy. Of note, cell therapy had limited impact on alleviating pain in patients with critical limb ischemia-related cutaneous ulcers.<sup>68</sup> </p> <h3>Final Thoughts</h3> <p>Wound care following amputation is a multidisciplinary endeavor, necessitating collaboration between many health care professionals. Dermatologists play a crucial role in providing routine care as well as addressing wound healing and related skin issues among amputation patients. As the field progresses, dermatologists are well positioned to make notable contributions and ensure enhanced outcomes, resulting in a better quality of life for patients facing the challenges of limb amputation and prosthetic use.</p> <h2>References</h2> <p class="reference"> 1. Brockes JP, Kumar A. Comparative aspects of animal regeneration. <i>Annu Rev Cell Dev Biol</i>. 2008;24:525-549. doi:10.1146/annurev.cellbio.24.110707.175336</p> <p class="reference"> 2. Eming SA, Hammerschmidt M, Krieg T, et al. Interrelation of immunity and tissue repair or regeneration. <i>Semin Cell Dev Biol</i>. 2009;20:517-527. doi:10.1016/j.semcdb.2009.04.009<br/><br/> 3. Eming SA. Evolution of immune pathways in regeneration and repair: recent concepts and translational perspectives. <i>Semin Immunol</i>. 2014;26:275-276. doi:10.1016/j.smim.2014.09.001<br/><br/> 4. Bolognia JL, Jorizzo JJ, Schaffer JV, et al. <i>Dermatology</i>. 4th edition. Elsevier; 2018.<br/><br/> 5. Wang PH, Huang BS, Horng HC, et al. Wound healing. <i>J Chin Med Assoc JCMA</i>. 2018;81:94-101. doi:10.1016/j.jcma.2017.11.002<br/><br/> 6. Velnar T, Bailey T, Smrkolj V. The wound healing process: an overview of the cellular and molecular mechanisms. <i>J Int Med Res</i>. 2009;37:1528-1542. doi:10.1177/147323000903700531<br/><br/> 7. Gurtner GC, Werner S, Barrandon Y, et al. Wound repair and regeneration. <i>Nature</i>. 2008;453:314-321. doi:10.1038/nature07039<br/><br/> 8. Eming SA, Martin P, Tomic-Canic M. Wound repair and regeneration: mechanisms, signaling, and translation. <i>Sci Transl Med</i>. 2014;6:265sr6. doi:10.1126/scitranslmed.3009337<br/><br/> 9. Eming SA, Brachvogel B, Odorisio T, et al. Regulation of angiogenesis: wound healing as a model. <i>Prog Histochem Cytochem</i>. 2007;42:115-170. doi:10.1016/j.proghi.2007.06.001<br/><br/>10. Janis JE, Harrison B. Wound healing: part I. basic science. <i>Plast Reconstr Surg</i>. 2016;138(3 suppl):9S-17S. doi:10.1097/PRS.0000000000002773<br/><br/>11. Profyris C, Tziotzios C, Do Vale I. Cutaneous scarring: pathophysiology, molecular mechanisms, and scar reduction therapeutics. part I: the molecular basis of scar formation. <i>J Am Acad Dermatol</i>. 2012;66:1-10; quiz 11-12. doi:10.1016/j.jaad.2011.05.055<br/><br/>12. Kwan P, Ding J, Tredget EE. MicroRNA 181b regulates decorin production by dermal fibroblasts and may be a potential therapy for hypertrophic scar. <i>PLoS One</i>. 2015;10:e0123054. doi:10.1371/journal.pone.0123054<br/><br/>13. Ben W, Yang Y, Yuan J, et al. Human papillomavirus 16 E6 modulates the expression of host microRNAs in cervical cancer. <i>Taiwan J Obstet Gyneco</i>l. 2015;54:364-370. doi:10.1016/j.tjog.2014.06.007<br/><br/>14. Yu EH, Tu HF, Wu CH, et al. MicroRNA-21 promotes perineural invasion and impacts survival in patients with oral carcinoma. <i>J Chin Med Assoc JCMA</i>. 2017;80:383-388. doi:10.1016/j.jcma.2017.01.003</p> <p class="reference">15. Wen KC, Sung PL, Yen MS, et al. MicroRNAs regulate several functions of normal tissues and malignancies. <i>Taiwan J Obstet Gynecol</i>. 2013;52:465-469. doi:10.1016/j.tjog.2013.10.002</p> <p class="reference">16. Babalola O, Mamalis A, Lev-Tov H, et al. The role of microRNAs in skin fibrosis. <i>Arch Dermatol Res</i>. 2013;305:763-776. doi:10.1007/s00403-013-1410-1<br/><br/>17. Hofer M, Hoferová Z, Falk M. Pharmacological modulation of radiation damage. does it exist a chance for other substances than hematopoietic growth factors and cytokines? <i>Int J Mol Sci</i>. 2017;18:1385. doi:10.3390/ijms18071385<br/><br/>18. Darby IA, Weller CD. Aspirin treatment for chronic wounds: potential beneficial and inhibitory effects. <i>Wound Repair Regen</i>. 2017;25:7-12. doi:10.1111/wrr.12502<br/><br/>19. Khalid KA, Nawi AFM, Zulkifli N, et al. Aging and wound healing of the skin: a review of clinical and pathophysiological hallmarks. <i>Life</i>. 2022;12:2142. doi:10.3390/life12122142<br/><br/>20. Peacock HM, Gilbert EAB, Vickaryous MK. Scar‐free cutaneous wound healing in the leopard gecko, Eublepharis macularius. <i>J Anat</i>. 2015;227:596-610. doi:10.1111/joa.12368<br/><br/>21. Delorme SL, Lungu IM, Vickaryous MK. Scar‐free wound healing and regeneration following tail loss in the leopard gecko, Eublepharis macularius. <i>Anat Rec</i>. 2012;295:1575-1595. doi:10.1002/ar.22490<br/><br/>22. Brunauer R, Xia IG, Asrar SN, et al. Aging delays epimorphic regeneration in mice. <i>J Gerontol Ser A Biol Sci Med Sci</i>. 2021;76:1726-1733. doi:10.1093/gerona/glab131<br/><br/>23. Dolan CP, Yang TJ, Zimmel K, et al. Epimorphic regeneration of the mouse digit tip is finite. <i>Stem Cell Res Ther</i>. 2022;13:62. doi:10.1186/s13287-022-02741-2<br/><br/>24. Simkin J, Han M, Yu L, et al. The mouse digit tip: from wound healing to regeneration. <i>Methods Mol Biol Clifton NJ</i>. 2013;1037:419-435. doi:10.1007/978-1-62703-505-7_24<br/><br/>25. Ziegler-Graham K, MacKenzie EJ, Ephraim PL, et al. Estimating the prevalence of limb loss in the United States: 2005 to 2050. <i>Arch Phys Med Rehabil</i>. 2008;89:422-429. doi:10.1016/j.apmr.2007.11.005<br/><br/>26. Dudek NL, Marks MB, Marshall SC, et al. Dermatologic conditions associated with use of a lower-extremity prosthesis. <i>Arch Phys Med Rehabil</i>. 2005;86:659-663. doi:10.1016/j.apmr.2004.09.003<br/><br/>27. Lannan FM, Meyerle JH. The dermatologist’s role in amputee skin care. <i>Cutis</i>. 2019;103:86-90.<br/><br/>28. Dougherty AL, Mohrle CR, Galarneau MR, et al. Battlefield extremity injuries in Operation Iraqi Freedom. <i>Injury</i>. 2009;40:772-777. doi:10.1016/j.injury.2009.02.014<br/><br/>29. Epstein RA, Heinemann AW, McFarland LV. Quality of life for veterans and servicemembers with major traumatic limb loss from Vietnam and OIF/OEF conflicts. <i>J Rehabil Res Dev. </i>2010;47:373-385. doi:10.1682/jrrd.2009.03.0023<br/><br/>30. Pinzur MS, Gold J, Schwartz D, et al. Energy demands for walking in dysvascular amputees as related to the level of amputation. <i>Orthopedic</i>s. 1992;15:1033-1036; discussion 1036-1037. doi:10.3928/0147-7447-19920901-07<br/><br/>31. Robinson V, Sansam K, Hirst L, et al. Major lower limb amputation–what, why and how to achieve the best results. <i>Orthop Trauma</i>. 2010;24:276-285. doi:10.1016/j.mporth.2010.03.017<br/><br/>32. Lu S, Wang C, Zhong W, et al. Amputation stump revision using a free sural neurocutaneous perforator flap. <i>Ann Plast Surg</i>. 2016;76:83-87. doi:10.1097/SAP.0000000000000211<br/><br/>33. Kim SW, Jeon SB, Hwang KT, et al. Coverage of amputation stumps using a latissimus dorsi flap with a serratus anterior muscle flap: a comparative study. <i>Ann Plast Surg. </i>2016;76:88-93. doi:10.1097/SAP.0000000000000220<br/><br/>34. Pavey GJ, Formby PM, Hoyt BW, et al. Intrawound antibiotic powder decreases frequency of deep infection and severity of heterotopic ossification in combat lower extremity amputations. <i>Clin Orthop</i>. 2019;477:802-810. doi:10.1007/s11999.0000000000000090<br/><br/>35. Dunkel N, Belaieff W, Assal M, et al. Wound dehiscence and stump infection after lower limb amputation: risk factors and association with antibiotic use. <i>J Orthop Sci Off J Jpn </i>Orthop <i>Assoc</i>. 2012;17:588-594. doi:10.1007/s00776-012-0245-5<br/><br/>36. Rubin G, Orbach H, Rinott M, et al. The use of prophylactic antibiotics in treatment of fingertip amputation: a randomized prospective trial. <i>Am J Emerg Med</i>. 2015;33:645-647. doi:10.1016/j.ajem.2015.02.002<br/><br/>37. Azarbal AF, Harris S, Mitchell EL, et al. Nasal methicillin-resistant Staphylococcus aureus colonization is associated with increased wound occurrence after major lower extremity amputation. <i>J Vasc Surg</i>. 2015;62:401-405. doi:10.1016/j.jvs.2015.02.052<br/><br/>38. Kwasniewski M, Mitchel D. Post amputation skin and wound care. <i>Phys Med Rehabil Clin N Am</i>. 2022;33:857-870. doi:10.1016/j.pmr.2022.06.010<br/><br/>39. Chang H, Maldonado TS, Rockman CB, et al. Closed incision negative pressure wound therapy may decrease wound complications in major lower extremity amputations. <i>J Vasc Sur</i>g. 2021;73:1041-1047. doi:10.1016/j.jvs.2020.07.061<br/><br/>40. Kalliainen LK, Gordillo GM, Schlanger R, et al. Topical oxygen as an adjunct to wound healing: a clinical case series. <i>Pathophysiol Off J Int Soc Pathophysio</i>l. 2003;9:81-87. doi:10.1016/s0928-4680(02)00079-2<br/><br/>41. Reichmann JP, Stevens PM, Rheinstein J, et al. Removable rigid dressings for postoperative management of transtibial amputations: a review of published evidence.<i> PM R. </i>2018;10:516-523. doi:10.1016/j.pmrj.2017.10.002<br/><br/>42. MacLean N, Fick GH. The effect of semirigid dressings on below-knee amputations. <i>Phys Ther</i>. 1994;74:668-673. doi:10.1093/ptj/74.7.668<br/><br/>43. Koonalinthip N, Sukthongsa A, Janchai S. Comparison of removable rigid dressing and elastic bandage for residual limb maturation in transtibial amputees: a randomized controlled trial. <i>Arch Phys Med Rehabil</i>. 2020;101:1683-1688. doi:10.1016/j.apmr.2020.05.009<br/><br/>44. Taylor L, Cavenett S, Stepien JM, et al. Removable rigid dressings: a retrospective case-note audit to determine the validity of post-amputation application. <i>Prosthet Orthot Int. </i>2008;32:223-230. doi:10.1080/03093640802016795<br/><br/>45. Sumpio B, Shine SR, Mahler D, et al. A comparison of immediate postoperative rigid and soft dressings for below-knee amputations. <i>Ann Vasc Surg</i>. 2013;27:774-780. doi:10.1016/j.avsg.2013.03.007<br/><br/>46. van Velzen AD, Nederhand MJ, Emmelot CH, et al. Early treatment of trans-tibial amputees: retrospective analysis of early fitting and elastic bandaging. <i>Prosthet Orthot Int. </i>2005;29:3-12. doi:10.1080/17461550500069588<br/><br/>47. Chin T, Toda M. Results of prosthetic rehabilitation on managing transtibial vascular amputation with silicone liner after wound closure. <i>J Int Med Res</i>. 2016;44:957-967. doi:10.1177/0300060516647554</p> <p class="reference">48. Hoskins RD, Sutton EE, Kinor D, et al. Using vacuum-assisted suspension to manage residual limb wounds in persons with transtibial amputation: a case series. <i>Prosthet Orthot In</i>t. 2014;38:68-74. doi:10.1177/0309364613487547<br/><br/>49. Johannesson A, Larsson GU, Oberg T, et al. Comparison of vacuum-formed removable rigid dressing with conventional rigid dressing after transtibial amputation: similar outcome in a randomized controlled trial involving 27 patients. <i>Acta Orthop</i>. 2008;79:361-369. doi:10.1080/17453670710015265<br/><br/>50. Alsancak S, Köse SK, Altınkaynak H. Effect of elastic bandaging and prosthesis on the decrease in stump volume. <i>Acta Orthop Traumatol Turc</i>. 2011;45:14-22. doi:10.3944/AOTT.2011.2365<br/><br/>51. Than MP, Smith RA, Hammond C, et al. Keratin-based wound care products for treatment of resistant vascular wounds. <i>J Clin Aesthetic Dermatol</i>. 2012;5:31-35.<br/><br/>52. Gurtner GC, Garcia AD, Bakewell K, et al. A retrospective matched‐cohort study of 3994 lower extremity wounds of multiple etiologies across 644 institutions comparing a bioactive human skin allograft, TheraSkin, plus standard of care, to standard of care alone. <i>Int Wound J</i>. 2020;17:55-64. doi:10.1111/iwj.13231<br/><br/>53. Buikema KES, Meyerle JH. Amputation stump: privileged harbor for infections, tumors, and immune disorders. <i>Clin Dermatol.</i> 2014;32:670-677. doi:10.1016/j.clindermatol.2014.04.015<br/><br/>54. Yang NB, Garza LA, Foote CE, et al. High prevalence of stump dermatoses 38 years or more after amputation. <i>Arch Dermatol</i>. 2012;148:1283-1286. doi:10.1001/archdermatol.2012.3004<br/><br/>55. Potter BK, Burns TC, Lacap AP, et al. Heterotopic ossification following traumatic and combat-related amputations. Prevalence, risk factors, and preliminary results of excision. <i>J Bone Joint Surg Am</i>. 2007;89:476-486. doi:10.2106/JBJS.F.00412<br/><br/>56. Edwards DS, Kuhn KM, Potter BK, et al. Heterotopic ossification: a review of current understanding, treatment, and future. <i>J Orthop Trauma</i>. 2016;30(suppl 3):S27-S30. doi:10.1097/BOT.0000000000000666<br/><br/>57. Tintle SM, Shawen SB, Forsberg JA, et al. Reoperation after combat-related major lower extremity amputations.<i> J Orthop Trauma.</i> 2014;28:232-237. doi:10.1097/BOT.0b013e3182a53130<br/><br/>58. Bui KM, Raugi GJ, Nguyen VQ, et al. Skin problems in individuals with lower-limb loss: literature review and proposed classification system. <i>J Rehabil Res Dev</i>. 2009;46:1085-1090. doi:10.1682/jrrd.2009.04.0052<br/><br/>59. Turan H, Bas¸kan EB, Adim SB, et al. Acroangiodermatitis in a below-knee amputation stump. <i>Clin Exp Dermatol.</i> 2011;36:560-561. doi:10.1111/j.1365-2230.2011.04037.x<br/><br/>60. Lin CH, Ma H, Chung MT, et al. Granulomatous cutaneous lesions associated with risperidone-induced hyperprolactinemia in an amputated upper limb. <i>Int J Dermatol</i>. 2012;51:75-78. doi:10.1111/j.1365-4632.2011.04906.x<br/><br/>61. Schwartz RA, Bagley MP, Janniger CK, et al. Verrucous carcinoma of a leg amputation stump. <i>Dermatologica</i>. 1991;182:193-195. doi:10.1159/00024778262. Campanati A, Diotallevi F, Radi G, et al. Efficacy and safety of botulinum toxin B in focal hyperhidrosis: a narrative review. <i>Toxins</i>. 2023;15:147. doi:10.3390/toxins15020147<br/><br/>63. Anderson RR, Donelan MB, Hivnor C, et al. Laser treatment of traumatic scars with an emphasis on ablative fractional laser resurfacing: consensus report. <i>JAMA Dermatol. </i>2014;150:187-193. doi:10.1001/jamadermatol.2013.7761<br/><br/>64. McGrath M, McCarthy J, Gallego A, et al. The influence of perforated prosthetic liners on residual limb wound healing: a case report. <i>Can Prosthet Orthot J</i>. 2019;2:32723. doi:10.33137/cpoj.v2i1.32723<br/><br/>65. Abu El Hawa AA, Klein D, Bekeny JC, et al. The impact of statins on wound healing: an ally in treating the highly comorbid patient. <i>J Wound Care</i>. 2022;31(suppl 2):S36-S41. doi:10.12968/jowc.2022.31.Sup2.S36<br/><br/>66. Nasseri S, Sharifi M. Therapeutic potential of antimicrobial peptides for wound healing. <i>Int J Pept Res Ther</i>. 2022;28:38. doi:10.1007/s10989-021-10350-5<br/><br/>67. Lee JV, Engel C, Tay S, et al. N-Acetyl-Cysteine treatment after lower extremity amputation improves areas of perfusion defect and wound healing outcomes. <i>J Vasc Surg</i>. 2021;73:39-40. doi:10.1016/j.jvs.2020.12.025<br/><br/>68. Dong Y, Yang Q, Sun X. Comprehensive analysis of cell therapy on chronic skin wound healing: a meta-analysis. <i>Hum Gene Ther</i>. 2021;32:787-795. doi:10.1089/hum.2020.275</p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>bio</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="disclosure">From the Naval Medical Center San Diego, California.</p> <p class="disclosure">The authors report no conflict of interest. <br/><br/>All authors are military service members. This work was prepared as part of their official duties. Title 17 U.S.C. 105 provides that “Copyright protection under this title is not available for any work of the United States Government.” Title 17 U.S.C. 101 defines a United States Government work as a work prepared by a military service member or employee of the United States Government as part of that person’s official duties. <br/><br/>The views expressed in this article are those of the authors and do not necessarily reflect the official policy or position of the Department of the Navy, the Department of Defense, or the US Government. <br/><br/>Correspondence: David S. Kirwin, MD, Naval Medical Center San Diego Dermatology Department, 1261 34th St, Unit 31, San Diego, CA 92102 (dsk247@cornell.edu).<br/><br/>doi:10.12788/cutis.0970 </p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>in</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="insidehead">Practice <strong>Points</strong></p> <ul class="insidebody"> <li>Wound healing in adults is a complex dynamic process that usually takes the greater part of 1 year to completely resolve and is marked by the end of scar formation. </li> <li>Postamputation residual limbs are subject to mechanical and biophysical stress to which the overlying skin is not accustomed. Skin treatment aims at mitigating these stresses. </li> <li>The major dermatologic barriers to successful wound healing following amputation include infection, skin breakdown, formation of chronic wounds and granulation tissue, heterotopic ossification, and hyperhidrosis. </li> </ul> </itemContent> </newsItem> </itemSet></root>
Inside the Article

Practice Points

  • Wound healing in adults is a complex dynamic process that usually takes the greater part of 1 year to completely resolve and is marked by the end of scar formation.
  • Postamputation residual limbs are subject to mechanical and biophysical stress to which the overlying skin is not accustomed. Skin treatment aims at mitigating these stresses.
  • The major dermatologic barriers to successful wound healing following amputation include infection, skin breakdown, formation of chronic wounds and granulation tissue, heterotopic ossification, and hyperhidrosis.
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
Image
Teambase ID
180026DF.SIG
Disable zoom
Off

Study: Lifetime Cost of Vyjuvek Gene Therapy for DEB Could Be $15-$22 Million

Article Type
Changed
Mon, 02/12/2024 - 09:52

The lifetime cost of the new topical gene therapy Vyjuvek (beremagene geperpavec, formerly known as B-VEC) could be as much as $15-$22 million per patient, a figure that may give payers, especially federal programs like Medicaid, pause, according to the authors of a new study.

The US Food and Drug Administration (FDA) approved Vyjuvek (Krystal Biotech) in May 2023 for the treatment of wounds in patients ages 6 months and older with dystrophic epidermolysis bullosa (DEB), which includes two types, the most severe form (autosomal recessive, or RDEB) and the autosomal dominant form of DEB (DDEB), which tends to be milder.

Raymakers_Adam_Boston_web.jpg
%3Cp%3E%26nbsp%3B%3C%2Fp%3E%3Cp%3EDr.%20Raymakers%3C%2Fp%3E


Treatment with Vyjuvek “represents an important advance in the treatment of RDEB,” wrote Adam J.N. Raymakers, PhD, and colleagues at the Program on Regulation, Therapeutics, and Law; the Department of Dermatology; and the Division of Pulmonary and Critical Care Medicine at Brigham & Women’s Hospital in Boston, Massachusetts, in their paper, published in JAMA Dermatology. But the price “will be high, potentially limiting patients’ access to it,” they added. Evidence to support it in DDEB “is less conclusive,” they wrote, noting that the pivotal phase 3 study that led to approval included one patient with DDEB.

“The wider indication granted by the FDA may lead to friction between payers on the one hand and patients and physicians on the other,” they wrote, noting a potential minimum price of $300,000 per patient a year, which was based on Krystal’s regulatory filings.

There is no cure for DEB. Vyjuvek, applied as a gel on an ongoing basis, uses a nonreplicating herpes simplex virus type 1 vector to deliver the COL7A1 gene directly to skin cells, restoring the COL7 protein fibrils that stabilize skin structure.

The authors estimated that in the United States, 894 individuals – largely children – with both forms of the disease would be eligible for Vyjuvek treatment in the first year. Based on the $300,000 price, spending on gene therapy could range from $179 million to $357 million for those 894 patients, they reported in the study.

Over the first 3 years, spending could range as high as $1 billion, the authors estimated. Even if patients with only the most severe disease (RDEB) — an estimated 442 patients — received treatment, spending could be $132 million and up to $400 million or more over the first 3 years, they wrote.

Some media outlets have reported that Vyjuvek could cost as much as $600,000, said Dr. Raymakers, a research fellow. That price “would double all of our estimates,” he told this news organization.

The study assumed that patients with RDEB would only live to age 50, which led to a lifetime cost estimate of $15 million. But that is likely a conservative estimate, he and his coauthors wrote, noting that many patients with RDEB die from squamous cell carcinoma, but that Vyjuvek could, by attenuating skin damage, also potentially prevent skin cancer.

Dr. Raymakers said he and his colleagues began their study when Vyjuvek was approved, and thus they did not have any real-world data on the price or payer responses. Their estimates also did not include differing dosing regimens, which also could change their spending figures.

Krystal Biotech recently reported that in its third quarter of 2023 – representing just 1 month of Vyjuvek availability – it received requests to begin treatment for 284 patients from 136 unique clinicians. Twenty percent of the start requests were for patients with the milder form (DDEB), and a third of all the requests were for patients 10 years of age or younger. The company also said that it had “received positive coverage determinations from all major commercial national health plans” and that it was on track to receive approval from most state Medicaid plans.

In 1 month, Krystal reported net Vyjuvek revenues of $8.6 million.

The authors suggested that one way to evaluate Vyjuvek’s value — especially for those with DDEB — would be through a cost-effectiveness study. While important, a cost-effectiveness study would not get at the impact on a payer, said Dr. Raymakers. “Something can be cost-effective but unaffordable to the system,” he said.

“When there’s one of these very expensive therapies, that’s one thing,” he said. “But when there’s more and more coming to market, you wonder how much can be tolerated,” said Dr. Raymakers.

 

 

CMS Launching Gene Therapy Program

The Biden administration recently announced that it was launching a program aimed at increasing access, curbing costs, and ensuring value of gene therapies, starting with sickle cell disease. The program will begin in early 2025. Among other aspects, the federal government will negotiate the price of the product with the manufacturer.

“The goal of the Cell and Gene Therapy Access Model is to increase access to innovative cell and gene therapies for people with Medicaid by making it easier for states to pay for these therapies,” said Liz Fowler, CMS Deputy Administrator and Director of the CMS Innovation Center, in a statement announcing the program.

Whether the new program takes a look at Vyjuvek – and when – is not clear.

[embed:render:related:node:263130]

But the authors of the study noted that the lifetime costs of treating a patient with Vyjuvek “exceed the costs of all other one-time gene therapies for other diseases.” And they wrote, even at the most conservative estimates, Vyjuvek “will be the most expensive gene therapy currently marketed in the US.”

The study was funded by a grant from Arnold Ventures, grants from the Kaiser Permanente Institute for Health Policy, the Commonwealth Fund, and the National Heart, Lung, and Blood Institute. Dr. Raymakers and co-authors reported no financial relationships relevant to the work.

Publications
Topics
Sections

The lifetime cost of the new topical gene therapy Vyjuvek (beremagene geperpavec, formerly known as B-VEC) could be as much as $15-$22 million per patient, a figure that may give payers, especially federal programs like Medicaid, pause, according to the authors of a new study.

The US Food and Drug Administration (FDA) approved Vyjuvek (Krystal Biotech) in May 2023 for the treatment of wounds in patients ages 6 months and older with dystrophic epidermolysis bullosa (DEB), which includes two types, the most severe form (autosomal recessive, or RDEB) and the autosomal dominant form of DEB (DDEB), which tends to be milder.

Raymakers_Adam_Boston_web.jpg
%3Cp%3E%26nbsp%3B%3C%2Fp%3E%3Cp%3EDr.%20Raymakers%3C%2Fp%3E


Treatment with Vyjuvek “represents an important advance in the treatment of RDEB,” wrote Adam J.N. Raymakers, PhD, and colleagues at the Program on Regulation, Therapeutics, and Law; the Department of Dermatology; and the Division of Pulmonary and Critical Care Medicine at Brigham & Women’s Hospital in Boston, Massachusetts, in their paper, published in JAMA Dermatology. But the price “will be high, potentially limiting patients’ access to it,” they added. Evidence to support it in DDEB “is less conclusive,” they wrote, noting that the pivotal phase 3 study that led to approval included one patient with DDEB.

“The wider indication granted by the FDA may lead to friction between payers on the one hand and patients and physicians on the other,” they wrote, noting a potential minimum price of $300,000 per patient a year, which was based on Krystal’s regulatory filings.

There is no cure for DEB. Vyjuvek, applied as a gel on an ongoing basis, uses a nonreplicating herpes simplex virus type 1 vector to deliver the COL7A1 gene directly to skin cells, restoring the COL7 protein fibrils that stabilize skin structure.

The authors estimated that in the United States, 894 individuals – largely children – with both forms of the disease would be eligible for Vyjuvek treatment in the first year. Based on the $300,000 price, spending on gene therapy could range from $179 million to $357 million for those 894 patients, they reported in the study.

Over the first 3 years, spending could range as high as $1 billion, the authors estimated. Even if patients with only the most severe disease (RDEB) — an estimated 442 patients — received treatment, spending could be $132 million and up to $400 million or more over the first 3 years, they wrote.

Some media outlets have reported that Vyjuvek could cost as much as $600,000, said Dr. Raymakers, a research fellow. That price “would double all of our estimates,” he told this news organization.

The study assumed that patients with RDEB would only live to age 50, which led to a lifetime cost estimate of $15 million. But that is likely a conservative estimate, he and his coauthors wrote, noting that many patients with RDEB die from squamous cell carcinoma, but that Vyjuvek could, by attenuating skin damage, also potentially prevent skin cancer.

Dr. Raymakers said he and his colleagues began their study when Vyjuvek was approved, and thus they did not have any real-world data on the price or payer responses. Their estimates also did not include differing dosing regimens, which also could change their spending figures.

Krystal Biotech recently reported that in its third quarter of 2023 – representing just 1 month of Vyjuvek availability – it received requests to begin treatment for 284 patients from 136 unique clinicians. Twenty percent of the start requests were for patients with the milder form (DDEB), and a third of all the requests were for patients 10 years of age or younger. The company also said that it had “received positive coverage determinations from all major commercial national health plans” and that it was on track to receive approval from most state Medicaid plans.

In 1 month, Krystal reported net Vyjuvek revenues of $8.6 million.

The authors suggested that one way to evaluate Vyjuvek’s value — especially for those with DDEB — would be through a cost-effectiveness study. While important, a cost-effectiveness study would not get at the impact on a payer, said Dr. Raymakers. “Something can be cost-effective but unaffordable to the system,” he said.

“When there’s one of these very expensive therapies, that’s one thing,” he said. “But when there’s more and more coming to market, you wonder how much can be tolerated,” said Dr. Raymakers.

 

 

CMS Launching Gene Therapy Program

The Biden administration recently announced that it was launching a program aimed at increasing access, curbing costs, and ensuring value of gene therapies, starting with sickle cell disease. The program will begin in early 2025. Among other aspects, the federal government will negotiate the price of the product with the manufacturer.

“The goal of the Cell and Gene Therapy Access Model is to increase access to innovative cell and gene therapies for people with Medicaid by making it easier for states to pay for these therapies,” said Liz Fowler, CMS Deputy Administrator and Director of the CMS Innovation Center, in a statement announcing the program.

Whether the new program takes a look at Vyjuvek – and when – is not clear.

[embed:render:related:node:263130]

But the authors of the study noted that the lifetime costs of treating a patient with Vyjuvek “exceed the costs of all other one-time gene therapies for other diseases.” And they wrote, even at the most conservative estimates, Vyjuvek “will be the most expensive gene therapy currently marketed in the US.”

The study was funded by a grant from Arnold Ventures, grants from the Kaiser Permanente Institute for Health Policy, the Commonwealth Fund, and the National Heart, Lung, and Blood Institute. Dr. Raymakers and co-authors reported no financial relationships relevant to the work.

The lifetime cost of the new topical gene therapy Vyjuvek (beremagene geperpavec, formerly known as B-VEC) could be as much as $15-$22 million per patient, a figure that may give payers, especially federal programs like Medicaid, pause, according to the authors of a new study.

The US Food and Drug Administration (FDA) approved Vyjuvek (Krystal Biotech) in May 2023 for the treatment of wounds in patients ages 6 months and older with dystrophic epidermolysis bullosa (DEB), which includes two types, the most severe form (autosomal recessive, or RDEB) and the autosomal dominant form of DEB (DDEB), which tends to be milder.

Raymakers_Adam_Boston_web.jpg
%3Cp%3E%26nbsp%3B%3C%2Fp%3E%3Cp%3EDr.%20Raymakers%3C%2Fp%3E


Treatment with Vyjuvek “represents an important advance in the treatment of RDEB,” wrote Adam J.N. Raymakers, PhD, and colleagues at the Program on Regulation, Therapeutics, and Law; the Department of Dermatology; and the Division of Pulmonary and Critical Care Medicine at Brigham & Women’s Hospital in Boston, Massachusetts, in their paper, published in JAMA Dermatology. But the price “will be high, potentially limiting patients’ access to it,” they added. Evidence to support it in DDEB “is less conclusive,” they wrote, noting that the pivotal phase 3 study that led to approval included one patient with DDEB.

“The wider indication granted by the FDA may lead to friction between payers on the one hand and patients and physicians on the other,” they wrote, noting a potential minimum price of $300,000 per patient a year, which was based on Krystal’s regulatory filings.

There is no cure for DEB. Vyjuvek, applied as a gel on an ongoing basis, uses a nonreplicating herpes simplex virus type 1 vector to deliver the COL7A1 gene directly to skin cells, restoring the COL7 protein fibrils that stabilize skin structure.

The authors estimated that in the United States, 894 individuals – largely children – with both forms of the disease would be eligible for Vyjuvek treatment in the first year. Based on the $300,000 price, spending on gene therapy could range from $179 million to $357 million for those 894 patients, they reported in the study.

Over the first 3 years, spending could range as high as $1 billion, the authors estimated. Even if patients with only the most severe disease (RDEB) — an estimated 442 patients — received treatment, spending could be $132 million and up to $400 million or more over the first 3 years, they wrote.

Some media outlets have reported that Vyjuvek could cost as much as $600,000, said Dr. Raymakers, a research fellow. That price “would double all of our estimates,” he told this news organization.

The study assumed that patients with RDEB would only live to age 50, which led to a lifetime cost estimate of $15 million. But that is likely a conservative estimate, he and his coauthors wrote, noting that many patients with RDEB die from squamous cell carcinoma, but that Vyjuvek could, by attenuating skin damage, also potentially prevent skin cancer.

Dr. Raymakers said he and his colleagues began their study when Vyjuvek was approved, and thus they did not have any real-world data on the price or payer responses. Their estimates also did not include differing dosing regimens, which also could change their spending figures.

Krystal Biotech recently reported that in its third quarter of 2023 – representing just 1 month of Vyjuvek availability – it received requests to begin treatment for 284 patients from 136 unique clinicians. Twenty percent of the start requests were for patients with the milder form (DDEB), and a third of all the requests were for patients 10 years of age or younger. The company also said that it had “received positive coverage determinations from all major commercial national health plans” and that it was on track to receive approval from most state Medicaid plans.

In 1 month, Krystal reported net Vyjuvek revenues of $8.6 million.

The authors suggested that one way to evaluate Vyjuvek’s value — especially for those with DDEB — would be through a cost-effectiveness study. While important, a cost-effectiveness study would not get at the impact on a payer, said Dr. Raymakers. “Something can be cost-effective but unaffordable to the system,” he said.

“When there’s one of these very expensive therapies, that’s one thing,” he said. “But when there’s more and more coming to market, you wonder how much can be tolerated,” said Dr. Raymakers.

 

 

CMS Launching Gene Therapy Program

The Biden administration recently announced that it was launching a program aimed at increasing access, curbing costs, and ensuring value of gene therapies, starting with sickle cell disease. The program will begin in early 2025. Among other aspects, the federal government will negotiate the price of the product with the manufacturer.

“The goal of the Cell and Gene Therapy Access Model is to increase access to innovative cell and gene therapies for people with Medicaid by making it easier for states to pay for these therapies,” said Liz Fowler, CMS Deputy Administrator and Director of the CMS Innovation Center, in a statement announcing the program.

Whether the new program takes a look at Vyjuvek – and when – is not clear.

[embed:render:related:node:263130]

But the authors of the study noted that the lifetime costs of treating a patient with Vyjuvek “exceed the costs of all other one-time gene therapies for other diseases.” And they wrote, even at the most conservative estimates, Vyjuvek “will be the most expensive gene therapy currently marketed in the US.”

The study was funded by a grant from Arnold Ventures, grants from the Kaiser Permanente Institute for Health Policy, the Commonwealth Fund, and the National Heart, Lung, and Blood Institute. Dr. Raymakers and co-authors reported no financial relationships relevant to the work.

Publications
Publications
Topics
Article Type
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>166897</fileName> <TBEID>0C04E7D6.SIG</TBEID> <TBUniqueIdentifier>MD_0C04E7D6</TBUniqueIdentifier> <newsOrJournal>News</newsOrJournal> <publisherName>Frontline Medical Communications</publisherName> <storyname/> <articleType>2</articleType> <TBLocation>QC Done-All Pubs</TBLocation> <QCDate>20240209T162635</QCDate> <firstPublished>20240209T163310</firstPublished> <LastPublished>20240209T163310</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240209T163310</CMSDate> <articleSource>FROM JAMA DERMATOLOGY</articleSource> <facebookInfo/> <meetingNumber/> <byline>Alicia Ault</byline> <bylineText>ALICIA AULT</bylineText> <bylineFull>ALICIA AULT</bylineFull> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType>News</newsDocType> <journalDocType/> <linkLabel/> <pageRange/> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:imng"> <name>IMNG Medical Media</name> <rightsInfo> <copyrightHolder> <name>Frontline Medical News</name> </copyrightHolder> <copyrightNotice>Copyright (c) 2015 Frontline Medical News, a Frontline Medical Communications Inc. company. All rights reserved. This material may not be published, broadcast, copied, or otherwise reproduced or distributed without the prior written permission of Frontline Medical Communications Inc.</copyrightNotice> </rightsInfo> </provider> <abstract/> <metaDescription>The lifetime cost of the new topical gene therapy Vyjuvek (beremagene geperpavec, formerly known as B-VEC) could be as much as $15-$22 million per patient, a fi</metaDescription> <articlePDF/> <teaserImage/> <title>Study: Lifetime Cost of Vyjuvek Gene Therapy for DEB Could Be $15-$22 Million</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear/> <pubPubdateMonth/> <pubPubdateDay/> <pubVolume/> <pubNumber/> <wireChannels/> <primaryCMSID/> <CMSIDs/> <keywords/> <seeAlsos/> <publications_g> <publicationData> <publicationCode>skin</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> <publicationData> <publicationCode>fp</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> <publicationData> <publicationCode>im</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> <publicationData> <publicationCode>pn</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> </publications_g> <publications> <term canonical="true">13</term> <term>15</term> <term>21</term> <term>25</term> </publications> <sections> <term>27970</term> <term canonical="true">39313</term> </sections> <topics> <term>285</term> <term canonical="true">271</term> <term>203</term> <term>313</term> <term>38029</term> </topics> <links/> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Study: Lifetime Cost of Vyjuvek Gene Therapy for DEB Could Be $15-$22 Million</title> <deck/> </itemMeta> <itemContent> <p><span class="tag metaDescription">The lifetime cost of the new topical gene therapy Vyjuvek (<span class="Hyperlink"><a href="https://reference.medscape.com/drug/vyjuvek-beremagene-geperpavec-4000316">beremagene geperpavec</a></span>, formerly known as B-VEC) could be as much as $15-$22 million per patient, a figure that may give payers, especially federal programs like Medicaid, pause,</span> according to the authors of a new study.</p> <p>The US Food and Drug Administration (FDA) <span class="Hyperlink"><a href="https://www.fda.gov/news-events/press-announcements/fda-approves-first-topical-gene-therapy-treatment-wounds-patients-dystrophic-epidermolysis-bullosa">approved Vyjuvek</a></span> (Krystal Biotech) in May 2023 for the treatment of wounds in patients ages 6 months and older with dystrophic <span class="Hyperlink"><a href="https://emedicine.medscape.com/article/1062939-overview">epidermolysis bullosa</a></span> (DEB), which includes two types, the most severe form (autosomal recessive, or RDEB) and the autosomal dominant form of DEB (DDEB), which tends to be milder.<br/><br/>Treatment with Vyjuvek “represents an important advance in the treatment of RDEB,” wrote Adam J.N. Raymakers, PhD, and colleagues at the Program on Regulation, Therapeutics, and Law; the Department of Dermatology; and the Division of Pulmonary and Critical Care Medicine at Brigham &amp; Women’s Hospital in Boston, Massachusetts, in their <span class="Hyperlink"><a href="https://jamanetwork.com/journals/jamadermatology/fullarticle/2814373">paper</a></span>, published in <em>JAMA Dermatology</em>. But the price “will be high, potentially limiting patients’ access to it,” they added. Evidence to support it in DDEB “is less conclusive,” they wrote, noting that the pivotal phase 3 study that led to approval included one patient with DDEB.<br/><br/>“The wider indication granted by the FDA may lead to friction between payers on the one hand and patients and physicians on the other,” they wrote, noting a potential minimum price of $300,000 per patient a year, which was based on Krystal’s regulatory filings.<br/><br/>There is no cure for DEB. <span class="Hyperlink"><a href="https://reference.medscape.com/drug/vyjuvek-beremagene-geperpavec-4000316">Vyjuvek</a></span>, applied as a gel on an ongoing basis, uses a nonreplicating <span class="Hyperlink"><a href="https://emedicine.medscape.com/article/218580-overview">herpes simplex</a></span> virus type 1 vector to deliver the COL7A1 gene directly to skin cells, restoring the COL7 protein fibrils that stabilize skin structure.<br/><br/>The authors estimated that in the United States, 894 individuals – largely children – with both forms of the disease would be eligible for Vyjuvek treatment in the first year. Based on the $300,000 price, spending on gene therapy could range from $179 million to $357 million for those 894 patients, they reported in the study.<br/><br/>Over the first 3 years, spending could range as high as $1 billion, the authors estimated. Even if patients with only the most severe disease (RDEB) — an estimated 442 patients — received treatment, spending could be $132 million and up to $400 million or more over the first 3 years, they wrote.<br/><br/>Some media outlets have reported that Vyjuvek could cost as much as $600,000, said Dr. Raymakers, a research fellow. That price “would double all of our estimates,” he told this news organization.<br/><br/>The study assumed that patients with RDEB would only live to age 50, which led to a lifetime cost estimate of $15 million. But that is likely a conservative estimate, he and his coauthors wrote, noting that many patients with RDEB die from squamous cell carcinoma, but that Vyjuvek could, by attenuating skin damage, also potentially prevent skin cancer.<br/><br/>Dr. Raymakers said he and his colleagues began their study when Vyjuvek was approved, and thus they did not have any real-world data on the price or payer responses. Their estimates also did not include differing dosing regimens, which also could change their spending figures.<br/><br/>Krystal Biotech recently <span class="Hyperlink"><a href="https://ir.krystalbio.com/news-releases/news-release-details/krystal-biotech-announces-third-quarter-2023-financial-results">reported that in its third quarter of 2023</a></span> – representing just 1 month of Vyjuvek availability – it received requests to begin treatment for 284 patients from 136 unique clinicians. Twenty percent of the start requests were for patients with the milder form (DDEB), and a third of all the requests were for patients 10 years of age or younger. The company also said that it had “received positive coverage determinations from all major commercial national health plans” and that it was on track to receive approval from most state Medicaid plans.<br/><br/>In 1 month, Krystal reported net Vyjuvek revenues of $8.6 million.<br/><br/>The authors suggested that one way to evaluate Vyjuvek’s value — especially for those with DDEB — would be through a cost-effectiveness study. While important, a cost-effectiveness study would not get at the impact on a payer, said Dr. Raymakers. “Something can be cost-effective but unaffordable to the system,” he said.<br/><br/>“When there’s one of these very expensive therapies, that’s one thing,” he said. “But when there’s more and more coming to market, you wonder how much can be tolerated,” said Dr. Raymakers.<br/><br/><br/><br/></p> <h2>CMS Launching Gene Therapy Program</h2> <p>The Biden administration recently announced that it was launching a program aimed at increasing access, curbing costs, and ensuring value of gene therapies, <span class="Hyperlink"><a href="https://www.hhs.gov/about/news/2024/01/30/biden-harris-administration-announces-action-increase-access-sickle-cell-disease-treatments.html">starting with sickle cell disease</a></span>. The program will begin in early 2025. Among other aspects, the federal government will negotiate the price of the product with the manufacturer.</p> <p>“The goal of the Cell and Gene Therapy Access Model is to increase access to innovative cell and gene therapies for people with Medicaid by making it easier for states to pay for these therapies,” said Liz Fowler, CMS Deputy Administrator and Director of the CMS Innovation Center, <span class="Hyperlink"><a href="https://www.hhs.gov/about/news/2024/01/30/biden-harris-administration-announces-action-increase-access-sickle-cell-disease-treatments.html">in a statement</a></span> announcing the program.<br/><br/>Whether the new program takes a look at Vyjuvek – and when – is not clear.<br/><br/>But the authors of the study noted that the lifetime costs of treating a patient with Vyjuvek “exceed the costs of all other one-time gene therapies for other diseases.” And they wrote, even at the most conservative estimates, Vyjuvek “will be the most expensive gene therapy currently marketed in the US.”<br/><br/>The study was funded by a grant from Arnold Ventures, grants from the Kaiser Permanente Institute for Health Policy, the Commonwealth Fund, and the National Heart, Lung, and Blood Institute. Dr. Raymakers and co-authors reported no financial relationships relevant to the work.<span class="end"/></p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>teaser</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p>“The wider indication granted by the FDA may lead to friction between payers on the one hand and patients and physicians on the other,” the authors wrote.</p> </itemContent> </newsItem> </itemSet></root>
Article Source

FROM JAMA DERMATOLOGY

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

Rosemary, Part 1

Article Type
Changed
Tue, 02/27/2024 - 09:19

A member of the Lamiaceae family, Salvia rosmarinus (rosemary),* an aromatic plant native to the Mediterranean region and now cultivated globally, has been used for centuries in cuisine and medicine, with several well-established biological activities.1-3 Thought to contribute to preventing hair loss, rosemary oil was also used for hundreds of years in hair rinses in the Mediterranean area.4 In traditional Iranian medicine, rosemary essential oil has been topically applied as an analgesic, anti-inflammatory, and anti-acne remedy.5 Rosemary is known to absorb UV light well and to impart antibacterial and antifungal activity, as well as help maintain skin homeostasis.3 It is also used and under further study for its anti-inflammatory, antioxidant, anti-infective, and anticancer activity.2,6-9 The health benefits of rosemary are typically ascribed to its constituent carnosol/carnosic and ursolic acids.7In part 1 of this update on rosemary, the focus will be on chemical constituents, wound healing, anticancer activity, and hair care potential.

Chemical Constituents

The key chemical components of S. rosmarinus include bitter principle, resin, tannic acid, flavonoids, and volatile oils (made up of borneol, bornyl acetate, camphene, cineol, pinene, and camphor).10 Other important constituents of rosemary oil, in particular, include p-Cymene, linalool, gamma-terpinene, thymol, beta-pinene, alpha-pinene, eucalyptol, and carnosic acid.9 Volatile oils of rosemary have been used in various oils and lotions to treat wounds and with the intention of stimulating hair growth.10

Wound Healing

In a 2022 study in 60 adult male rats, Bulhões and colleagues found that the use of rosemary leaf essential oil-based ointments on skin lesions spurred wound healing, decreased inflammation, and enhanced angiogenesis as well as collagen fiber density.11

Three years earlier, Labib and colleagues studied the wound healing capacity of three chitosan-based topical formulations containing either tea tree essential oil, rosemary essential oil, or a mixture of both oils in an excision wound model in rats.

Rosemary_Oil_1316967048_web.jpg

The combination preparation was found to be the most effective in fostering various stages of wound healing, with significant increases in wound contraction percentage observed in the combination group compared with either group treated using individual essential oils or the untreated animals.12

A 2010 in vivo study by Abu-Al-Basal using BALB/c mice with diabetes revealed that the topical application of rosemary essential oil for three days reduced inflammation, enhanced wound contraction and re-epithelialization, and promoted angiogenesis, granulation tissue regeneration, and collagen deposition.13

Anticancer Activity

Using a 7,12-dimethlybenz(a)anthracene (DMBA)-initiated and croton oil-promoted model in 2006, Sancheti and Goyal determined that rosemary extract administered orally at a dose rate of 500 mg/kg body weight/mouse significantly inhibited two-stage skin tumorigenesis in mice.14 Nearly a decade later, Cattaneo and colleagues determined that a rosemary hydroalcoholic extract displayed antiproliferative effects on the human melanoma A375 cell line.8

The polyphenols carnosic acid and rosmarinic acid are most often cited as the sources of the reputed anticancer effects of rosemary.15

Hair Health

Early in 2023, Begum and colleagues developed a 1% hair lotion including a methanolic extract of the aerial part of S. rosmarinus that they assessed for potential hair growth activity in C57BL/6 mice. Using water as a control and 2% minoxidil hair lotion as standard, the investigators determined that their rosemary hair lotion demonstrated significant hair growth promotion, exceeding that seen in the mice treated with the drug standard.1

Baumann_Leslie_S_USE_web.jpg
Dr. Leslie S. Baumann

In a randomized controlled study in C57BL/6NCrSlc mice a decade earlier, Murata and colleagues evaluated the anti-androgenic activity and hair growth potential imparted by topical rosemary oil compared with finasteride and minoxidil. Rosemary oil leaf extract, with 12-O-methylcarnosic acid as its most active component, robustly suppressed 5alpha-reductase and stimulated hair growth in vivo in both the androgenetic alopecia/testosterone-treated mouse model, as well as the hair growth activating mouse model as compared with minoxidil. Further, the inhibitory activity of rosemary was 82.4% and 94.6% at 200 mcg/mL and 500 mcg/mL, respectively, whereas finasteride demonstrated 81.9% at 250 nM.16

A human study two years later was even more encouraging. Panahi and colleagues conducted a randomized comparative trial with 100 patients to investigate the effects of rosemary oil as opposed to minoxidil 2% for the treatment of androgenetic alopecia over 6 months. By 6 months, significantly greater hair counts were observed in both groups compared with baseline and 3-month readings, but no significant variations between groups. No differences were found in the frequency of dryness, greasiness, or dandruff at any time point or between groups. Scalp itching was significantly greater at the 3- and 6-month points in both groups, particularly in the minoxidil group at both of those time points. The investigators concluded that rosemary oil compared well with minoxidil as androgenetic alopecia therapy.17

 

 

Conclusion

Rosemary has been used in traditional medicine for hundreds of years and it has been a common ingredient in cosmetic and cosmeceutical formulations for more than 20 years. Recent findings suggest a broad array of applications in modern medicine, particularly dermatology. The next column will focus on the most recent studies pertaining to the antioxidant and anti-aging activity of this aromatic shrub.

Dr. Baumann is a private practice dermatologist, researcher, author, and entrepreneur in Miami. She founded the division of cosmetic dermatology at the University of Miami in 1997. The third edition of her bestselling textbook, “Cosmetic Dermatology,” was published in 2022. Dr. Baumann has received funding for advisory boards and/or clinical research trials from Allergan, Galderma, Johnson & Johnson, and Burt’s Bees. She is the CEO of Skin Type Solutions Inc., a SaaS company used to generate skin care routines in office and as a ecommerce solution. Write to her at dermnews@mdedge.com.

References

1. Begum A et al. Adv Biomed Res. 2023 Mar 21;12:60.

2. de Oliveira JR et al. J Biomed Sci. 2019 Jan 9;26(1):5.

3. González-Minero FJ et al. Cosmetics. 2020 Oct 3;7(4):77.

4. Dinkins J et al. Int J Dermatol. 2023 Aug;62(8):980-5.

5. Akbari J et al. Pharm Biol. 2015;53(10):1442-7.

6. Allegra A et al. Nutrients. 2020 Jun 10;12(6):1739.

7. de Macedo LM et al. Plants (Basel). 2020 May 21;9(5):651.

8. Cattaneo L et al. PLoS One. 2015 Jul 15;10(7):e0132439.

9. Borges RS et al. J Ethnopharmacol. 2019 Jan 30;229:29-45.

10. Begum A et al. Acta Sci Pol Technol Aliment. 2013 Jan-Mar;12(1):61-73.

11. Bulhões AAVC et al. Acta Cir Bras. 2022 Apr 8;37(1):e370104.

12. Labib RM et al. PLoS One. 2019 Sep 16;14(9):e0219561.

13. Abu-Al-Basal MA. J Ethnopharmacol. 2010 Sep 15;131(2):443-50.

14. Sancheti G and Goyal PK. Phytother Res. 2006 Nov;20(11):981-6.

15. Moore J et al. Nutrients. 2016 Nov 17;8(11):731.

16. Murata K et al. Phytother Res. 2013 Feb;27(2):212-7.

17. Panahi Y et al. Skinmed. 2015 Jan-Feb;13(1):15-21.

*Correction, 2/27: This column was updated with the more recent name for rosemary, Salvia rosmarinus.

Publications
Topics
Sections

A member of the Lamiaceae family, Salvia rosmarinus (rosemary),* an aromatic plant native to the Mediterranean region and now cultivated globally, has been used for centuries in cuisine and medicine, with several well-established biological activities.1-3 Thought to contribute to preventing hair loss, rosemary oil was also used for hundreds of years in hair rinses in the Mediterranean area.4 In traditional Iranian medicine, rosemary essential oil has been topically applied as an analgesic, anti-inflammatory, and anti-acne remedy.5 Rosemary is known to absorb UV light well and to impart antibacterial and antifungal activity, as well as help maintain skin homeostasis.3 It is also used and under further study for its anti-inflammatory, antioxidant, anti-infective, and anticancer activity.2,6-9 The health benefits of rosemary are typically ascribed to its constituent carnosol/carnosic and ursolic acids.7In part 1 of this update on rosemary, the focus will be on chemical constituents, wound healing, anticancer activity, and hair care potential.

Chemical Constituents

The key chemical components of S. rosmarinus include bitter principle, resin, tannic acid, flavonoids, and volatile oils (made up of borneol, bornyl acetate, camphene, cineol, pinene, and camphor).10 Other important constituents of rosemary oil, in particular, include p-Cymene, linalool, gamma-terpinene, thymol, beta-pinene, alpha-pinene, eucalyptol, and carnosic acid.9 Volatile oils of rosemary have been used in various oils and lotions to treat wounds and with the intention of stimulating hair growth.10

Wound Healing

In a 2022 study in 60 adult male rats, Bulhões and colleagues found that the use of rosemary leaf essential oil-based ointments on skin lesions spurred wound healing, decreased inflammation, and enhanced angiogenesis as well as collagen fiber density.11

Three years earlier, Labib and colleagues studied the wound healing capacity of three chitosan-based topical formulations containing either tea tree essential oil, rosemary essential oil, or a mixture of both oils in an excision wound model in rats.

Rosemary_Oil_1316967048_web.jpg

The combination preparation was found to be the most effective in fostering various stages of wound healing, with significant increases in wound contraction percentage observed in the combination group compared with either group treated using individual essential oils or the untreated animals.12

A 2010 in vivo study by Abu-Al-Basal using BALB/c mice with diabetes revealed that the topical application of rosemary essential oil for three days reduced inflammation, enhanced wound contraction and re-epithelialization, and promoted angiogenesis, granulation tissue regeneration, and collagen deposition.13

Anticancer Activity

Using a 7,12-dimethlybenz(a)anthracene (DMBA)-initiated and croton oil-promoted model in 2006, Sancheti and Goyal determined that rosemary extract administered orally at a dose rate of 500 mg/kg body weight/mouse significantly inhibited two-stage skin tumorigenesis in mice.14 Nearly a decade later, Cattaneo and colleagues determined that a rosemary hydroalcoholic extract displayed antiproliferative effects on the human melanoma A375 cell line.8

The polyphenols carnosic acid and rosmarinic acid are most often cited as the sources of the reputed anticancer effects of rosemary.15

Hair Health

Early in 2023, Begum and colleagues developed a 1% hair lotion including a methanolic extract of the aerial part of S. rosmarinus that they assessed for potential hair growth activity in C57BL/6 mice. Using water as a control and 2% minoxidil hair lotion as standard, the investigators determined that their rosemary hair lotion demonstrated significant hair growth promotion, exceeding that seen in the mice treated with the drug standard.1

Baumann_Leslie_S_USE_web.jpg
Dr. Leslie S. Baumann

In a randomized controlled study in C57BL/6NCrSlc mice a decade earlier, Murata and colleagues evaluated the anti-androgenic activity and hair growth potential imparted by topical rosemary oil compared with finasteride and minoxidil. Rosemary oil leaf extract, with 12-O-methylcarnosic acid as its most active component, robustly suppressed 5alpha-reductase and stimulated hair growth in vivo in both the androgenetic alopecia/testosterone-treated mouse model, as well as the hair growth activating mouse model as compared with minoxidil. Further, the inhibitory activity of rosemary was 82.4% and 94.6% at 200 mcg/mL and 500 mcg/mL, respectively, whereas finasteride demonstrated 81.9% at 250 nM.16

A human study two years later was even more encouraging. Panahi and colleagues conducted a randomized comparative trial with 100 patients to investigate the effects of rosemary oil as opposed to minoxidil 2% for the treatment of androgenetic alopecia over 6 months. By 6 months, significantly greater hair counts were observed in both groups compared with baseline and 3-month readings, but no significant variations between groups. No differences were found in the frequency of dryness, greasiness, or dandruff at any time point or between groups. Scalp itching was significantly greater at the 3- and 6-month points in both groups, particularly in the minoxidil group at both of those time points. The investigators concluded that rosemary oil compared well with minoxidil as androgenetic alopecia therapy.17

 

 

Conclusion

Rosemary has been used in traditional medicine for hundreds of years and it has been a common ingredient in cosmetic and cosmeceutical formulations for more than 20 years. Recent findings suggest a broad array of applications in modern medicine, particularly dermatology. The next column will focus on the most recent studies pertaining to the antioxidant and anti-aging activity of this aromatic shrub.

Dr. Baumann is a private practice dermatologist, researcher, author, and entrepreneur in Miami. She founded the division of cosmetic dermatology at the University of Miami in 1997. The third edition of her bestselling textbook, “Cosmetic Dermatology,” was published in 2022. Dr. Baumann has received funding for advisory boards and/or clinical research trials from Allergan, Galderma, Johnson & Johnson, and Burt’s Bees. She is the CEO of Skin Type Solutions Inc., a SaaS company used to generate skin care routines in office and as a ecommerce solution. Write to her at dermnews@mdedge.com.

References

1. Begum A et al. Adv Biomed Res. 2023 Mar 21;12:60.

2. de Oliveira JR et al. J Biomed Sci. 2019 Jan 9;26(1):5.

3. González-Minero FJ et al. Cosmetics. 2020 Oct 3;7(4):77.

4. Dinkins J et al. Int J Dermatol. 2023 Aug;62(8):980-5.

5. Akbari J et al. Pharm Biol. 2015;53(10):1442-7.

6. Allegra A et al. Nutrients. 2020 Jun 10;12(6):1739.

7. de Macedo LM et al. Plants (Basel). 2020 May 21;9(5):651.

8. Cattaneo L et al. PLoS One. 2015 Jul 15;10(7):e0132439.

9. Borges RS et al. J Ethnopharmacol. 2019 Jan 30;229:29-45.

10. Begum A et al. Acta Sci Pol Technol Aliment. 2013 Jan-Mar;12(1):61-73.

11. Bulhões AAVC et al. Acta Cir Bras. 2022 Apr 8;37(1):e370104.

12. Labib RM et al. PLoS One. 2019 Sep 16;14(9):e0219561.

13. Abu-Al-Basal MA. J Ethnopharmacol. 2010 Sep 15;131(2):443-50.

14. Sancheti G and Goyal PK. Phytother Res. 2006 Nov;20(11):981-6.

15. Moore J et al. Nutrients. 2016 Nov 17;8(11):731.

16. Murata K et al. Phytother Res. 2013 Feb;27(2):212-7.

17. Panahi Y et al. Skinmed. 2015 Jan-Feb;13(1):15-21.

*Correction, 2/27: This column was updated with the more recent name for rosemary, Salvia rosmarinus.

A member of the Lamiaceae family, Salvia rosmarinus (rosemary),* an aromatic plant native to the Mediterranean region and now cultivated globally, has been used for centuries in cuisine and medicine, with several well-established biological activities.1-3 Thought to contribute to preventing hair loss, rosemary oil was also used for hundreds of years in hair rinses in the Mediterranean area.4 In traditional Iranian medicine, rosemary essential oil has been topically applied as an analgesic, anti-inflammatory, and anti-acne remedy.5 Rosemary is known to absorb UV light well and to impart antibacterial and antifungal activity, as well as help maintain skin homeostasis.3 It is also used and under further study for its anti-inflammatory, antioxidant, anti-infective, and anticancer activity.2,6-9 The health benefits of rosemary are typically ascribed to its constituent carnosol/carnosic and ursolic acids.7In part 1 of this update on rosemary, the focus will be on chemical constituents, wound healing, anticancer activity, and hair care potential.

Chemical Constituents

The key chemical components of S. rosmarinus include bitter principle, resin, tannic acid, flavonoids, and volatile oils (made up of borneol, bornyl acetate, camphene, cineol, pinene, and camphor).10 Other important constituents of rosemary oil, in particular, include p-Cymene, linalool, gamma-terpinene, thymol, beta-pinene, alpha-pinene, eucalyptol, and carnosic acid.9 Volatile oils of rosemary have been used in various oils and lotions to treat wounds and with the intention of stimulating hair growth.10

Wound Healing

In a 2022 study in 60 adult male rats, Bulhões and colleagues found that the use of rosemary leaf essential oil-based ointments on skin lesions spurred wound healing, decreased inflammation, and enhanced angiogenesis as well as collagen fiber density.11

Three years earlier, Labib and colleagues studied the wound healing capacity of three chitosan-based topical formulations containing either tea tree essential oil, rosemary essential oil, or a mixture of both oils in an excision wound model in rats.

Rosemary_Oil_1316967048_web.jpg

The combination preparation was found to be the most effective in fostering various stages of wound healing, with significant increases in wound contraction percentage observed in the combination group compared with either group treated using individual essential oils or the untreated animals.12

A 2010 in vivo study by Abu-Al-Basal using BALB/c mice with diabetes revealed that the topical application of rosemary essential oil for three days reduced inflammation, enhanced wound contraction and re-epithelialization, and promoted angiogenesis, granulation tissue regeneration, and collagen deposition.13

Anticancer Activity

Using a 7,12-dimethlybenz(a)anthracene (DMBA)-initiated and croton oil-promoted model in 2006, Sancheti and Goyal determined that rosemary extract administered orally at a dose rate of 500 mg/kg body weight/mouse significantly inhibited two-stage skin tumorigenesis in mice.14 Nearly a decade later, Cattaneo and colleagues determined that a rosemary hydroalcoholic extract displayed antiproliferative effects on the human melanoma A375 cell line.8

The polyphenols carnosic acid and rosmarinic acid are most often cited as the sources of the reputed anticancer effects of rosemary.15

Hair Health

Early in 2023, Begum and colleagues developed a 1% hair lotion including a methanolic extract of the aerial part of S. rosmarinus that they assessed for potential hair growth activity in C57BL/6 mice. Using water as a control and 2% minoxidil hair lotion as standard, the investigators determined that their rosemary hair lotion demonstrated significant hair growth promotion, exceeding that seen in the mice treated with the drug standard.1

Baumann_Leslie_S_USE_web.jpg
Dr. Leslie S. Baumann

In a randomized controlled study in C57BL/6NCrSlc mice a decade earlier, Murata and colleagues evaluated the anti-androgenic activity and hair growth potential imparted by topical rosemary oil compared with finasteride and minoxidil. Rosemary oil leaf extract, with 12-O-methylcarnosic acid as its most active component, robustly suppressed 5alpha-reductase and stimulated hair growth in vivo in both the androgenetic alopecia/testosterone-treated mouse model, as well as the hair growth activating mouse model as compared with minoxidil. Further, the inhibitory activity of rosemary was 82.4% and 94.6% at 200 mcg/mL and 500 mcg/mL, respectively, whereas finasteride demonstrated 81.9% at 250 nM.16

A human study two years later was even more encouraging. Panahi and colleagues conducted a randomized comparative trial with 100 patients to investigate the effects of rosemary oil as opposed to minoxidil 2% for the treatment of androgenetic alopecia over 6 months. By 6 months, significantly greater hair counts were observed in both groups compared with baseline and 3-month readings, but no significant variations between groups. No differences were found in the frequency of dryness, greasiness, or dandruff at any time point or between groups. Scalp itching was significantly greater at the 3- and 6-month points in both groups, particularly in the minoxidil group at both of those time points. The investigators concluded that rosemary oil compared well with minoxidil as androgenetic alopecia therapy.17

 

 

Conclusion

Rosemary has been used in traditional medicine for hundreds of years and it has been a common ingredient in cosmetic and cosmeceutical formulations for more than 20 years. Recent findings suggest a broad array of applications in modern medicine, particularly dermatology. The next column will focus on the most recent studies pertaining to the antioxidant and anti-aging activity of this aromatic shrub.

Dr. Baumann is a private practice dermatologist, researcher, author, and entrepreneur in Miami. She founded the division of cosmetic dermatology at the University of Miami in 1997. The third edition of her bestselling textbook, “Cosmetic Dermatology,” was published in 2022. Dr. Baumann has received funding for advisory boards and/or clinical research trials from Allergan, Galderma, Johnson & Johnson, and Burt’s Bees. She is the CEO of Skin Type Solutions Inc., a SaaS company used to generate skin care routines in office and as a ecommerce solution. Write to her at dermnews@mdedge.com.

References

1. Begum A et al. Adv Biomed Res. 2023 Mar 21;12:60.

2. de Oliveira JR et al. J Biomed Sci. 2019 Jan 9;26(1):5.

3. González-Minero FJ et al. Cosmetics. 2020 Oct 3;7(4):77.

4. Dinkins J et al. Int J Dermatol. 2023 Aug;62(8):980-5.

5. Akbari J et al. Pharm Biol. 2015;53(10):1442-7.

6. Allegra A et al. Nutrients. 2020 Jun 10;12(6):1739.

7. de Macedo LM et al. Plants (Basel). 2020 May 21;9(5):651.

8. Cattaneo L et al. PLoS One. 2015 Jul 15;10(7):e0132439.

9. Borges RS et al. J Ethnopharmacol. 2019 Jan 30;229:29-45.

10. Begum A et al. Acta Sci Pol Technol Aliment. 2013 Jan-Mar;12(1):61-73.

11. Bulhões AAVC et al. Acta Cir Bras. 2022 Apr 8;37(1):e370104.

12. Labib RM et al. PLoS One. 2019 Sep 16;14(9):e0219561.

13. Abu-Al-Basal MA. J Ethnopharmacol. 2010 Sep 15;131(2):443-50.

14. Sancheti G and Goyal PK. Phytother Res. 2006 Nov;20(11):981-6.

15. Moore J et al. Nutrients. 2016 Nov 17;8(11):731.

16. Murata K et al. Phytother Res. 2013 Feb;27(2):212-7.

17. Panahi Y et al. Skinmed. 2015 Jan-Feb;13(1):15-21.

*Correction, 2/27: This column was updated with the more recent name for rosemary, Salvia rosmarinus.

Publications
Publications
Topics
Article Type
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>166527</fileName> <TBEID>0C04DFD6.SIG</TBEID> <TBUniqueIdentifier>MD_0C04DFD6</TBUniqueIdentifier> <newsOrJournal>News</newsOrJournal> <publisherName>Frontline Medical Communications</publisherName> <storyname>Feb Cosmecuetical Critique</storyname> <articleType>353</articleType> <TBLocation>QC Done-All Pubs</TBLocation> <QCDate>20240118T164445</QCDate> <firstPublished>20240119T092915</firstPublished> <LastPublished>20240119T092915</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240119T092915</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>Leslie S. Baumann</byline> <bylineText>LESLIE S. BAUMANN, MD</bylineText> <bylineFull>LESLIE S. BAUMANN, MD</bylineFull> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType>Column</newsDocType> <journalDocType/> <linkLabel/> <pageRange/> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:imng"> <name>IMNG Medical Media</name> <rightsInfo> <copyrightHolder> <name>Frontline Medical News</name> </copyrightHolder> <copyrightNotice>Copyright (c) 2015 Frontline Medical News, a Frontline Medical Communications Inc. company. All rights reserved. This material may not be published, broadcast, copied, or otherwise reproduced or distributed without the prior written permission of Frontline Medical Communications Inc.</copyrightNotice> </rightsInfo> </provider> <abstract/> <metaDescription>In part 1 of this update on rosemary, the focus will be on chemical constituents, wound healing, anticancer activity, and hair care potential</metaDescription> <articlePDF/> <teaserImage>299986</teaserImage> <title>Rosemary, Part 1</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear/> <pubPubdateMonth/> <pubPubdateDay/> <pubVolume/> <pubNumber/> <wireChannels/> <primaryCMSID/> <CMSIDs/> <keywords/> <seeAlsos/> <publications_g> <publicationData> <publicationCode>skin</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> </publications_g> <publications> <term canonical="true">13</term> </publications> <sections> <term>52</term> <term canonical="true">27928</term> </sections> <topics> <term>177</term> <term canonical="true">219</term> <term>313</term> </topics> <links> <link> <itemClass qcode="ninat:picture"/> <altRep contenttype="image/jpeg">images/240125be.jpg</altRep> <description role="drol:caption"/> <description role="drol:credit">HUIZENG HU/Moment/Getty Images</description> </link> <link> <itemClass qcode="ninat:picture"/> <altRep contenttype="image/jpeg">images/2400b666.jpg</altRep> <description role="drol:caption">Dr. Leslie S. Baumann</description> <description role="drol:credit">Baumann Cosmetic &amp; Research Institute</description> </link> </links> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Rosemary, Part 1</title> <deck/> </itemMeta> <itemContent> <p>A member of the Lamiaceae family, <em>Rosmarinus officinalis</em> (rosemary), an aromatic plant native to the Mediterranean region and now cultivated globally, has been used for centuries in cuisine and medicine, with several well-established biological activities.<sup>1-3</sup> Thought to contribute to preventing hair loss, rosemary oil was also used for hundreds of years in hair rinses in the Mediterranean area.<sup>4</sup> In traditional Iranian medicine, rosemary essential oil has been topically applied as an analgesic, anti-inflammatory, and anti-acne remedy.<sup>5</sup> Rosemary is known to absorb UV light well and to impart antibacterial and antifungal activity, as well as help maintain skin homeostasis.<sup>3</sup> It is also used and under further study for its anti-inflammatory, antioxidant, anti-infective, and anticancer activity.<sup>2,6-9</sup> The health benefits of rosemary are typically ascribed to its constituent carnosol/carnosic and ursolic acids.<sup>7</sup> <span class="tag metaDescription">In part 1 of this update on rosemary, the focus will be on chemical constituents, wound healing, anticancer activity, and hair care potential</span>.</p> <h2>Chemical Constituents</h2> <p>The key chemical components of <em>R. officinalis</em> include bitter principle, resin, tannic acid, flavonoids, and volatile oils (made up of borneol, bornyl acetate, camphene, cineol, pinene, and camphor).<sup>10</sup> Other important constituents of rosemary oil, in particular, include p-Cymene, linalool, gamma-terpinene, thymol, beta-pinene, alpha-pinene, eucalyptol, and carnosic acid.<sup>9</sup> Volatile oils of rosemary have been used in various oils and lotions to treat wounds and with the intention of stimulating hair growth.<sup>10</sup></p> <h2>Wound Healing</h2> <p>In a 2022 study in 60 adult male rats, Bulhões and colleagues found that the use of rosemary leaf essential oil-based ointments on skin lesions spurred wound healing, decreased inflammation, and enhanced angiogenesis as well as collagen fiber density.<sup>11</sup><br/><br/>Three years earlier, Labib and colleagues studied the wound healing capacity of three chitosan-based topical formulations containing either tea tree essential oil, rosemary essential oil, or a mixture of both oils in an excision wound model in rats. [[{"fid":"299986","view_mode":"medstat_image_flush_right","fields":{"format":"medstat_image_flush_right","field_file_image_alt_text[und][0][value]":"Rosemary essential oil and fresh twig.","field_file_image_credit[und][0][value]":"HUIZENG HU/Moment/Getty Images","field_file_image_caption[und][0][value]":""},"type":"media","attributes":{"class":"media-element file-medstat_image_flush_right"}}]]The combination preparation was found to be the most effective in fostering various stages of wound healing, with significant increases in wound contraction percentage observed in the combination group compared with either group treated using individual essential oils or the untreated animals.<sup>12</sup><br/><br/>A 2010 in vivo study by Abu-Al-Basal using BALB/c mice with diabetes revealed that the topical application of rosemary essential oil for three days reduced inflammation, enhanced wound contraction and re-epithelialization, and promoted angiogenesis, granulation tissue regeneration, and collagen deposition.<sup>13</sup></p> <h2>Anticancer Activity</h2> <p>Using a 7,12-dimethlybenz(a)anthracene (DMBA)-initiated and croton oil-promoted model in 2006, Sancheti and Goyal determined that rosemary extract administered orally at a dose rate of 500 mg/kg body weight/mouse significantly inhibited two-stage skin tumorigenesis in mice.<sup>14</sup> Nearly a decade later, Cattaneo and colleagues determined that a rosemary hydroalcoholic extract displayed antiproliferative effects on the human melanoma A375 cell line.<sup>8</sup> <br/><br/>The polyphenols carnosic acid and rosmarinic acid are most often cited as the sources of the reputed anticancer effects of rosemary.<sup>15</sup></p> <h2>Hair Health</h2> <p>Early in 2023, Begum and colleagues developed a 1% hair lotion including a methanolic extract of the aerial part of <em>R. officinalis</em> that they assessed for potential hair growth activity in C57BL/6 mice. Using water as a control and 2% minoxidil hair lotion as standard, the investigators determined that their rosemary hair lotion demonstrated significant hair growth promotion, exceeding that seen in the mice treated with the drug standard.<sup>1</sup><br/><br/>[[{"fid":"239756","view_mode":"medstat_image_flush_left","fields":{"format":"medstat_image_flush_left","field_file_image_alt_text[und][0][value]":"Dr. Leslie S. Baumann, a dermatologist, researcher, author, and entrepreneur who practices in Miami.","field_file_image_credit[und][0][value]":"Baumann Cosmetic &amp; Research Institute","field_file_image_caption[und][0][value]":"Dr. Leslie S. Baumann"},"type":"media","attributes":{"class":"media-element file-medstat_image_flush_left"}}]]In a randomized controlled study in C57BL/6NCrSlc mice a decade earlier, Murata and colleagues evaluated the anti-androgenic activity and hair growth potential imparted by topical rosemary oil compared with finasteride and minoxidil. Rosemary oil leaf extract, with 12-O-methylcarnosic acid as its most active component, robustly suppressed 5alpha-reductase and stimulated hair growth in vivo in both the androgenetic alopecia/testosterone-treated mouse model, as well as the hair growth activating mouse model as compared with minoxidil. Further, the inhibitory activity of rosemary was 82.4% and 94.6% at 200 mcg/mL and 500 mcg/mL, respectively, whereas finasteride demonstrated 81.9% at 250 nM.<sup>16</sup><br/><br/>A human study two years later was even more encouraging. Panahi and colleagues conducted a randomized comparative trial with 100 patients to investigate the effects of rosemary oil as opposed to minoxidil 2% for the treatment of androgenetic alopecia over 6 months. By 6 months, significantly greater hair counts were observed in both groups compared with baseline and 3-month readings, but no significant variations between groups. No differences were found in the frequency of dryness, greasiness, or dandruff at any time point or between groups. Scalp itching was significantly greater at the 3- and 6-month points in both groups, particularly in the minoxidil group at both of those time points. The investigators concluded that rosemary oil compared well with minoxidil as androgenetic alopecia therapy.<sup>17</sup></p> <h2>Conclusion</h2> <p>Rosemary has been used in traditional medicine for hundreds of years and it has been a common ingredient in cosmetic and cosmeceutical formulations for more than 20 years. Recent findings suggest a broad array of applications in modern medicine, particularly dermatology. The next column will focus on the most recent studies pertaining to the antioxidant and anti-aging activity of this aromatic shrub.</p> <p> <em><span class="Hyperlink"><a href="https://lesliebaumannmd.com/">Dr. Baumann</a></span> is a private practice dermatologist, researcher, author, and entrepreneur in Miami. She founded the division of cosmetic dermatology at the University of Miami in 1997. The third edition of her bestselling textbook, “Cosmetic Dermatology,” was published in 2022. Dr. Baumann has received funding for advisory boards and/or clinical research trials from Allergan, Galderma, Johnson &amp; Johnson, and Burt’s Bees. She is the CEO of Skin Type Solutions Inc., a SaaS company used to generate skin care routines in office and as a ecommerce solution. Write to her at <span class="Hyperlink"><a href="mailto:dermnews%40mdedge.com?subject=">dermnews@mdedge.com</a></span>.</em> </p> <h2>References</h2> <p>1. Begum A et al. <span class="Hyperlink"><a href="https://journals.lww.com/adbm/fulltext/2023/03210/evaluation_of_herbal_hair_lotion_loaded_with.60.aspx">Adv Biomed Res. 2023 Mar 21;12:60</a>.</span><br/><br/>2. de Oliveira JR et al. <span class="Hyperlink"><a href="https://jbiomedsci.biomedcentral.com/articles/10.1186/s12929-019-0499-8">J Biomed Sci. 2019 Jan 9;26(1):5</a></span>.<br/><br/>3. González-Minero FJ et al. <span class="Hyperlink"><a href="https://www.mdpi.com/2079-9284/7/4/77">Cosmetics. 2020 Oct 3;7(4):77</a></span>.<br/><br/>4. Dinkins J et al. <span class="Hyperlink"><a href="https://onlinelibrary.wiley.com/doi/10.1111/ijd.16657">Int J Dermatol. 2023 Aug;62(8):980-5</a></span>.<br/><br/>5. Akbari J et al. <span class="Hyperlink"><a href="https://www.tandfonline.com/doi/full/10.3109/13880209.2014.984855">Pharm Biol. 2015;53(10):1442-7</a></span>.<br/><br/>6. Allegra A et al. <span class="Hyperlink"><a href="https://www.mdpi.com/2072-6643/12/6/1739">Nutrients. 2020 Jun 10;12(6):1739</a></span>.<br/><br/>7. de Macedo LM et al. <span class="Hyperlink"><a href="https://www.mdpi.com/2223-7747/9/5/651">Plants (Basel). 2020 May 21;9(5):651</a></span>.<br/><br/>8. Cattaneo L et al. <span class="Hyperlink"><a href="https://journals.plos.org/plosone/article?id=10.1371/journal.pone.0132439">PLoS One. 2015 Jul 15;10(7):e0132439</a></span>.<br/><br/>9. Borges RS et al. <span class="Hyperlink"><a href="https://www.sciencedirect.com/science/article/pii/S0378874118314107?via%3Dihub">J Ethnopharmacol. 2019 Jan 30;229:29-45</a></span>.<br/><br/>10. Begum A et al. <span class="Hyperlink"><a href="https://pubmed.ncbi.nlm.nih.gov/24584866/">Acta Sci Pol Technol Aliment. 2013 Jan-Mar;12(1):61-73</a></span>.<br/><br/>11. Bulhões AAVC et al. <span class="Hyperlink"><a href="https://www.scielo.br/j/acb/a/3q4xdwv7cL3V6rSxrSgSCMG/?lang=en">Acta Cir Bras. 2022 Apr 8;37(1):e370104</a></span>.<br/><br/>12. Labib RM et al. <span class="Hyperlink"><a href="https://pubmed.ncbi.nlm.nih.gov/31525200/">PLoS One. 2019 Sep 16;14(9):e0219561</a></span>.<br/><br/>13. Abu-Al-Basal MA. <span class="Hyperlink"><a href="https://pubmed.ncbi.nlm.nih.gov/20633625/">J Ethnopharmacol. 2010 Sep 15;131(2):443-50.</a></span><br/><br/>14. Sancheti G and Goyal PK. <span class="Hyperlink"><a href="https://pubmed.ncbi.nlm.nih.gov/16927448/">Phytother Res. 2006 Nov;20(11):981-6</a></span>.<br/><br/>15. Moore J et al. <span class="Hyperlink"><a href="https://www.mdpi.com/2072-6643/8/11/731">Nutrients. 2016 Nov 17;8(11):731</a></span>.<br/><br/>16. Murata K et al. <span class="Hyperlink"><a href="https://pubmed.ncbi.nlm.nih.gov/22517595/">Phytother Res. 2013 Feb;27(2):212-7.</a></span><br/><br/>17. Panahi Y et al. <span class="Hyperlink"><a href="https://pubmed.ncbi.nlm.nih.gov/25842469/">Skinmed. 2015 Jan-Feb;13(1):15-21</a></span>.</p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>teaser</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p>Recent findings suggest a broad array of applications in modern medicine, particularly dermatology.</p> </itemContent> </newsItem> </itemSet></root>
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

FDA Approves Topical Gel For Wounds Associated With JEB and DEB

Article Type
Changed
Tue, 01/02/2024 - 15:36

The FDA has approved a topical gel containing birch triterpenes for the treatment of partial thickness wounds in patients 6 months and older with junctional epidermolysis bullosa (JEB) and dystrophic epidermolysis bullosa (DEB).

The gel is marketed under the name Filsuvez. It is the first approved treatment for wounds associated with JEB and the second for patients with DEB, following the approval of Vyjuvek (Krystal Biotech), a topical gene therapy gel, in May 2023.

First developed by Amryt Pharma and intended for home use, Filsuvez is now marketed by Chiesi Global Rare Diseases, which acquired Amryt in January 2023. The gel is applied topically to the wound at each dressing change.

[embed:render:related:node:264410]

The approval of Filsuvez is based on results from the Efficacy and Safety Study of Oleogel-S10 in Epidermolysis Bullosa (EASE), a randomized, placebo-controlled study of 223 people, the largest-ever phase 3 clinical trial for the treatment of EB, according to the Chiesi news release. The gel was well tolerated and met the primary endpoint with statistical significance, with 41.3% of patients achieving first complete target wound closure within 45 days (compared with 28.9% on placebo).

“I am so excited to say that this is another hurdle cleared and milestone achieved for the EB Community,” Brett Kopelan, executive director at debra of America said in a blog post. “We are now on the road to being able to treat EB more effectively, and to make the worst disease you’ve never heard of chronic, but livable, by making use of multiple therapeutic options in conjunction with each other.”

Publications
Topics
Sections

The FDA has approved a topical gel containing birch triterpenes for the treatment of partial thickness wounds in patients 6 months and older with junctional epidermolysis bullosa (JEB) and dystrophic epidermolysis bullosa (DEB).

The gel is marketed under the name Filsuvez. It is the first approved treatment for wounds associated with JEB and the second for patients with DEB, following the approval of Vyjuvek (Krystal Biotech), a topical gene therapy gel, in May 2023.

First developed by Amryt Pharma and intended for home use, Filsuvez is now marketed by Chiesi Global Rare Diseases, which acquired Amryt in January 2023. The gel is applied topically to the wound at each dressing change.

[embed:render:related:node:264410]

The approval of Filsuvez is based on results from the Efficacy and Safety Study of Oleogel-S10 in Epidermolysis Bullosa (EASE), a randomized, placebo-controlled study of 223 people, the largest-ever phase 3 clinical trial for the treatment of EB, according to the Chiesi news release. The gel was well tolerated and met the primary endpoint with statistical significance, with 41.3% of patients achieving first complete target wound closure within 45 days (compared with 28.9% on placebo).

“I am so excited to say that this is another hurdle cleared and milestone achieved for the EB Community,” Brett Kopelan, executive director at debra of America said in a blog post. “We are now on the road to being able to treat EB more effectively, and to make the worst disease you’ve never heard of chronic, but livable, by making use of multiple therapeutic options in conjunction with each other.”

The FDA has approved a topical gel containing birch triterpenes for the treatment of partial thickness wounds in patients 6 months and older with junctional epidermolysis bullosa (JEB) and dystrophic epidermolysis bullosa (DEB).

The gel is marketed under the name Filsuvez. It is the first approved treatment for wounds associated with JEB and the second for patients with DEB, following the approval of Vyjuvek (Krystal Biotech), a topical gene therapy gel, in May 2023.

First developed by Amryt Pharma and intended for home use, Filsuvez is now marketed by Chiesi Global Rare Diseases, which acquired Amryt in January 2023. The gel is applied topically to the wound at each dressing change.

[embed:render:related:node:264410]

The approval of Filsuvez is based on results from the Efficacy and Safety Study of Oleogel-S10 in Epidermolysis Bullosa (EASE), a randomized, placebo-controlled study of 223 people, the largest-ever phase 3 clinical trial for the treatment of EB, according to the Chiesi news release. The gel was well tolerated and met the primary endpoint with statistical significance, with 41.3% of patients achieving first complete target wound closure within 45 days (compared with 28.9% on placebo).

“I am so excited to say that this is another hurdle cleared and milestone achieved for the EB Community,” Brett Kopelan, executive director at debra of America said in a blog post. “We are now on the road to being able to treat EB more effectively, and to make the worst disease you’ve never heard of chronic, but livable, by making use of multiple therapeutic options in conjunction with each other.”

Publications
Publications
Topics
Article Type
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>166385</fileName> <TBEID>0C04DCDE.SIG</TBEID> <TBUniqueIdentifier>MD_0C04DCDE</TBUniqueIdentifier> <newsOrJournal>News</newsOrJournal> <publisherName>Frontline Medical Communications</publisherName> <storyname/> <articleType>2</articleType> <TBLocation>QC Done-All Pubs</TBLocation> <QCDate>20231221T180754</QCDate> <firstPublished>20231222T090307</firstPublished> <LastPublished>20231222T090307</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20231222T090307</CMSDate> <articleSource/> <facebookInfo>Carol Morton first edit</facebookInfo> <meetingNumber/> <byline>Doug Brunk</byline> <bylineText>DOUG BRUNK</bylineText> <bylineFull>DOUG BRUNK</bylineFull> <bylineTitleText>MDedge News</bylineTitleText> <USOrGlobal/> <wireDocType/> <newsDocType>News</newsDocType> <journalDocType/> <linkLabel/> <pageRange/> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:imng"> <name>IMNG Medical Media</name> <rightsInfo> <copyrightHolder> <name>Frontline Medical News</name> </copyrightHolder> <copyrightNotice>Copyright (c) 2015 Frontline Medical News, a Frontline Medical Communications Inc. company. All rights reserved. This material may not be published, broadcast, copied, or otherwise reproduced or distributed without the prior written permission of Frontline Medical Communications Inc.</copyrightNotice> </rightsInfo> </provider> <abstract/> <metaDescription>The FDA has approved a topical gel containing birch triterpenes for the treatment of partial thickness wounds in patients 6 months and older with junctional epi</metaDescription> <articlePDF/> <teaserImage/> <title>FDA Approves Topical Gel For Wounds Associated With JEB and DEB</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear/> <pubPubdateMonth/> <pubPubdateDay/> <pubVolume/> <pubNumber/> <wireChannels/> <primaryCMSID/> <CMSIDs/> <keywords/> <seeAlsos/> <publications_g> <publicationData> <publicationCode>skin</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> <publicationData> <publicationCode>im</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> <publicationData> <publicationCode>pn</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> <publicationData> <publicationCode>fp</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> </publications_g> <publications> <term canonical="true">13</term> <term>21</term> <term>25</term> <term>15</term> </publications> <sections> <term canonical="true">39313</term> </sections> <topics> <term>271</term> <term canonical="true">285</term> <term>313</term> <term>203</term> </topics> <links/> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>FDA Approves Topical Gel For Wounds Associated With JEB and DEB</title> <deck/> </itemMeta> <itemContent> <p>The FDA has approved a topical gel containing birch triterpenes for the treatment of partial thickness wounds in patients 6 months and older with junctional epidermolysis bullosa (JEB) and dystrophic epidermolysis bullosa (DEB).</p> <p>The gel is marketed under the name <span class="Hyperlink"><a href="https://www.globenewswire.com/news-release/2023/12/19/2798751/0/en/Chiesi-Global-Rare-Diseases-Receives-FDA-Approval-for-FILSUVEZ-birch-triterpenes-topical-gel-for-the-Treatment-of-Epidermolysis-Bullosa.html">Filsuvez</a></span>. It is the first approved treatment for wounds associated with JEB and the second for patients with DEB, following the approval of Vyjuvek (Krystal Biotech), a topical gene therapy gel, in May 2023.<br/><br/>First developed by Amryt Pharma and intended for home use, Filsuvez is now marketed by Chiesi Global Rare Diseases, which acquired Amryt in January 2023. The gel is applied topically to the wound at each dressing change.<br/><br/>The approval of Filsuvez is based on <span class="Hyperlink"><a href="https://www.clinicaltrials.gov/study/NCT03068780">results from</a></span> the <span class="Hyperlink">Efficacy and Safety Study of Oleogel-S10 in Epidermolysis Bullosa (EASE)</span>, a randomized, placebo-controlled study of 223 people, the largest-ever phase 3 clinical trial for the treatment of EB, according to the Chiesi news release. The gel was well tolerated and met the primary endpoint with statistical significance, with 41.3% of patients achieving first complete target wound closure within 45 days (compared with 28.9% on placebo). <br/><br/>“I am so excited to say that this is another hurdle cleared and milestone achieved for the EB Community,” Brett Kopelan, executive director at debra of America said in a <span class="Hyperlink"><a href="https://www.debra.org/blog/us-fda-approves-filsuvez-topical-gel-jeb-and-deb">blog post</a></span>. “We are now on the road to being able to treat EB more effectively, and to make the worst disease you’ve never heard of chronic, but livable, by making use of multiple therapeutic options in conjunction with each other.”<span class="end"/></p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>teaser</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent/> </newsItem> </itemSet></root>
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